1
|
Cadena-Cruz C, De-Avila-Arias M, Costello HM, Hurtado-Gomez L, Martínez-De-La-Rosa W, Macchia-Ceballos G, Rosales-Rada W, Valencia-Villa G, Villalba-Amarís P, Kararoudi MN, Peeples ME, San-Juan-Vergara H. Respiratory syncytial virus fuses with plasma membrane to infect primary cultures of bronchial epithelial cells. Front Microbiol 2025; 16:1498955. [PMID: 40099186 PMCID: PMC11911548 DOI: 10.3389/fmicb.2025.1498955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025] Open
Abstract
Background Respiratory syncytial virus (RSV) is a common cause of bronchiolitis in children under the age of five. RSV infection proceeds by fusion of the viral envelope with the target cell membrane, but it is unclear whether fusion occurs with plasma or endosomal membranes. Methods Entry and/or infection was studied in undifferentiated primary cultures of human bronchial epithelial cells. Synchronization of viral entry or infection was achieved by attaching the virus to the plasma membrane at temperatures of 4°C or 22°C. Cells in which entry events had occurred were identified by the enzymatic action of beta-lactamase M (BlaM) fused to the RSV P protein (BlaM-P) carried by rgRSV virions. BlaM cleaves the beta-lactam ring of CCF2 loaded into the cells, disrupting FRET and allowing blue light to be emitted. Green fluorescent protein (GFP) expression, encoded by the rgRSV genome, was used to identify infected cells. Results We found that adsorption of RSV at 4°C favors entry via endocytosis, whereas binding of the virus to the membrane at 22°C favors RSV entry via the plasma membrane. The induction of endocytosis by synchronization at 4°C is, therefore, an artifact. In addition, we found that all drugs that interfered with RSV infection reduced cell membrane deformations such as filopodia and lamellipodia, suggesting a mechanism by which they may interfere with RSV fusion with the cell membrane. Discussion In conclusion, RSV enters the cell by direct fusion of its envelope with the plasma membrane.
Collapse
Affiliation(s)
- Christian Cadena-Cruz
- Departamento de Medicina, Universidad del Norte, Barranquilla, Colombia
- Programa de Bacteriología, Universidad Libre Seccional, Barranquilla, Colombia
| | | | - Heather M Costello
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | | | | | | | - Wendy Rosales-Rada
- Departamento de Medicina, Universidad del Norte, Barranquilla, Colombia
- Grupo de Investigación Avanzada en Biomedicina, Programa de Microbiología, Universidad Libre de Colombia, Barranquilla, Atlántico, Colombia
| | | | | | - Meisam Naeimi Kararoudi
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Mark E Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States
- Infectious Disease Institute, The Ohio State University, Columbus, OH, United States
| | | |
Collapse
|
2
|
García-Arcos JM, Ziegler J, Grigolon S, Reymond L, Shajepal G, Cattin CJ, Lomakin A, Müller DJ, Ruprecht V, Wieser S, Voituriez R, Piel M. Rigidity percolation and active advection synergize in the actomyosin cortex to drive amoeboid cell motility. Dev Cell 2024; 59:2990-3007.e7. [PMID: 39047738 DOI: 10.1016/j.devcel.2024.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/11/2023] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Spontaneous locomotion is a common feature of most metazoan cells, generally attributed to the properties of actomyosin networks. This force-producing machinery has been studied down to the most minute molecular details, especially in lamellipodium-driven migration. Nevertheless, how actomyosin networks work inside contraction-driven amoeboid cells still lacks unifying principles. Here, using stable motile blebs from HeLa cells as a model amoeboid motile system, we imaged the dynamics of the actin cortex at the single filament level and revealed the co-existence of three distinct rheological phases. We introduce "advected percolation," a process where rigidity percolation and active advection synergize, spatially organizing the actin network's mechanical properties into a minimal and generic locomotion mechanism. Expanding from our observations on simplified systems, we speculate that this model could explain, down to the single actin filament level, how amoeboid cells, such as cancer or immune cells, can propel efficiently through complex 3D environments.
Collapse
Affiliation(s)
- Juan Manuel García-Arcos
- Institut Pierre Gilles de Gennes, PSL Research University, 6 rue Jean Calvin, 75005 Paris, France; Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Johannes Ziegler
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860 Castelldefels, Spain
| | - Silvia Grigolon
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP), 75005 Paris, France
| | - Loïc Reymond
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860 Castelldefels, Spain; Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Gaurav Shajepal
- Institut Pierre Gilles de Gennes, PSL Research University, 6 rue Jean Calvin, 75005 Paris, France
| | - Cédric J Cattin
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Alexis Lomakin
- Center for Pathobiochemistry and Genetics, Institute of Medical Chemistry, Medical University of Vienna, Währingerstraße 10, 1090 Vienna, Austria; Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Währingerstraße 10, 1090 Vienna, Austria
| | - Daniel J Müller
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Verena Ruprecht
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; ICREA, Pg. Lluis Companys 23, 08010 Barcelona, Spain
| | - Stefan Wieser
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860 Castelldefels, Spain
| | - Raphael Voituriez
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP), 75005 Paris, France; Laboratoire de Physique Théorique de la Matière Condensée, CNRS/Sorbonne Université, 4 Place Jussieu, 75005 Paris, France.
| | - Matthieu Piel
- Institut Pierre Gilles de Gennes, PSL Research University, 6 rue Jean Calvin, 75005 Paris, France; Institut Curie, PSL Research University, CNRS UMR 144, Paris, France.
| |
Collapse
|
3
|
García-Arcos JM, Jha A, Waterman CM, Piel M. Blebology: principles of bleb-based migration. Trends Cell Biol 2024; 34:838-853. [PMID: 38538441 PMCID: PMC11424778 DOI: 10.1016/j.tcb.2024.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/09/2024] [Accepted: 02/21/2024] [Indexed: 09/27/2024]
Abstract
Bleb-based migration, a conserved cell motility mode, has a crucial role in both physiological and pathological processes. Unlike the well-elucidated mechanisms of lamellipodium-based mesenchymal migration, the dynamics of bleb-based migration remain less understood. In this review, we highlight in a systematic way the establishment of front-rear polarity, bleb formation and extension, and the distinct regimes of bleb dynamics. We emphasize new evidence proposing a regulatory role of plasma membrane-cortex interactions in blebbing behavior and discuss the generation of force and its transmission during migration. Our analysis aims to deepen the understanding of the physical and molecular mechanisms of bleb-based migration, shedding light on its implications and significance for health and disease.
Collapse
Affiliation(s)
| | - Ankita Jha
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Clare M Waterman
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthieu Piel
- Institut Curie, UMR144, CNRS, PSL University, Paris, France; Institut Pierre Gilles de Gennes, PSL University, Paris, France.
| |
Collapse
|
4
|
Fujii Y, Ikenouchi J. Cytoplasmic zoning in membrane blebs. J Biochem 2024; 175:133-140. [PMID: 37943501 DOI: 10.1093/jb/mvad084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/23/2023] [Indexed: 11/10/2023] Open
Abstract
Blebs are membrane structures formed by the detachment of the plasma membrane from the underlying actin cytoskeleton. It is now clear that a wide variety of cells, including cancer cells, actively form blebs for cell migration and cell survival. The expansion of blebs has been regarded as the passive ballooning of the plasma membrane by an abrupt increase in intracellular pressure. However, recent studies revealed the importance of 'cytoplasmic zoning', i.e. local changes in the hydrodynamic properties and the ionic and protein content of the cytoplasm. In this review, we summarize the current understanding of the molecular mechanisms behind cytoplasmic zoning and its role in bleb expansion.
Collapse
Affiliation(s)
- Yuki Fujii
- Department of Biology, Faculty of Sciences, Kyushu University, Nishi-Ku, Fukuoka 819-0395, Japan
| | - Junichi Ikenouchi
- Department of Biology, Faculty of Sciences, Kyushu University, Nishi-Ku, Fukuoka 819-0395, Japan
| |
Collapse
|
5
|
Crater JM, Dunn D, Nixon DF, O’Brien RLF. HIV-1 Mediated Cortical Actin Disruption Mirrors ARP2/3 Defects Found in Primary T Cell Immunodeficiencies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.27.550856. [PMID: 38405733 PMCID: PMC10888893 DOI: 10.1101/2023.07.27.550856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
During cell movement, cortical actin balances mechanical and osmotic forces to maintain cell function while providing the scaffold for cell shape. Migrating CD4+ T cells have a polarized structure with a leading edge containing dynamic branched and linear F-actin structures that bridge intracellular components to surface adhesion molecules. These actin structures are complemented with a microtubular network beaded with membrane bound organelles in the trailing uropod. Disruption of actin structures leads to dysregulated migration and changes in morphology of affected cells. In HIV-1 infection, CD4+ T cells have dysregulated movement. However, the precise mechanisms by which HIV-1 affects CD4+ T cell movement are unknown. Here, we show that HIV-1 infection of primary CD4+ T cells causes at least four progressive morphological differences as a result of virally induced cortical cytoskeleton disruption, shown by ultrastructural and time lapse imaging. Infection with a ΔNef virus partially abrogated the dysfunctional phenotype in infected cells and partially restored a wild-type shape. The pathological morphologies after HIV-1 infection phenocopy leukocytes which contain genetic determinants of specific T cell Inborn Errors of Immunity (IEI) or Primary Immunodeficiencies (PID) that affect the actin cytoskeleton. To identify potential actin regulatory pathways that may be linked to the morphological deformities, uninfected CD4+ T cell morphology was characterized following addition of small molecule chemical inhibitors. The ARP2/3 inhibitor CK-666 recapitulated three of the four abnormal morphologies we observed in HIV-1 infected cells. Restoring ARP2/3 function and cortical actin integrity in people living with HIV-1 infection is a new avenue of investigation to eradicate HIV-1 infected cells from the body.
Collapse
Affiliation(s)
- Jacqueline M. Crater
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - Daniel Dunn
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - Douglas F. Nixon
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - Robert L. Furler O’Brien
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
6
|
Parent SE, Luu O, Bruce AEE, Winklbauer R. Two-phase kinetics and cell cortex elastic behavior in Xenopus gastrula cell-cell adhesion. Dev Cell 2024; 59:141-155.e6. [PMID: 38091998 DOI: 10.1016/j.devcel.2023.11.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 06/21/2023] [Accepted: 11/13/2023] [Indexed: 01/11/2024]
Abstract
Morphogenetic movements during animal development involve repeated making and breaking of cell-cell contacts. Recent biophysical models of cell-cell adhesion integrate adhesion molecule interactions and cortical cytoskeletal tension modulation, describing equilibrium states for established contacts. We extend this emerging unified concept of adhesion to contact formation kinetics, showing that aggregating Xenopus embryonic cells rapidly achieve Ca2+-independent low-contact states. Subsequent transitions to cadherin-dependent high-contact states show rapid decreases in contact cortical F-actin levels but slow contact area growth. We developed a biophysical model that predicted contact growth quantitatively from known cellular and cytoskeletal parameters, revealing that elastic resistance to deformation and cytoskeletal network turnover are essential determinants of adhesion kinetics. Characteristic time scales of contact growth to low and high states differ by an order of magnitude, being at a few minutes and tens of minutes, respectively, thus providing insight into the timescales of cell-rearrangement-dependent tissue movements.
Collapse
Affiliation(s)
- Serge E Parent
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.
| | - Olivia Luu
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Ashley E E Bruce
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.
| | - Rudolf Winklbauer
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.
| |
Collapse
|
7
|
Chikina AS, Zholudeva AO, Lomakina ME, Kireev II, Dayal AA, Minin AA, Maurin M, Svitkina TM, Alexandrova AY. Plasma Membrane Blebbing Is Controlled by Subcellular Distribution of Vimentin Intermediate Filaments. Cells 2024; 13:105. [PMID: 38201309 PMCID: PMC10778383 DOI: 10.3390/cells13010105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/18/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
The formation of specific cellular protrusions, plasma membrane blebs, underlies the amoeboid mode of cell motility, which is characteristic for free-living amoebae and leukocytes, and can also be adopted by stem and tumor cells to bypass unfavorable migration conditions and thus facilitate their long-distance migration. Not all cells are equally prone to bleb formation. We have previously shown that membrane blebbing can be experimentally induced in a subset of HT1080 fibrosarcoma cells, whereas other cells in the same culture under the same conditions retain non-blebbing mesenchymal morphology. Here we show that this heterogeneity is associated with the distribution of vimentin intermediate filaments (VIFs). Using different approaches to alter the VIF organization, we show that blebbing activity is biased toward cell edges lacking abundant VIFs, whereas the VIF-rich regions of the cell periphery exhibit low blebbing activity. This pattern is observed both in interphase fibroblasts, with and without experimentally induced blebbing, and during mitosis-associated blebbing. Moreover, the downregulation of vimentin expression or displacement of VIFs away from the cell periphery promotes blebbing even in cells resistant to bleb-inducing treatments. Thus, we reveal a new important function of VIFs in cell physiology that involves the regulation of non-apoptotic blebbing essential for amoeboid cell migration and mitosis.
Collapse
Affiliation(s)
- Aleksandra S. Chikina
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoe Shosse, Moscow 115478, Russia; (A.S.C.); (A.O.Z.); (M.E.L.)
- Dynamics of Immune Responses Team, INSERM-U1223 Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Anna O. Zholudeva
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoe Shosse, Moscow 115478, Russia; (A.S.C.); (A.O.Z.); (M.E.L.)
| | - Maria E. Lomakina
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoe Shosse, Moscow 115478, Russia; (A.S.C.); (A.O.Z.); (M.E.L.)
| | - Igor I. Kireev
- Department of Biology and A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, 1 Leninskie Gory, Moscow 119992, Russia;
| | - Alexander A. Dayal
- Institute of Protein Research, Department of Cell Biology, Russian Academy of Sciences, Moscow 119988, Russia; (A.A.D.); (A.A.M.)
| | - Alexander A. Minin
- Institute of Protein Research, Department of Cell Biology, Russian Academy of Sciences, Moscow 119988, Russia; (A.A.D.); (A.A.M.)
| | - Mathieu Maurin
- Institut Curie, PSL Research University, INSERM U932, 26 rue d’Ulm, 75248 Paris, France;
| | - Tatyana M. Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Antonina Y. Alexandrova
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoe Shosse, Moscow 115478, Russia; (A.S.C.); (A.O.Z.); (M.E.L.)
| |
Collapse
|
8
|
Zholudeva AO, Potapov NS, Kozlova EA, Lomakina ME, Alexandrova AY. Impairment of Assembly of the Vimentin Intermediate Filaments Leads to Suppression of Formation and Maturation of Focal Contacts and Alteration of the Type of Cellular Protrusions. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:184-195. [PMID: 38467554 DOI: 10.1134/s0006297924010127] [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: 09/04/2023] [Revised: 12/01/2023] [Accepted: 12/03/2023] [Indexed: 03/13/2024]
Abstract
Cell migration is largely determined by the type of protrusions formed by the cell. Mesenchymal migration is accomplished by formation of lamellipodia and/or filopodia, while amoeboid migration is based on bleb formation. Changing of migrational conditions can lead to alteration in the character of cell movement. For example, inhibition of the Arp2/3-dependent actin polymerization by the CK-666 inhibitor leads to transition from mesenchymal to amoeboid motility mode. Ability of the cells to switch from one type of motility to another is called migratory plasticity. Cellular mechanisms regulating migratory plasticity are poorly understood. One of the factors determining the possibility of migratory plasticity may be the presence and/or organization of vimentin intermediate filaments (VIFs). To investigate whether organization of the VIF network affects the ability of fibroblasts to form membrane blebs, we used rat embryo fibroblasts REF52 with normal VIF organization, fibroblasts with vimentin knockout (REF-/-), and fibroblasts with mutation inhibiting assembly of the full-length VIFs (REF117). Blebs formation was induced by treatment of cells with CK-666. Vimentin knockout did not lead to statistically significant increase in the number of cells with blebs. The fibroblasts with short fragments of vimentin demonstrate the significant increase in number of cells forming blebs both spontaneously and in the presence of CK-666. Disruption of the VIF organization did not lead to the significant changes in the microtubules network or the level of myosin light chain phosphorylation, but caused significant reduction in the focal contact system. The most pronounced and statistically significant decrease in both size and number of focal adhesions were observed in the REF117 cells. We believe that regulation of the membrane blebbing by VIFs is mediated by their effect on the focal adhesion system. Analysis of migration of fibroblasts with different organization of VIFs in a three-dimensional collagen gel showed that organization of VIFs determines the type of cell protrusions, which, in turn, determines the character of cell movement. A novel role of VIFs as a regulator of membrane blebbing, essential for manifestation of the migratory plasticity, is shown.
Collapse
Affiliation(s)
- Anna O Zholudeva
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| | - Nikolay S Potapov
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Ekaterina A Kozlova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Maria E Lomakina
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| | - Antonina Y Alexandrova
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia.
| |
Collapse
|
9
|
Asante-Asamani E, Dalton M, Brazill D, Strychalski W. Modeling the dynamics of actin and myosin during bleb stabilization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.564082. [PMID: 37961169 PMCID: PMC10634845 DOI: 10.1101/2023.10.26.564082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The actin cortex is very dynamic during migration of eukaryotes. In cells that use blebs as leading-edge protrusions, the cortex reforms beneath the cell membrane (bleb cortex) and completely disassembles at the site of bleb initiation. Remnants of the actin cortex at the site of bleb nucleation are referred to as the actin scar. We refer to the combined process of cortex reformation along with the degradation of the actin scar during bleb-based cell migration as bleb stabilization. The molecular factors that regulate the dynamic reorganization of the cortex are not fully understood. Myosin motor protein activity has been shown to be necessary for blebbing, with its major role associated with pressure generation to drive bleb expansion. Here, we examine the role of myosin in regulating cortex dynamics during bleb stabilization. Analysis of microscopy data from protein localization experiments in Dictyostelium discoideum cells reveals a rapid formation of the bleb's cortex with a delay in myosin accumulation. In the degrading actin scar, myosin is observed to accumulate before active degradation of the cortex begins. Through a combination of mathematical modeling and data fitting, we identify that myosin helps regulate the equilibrium concentration of actin in the bleb cortex during its reformation by increasing its dissasembly rate. Our modeling and analysis also suggests that cortex degradation is driven primarily by an exponential decrease in actin assembly rate rather than increased myosin activity. We attribute the decrease in actin assembly to the separation of the cell membrane from the cortex after bleb nucleation.
Collapse
Affiliation(s)
| | - Mackenzie Dalton
- Department of Mathematics, Clarkson University, Clarkson, Potsdam, NY 13699
| | | | - Wanda Strychalski
- Department of Mathematics, Applied Mathematics, and Statistics, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
10
|
Kapustina M, Li D, Zhu J, Wall B, Weinreb V, Cheney RE. Changes in cell surface excess are coordinated with protrusion dynamics during 3D motility. Biophys J 2023; 122:3656-3677. [PMID: 37207658 PMCID: PMC10541482 DOI: 10.1016/j.bpj.2023.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 03/23/2023] [Accepted: 04/20/2023] [Indexed: 05/21/2023] Open
Abstract
To facilitate rapid changes in morphology without endangering cell integrity, each cell possesses a substantial amount of cell surface excess (CSE) that can be promptly deployed to cover cell extensions. CSE can be stored in different types of small surface projections such as filopodia, microvilli, and ridges, with rounded bleb-like projections being the most common and rapidly achieved form of storage. We demonstrate that, similar to rounded cells in 2D culture, rounded cells in 3D collagen contain large amounts of CSE and use it to cover developing protrusions. Upon retraction of a protrusion, the CSE this produces is stored over the cell body similar to the CSE produced by cell rounding. We present high-resolution imaging of F-actin and microtubules (MTs) for different cell lines in a 3D environment and demonstrate the correlated changes between CSE and protrusion dynamics. To coordinate CSE storage and release with protrusion formation and motility, we expect cells to have specific mechanisms for regulating CSE, and we hypothesize that MTs play a substantial role in this mechanism by reducing cell surface dynamics and stabilizing CSE. We also suggest that different effects of MT depolymerization on cell motility, such as inhibiting mesenchymal motility and enhancing amoeboid, can be explained by this role of MTs in CSE regulation.
Collapse
Affiliation(s)
- Maryna Kapustina
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| | - Donna Li
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - James Zhu
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Brittany Wall
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Violetta Weinreb
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Richard E Cheney
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
11
|
Pugliese LA, De Lorenzi V, Bernardi M, Ghignoli S, Tesi M, Marchetti P, Pesce L, Cardarelli F. Unveiling nanoscale optical signatures of cytokine-induced β-cell dysfunction. Sci Rep 2023; 13:13342. [PMID: 37587148 PMCID: PMC10432522 DOI: 10.1038/s41598-023-40272-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/08/2023] [Indexed: 08/18/2023] Open
Abstract
Pro-inflammatory cytokines contribute to β-cell failure in both Type-1 and Type-2 Diabetes. Data collected so far allowed to dissect the genomic, transcriptomic, proteomic and biochemical landscape underlying cytokine-induced β-cell progression through dysfunction. Yet, no report thus far complemented such molecular information with the direct optical nanoscopy of the β-cell subcellular environment. Here we tackle this issue in Insulinoma 1E (INS-1E) β-cells by label-free fluorescence lifetime imaging microscopy (FLIM) and fluorescence-based super resolution imaging by expansion microscopy (ExM). It is found that 24-h exposure to IL-1β and IFN-γ is associated with a neat modification of the FLIM signature of cell autofluorescence due to the increase of either enzyme-bound NAD(P)H molecules and of oxidized lipid species. At the same time, ExM-based direct imaging unveils neat alteration of mitochondrial morphology (i.e. ~ 80% increase of mitochondrial circularity), marked degranulation (i.e. ~ 40% loss of insulin granules, with mis-localization of the surviving pool), appearance of F-actin-positive membrane blebs and an hitherto unknown extensive fragmentation of the microtubules network (e.g. ~ 37% reduction in the number of branches). Reported observations provide an optical-microscopy framework to interpret the amount of molecular information collected so far on β-cell dysfunction and pave the way to future ex-vivo and in-vivo investigations.
Collapse
Affiliation(s)
- Licia Anna Pugliese
- NEST Laboratory - Scuola Normale Superiore, Piazza San Silvestro 12, Pisa, Italy.
| | - Valentina De Lorenzi
- NEST Laboratory - Scuola Normale Superiore, Piazza San Silvestro 12, Pisa, Italy
| | - Mario Bernardi
- NEST Laboratory - Scuola Normale Superiore, Piazza San Silvestro 12, Pisa, Italy
| | - Samuele Ghignoli
- NEST Laboratory - Scuola Normale Superiore, Piazza San Silvestro 12, Pisa, Italy
| | - Marta Tesi
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Luca Pesce
- NEST Laboratory - Scuola Normale Superiore, Piazza San Silvestro 12, Pisa, Italy.
| | - Francesco Cardarelli
- NEST Laboratory - Scuola Normale Superiore, Piazza San Silvestro 12, Pisa, Italy.
| |
Collapse
|
12
|
Fernandes AR, Martins JP, Gomes ER, Mendes CS, Teodoro RO. Drosophila motor neuron boutons remodel through membrane blebbing coupled with muscle contraction. Nat Commun 2023; 14:3352. [PMID: 37291089 PMCID: PMC10250368 DOI: 10.1038/s41467-023-38421-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/26/2023] [Indexed: 06/10/2023] Open
Abstract
Wired neurons form new presynaptic boutons in response to increased synaptic activity, however the mechanism(s) by which this occurs remains uncertain. Drosophila motor neurons (MNs) have clearly discernible boutons that display robust structural plasticity, being therefore an ideal system in which to study activity-dependent bouton genesis. Here, we show that in response to depolarization and in resting conditions, MNs form new boutons by membrane blebbing, a pressure-driven mechanism that occurs in 3-D cell migration, but to our knowledge not previously described to occur in neurons. Accordingly, F-actin is decreased in boutons during outgrowth, and non-muscle myosin-II is dynamically recruited to newly formed boutons. Furthermore, muscle contraction plays a mechanical role, which we hypothesize promotes bouton addition by increasing MN confinement. Overall, we identified a mechanism by which established circuits form new boutons allowing their structural expansion and plasticity, using trans-synaptic physical forces as the main driving force.
Collapse
Affiliation(s)
- Andreia R Fernandes
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - João P Martins
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Edgar R Gomes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - César S Mendes
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Rita O Teodoro
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal.
| |
Collapse
|
13
|
Lin H, Fan Y, Zhi Z, Pang L, Sun D. Short-hairpin RNA-mediated suppression of cortactin may inhibit the migration and invasion abilities of endometrial cancer cells by reducing lamellipodia. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:1390-1399. [PMID: 37970440 PMCID: PMC10634056 DOI: 10.22038/ijbms.2023.67633.14863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 05/30/2023] [Indexed: 11/17/2023]
Abstract
Objectives The prognosis of endometrial cancer (EC) is significantly affected by tumor infiltration and metastasis. Cortactin (CTTN) regulates infiltration and metastasis in other tumors. Studies on the role and mechanism of CTTN in EC are limited and further studies are needed. Materials and Methods Quantitative PCR and immunohistochemistry were used to detect Ras-associated C3 botulinum toxin substrate 1 (Rac1) and CTTN in EC and normal tissues. The relationship between the expression of these two genes and their prognostic factors was analyzed. A CTTN-RNAi lentiviral system was constructed and transfected into EC cells. Migration and invasion were evaluated by scratch assay, transwell migration, and invasion assays. Pseudopodia formation was observed by immunofluorescence staining. Western blotting was performed to detect the expression of Rac1. Results The expression levels of Rac1 and CTTN in EC tissues were significantly higher than those in normal tissues. In the EC group, Rac1 and CTTN levels were correlated. The protein expression levels of Rac1 and CTTN were related to myometrial invasion and stage. After CTTN knockdown, the migration rate, invasiveness, and migratory ability of EC cells decreased significantly. Lamellipodia was observed to disappear with the appearance of blebs. Rac1 protein expression was decreased after CTTN knockdown. Conclusion CTTN may promote the invasion and migration of EC by lamellipodia. This effect may be related to the regulation of Rac1 by CTTN.
Collapse
Affiliation(s)
- Huisi Lin
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- These authors contributed eqully to this work
| | - Yujuan Fan
- Department of Gynecology and Obstetrics, University of the Chinese Academy of Sciences, Shenzhen Hospital, Shenzhen, China
- These authors contributed eqully to this work
| | - Zhifu Zhi
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lihong Pang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Dan Sun
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
14
|
Nguyen LTS, Robinson DN. The lectin Discoidin I acts in the cytoplasm to help assemble the contractile machinery. J Cell Biol 2022; 221:213504. [PMID: 36165849 PMCID: PMC9523886 DOI: 10.1083/jcb.202202063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 06/11/2022] [Accepted: 08/09/2022] [Indexed: 11/22/2022] Open
Abstract
Cellular functions, such as division and migration, require cells to undergo robust shape changes. Through their contractility machinery, cells also sense, respond, and adapt to their physical surroundings. In the cytoplasm, the contractility machinery organizes into higher order assemblies termed contractility kits (CKs). Using Dictyostelium discoideum, we previously identified Discoidin I (DscI), a classic secreted lectin, as a CK component through its physical interactions with the actin crosslinker Cortexillin I (CortI) and the scaffolding protein IQGAP2. Here, we find that DscI ensures robust cytokinesis through regulating intracellular components of the contractile machinery. Specifically, DscI is necessary for normal cytokinesis, cortical tension, membrane-cortex connections, and cortical distribution and mechanoresponsiveness of CortI. The dscI deletion mutants also have complex genetic epistatic relationships with CK components, acting as a genetic suppressor of cortI and iqgap1, but as an enhancer of iqgap2. This work underscores the fact that proteins like DiscI contribute in diverse ways to the activities necessary for optimal cell function.
Collapse
Affiliation(s)
- Ly T S Nguyen
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Douglas N Robinson
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
15
|
Alexandrova A, Lomakina M. How does plasticity of migration help tumor cells to avoid treatment: Cytoskeletal regulators and potential markers. Front Pharmacol 2022; 13:962652. [PMID: 36278174 PMCID: PMC9582651 DOI: 10.3389/fphar.2022.962652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/16/2022] [Indexed: 11/13/2022] Open
Abstract
Tumor shrinkage as a result of antitumor therapy is not the only and sufficient indicator of treatment success. Cancer progression leads to dissemination of tumor cells and formation of metastases - secondary tumor lesions in distant organs. Metastasis is associated with acquisition of mobile phenotype by tumor cells as a result of epithelial-to-mesenchymal transition and further cell migration based on cytoskeleton reorganization. The main mechanisms of individual cell migration are either mesenchymal, which depends on the activity of small GTPase Rac, actin polymerization, formation of adhesions with extracellular matrix and activity of proteolytic enzymes or amoeboid, which is based on the increase in intracellular pressure caused by the enhancement of actin cortex contractility regulated by Rho-ROCK-MLCKII pathway, and does not depend on the formation of adhesive structures with the matrix, nor on the activity of proteases. The ability of tumor cells to switch from one motility mode to another depending on cell context and environmental conditions, termed migratory plasticity, contributes to the efficiency of dissemination and often allows the cells to avoid the applied treatment. The search for new therapeutic targets among cytoskeletal proteins offers an opportunity to directly influence cell migration. For successful treatment it is important to assess the likelihood of migratory plasticity in a particular tumor. Therefore, the search for specific markers that can indicate a high probability of migratory plasticity is very important.
Collapse
|
16
|
Asante-Asamani E, Grange D, Rawal D, Santiago Z, Loustau J, Brazill D. A role for myosin II clusters and membrane energy in cortex rupture for Dictyostelium discoideum. PLoS One 2022; 17:e0265380. [PMID: 35468148 PMCID: PMC9037949 DOI: 10.1371/journal.pone.0265380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 03/01/2022] [Indexed: 11/18/2022] Open
Abstract
Blebs, pressure driven protrusions of the cell membrane, facilitate the movement of eukaryotic cells such as the soil amoeba Dictyostelium discoideum, white blood cells and cancer cells. Blebs initiate when the cell membrane separates from the underlying cortex. A local rupture of the cortex, has been suggested as a mechanism by which blebs are initiated. However, much clarity is still needed about how cells inherently regulate rupture of the cortex in locations where blebs are expected to form. In this work, we examine the role of membrane energy and the motor protein myosin II (myosin) in facilitating the cell driven rupture of the cortex. We perform under-agarose chemotaxis experiments, using Dictyostelium discoideum cells, to visualize the dynamics of myosin and calculate changes in membrane energy in the blebbing region. To facilitate a rapid detection of blebs and analysis of the energy and myosin distribution at the cell front, we introduce an autonomous bleb detection algorithm that takes in discrete cell boundaries and returns the coordinate location of blebs with its shape characteristics. We are able to identify by microscopy naturally occurring gaps in the cortex prior to membrane detachment at sites of bleb nucleation. These gaps form at positions calculated to have high membrane energy, and are associated with areas of myosin enrichment. Myosin is also shown to accumulate in the cortex prior to bleb initiation and just before the complete disassembly of the cortex. Together our findings provide direct spatial and temporal evidence to support cortex rupture as an intrinsic bleb initiation mechanism and suggests that myosin clusters are associated with regions of high membrane energy where its contractile activity leads to a rupture of the cortex at points of maximal energy.
Collapse
Affiliation(s)
| | - Daniel Grange
- Department of Applied Mathematics, Stony Brook University, New York, New York, United States of America
| | - Devarshi Rawal
- Mathematics and Statistics Department, Hunter College, Manhattan, New York, United States of America
| | - Zully Santiago
- Department of Natural Science, Baruch College, New York, New York, United States of America
| | - John Loustau
- Mathematics and Statistics Department, Hunter College, Manhattan, New York, United States of America
| | - Derrick Brazill
- Biological Science Department, Hunter College, Manhattan, New York, United States of America
- * E-mail:
| |
Collapse
|
17
|
Abstract
The cytoskeleton is a complex of detergent-insoluble components of the cytoplasm playing critical roles in cell motility, shape generation, and mechanical properties of a cell. Fibrillar polymers-actin filaments, microtubules, and intermediate filaments-are major constituents of the cytoskeleton, which constantly change their organization during cellular activities. The actin cytoskeleton is especially polymorphic, as actin filaments can form multiple higher-order assemblies performing different functions. Structural information about cytoskeleton organization is critical for understanding its functions and mechanisms underlying various forms of cellular activity. Because of the nanometer-scale thickness of cytoskeletal fibers, electron microscopy (EM) is a key tool to determine the structure of the cytoskeleton.This article describes application of rotary shadowing (or platinum replica ) EM (PREM) for visualization of the cytoskeleton . The procedure is applicable to thin cultured cells growing on glass coverslips and consists of detergent extraction (or mechanical "unroofing") of cells to expose their cytoskeleton , chemical fixation to provide stability, ethanol dehydration and critical point drying to preserve three-dimensionality, rotary shadowing with platinum to create contrast, and carbon coating to stabilize replicas. This technique provides easily interpretable three-dimensional images, in which individual cytoskeletal fibers are clearly resolved and individual proteins can be identified by immunogold labeling. More importantly, PREM is easily compatible with live cell imaging, so that one can correlate the dynamics of a cell or its components, e.g., expressed fluorescent proteins, with high-resolution structural organization of the cytoskeleton in the same cell.
Collapse
Affiliation(s)
- Tatyana Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
18
|
Actin Cytoskeletal Dynamics in Single-Cell Wound Repair. Int J Mol Sci 2021; 22:ijms221910886. [PMID: 34639226 PMCID: PMC8509258 DOI: 10.3390/ijms221910886] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 11/17/2022] Open
Abstract
The plasma membrane protects the eukaryotic cell from its surroundings and is essential for cell viability; thus, it is crucial that membrane disruptions are repaired quickly to prevent immediate dyshomeostasis and cell death. Accordingly, cells have developed efficient repair mechanisms to rapidly reseal ruptures and reestablish membrane integrity. The cortical actin cytoskeleton plays an instrumental role in both plasma membrane resealing and restructuring in response to damage. Actin directly aids membrane repair or indirectly assists auxiliary repair mechanisms. Studies investigating single-cell wound repair have often focused on the recruitment and activation of specialized repair machinery, despite the undeniable need for rapid and dynamic cortical actin modulation; thus, the role of the cortical actin cytoskeleton during wound repair has received limited attention. This review aims to provide a comprehensive overview of membrane repair mechanisms directly or indirectly involving cortical actin cytoskeletal remodeling.
Collapse
|
19
|
Halder D, Mallick D, Chatterjee A, Jana SS. Nonmuscle Myosin II in cancer cell migration and mechanotransduction. Int J Biochem Cell Biol 2021; 139:106058. [PMID: 34400319 DOI: 10.1016/j.biocel.2021.106058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 07/16/2021] [Accepted: 08/11/2021] [Indexed: 11/16/2022]
Abstract
Cell migration is a key step of cancer metastasis, immune-cell navigation, homing of stem cells and development. What adds complexity to it is the heterogeneity of the tissue environment that gives rise to a vast diversity of migratory mechanisms utilized by cells. A majority of cell motility mechanisms reported elsewhere largely converge in depicting the importance of the activity and complexity of actomyosin networks in the cell. In this review, we highlight the less discussed functional diversity of these actomyosin complexes and describe in detail how the major cellular actin-binding molecular motor proteins, nonmuscle myosin IIs are regulated and how they participate and mechanically reciprocate to changes in the microenvironment during cancer cell migration and tumor progression. Understanding the role of nonmuscle myosin IIs in the cancer cell is important for designing efficient therapeutic strategies to prevent cancer metastasis.
Collapse
Affiliation(s)
- Debdatta Halder
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata, India; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel(2)
| | - Ditipriya Mallick
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata, India
| | - Ananya Chatterjee
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata, India
| | - Siddhartha S Jana
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata, India.
| |
Collapse
|
20
|
Yu Y, Yoshimura SH. Investigating the morphological dynamics of the plasma membrane by high-speed atomic force microscopy. J Cell Sci 2021; 134:272010. [PMID: 34468000 DOI: 10.1242/jcs.243584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Despite numerous recent developments in bioimaging techniques, nanoscale and live-cell imaging of the plasma membrane has been challenging because of the insufficient z-resolution of optical microscopes, as well as the lack of fluorescent probes to specifically label small membrane structures. High-speed atomic force microscopy (HS-AFM) is a powerful tool for visualising the dynamics of a specimen surface and is therefore suitable for observing plasma membrane dynamics. Recent developments in HS-AFM for live-cell imaging have enabled the visualisation of the plasma membrane and the network of cortical actin underneath the membrane in a living cell. Furthermore, correlative imaging with fluorescence microscopy allows for the direct visualisation of morphological changes of the plasma membrane together with the dynamic assembly or disassembly of proteins during the entire course of endocytosis in a living cell. Here, we review these recent advances in HS-AFM in order to analyse various cellular events occurring at the cell surface.
Collapse
Affiliation(s)
- Yiming Yu
- Graduate School of Biostudies, Kyoto University, Yoshida-konoe, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shige H Yoshimura
- Graduate School of Biostudies, Kyoto University, Yoshida-konoe, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
21
|
Flormann DAD, Schu M, Terriac E, Thalla D, Kainka L, Koch M, Gad AKB, Lautenschläger F. A novel universal algorithm for filament network tracing and cytoskeleton analysis. FASEB J 2021; 35:e21582. [PMID: 33835502 DOI: 10.1096/fj.202100048r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/06/2021] [Accepted: 03/25/2021] [Indexed: 01/01/2023]
Abstract
The rapid development of advanced microscopy techniques over recent decades has significantly increased the quality of imaging and our understanding of subcellular structures, such as the organization of the filaments of the cytoskeleton using fluorescence and electron microscopy. However, these recent improvements in imaging techniques have not been matched by similar development of techniques for computational analysis of the images of filament networks that can now be obtained. Hence, for a wide range of applications, reliable computational analysis of such two-dimensional methods remains challenging. Here, we present a new algorithm for tracing of filament networks. This software can extract many important parameters from grayscale images of filament networks, including the mesh hole size, and filament length and connectivity (also known as Coordination Number). In addition, the method allows sub-networks to be distinguished in two-dimensional images using intensity thresholding. We show that the algorithm can be used to analyze images of cytoskeleton networks obtained using different advanced microscopy methods. We have thus developed a new improved method for computational analysis of two-dimensional images of filamentous networks that has wide applications for existing imaging techniques. The algorithm is available as open-source software.
Collapse
Affiliation(s)
- Daniel A D Flormann
- Department of Physics, Saarland University, Saarbruecken, Germany.,INM - Leibniz Institute for New Materials, Saarbruecken, Germany
| | - Moritz Schu
- Department of Physics, Saarland University, Saarbruecken, Germany
| | - Emmanuel Terriac
- INM - Leibniz Institute for New Materials, Saarbruecken, Germany
| | - Divyendu Thalla
- Department of Physics, Saarland University, Saarbruecken, Germany.,INM - Leibniz Institute for New Materials, Saarbruecken, Germany
| | - Lucina Kainka
- Department of Physics, Saarland University, Saarbruecken, Germany.,INM - Leibniz Institute for New Materials, Saarbruecken, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, Saarbruecken, Germany
| | - Annica K B Gad
- Department of Oncology and Metabolism, The Medical School, Weston Park Cancer Centre, Sheffield, UK.,Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal
| | - Franziska Lautenschläger
- Department of Physics, Saarland University, Saarbruecken, Germany.,INM - Leibniz Institute for New Materials, Saarbruecken, Germany
| |
Collapse
|
22
|
Molecular basis of functional exchangeability between ezrin and other actin-membrane associated proteins during cytokinesis. Exp Cell Res 2021; 403:112600. [PMID: 33862101 DOI: 10.1016/j.yexcr.2021.112600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/03/2021] [Accepted: 04/05/2021] [Indexed: 01/09/2023]
Abstract
The mechanism that mediates the interaction between the contractile ring and the plasma membrane during cytokinesis remains elusive. We previously found that ERM (Ezrin/Radixin/Moesin) proteins, which usually mediate cellular pole contraction, become over-accumulated at the cell equator and support furrow ingression upon the loss of other actin-membrane associated proteins, anillin and supervillin. In this study, we addressed the molecular basis of the exchangeability between ezrin and other actin-membrane associated proteins in mediating cortical contraction during cytokinesis. We found that depletion of anillin and supervillin caused over-accumulation of the membrane-associated FERM domain and actin-binding C-terminal domain (C-term) of ezrin at the cleavage furrow, respectively. This finding suggests that ezrin differentially shares its binding sites with these proteins on the actin cytoskeleton or inner membrane surface. Using chimeric mutants, we found that ezrin C-term, but not the FERM domain, can substitute for the corresponding anillin domains in cytokinesis and cell proliferation. On the other hand, either the membrane-associated or the actin/myosin-binding domains of anillin could not substitute for the corresponding ezrin domains in controlling cortical blebbing at the cell poles. Our results highlight specific designs of actin- or membrane-associated moieties of different actin-membrane associated proteins with limited exchangeability, which enables them to support diverse cortical activities on the shared actin-membrane interface during cytokinesis.
Collapse
|
23
|
Jia C, Shi J, Han T, Yu ACH, Qin P. Plasma Membrane Blebbing Dynamics Involved in the Reversibly Perforated Cell by Ultrasound-Driven Microbubbles. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:733-750. [PMID: 33358511 DOI: 10.1016/j.ultrasmedbio.2020.11.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 11/13/2020] [Accepted: 11/24/2020] [Indexed: 06/12/2023]
Abstract
The perforation of plasma membrane by ultrasound-driven microbubbles (i.e., sonoporation) provides a temporary window for transporting macromolecules into the cytoplasm that is promising with respect to drug delivery and gene therapy. To improve the efficacy of delivery while ensuring biosafety, membrane resealing and cell recovery are required to help sonoporated cells defy membrane injury and regain their normal function. Blebs are found to accompany the recovery of sonoporated cells. However, the spatiotemporal characteristics of blebs and the underlying mechanisms remain unclear. With a customized platform for ultrasound exposure and 2-D/3-D live single-cell imaging, localized membrane perforation was induced with ultrasound-driven microbubbles, and the cellular responses were monitored using multiple fluorescent probes. The results indicated that localized blebs undergoing four phases (nucleation, expansion, pausing and retraction) on a time scale of tens of seconds to minutes were specifically involved in the reversibly sonoporated cells. The blebs spatially correlated with the membrane perforation site and temporally lagged (about tens of seconds to minutes) the resealing of perforated membrane. Their diameter (about several microns) and lifetime (about tens of seconds to minutes) positively correlated with the degree of sonoporation. Further studies revealed that intracellular calcium transients might be an upstream signal for triggering blebbing nucleation; exocytotic lysosomes not only contributed to resealing of the perforated membrane, but also to the increasing bleb volume during expansion; and actin components accumulation facilitated bleb retraction. These results provide new insight into the short-term strategies that the sonoporated cell employs to recover on membrane perforation and to remodel membrane structure and a biophysical foundation for sonoporation-based therapy.
Collapse
Affiliation(s)
- Caixia Jia
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jianmin Shi
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Han
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Alfred C H Yu
- Schlegel Research Institute for Aging, University of Waterloo, Waterloo, ON, Canada
| | - Peng Qin
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
24
|
Graybill PM, Jana A, Kapania RK, Nain AS, Davalos RV. Single Cell Forces after Electroporation. ACS NANO 2021; 15:2554-2568. [PMID: 33236888 PMCID: PMC10949415 DOI: 10.1021/acsnano.0c07020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Exogenous high-voltage pulses increase cell membrane permeability through a phenomenon known as electroporation. This process may also disrupt the cell cytoskeleton causing changes in cell contractility; however, the contractile signature of cell force after electroporation remains unknown. Here, single-cell forces post-electroporation are measured using suspended extracellular matrix-mimicking nanofibers that act as force sensors. Ten, 100 μs pulses are delivered at three voltage magnitudes (500, 1000, and 1500 V) and two directions (parallel and perpendicular to cell orientation), exposing glioblastoma cells to electric fields between 441 V cm-1 and 1366 V cm-1. Cytoskeletal-driven force loss and recovery post-electroporation involves three distinct stages. Low electric field magnitudes do not cause disruption, but higher fields nearly eliminate contractility 2-10 min post-electroporation as cells round following calcium-mediated retraction (stage 1). Following rounding, a majority of analyzed cells enter an unusual and unexpected biphasic stage (stage 2) characterized by increased contractility tens of minutes post-electroporation, followed by force relaxation. The biphasic stage is concurrent with actin disruption-driven blebbing. Finally, cells elongate and regain their pre-electroporation morphology and contractility in 1-3 h (stage 3). With increasing voltages applied perpendicular to cell orientation, we observe a significant drop in cell viability. Experiments with multiple healthy and cancerous cell lines demonstrate that contractile force is a more dynamic and sensitive metric than cell shape to electroporation. A mechanobiological understanding of cell contractility post-electroporation will deepen our understanding of the mechanisms that drive recovery and may have implications for molecular medicine, genetic engineering, and cellular biophysics.
Collapse
Affiliation(s)
- Philip M Graybill
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Aniket Jana
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Rakesh K Kapania
- Department of Aerospace and Ocean Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Amrinder S Nain
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, Virginia 24061, United States
| | - Rafael V Davalos
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, Virginia 24061, United States
| |
Collapse
|
25
|
Phenotypic Plasticity of Cancer Cells Based on Remodeling of the Actin Cytoskeleton and Adhesive Structures. Int J Mol Sci 2021; 22:ijms22041821. [PMID: 33673054 PMCID: PMC7918886 DOI: 10.3390/ijms22041821] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 02/08/2023] Open
Abstract
There is ample evidence that, instead of a binary switch, epithelial-mesenchymal transition (EMT) in cancer results in a flexible array of phenotypes, each one uniquely suited to a stage in the invasion-metastasis cascade. The phenotypic plasticity of epithelium-derived cancer cells gives them an edge in surviving and thriving in alien environments. This review describes in detail the actin cytoskeleton and E-cadherin-based adherens junction rearrangements that cancer cells need to implement in order to achieve the advantageous epithelial/mesenchymal phenotype and plasticity of migratory phenotypes that can arise from partial EMT.
Collapse
|
26
|
Ren W, Zhao W, Cao L, Huang J. Involvement of the Actin Machinery in Programmed Cell Death. Front Cell Dev Biol 2021; 8:634849. [PMID: 33634110 PMCID: PMC7900405 DOI: 10.3389/fcell.2020.634849] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 12/28/2020] [Indexed: 12/15/2022] Open
Abstract
Programmed cell death (PCD) depicts a genetically encoded and an orderly mode of cellular mortality. When triggered by internal or external stimuli, cells initiate PCDs through evolutionary conserved regulatory mechanisms. Actin, as a multifunctional cytoskeleton protein that forms microfilament, its integrity and dynamics are essential for a variety of cellular processes (e.g., morphogenesis, membrane blebbing and intracellular transport). Decades of work have broadened our knowledge about different types of PCDs and their distinguished signaling pathways. However, an ever-increasing pool of evidences indicate that the delicate relationship between PCDs and the actin cytoskeleton is beginning to be elucidated. The purpose of this article is to review the current understanding of the relationships between different PCDs and the actin machinery (actin, actin-binding proteins and proteins involved in different actin signaling pathways), in the hope that this attempt can shed light on ensuing studies and the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Weida Ren
- Key Laboratory for Regenerative Medicine, Ministry of Education, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Wanyu Zhao
- Key Laboratory for Regenerative Medicine, Ministry of Education, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Lingbo Cao
- Key Laboratory for Regenerative Medicine, Ministry of Education, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Junqi Huang
- Key Laboratory for Regenerative Medicine, Ministry of Education, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
27
|
Ghosh I, Singh RK, Mishra M, Kapoor S, Jana SS. Switching between blebbing and lamellipodia depends on the degree of non-muscle myosin II activity. J Cell Sci 2021; 134:jcs.248732. [PMID: 33298514 DOI: 10.1242/jcs.248732] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 11/19/2020] [Indexed: 01/09/2023] Open
Abstract
Cells can adopt both mesenchymal and amoeboid modes of migration through membrane protrusive activities, namely formation of lamellipodia and blebbing. How the molecular players control the transition between lamellipodia and blebs is yet to be explored. Here, we show that addition of the ROCK inhibitor Y27632 or low doses of blebbistatin, an inhibitor of non-muscle myosin II (NMII) ATPase activity and filament partitioning, induces blebbing to lamellipodia conversion (BLC), whereas addition of low doses of ML7, an inhibitor of myosin light chain kinase (MLCK), induces lamellipodia to blebbing conversion (LBC) in human MDA-MB-231 cells. Similarly, siRNA-mediated knockdown of ROCK and MLCK induces BLC and LBC, respectively. Interestingly, both blebs and lamellipodia membrane protrusions are able to maintain the ratio of phosphorylated to unphosphorylated regulatory light chain at cortices when MLCK and ROCK, respectively, are inhibited either pharmacologically or genetically, suggesting that MLCK and ROCK activities are interlinked in BLC and LBC. Such BLCs and LBCs are also inducible in other cell lines, including MCF7 and MCF10A. These studies reveal that the relative activity of ROCK and MLCK, which controls both the ATPase activity and filament-forming property of NMII, is a determining factor in whether a cell exhibits blebbing or lamellipodia.
Collapse
Affiliation(s)
- Indranil Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Raman K Singh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India.,Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Manjari Mishra
- Department of Chemistry, Indian Institute of Technology - Bombay, Mumbai 400076, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology - Bombay, Mumbai 400076, India
| | - Siddhartha S Jana
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| |
Collapse
|
28
|
Morone N, Usukura E, Narita A, Usukura J. Improved unroofing protocols for cryo-electron microscopy, atomic force microscopy and freeze-etching electron microscopy and the associated mechanisms. Microscopy (Oxf) 2020; 69:350-359. [PMID: 32447402 DOI: 10.1093/jmicro/dfaa028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/05/2020] [Accepted: 05/18/2020] [Indexed: 11/13/2022] Open
Abstract
Unroofing, which is the mechanical shearing of a cell to expose the cytoplasmic surface of the cell membrane, is a unique preparation method that allows membrane cytoskeletons to be observed by cryo-electron microscopy, atomic force microscopy, freeze-etching electron microscopy and other methods. Ultrasound and adhesion have been known to mechanically unroof cells. In this study, unroofing using these two means was denoted sonication unroofing and adhesion unroofing, respectively. We clarified the mechanisms by which cell membranes are removed in these unroofing procedures and established efficient protocols for each based on the mechanisms. In sonication unroofing, fine bubbles generated by sonication adhered electrostatically to apical cell surfaces and then removed the apical (dorsal) cell membrane with the assistance of buoyancy and water flow. The cytoplasmic surface of the ventral cell membrane remaining on the grids became observable by this method. In adhesion unroofing, grids charged positively by coating with Alcian blue were pressed onto the cells, thereby tightly adsorbing the dorsal cell membrane. Subsequently, a part of the cell membrane strongly adhered to the grids was peeled from the cells and transferred onto the grids when the grids were lifted. This method thus allowed the visualization of the cytoplasmic surface of the dorsal cell membrane. This paper describes robust, improved protocols for the two unroofing methods in detail. In addition, micro-unroofing (perforation) likely due to nanobubbles is introduced as a new method to make cells transparent to electron beams.
Collapse
Affiliation(s)
- Nobuhiro Morone
- Medical Research Council Toxicology Unit, University of Cambridge, Lancaster Road, Leicester LE1 9HN, UK
| | - Eiji Usukura
- Institute of Materials and Systems for Sustainability, Nagoya University, Chikusa-ku, Nagoya 464-8601, Japan
| | - Akihiro Narita
- Structural Biology Research Centre, Graduate School of Science, Nagoya University, Chikusa-ku,Nagoya,464-8601, Japan
| | - Jiro Usukura
- Institute of Materials and Systems for Sustainability, Nagoya University, Chikusa-ku, Nagoya 464-8601, Japan
| |
Collapse
|
29
|
Marcksl1 modulates endothelial cell mechanoresponse to haemodynamic forces to control blood vessel shape and size. Nat Commun 2020; 11:5476. [PMID: 33127887 PMCID: PMC7603353 DOI: 10.1038/s41467-020-19308-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 10/02/2020] [Indexed: 11/16/2022] Open
Abstract
The formation of vascular tubes is driven by extensive changes in endothelial cell (EC) shape. Here, we have identified a role of the actin-binding protein, Marcksl1, in modulating the mechanical properties of EC cortex to regulate cell shape and vessel structure during angiogenesis. Increasing and depleting Marcksl1 expression level in vivo results in an increase and decrease, respectively, in EC size and the diameter of microvessels. Furthermore, endothelial overexpression of Marcksl1 induces ectopic blebbing on both apical and basal membranes, during and after lumen formation, that is suppressed by reduced blood flow. High resolution imaging reveals that Marcksl1 promotes the formation of linear actin bundles and decreases actin density at the EC cortex. Our findings demonstrate that a balanced network of linear and branched actin at the EC cortex is essential in conferring cortical integrity to resist the deforming forces of blood flow to regulate vessel structure. During lumen formation in blood vessels, endothelial cells become exposed to hemodynamic forces that induce membrane blebbing and changes in cell shape. Here, the authors show endothelial cells develop an actin-based protective mechanism in the cell cortex that prevents excessive blebbing to control cell shape and vessel diameter.
Collapse
|
30
|
Alexandrova AY, Chikina AS, Svitkina TM. Actin cytoskeleton in mesenchymal-to-amoeboid transition of cancer cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:197-256. [PMID: 33066874 DOI: 10.1016/bs.ircmb.2020.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During development of metastasis, tumor cells migrate through different tissues and encounter different extracellular matrices. An ability of cells to adapt mechanisms of their migration to these diverse environmental conditions, called migration plasticity, gives tumor cells an advantage over normal cells for long distant dissemination. Different modes of individual cell motility-mesenchymal and amoeboid-are driven by different molecular mechanisms, which largely depend on functions of the actin cytoskeleton that can be modulated in a wide range by cellular signaling mechanisms in response to environmental conditions. Various triggers can switch one motility mode to another, but regulations of these transitions are incompletely understood. However, understanding of the mechanisms driving migration plasticity is instrumental for finding anti-cancer treatment capable to stop cancer metastasis. In this review, we discuss cytoskeletal features, which allow the individually migrating cells to switch between mesenchymal and amoeboid migrating modes, called mesenchymal-to-amoeboid transition (MAT). We briefly describe main characteristics of different cell migration modes, and then discuss the triggering factors that initiate MAT with special attention to cytoskeletal features essential for migration plasticity.
Collapse
Affiliation(s)
- Antonina Y Alexandrova
- Laboratory of Mechanisms of Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia.
| | - Aleksandra S Chikina
- Cell Migration and Invasion and Spatio-Temporal Regulation of Antigen Presentation teams, UMR144/U932 Institut Curie, Paris, France
| | - Tatyana M Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
31
|
Svitkina TM. Actin Cell Cortex: Structure and Molecular Organization. Trends Cell Biol 2020; 30:556-565. [PMID: 32278656 PMCID: PMC7566779 DOI: 10.1016/j.tcb.2020.03.005] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/11/2020] [Accepted: 03/19/2020] [Indexed: 12/20/2022]
Abstract
The actin cytoskeleton consists of structurally and biochemically different actin filament arrays. Among them, the actin cortex is thought to have key roles in cell mechanics, but remains a poorly characterized part of the actin cytoskeleton. The cell cortex is typically defined as a thin layer of actin meshwork that uniformly underlies the plasma membrane of the entire cell. However, this definition applies only to specific cases. In general, the cortex structure and subcellular distribution vary significantly across cell types and physiological states of the cell. In this review, I focus on our current knowledge of the structure and molecular composition of the cell cortex.
Collapse
Affiliation(s)
- Tatyana M Svitkina
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
32
|
Novák J, Vopálenský V, Pospíšek M, Vedeler A. Co-localization of Interleukin-1α and Annexin A2 at the plasma membrane in response to oxidative stress. Cytokine 2020; 133:155141. [PMID: 32615410 DOI: 10.1016/j.cyto.2020.155141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/11/2020] [Accepted: 05/15/2020] [Indexed: 12/19/2022]
Abstract
Interleukin-1α (IL-1α) and Annexin A2 (AnxA2) are pleiotropic molecules with both intracellular and extracellular roles. They share several characteristics including unconventional secretion aided by S100 proteins, anchoring of the externalized proteins at the outer surface of the plasma membrane and response to oxidative stress. Although IL-1α and AnxA2 have been implicated in a variety of biological processes, including cancer, little is known about the mechanisms of their cellular release. In the present study, employing the non-cancerous breast epithelial MCF10A cells, we demonstrate that IL-1α and AnxA2 establish a close association in response to oxidative stress. Stress conditions lead to translocation of both proteins towards lamellipodia rich in vimentin and association of full-length IL-1α and Tyr23 phosphorylated AnxA2 with the plasma membrane at peripheral sites depleted of F-actin. Notably, membrane-associated IL-1α and AnxA2 preferentially localize to the outer edges of the MCF10A cell islands, suggesting that the two proteins participate in the communication of these epithelial cells with their neighboring cells. Similarly, in U2OS osteosarcoma cell line both endogenous IL-1α and transiently produced IL-1α/EGFP associate with the plasma membrane. While benign MFC10A cells present membrane-associated IL-1α and AnxA2 at the edges of their cell islands, the aggressive cancerous U2OS cells communicate in such manner also with distant cells.
Collapse
Affiliation(s)
- Josef Novák
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic; Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway.
| | - Václav Vopálenský
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Martin Pospíšek
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Anni Vedeler
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
33
|
Abstract
Cell migration is an essential process, both in unicellular organisms such as amoeba and as individual or collective motility in highly developed multicellular organisms like mammals. It is controlled by a variety of activities combining protrusive and contractile forces, normally generated by actin filaments. Here, we summarize actin filament assembly and turnover processes, and how respective biochemical activities translate into different protrusion types engaged in migration. These actin-based plasma membrane protrusions include actin-related protein 2/3 complex-dependent structures such as lamellipodia and membrane ruffles, filopodia as well as plasma membrane blebs. We also address observed antagonisms between these protrusion types, and propose a model - also inspired by previous literature - in which a complex balance between specific Rho GTPase signaling pathways dictates the protrusion mechanism employed by cells. Furthermore, we revisit published work regarding the fascinating antagonism between Rac and Rho GTPases, and how this intricate signaling network can define cell behavior and modes of migration. Finally, we discuss how the assembly of actin filament networks can feed back onto their regulators, as exemplified for the lamellipodial factor WAVE regulatory complex, tightly controlling accumulation of this complex at specific subcellular locations as well as its turnover.
Collapse
|
34
|
Peropadre A, Hazen MJ, Pérez Martín JM, Fernández Freire P. An acute exposure to perfluorooctanoic acid causes non-reversible plasma membrane injury in HeLa cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 260:114008. [PMID: 31995777 DOI: 10.1016/j.envpol.2020.114008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 12/17/2019] [Accepted: 01/16/2020] [Indexed: 06/10/2023]
Abstract
Health and environmental risks regarding perfluorooctanoic acid, a well-known perfluorinated compound, are still a subject of great concern. Ubiquitous exposure and disparity of results make it difficult to determine the underlying mechanism of action, especially at the cellular level. This study proposes an experimental design to assess the reversibility of adverse effects after a one-time exposure to the compound, in comparison with other more conventional timings. Complementary endpoints including total protein content, neutral red uptake and MTT reduction tests along with division rates and microscopic observations were evaluated in HeLa cells. In addition, PFOA quantification inside the cells was performed. The cellular effects exerted after 24 h exposure to perfluorooctanoic acid are non-reversible after a 48 h recovery period. In addition, we describe for the first time the induction of plasma membrane blebbing and the activation of membrane repair mechanisms after recovery from non-cytotoxic treatments with the compound. This experimental design has provided relevant information regarding the toxicity of this perfluorinated compound, relating all the adverse effects detected to its interaction with the plasma membrane.
Collapse
Affiliation(s)
- Ana Peropadre
- Department of Biology (Lab A-110), Faculty of Sciences, Universidad Autónoma de Madrid, C/Darwin 2, 28049, Madrid, Spain
| | - Maria José Hazen
- Department of Biology (Lab A-110), Faculty of Sciences, Universidad Autónoma de Madrid, C/Darwin 2, 28049, Madrid, Spain
| | - José Manuel Pérez Martín
- Department of Biology (Lab A-110), Faculty of Sciences, Universidad Autónoma de Madrid, C/Darwin 2, 28049, Madrid, Spain
| | - Paloma Fernández Freire
- Department of Biology (Lab A-110), Faculty of Sciences, Universidad Autónoma de Madrid, C/Darwin 2, 28049, Madrid, Spain.
| |
Collapse
|
35
|
Polesskaya A, Vicente-Manzanares M. Meeting Report - Workshop 'Actin-based mechanosensation and force generation in health and disease'. J Cell Sci 2020; 133:133/6/jcs244319. [PMID: 32184275 DOI: 10.1242/jcs.244319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
International experts in the fields of cellular motility, force generation and mechanosensation met in Baeza, a UNESCO World Heritage city, from the 10th to the 13th of November, 2019. The meeting, part of the 'Current Trends in Biomedicine' series, took place at the 'Sede Antonio Machado', a beautiful 17th century building turned into a conference center of the Universidad Internacional de Andalucía (UNIA), which sponsored the event. The meeting was organized by Alexis Gautreau, Pekka Lappalainen and Miguel Vicente-Manzanares, with the support of the European Molecular Biology Organization (EMBO) and the Spanish-based company IMPETUX. Fifty scientists presented recent results during the talks, poster sessions and thematic discussions. As Baeza itself served as a crossroads of medieval Christian, Moorish and Jewish cultures, the meeting brought together cell biologists, biochemists, biophysicists and engineers from around the world that provided an integrated vision of the role of the actin cytoskeleton, force generation and mechanosensation in diverse physiological processes and pathologies.
Collapse
Affiliation(s)
- Anna Polesskaya
- CNRS UMR7654, Institut Polytechnique de Paris, 91120 Palaiseau, France
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 370-07 Salamanca, Spain
| |
Collapse
|
36
|
Liu H, Zhu L, Dudiki T, Gabanic B, Good L, Podrez EA, Cherepanova OA, Qin J, Byzova TV. Macrophage Migration and Phagocytosis Are Controlled by Kindlin-3's Link to the Cytoskeleton. THE JOURNAL OF IMMUNOLOGY 2020; 204:1954-1967. [PMID: 32094207 DOI: 10.4049/jimmunol.1901134] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 01/15/2020] [Indexed: 01/08/2023]
Abstract
Major myeloid cell functions from adhesion to migration and phagocytosis are mediated by integrin adhesion complexes, also known as adhesome. The presence of a direct integrin binding partner Kindlin-3 is crucial for these functions, and its lack causes severe immunodeficiency in humans. However, how Kindlin-3 is incorporated into the adhesome and how its function is regulated is poorly understood. In this study, using nuclear magnetic resonance spectroscopy, we show that Kindlin-3 directly interacts with paxillin (PXN) and leupaxin (LPXN) via G43/L47 within its F0 domain. Surprisingly, disruption of Kindlin-3-PXN/LPXN interactions in Raw 264.7 macrophages promoted cell spreading and polarization, resulting in upregulation of both general cell motility and directed cell migration, which is in a drastic contrast to the consequences of Kindlin-3 knockout. Moreover, disruption of Kindlin-3-PXN/LPXN binding promoted the transition from mesenchymal to amoeboid mode of movement as well as augmented phagocytosis. Thus, these novel links between Kindlin-3 and key adhesome members PXN/LPXN limit myeloid cell motility and phagocytosis, thereby providing an important immune regulatory mechanism.
Collapse
Affiliation(s)
- Huan Liu
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Liang Zhu
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Tejasvi Dudiki
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Benjamin Gabanic
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Logan Good
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Eugene A Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Olga A Cherepanova
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Jun Qin
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Tatiana V Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195;
| |
Collapse
|
37
|
Brito C, Mesquita FS, Bleck CKE, Sellers JR, Cabanes D, Sousa S. Perfringolysin O-Induced Plasma Membrane Pores Trigger Actomyosin Remodeling and Endoplasmic Reticulum Redistribution. Toxins (Basel) 2019; 11:toxins11070419. [PMID: 31319618 PMCID: PMC6669444 DOI: 10.3390/toxins11070419] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/10/2019] [Accepted: 07/15/2019] [Indexed: 01/28/2023] Open
Abstract
Clostridium perfringens produces an arsenal of toxins that act together to cause severe infections in humans and livestock animals. Perfringolysin O (PFO) is a cholesterol-dependent pore-forming toxin encoded in the chromosome of virtually all C. perfringens strains and acts in synergy with other toxins to determine the outcome of the infection. However, its individual contribution to the disease is poorly understood. Here, we intoxicated human epithelial and endothelial cells with purified PFO to evaluate the host cytoskeletal responses to PFO-induced damage. We found that, at sub-lytic concentrations, PFO induces a profound reorganization of the actomyosin cytoskeleton culminating into the assembly of well-defined cortical actomyosin structures at sites of plasma membrane (PM) remodeling. The assembly of such structures occurs concomitantly with the loss of the PM integrity and requires pore-formation, calcium influx, and myosin II activity. The recovery from the PM damage occurs simultaneously with the disassembly of cortical structures. PFO also targets the endoplasmic reticulum (ER) by inducing its disruption and vacuolation. ER-enriched vacuoles were detected at the cell cortex within the PFO-induced actomyosin structures. These cellular events suggest the targeting of the endothelium integrity at early stages of C. perfringens infection, in which secreted PFO is at sub-lytic concentrations.
Collapse
Affiliation(s)
- Cláudia Brito
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, 4099-002 Porto, Portugal
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel, Salazar, Universidade do Porto, 4099-002 Porto, Portugal
| | - Francisco S Mesquita
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, 4099-002 Porto, Portugal
| | - Christopher K E Bleck
- Electron Microscopy Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - James R Sellers
- Laboratory of Molecular Physiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Didier Cabanes
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, 4099-002 Porto, Portugal
| | - Sandra Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, 4099-002 Porto, Portugal.
| |
Collapse
|