1
|
Landino J, Misterovich E, van den Goor L, Adhikary B, Chumki S, Davidson LA, Miller AL. Neighbor cells restrain furrowing during Xenopus epithelial cytokinesis. Dev Cell 2025:S1534-5807(25)00157-1. [PMID: 40203834 DOI: 10.1016/j.devcel.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/15/2024] [Accepted: 03/17/2025] [Indexed: 04/11/2025]
Abstract
Cytokinesis challenges epithelial tissue homeostasis by generating forces that pull on neighboring cells. Junction reinforcement at the furrow in Xenopus epithelia regulates the speed of furrowing, suggesting that cytokinesis is subject to resistive forces from epithelial neighbors. We show that contractility factors accumulate near the furrow in neighboring cells, and increasing neighbor cell stiffness slows furrowing. Optogenetically increasing contractility in one or both neighbor cells slows furrowing or induces cytokinetic failure. Uncoupling mechanotransduction between dividing cells and their neighbors increases the furrow ingression rate, alters topological cell packing following cytokinesis, and impairs barrier function at the furrow. Computational modeling validates our findings and provides additional insights about epithelial mechanics during cytokinesis. We conclude that forces from the cytokinetic array must be carefully balanced with restraining forces generated by neighbor cells to regulate the speed and success of cytokinesis and maintain epithelial homeostasis.
Collapse
Affiliation(s)
- Jennifer Landino
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| | - Eileen Misterovich
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Lotte van den Goor
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Babli Adhikary
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Shahana Chumki
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Lance A Davidson
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ann L Miller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA; Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Craig Z, Arnold TR, Walworth K, Walkon A, Miller AL. Anillin tunes contractility and regulates barrier function during Rho flare-mediated tight junction remodeling. Mol Biol Cell 2025; 36:ar31. [PMID: 39841565 PMCID: PMC11974952 DOI: 10.1091/mbc.e24-11-0513] [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: 11/22/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
To preserve barrier function, cell-cell junctions must dynamically remodel during cell shape changes. We have previously described a rapid tight junction repair pathway characterized by local, transient activations of RhoA, termed "Rho flares," which repair leaks in tight junctions via promoting local actomyosin-mediated junction remodeling. In this pathway, junction elongation is a mechanical trigger that initiates RhoA activation through an influx of intracellular calcium and recruitment of p115RhoGEF. However, mechanisms that tune the level of RhoA activation and Myosin II contractility during the process remain uncharacterized. Here, we show that the scaffolding protein Anillin localizes to Rho flares and regulates RhoA activity and actomyosin contraction at flares. Knocking down Anillin results in Rho flares with increased intensity but shorter duration. These changes in active RhoA dynamics weaken downstream F-actin and Myosin II accumulation at the site of Rho flares, resulting in decreased junction contraction. Consequently, tight junction breaks are not reinforced following Rho flares. We show that Anillin-driven RhoA regulation is necessary for successfully repairing tight junction leaks and protecting junctions from repeated barrier damage. Together, these results uncover a novel regulatory role for Anillin during tight junction repair and barrier function maintenance.
Collapse
Affiliation(s)
- Zie Craig
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Torey R. Arnold
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Kelsey Walworth
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Alexander Walkon
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Ann L. Miller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
3
|
Raj N, Weiß MS, Vos BE, Weischer S, Brinkmann F, Betz T, Trappmann B, Gerke V. Membrane Tension Regulation is Required for Wound Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402317. [PMID: 39360573 DOI: 10.1002/advs.202402317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 09/17/2024] [Indexed: 10/04/2024]
Abstract
Disruptions of the eukaryotic plasma membrane due to chemical and mechanical challenges are frequent and detrimental and thus need to be repaired to maintain proper cell function and avoid cell death. However, the cellular mechanisms involved in wound resealing and restoration of homeostasis are diverse and contended. Here, it is shown that clathrin-mediated endocytosis is induced at later stages of plasma membrane wound repair following the actual resealing of the wound. This compensatory endocytosis occurs near the wound, predominantly at sites of previous early endosome exocytosis which is required in the initial stage of membrane resealing, suggesting a spatio-temporal co-ordination of exo- and endocytosis during wound repair. Using cytoskeletal alterations and modulations of membrane tension and membrane area, membrane tension is identified as a major regulator of the wounding-associated exo- and endocytic events that mediate efficient wound repair. Thus, membrane tension changes are a universal trigger for plasma membrane wound repair modulating the exocytosis of early endosomes required for resealing and subsequent clathrin-mediated endocytosis acting at later stages to restore cell homeostasis and function.
Collapse
Affiliation(s)
- Nikita Raj
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation (ZMBE), Multiscale Imaging Centre, Cells in Motion Interfaculty Center, University of Münster, 48149, Münster, Germany
| | - Martin S Weiß
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
| | - Bart E Vos
- Third Institute of Physics, University of Göttingen, 37077, Göttingen, Germany
| | - Sarah Weischer
- Multiscale Imaging Centre, Cells in Motion Interfaculty Center, University of Münster, 48149, Münster, Germany
| | - Frauke Brinkmann
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation (ZMBE), University of Münster, 48149, Münster, Germany
| | - Timo Betz
- Third Institute of Physics, University of Göttingen, 37077, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37077, Göttingen, Germany
| | - Britta Trappmann
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
- Department of Chemistry and Chemical Biology, TU Dortmund University, 44227, Dortmund, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation (ZMBE), Multiscale Imaging Centre, Cells in Motion Interfaculty Center, University of Münster, 48149, Münster, Germany
| |
Collapse
|
4
|
Gest AM, Sahan AZ, Zhong Y, Lin W, Mehta S, Zhang J. Molecular Spies in Action: Genetically Encoded Fluorescent Biosensors Light up Cellular Signals. Chem Rev 2024; 124:12573-12660. [PMID: 39535501 PMCID: PMC11613326 DOI: 10.1021/acs.chemrev.4c00293] [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: 04/17/2024] [Revised: 09/07/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024]
Abstract
Cellular function is controlled through intricate networks of signals, which lead to the myriad pathways governing cell fate. Fluorescent biosensors have enabled the study of these signaling pathways in living systems across temporal and spatial scales. Over the years there has been an explosion in the number of fluorescent biosensors, as they have become available for numerous targets, utilized across spectral space, and suited for various imaging techniques. To guide users through this extensive biosensor landscape, we discuss critical aspects of fluorescent proteins for consideration in biosensor development, smart tagging strategies, and the historical and recent biosensors of various types, grouped by target, and with a focus on the design and recent applications of these sensors in living systems.
Collapse
Affiliation(s)
- Anneliese
M. M. Gest
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Ayse Z. Sahan
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, California 92093, United States
| | - Yanghao Zhong
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Wei Lin
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Sohum Mehta
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Jin Zhang
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Shu
Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
5
|
Craig Z, Arnold TR, Walworth K, Walkon A, Miller AL. Anillin tunes contractility and regulates barrier function during Rho flare-mediated tight junction remodeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624537. [PMID: 39605712 PMCID: PMC11601591 DOI: 10.1101/2024.11.20.624537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
To preserve barrier function, cell-cell junctions must dynamically remodel during cell shape changes. We have previously described a rapid tight junction repair pathway characterized by local, transient activation of RhoA, termed 'Rho flares,' which repair leaks in tight junctions via promoting local actomyosin-mediated junction remodeling. In this pathway, junction elongation is a mechanical trigger that initiates RhoA activation through an influx of intracellular calcium and recruitment of p115RhoGEF. However, mechanisms that tune the level of RhoA activation and Myosin II contractility during the process remain uncharacterized. Here, we show that the scaffolding protein Anillin localizes to Rho flares and regulates RhoA activity and actomyosin contraction at flares. Knocking down Anillin results in Rho flares with increased intensity but shorter duration. These changes in active RhoA dynamics weaken downstream F-actin and Myosin II accumulation at the site of Rho flares, resulting in decreased junction contraction. Consequently, tight junction breaks are not reinforced following Rho flares. We show that Anillin-driven RhoA regulation is necessary for successfully repairing tight junction leaks and protecting junctions from repeated barrier damage. Together, these results uncover a novel regulatory role for Anillin during tight junction repair and barrier function maintenance.
Collapse
Affiliation(s)
- Zie Craig
- Department of Molecular, Cellular, and Developmental Biology; University of Michigan; Ann Arbor, Michigan, 48109; USA
| | - Torey R. Arnold
- Department of Molecular, Cellular, and Developmental Biology; University of Michigan; Ann Arbor, Michigan, 48109; USA
| | - Kelsey Walworth
- Department of Molecular, Cellular, and Developmental Biology; University of Michigan; Ann Arbor, Michigan, 48109; USA
| | - Alexander Walkon
- Department of Molecular, Cellular, and Developmental Biology; University of Michigan; Ann Arbor, Michigan, 48109; USA
| | - Ann L. Miller
- Department of Molecular, Cellular, and Developmental Biology; University of Michigan; Ann Arbor, Michigan, 48109; USA
| |
Collapse
|
6
|
Nakamura M, Parkhurst SM. Calcium influx rapidly establishes distinct spatial recruitments of Annexins to cell wounds. Genetics 2024; 227:iyae101. [PMID: 38874345 PMCID: PMC11304956 DOI: 10.1093/genetics/iyae101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/04/2024] [Accepted: 06/09/2024] [Indexed: 06/15/2024] Open
Abstract
To survive daily damage, the formation of actomyosin ring at the wound edge is required to rapidly close cell wounds. Calcium influx is one of the start signals for these cell wound repair events. Here, we find that the rapid recruitment of all 3 Drosophila calcium-responding and phospholipid-binding Annexin proteins (AnxB9, AnxB10, and AnxB11) to distinct regions around the wound is regulated by the quantity of calcium influx rather than their binding to specific phospholipids. The distinct recruitment patterns of these Annexins regulate the subsequent recruitment of RhoGEF2 and RhoGEF3 through actin stabilization to form a robust actomyosin ring. Surprisingly, while the wound does not close in the absence of calcium influx, we find that reduced calcium influx can still initiate repair processes, albeit leading to severe repair phenotypes. Thus, our results suggest that, in addition to initiating repair events, the quantity of calcium influx is important for precise Annexin spatiotemporal protein recruitment to cell wounds and efficient wound repair.
Collapse
Affiliation(s)
- Mitsutoshi Nakamura
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Susan M Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| |
Collapse
|
7
|
Quinn CJ, Cartwright EJ, Trafford AW, Dibb KM. On the role of dysferlin in striated muscle: membrane repair, t-tubules and Ca 2+ handling. J Physiol 2024; 602:1893-1910. [PMID: 38615232 DOI: 10.1113/jp285103] [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: 06/27/2023] [Accepted: 03/05/2024] [Indexed: 04/15/2024] Open
Abstract
Dysferlin is a 237 kDa membrane-associated protein characterised by multiple C2 domains with a diverse role in skeletal and cardiac muscle physiology. Mutations in DYSF are known to cause various types of human muscular dystrophies, known collectively as dysferlinopathies, with some patients developing cardiomyopathy. A myriad of in vitro membrane repair studies suggest that dysferlin plays an integral role in the membrane repair complex in skeletal muscle. In comparison, less is known about dysferlin in the heart, but mounting evidence suggests that dysferlin's role is similar in both muscle types. Recent findings have shown that dysferlin regulates Ca2+ handling in striated muscle via multiple mechanisms and that this becomes more important in conditions of stress. Maintenance of the transverse (t)-tubule network and the tight coordination of excitation-contraction coupling are essential for muscle contractility. Dysferlin regulates the maintenance and repair of t-tubules, and it is suspected that dysferlin regulates t-tubules and sarcolemmal repair through a similar mechanism. This review focuses on the emerging complexity of dysferlin's activity in striated muscle. Such insights will progress our understanding of the proteins and pathways that regulate basic heart and skeletal muscle function and help guide research into striated muscle pathology, especially that which arises due to dysferlin dysfunction.
Collapse
Affiliation(s)
- C J Quinn
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| | - E J Cartwright
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| | - A W Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| | - K M Dibb
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| |
Collapse
|
8
|
Bement WM, Goryachev AB, Miller AL, von Dassow G. Patterning of the cell cortex by Rho GTPases. Nat Rev Mol Cell Biol 2024; 25:290-308. [PMID: 38172611 DOI: 10.1038/s41580-023-00682-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2023] [Indexed: 01/05/2024]
Abstract
The Rho GTPases - RHOA, RAC1 and CDC42 - are small GTP binding proteins that regulate basic biological processes such as cell locomotion, cell division and morphogenesis by promoting cytoskeleton-based changes in the cell cortex. This regulation results from active (GTP-bound) Rho GTPases stimulating target proteins that, in turn, promote actin assembly and myosin 2-based contraction to organize the cortex. This basic regulatory scheme, well supported by in vitro studies, led to the natural assumption that Rho GTPases function in vivo in an essentially linear matter, with a given process being initiated by GTPase activation and terminated by GTPase inactivation. However, a growing body of evidence based on live cell imaging, modelling and experimental manipulation indicates that Rho GTPase activation and inactivation are often tightly coupled in space and time via signalling circuits and networks based on positive and negative feedback. In this Review, we present and discuss this evidence, and we address one of the fundamental consequences of coupled activation and inactivation: the ability of the Rho GTPases to self-organize, that is, direct their own transition from states of low order to states of high order. We discuss how Rho GTPase self-organization results in the formation of diverse spatiotemporal cortical patterns such as static clusters, oscillatory pulses, travelling wave trains and ring-like waves. Finally, we discuss the advantages of Rho GTPase self-organization and pattern formation for cell function.
Collapse
Affiliation(s)
- William M Bement
- Center for Quantitative Cell Imaging, Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Andrew B Goryachev
- Center for Engineering Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK.
| | - Ann L Miller
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| | | |
Collapse
|
9
|
Xu S, Yang TJ, Xu S, Gong YN. Plasma membrane repair empowers the necrotic survivors as innate immune modulators. Semin Cell Dev Biol 2024; 156:93-106. [PMID: 37648621 PMCID: PMC10872800 DOI: 10.1016/j.semcdb.2023.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/20/2023] [Accepted: 08/20/2023] [Indexed: 09/01/2023]
Abstract
The plasma membrane is crucial to the survival of animal cells, and damage to it can be lethal, often resulting in necrosis. However, cells possess multiple mechanisms for repairing the membrane, which allows them to maintain their integrity to some extent, and sometimes even survive. Interestingly, cells that survive a near-necrosis experience can recognize sub-lethal membrane damage and use it as a signal to secrete chemokines and cytokines, which activate the immune response. This review will present evidence of necrotic cell survival in both in vitro and in vivo systems, including in C. elegans, mouse models, and humans. We will also summarize the various membrane repair mechanisms cells use to maintain membrane integrity. Finally, we will propose a mathematical model to illustrate how near-death experiences can transform dying cells into innate immune modulators for their microenvironment. By utilizing their membrane repair activity, the biological effects of cell death can extend beyond the mere elimination of the cells.
Collapse
Affiliation(s)
- Shiqi Xu
- Center for Stem Cell and Regenerative Medicine and Department of Burn and Wound Repair of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine and the Zhejiang University-University of Edinburgh Institute, 718 East Haizhou Rd., Haining, Zhejiang 314400, China
| | - Tyler J Yang
- Departments of Biology and Advanced Placement Biology, White Station High School, Memphis, TN 38117, USA
| | - Suhong Xu
- Center for Stem Cell and Regenerative Medicine and Department of Burn and Wound Repair of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine and the Zhejiang University-University of Edinburgh Institute, 718 East Haizhou Rd., Haining, Zhejiang 314400, China.
| | - Yi-Nan Gong
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, 5115 Center Avenue, Pittsburgh, PA 15213, USA.
| |
Collapse
|
10
|
Sepaniac LA, Davenport NR, Bement WM. Bring the pain: wounding reveals a transition from cortical excitability to epithelial excitability in Xenopus embryos. Front Cell Dev Biol 2024; 11:1295569. [PMID: 38456169 PMCID: PMC10918254 DOI: 10.3389/fcell.2023.1295569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 12/08/2023] [Indexed: 03/09/2024] Open
Abstract
The cell cortex plays many critical roles, including interpreting and responding to internal and external signals. One behavior which supports a cell's ability to respond to both internal and externally-derived signaling is cortical excitability, wherein coupled positive and negative feedback loops generate waves of actin polymerization and depolymerization at the cortex. Cortical excitability is a highly conserved behavior, having been demonstrated in many cell types and organisms. One system well-suited to studying cortical excitability is Xenopus laevis, in which cortical excitability is easily monitored for many hours after fertilization. Indeed, recent investigations using X. laevis have furthered our understanding of the circuitry underlying cortical excitability and how it contributes to cytokinesis. Here, we describe the impact of wounding, which represents both a chemical and a physical signal, on cortical excitability. In early embryos (zygotes to early blastulae), we find that wounding results in a transient cessation ("freezing") of wave propagation followed by transport of frozen waves toward the wound site. We also find that wounding near cell-cell junctions results in the formation of an F-actin (actin filament)-based structure that pulls the junction toward the wound; at least part of this structure is based on frozen waves. In later embryos (late blastulae to gastrulae), we find that cortical excitability diminishes and is progressively replaced by epithelial excitability, a process in which wounded cells communicate with other cells via wave-like increases of calcium and apical F-actin. While the F-actin waves closely follow the calcium waves in space and time, under some conditions the actin wave can be uncoupled from the calcium wave, suggesting that they may be independently regulated by a common upstream signal. We conclude that as cortical excitability disappears from the level of the individual cell within the embryo, it is replaced by excitability at the level of the embryonic epithelium itself.
Collapse
Affiliation(s)
- Leslie A. Sepaniac
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, United States
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, United States
| | - Nicholas R. Davenport
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, United States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, United States
| | - William M. Bement
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, United States
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, United States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
11
|
Yumura S. Wound Repair of the Cell Membrane: Lessons from Dictyostelium Cells. Cells 2024; 13:341. [PMID: 38391954 PMCID: PMC10886852 DOI: 10.3390/cells13040341] [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: 12/20/2023] [Revised: 01/30/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
The cell membrane is frequently subjected to damage, either through physical or chemical means. The swift restoration of the cell membrane's integrity is crucial to prevent the leakage of intracellular materials and the uncontrolled influx of extracellular ions. Consequently, wound repair plays a vital role in cell survival, akin to the importance of DNA repair. The mechanisms involved in wound repair encompass a series of events, including ion influx, membrane patch formation, endocytosis, exocytosis, recruitment of the actin cytoskeleton, and the elimination of damaged membrane sections. Despite the absence of a universally accepted general model, diverse molecular models have been proposed for wound repair in different organisms. Traditional wound methods not only damage the cell membrane but also impact intracellular structures, including the underlying cortical actin networks, microtubules, and organelles. In contrast, the more recent improved laserporation selectively targets the cell membrane. Studies on Dictyostelium cells utilizing this method have introduced a novel perspective on the wound repair mechanism. This review commences by detailing methods for inducing wounds and subsequently reviews recent developments in the field.
Collapse
Affiliation(s)
- Shigehiko Yumura
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8511, Japan
| |
Collapse
|
12
|
Nanda S, Calderon A, Sachan A, Duong TT, Koch J, Xin X, Solouk-Stahlberg D, Wu YW, Nalbant P, Dehmelt L. Rho GTPase activity crosstalk mediated by Arhgef11 and Arhgef12 coordinates cell protrusion-retraction cycles. Nat Commun 2023; 14:8356. [PMID: 38102112 PMCID: PMC10724141 DOI: 10.1038/s41467-023-43875-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023] Open
Abstract
Rho GTPases play a key role in the spatio-temporal coordination of cytoskeletal dynamics during cell migration. Here, we directly investigate crosstalk between the major Rho GTPases Rho, Rac and Cdc42 by combining rapid activity perturbation with activity measurements in mammalian cells. These studies reveal that Rac stimulates Rho activity. Direct measurement of spatio-temporal activity patterns show that Rac activity is tightly and precisely coupled to local cell protrusions, followed by Rho activation during retraction. Furthermore, we find that the Rho-activating Lbc-type GEFs Arhgef11 and Arhgef12 are enriched at transient cell protrusions and retractions and recruited to the plasma membrane by active Rac. In addition, their depletion reduces activity crosstalk, cell protrusion-retraction dynamics and migration distance and increases migration directionality. Thus, our study shows that Arhgef11 and Arhgef12 facilitate exploratory cell migration by coordinating cell protrusion and retraction by coupling the activity of the associated regulators Rac and Rho.
Collapse
Affiliation(s)
- Suchet Nanda
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Abram Calderon
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Arya Sachan
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
| | - Thanh-Thuy Duong
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Johannes Koch
- Department of Molecular Cell Biology, Center of Medical Biotechnology, University of Duisburg-Essen, 45141, Essen, Germany
| | - Xiaoyi Xin
- SciLifeLab and Department of Chemistry, Umeå Centre for Microbial Research, Umeå University, 90187, Umeå, Sweden
| | - Djamschid Solouk-Stahlberg
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Yao-Wen Wu
- SciLifeLab and Department of Chemistry, Umeå Centre for Microbial Research, Umeå University, 90187, Umeå, Sweden
| | - Perihan Nalbant
- Department of Molecular Cell Biology, Center of Medical Biotechnology, University of Duisburg-Essen, 45141, Essen, Germany.
| | - Leif Dehmelt
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany.
| |
Collapse
|
13
|
Nakamura M, Parkhurst SM. Calcium influx rapidly establishes distinct spatial recruitments of Annexins to cell wounds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.03.569799. [PMID: 38105960 PMCID: PMC10723296 DOI: 10.1101/2023.12.03.569799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
To survive daily damage, the formation of actomyosin ring at the wound periphery is required to rapidly close cell wounds. Calcium influx is one of the start signals for these cell wound repair events. Here, we find that rapid recruitment of all three Drosophila calcium responding and phospholipid binding Annexin proteins (AnxB9, AnxB10, AnxB11) to distinct regions around the wound are regulated by the quantity of calcium influx rather than their binding to specific phospholipids. The distinct recruitment patterns of these Annexins regulate the subsequent recruitment of RhoGEF2 and RhoGEF3 through actin stabilization to form a robust actomyosin ring. Surprisingly, we find that reduced extracellular calcium and depletion of intracellular calcium affect cell wound repair differently, despite these two conditions exhibiting similar GCaMP signals. Thus, our results suggest that, in addition to initiating repair events, both the quantity and sources of calcium influx are important for precise Annexin spatiotemporal protein recruitment to cell wounds and efficient wound repair.
Collapse
Affiliation(s)
- Mitsutoshi Nakamura
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA 98109
| | - Susan M. Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA 98109
| |
Collapse
|
14
|
Shao Q, Wijaya CS, Wang S, Meng X, Yuan C, Ma C, Xu S. The SNARE complex formed by RIC-4/SEC-22/SYX-2 promotes C. elegans epidermal wound healing. Cell Rep 2023; 42:113349. [PMID: 37910502 DOI: 10.1016/j.celrep.2023.113349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/18/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023] Open
Abstract
Maintaining cellular viability relies on the integrity of the plasma membrane, which must be repaired upon damage. Soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE)-mediated membrane fusion is a crucial mechanism involved in membrane repair. In C. elegans epidermal cell hyp 7, syntaxin-2 (SYX-2) facilitates large membrane wound repair; however, the underlying molecular mechanism remains unclear. Here, we found that SNAP-25 protein RIC-4 and synaptobrevin protein SEC-22 are required for SYX-2 recruitment at the wound site. They interact to form a SNARE complex to promote membrane repair in vivo and fusion in vitro. Moreover, we found that SEC-22 localized in multiple intracellular compartments, including endosomes and the trans-Golgi network, which recruited to the wounds. Furthermore, inhibition of RAB-5 disrupted SEC-22 localization and prevented its interaction with SYX-2. Our findings suggest that RAB-5 facilitates the formation of the RIC-4/SEC-22/SYX-2 SNARE complex and provides valuable insights into the molecular mechanism of how cells repair large membrane wounds.
Collapse
Affiliation(s)
- Qingfang Shao
- International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine and the Zhejiang University-University of Edinburgh Institute, 718 East Haizhou Road, Haining, Zhejiang 314400, China; Center for Stem Cell and Regenerative Medicine and Department of Burn and Wound Repair of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Chandra Sugiarto Wijaya
- International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine and the Zhejiang University-University of Edinburgh Institute, 718 East Haizhou Road, Haining, Zhejiang 314400, China; School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xinan Meng
- International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine and the Zhejiang University-University of Edinburgh Institute, 718 East Haizhou Road, Haining, Zhejiang 314400, China; School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Cheng Yuan
- International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine and the Zhejiang University-University of Edinburgh Institute, 718 East Haizhou Road, Haining, Zhejiang 314400, China
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Suhong Xu
- International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine and the Zhejiang University-University of Edinburgh Institute, 718 East Haizhou Road, Haining, Zhejiang 314400, China; Center for Stem Cell and Regenerative Medicine and Department of Burn and Wound Repair of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
15
|
Zha D, Wang S, Monaghan-Nichols P, Qian Y, Sampath V, Fu M. Mechanisms of Endothelial Cell Membrane Repair: Progress and Perspectives. Cells 2023; 12:2648. [PMID: 37998383 PMCID: PMC10670313 DOI: 10.3390/cells12222648] [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: 09/08/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
Endothelial cells are the crucial inner lining of blood vessels, which are pivotal in vascular homeostasis and integrity. However, these cells are perpetually subjected to a myriad of mechanical, chemical, and biological stresses that can compromise their plasma membranes. A sophisticated repair system involving key molecules, such as calcium, annexins, dysferlin, and MG53, is essential for maintaining endothelial viability. These components orchestrate complex mechanisms, including exocytosis and endocytosis, to repair membrane disruptions. Dysfunctions in this repair machinery, often exacerbated by aging, are linked to endothelial cell death, subsequently contributing to the onset of atherosclerosis and the progression of cardiovascular diseases (CVD) and stroke, major causes of mortality in the United States. Thus, identifying the core machinery for endothelial cell membrane repair is critically important for understanding the pathogenesis of CVD and stroke and developing novel therapeutic strategies for combating CVD and stroke. This review summarizes the recent advances in understanding the mechanisms of endothelial cell membrane repair. The future directions of this research area are also highlighted.
Collapse
Affiliation(s)
- Duoduo Zha
- Department of Biomedical Science, School of Medicine, University of Missouri Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA; (D.Z.); (P.M.-N.)
- The National Engineering Research Center for Bioengineering Drugs and Technologies, Institute of Translational Medicine, Nanchang University, 1299 Xuefu Rd, Honggu District, Nanchang 330031, China;
| | - Shizhen Wang
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri Kansas City, 5009 Rockhill Road, Kansas City, MO 64110, USA;
| | - Paula Monaghan-Nichols
- Department of Biomedical Science, School of Medicine, University of Missouri Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA; (D.Z.); (P.M.-N.)
| | - Yisong Qian
- The National Engineering Research Center for Bioengineering Drugs and Technologies, Institute of Translational Medicine, Nanchang University, 1299 Xuefu Rd, Honggu District, Nanchang 330031, China;
| | - Venkatesh Sampath
- Department of Pediatric, Children’s Mercy Hospital, Children’s Mercy Research Institute, Kansas City, MO 64108, USA;
| | - Mingui Fu
- Department of Biomedical Science, School of Medicine, University of Missouri Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA; (D.Z.); (P.M.-N.)
| |
Collapse
|
16
|
Golding AE, Li W, Blank PS, Cologna SM, Zimmerberg J. Relative quantification of progressive changes in healthy and dysferlin-deficient mouse skeletal muscle proteomes. Muscle Nerve 2023; 68:805-816. [PMID: 37706611 DOI: 10.1002/mus.27975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023]
Abstract
INTRODUCTION/AIMS Individuals with dysferlinopathies, a group of genetic muscle diseases, experience delay in the onset of muscle weakness. The cause of this delay and subsequent muscle wasting are unknown, and there are currently no clinical interventions to limit or prevent muscle weakness. To better understand molecular drivers of dysferlinopathies, age-dependent changes in the proteomic profile of skeletal muscle (SM) in wild-type (WT) and dysferlin-deficient mice were identified. METHODS Quadriceps were isolated from 6-, 18-, 42-, and 77-wk-old C57BL/6 (WT, Dysf+/+ ) and BLAJ (Dysf-/- ) mice (n = 3, 2 male/1 female or 1 male/2 female, 24 total). Whole-muscle proteomes were characterized using liquid chromatography-mass spectrometry with relative quantification using TMT10plex isobaric labeling. Principle component analysis was utilized to detect age-dependent proteomic differences over the lifespan of, and between, WT and dysferlin-deficient SM. The biological relevance of proteins with significant variation was established using Ingenuity Pathway Analysis. RESULTS Over 3200 proteins were identified between 6-, 18-, 42-, and 77-wk-old mice. In total, 46 proteins varied in aging WT SM (p < .01), while 365 varied in dysferlin-deficient SM. However, 569 proteins varied between aged-matched WT and dysferlin-deficient SM. Proteins with significant variation in expression across all comparisons followed distinct temporal trends. DISCUSSION Proteins involved in sarcolemma repair and regeneration underwent significant changes in SM over the lifespan of WT mice, while those associated with immune infiltration and inflammation were overly represented over the lifespan of dysferlin-deficient mice. The proteins identified herein are likely to contribute to our overall understanding of SM aging and dysferlinopathy disease progression.
Collapse
Affiliation(s)
- Adriana E Golding
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Section on Intracellular Protein Trafficking, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Wenping Li
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois, USA
| | - Paul S Blank
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois, USA
| | - Joshua Zimmerberg
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
17
|
Bittel DC, Jaiswal JK. Early Endosomes Undergo Calcium-Triggered Exocytosis and Enable Repair of Diffuse and Focal Plasma Membrane Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300245. [PMID: 37705135 PMCID: PMC10667805 DOI: 10.1002/advs.202300245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/27/2023] [Indexed: 09/15/2023]
Abstract
Cells are routinely exposed to agents that cause plasma membrane (PM) injury. While pore-forming toxins (PFTs), and chemicals cause nanoscale holes dispersed throughout the PM, mechanical trauma causes focal lesions in the PM. To examine if these distinct injuries share common repair mechanism, membrane trafficking is monitored as the PM repairs from such injuries. During the course of repair, dispersed PM injury by the PFT Streptolysin O activates endocytosis, while focal mechanical injury to the PM inhibits endocytosis. Consequently, acute block of endocytosis prevents repair of diffuse, but not of focal injury. In contrast, a chronic block in endocytosis depletes cells of early endosomes and inhibits repair of focal injury. This study finds that both focal and diffuse PM injury activate Ca2+ -triggered exocytosis of early endosomes. The use of markers including endocytosed cargo, Rab5, Rab11, and VAMP3, all reveal injury-triggered exocytosis of early endosomes. Inhibiting Rab5 prevents injury-triggered early endosome exocytosis and phenocopies the failed PM repair of cells chronically depleted of early endosomes. These results identify early endosomes as a Ca2+ -regulated exocytic compartment, and uncover the requirement of their dual functions - endocytosis and regulated exocytosis, to differentially support PM repair based on the nature of the injury.
Collapse
Affiliation(s)
- Daniel C. Bittel
- Center for Genetic Medicine ResearchChildren's National Research Institute7144 13th Pl NWWashington, DC20012USA
| | - Jyoti K. Jaiswal
- Center for Genetic Medicine ResearchChildren's National Research Institute7144 13th Pl NWWashington, DC20012USA
- Department of Genomics and Precision MedicineGeorge Washington University School of Medicine and Health SciencesWashington, DC20012USA
| |
Collapse
|
18
|
Meng X, Wijaya CS, Shao Q, Xu S. Triggered Golgi membrane enrichment promotes PtdIns(4,5)P2 generation for plasma membrane repair. J Cell Biol 2023; 222:214098. [PMID: 37158801 PMCID: PMC10176212 DOI: 10.1083/jcb.202303017] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/06/2023] [Accepted: 04/20/2023] [Indexed: 05/10/2023] Open
Abstract
The maintenance of plasma membrane integrity and a capacity for efficiently repairing damaged membranes are essential for cell survival. Large-scale wounding depletes various membrane components at the wound sites, including phosphatidylinositols, yet little is known about how phosphatidylinositols are generated after depletion. Here, working with our in vivo C. elegans epidermal cell wounding model, we discovered phosphatidylinositol 4-phosphate (PtdIns4P) accumulation and local phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] generation at the wound site. We found that PtdIns(4,5)P2 generation depends on the delivery of PtdIns4P, PI4K, and PI4P 5-kinase PPK-1. In addition, we show that wounding triggers enrichment of the Golgi membrane to the wound site, and that is required for membrane repair. Moreover, genetic and pharmacological inhibitor experiments support that the Golgi membrane provides the PtdIns4P for PtdIns(4,5)P2 generation at the wounds. Our findings demonstrate how the Golgi apparatus facilitates membrane repair in response to wounding and offers a valuable perspective on cellular survival mechanisms upon mechanical stress in a physiological context.
Collapse
Affiliation(s)
- Xinan Meng
- International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine and the Zhejiang University-University of Edinburgh Institute , Haining Zhejiang, China
| | - Chandra Sugiarto Wijaya
- Department of Burn and Wound Repair of the Second Affiliated Hospital, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou Zhejiang, China
- School of Basic Medical Sciences, Zhejiang University School of Medicine , Hangzhou Zhejiang, China
| | - Qingfang Shao
- International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine and the Zhejiang University-University of Edinburgh Institute , Haining Zhejiang, China
| | - Suhong Xu
- International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine and the Zhejiang University-University of Edinburgh Institute , Haining Zhejiang, China
- Department of Burn and Wound Repair of the Second Affiliated Hospital, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou Zhejiang, China
- School of Basic Medical Sciences, Zhejiang University School of Medicine , Hangzhou Zhejiang, China
| |
Collapse
|
19
|
Landino J, Misterovich E, Chumki S, Miller AL. Neighbor cells restrain furrowing during epithelial cytokinesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544077. [PMID: 37333405 PMCID: PMC10274919 DOI: 10.1101/2023.06.08.544077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Cytokinesis challenges epithelial tissue homeostasis by generating forces that pull on neighboring cells via cell-cell junctions. Previous work has shown that junction reinforcement at the furrow in Xenopus laevis epithelia regulates the speed of furrowing1. This suggests the cytokinetic array that drives cell division is subject to resistive forces from epithelial neighbor cells. We show here that contractility factors accumulate in neighboring cells near the furrow during cytokinesis. Additionally, increasing neighbor cell stiffness, via ɑ-actinin overexpression, or contractility, through optogenetic Rho activation in one neighbor cell, slows or asymmetrically pauses furrowing, respectively. Notably, optogenetic stimulation of neighbor cell contractility on both sides of the furrow induces cytokinetic failure and binucleation. We conclude that forces from the cytokinetic array in the dividing cell are carefully balanced with restraining forces generated by neighbor cells, and neighbor cell mechanics regulate the speed and success of cytokinesis.
Collapse
Affiliation(s)
- Jennifer Landino
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor
| | - Eileen Misterovich
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor
| | - Shahana Chumki
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor
| | - Ann L. Miller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor
| |
Collapse
|
20
|
Raj N, Greune L, Kahms M, Mildner K, Franzkoch R, Psathaki OE, Zobel T, Zeuschner D, Klingauf J, Gerke V. Early Endosomes Act as Local Exocytosis Hubs to Repair Endothelial Membrane Damage. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300244. [PMID: 36938863 PMCID: PMC10161044 DOI: 10.1002/advs.202300244] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/21/2023] [Indexed: 05/06/2023]
Abstract
The plasma membrane of a cell is subject to stresses causing ruptures that must be repaired immediately to preserve membrane integrity and ensure cell survival. Yet, the spatio-temporal membrane dynamics at the wound site and the source of the membrane required for wound repair are poorly understood. Here, it is shown that early endosomes, previously only known to function in the uptake of extracellular material and its endocytic transport, are involved in plasma membrane repair in human endothelial cells. Using live-cell imaging and correlative light and electron microscopy, it is demonstrated that membrane injury triggers a previously unknown exocytosis of early endosomes that is induced by Ca2+ entering through the wound. This exocytosis is restricted to the vicinity of the wound site and mediated by the endosomal soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) VAMP2, which is crucial for efficient membrane repair. Thus, the newly identified Ca2+ -evoked and localized exocytosis of early endosomes supplies the membrane material required for rapid resealing of a damaged plasma membrane, thereby providing the first line of defense against damage in mechanically challenged endothelial cells.
Collapse
Affiliation(s)
- Nikita Raj
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation (ZMBE), Cells in Motion Interfaculty Center, University of Münster, 48149, Münster, Germany
| | - Lilo Greune
- Institute of Infectiology, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, 48149, Münster, Germany
| | - Martin Kahms
- Institute of Medical Physics and Biophysics, University of Münster, 48149, Münster, Germany
| | - Karina Mildner
- Electron Microscopy Facility, Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
| | - Rico Franzkoch
- Department of Biology, integrated Bioimaging Facility (iBiOs), Center of Cellular Nanoanalytics (CellNanO), University of Osnabrück, 49076, Osnabrück, Germany
| | - Olympia Ekaterini Psathaki
- Department of Biology, integrated Bioimaging Facility (iBiOs), Center of Cellular Nanoanalytics (CellNanO), University of Osnabrück, 49076, Osnabrück, Germany
| | - Thomas Zobel
- Imaging Network, Cells in Motion Interfaculty Centre, University of Münster, 48149, Münster, Germany
| | - Dagmar Zeuschner
- Electron Microscopy Facility, Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
| | - Jürgen Klingauf
- Institute of Medical Physics and Biophysics, University of Münster, 48149, Münster, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation (ZMBE), Cells in Motion Interfaculty Center, University of Münster, 48149, Münster, Germany
| |
Collapse
|
21
|
Mamun MAA, Cao W, Nakamura S, Maruyama JI. Large-scale identification of genes involved in septal pore plugging in multicellular fungi. Nat Commun 2023; 14:1418. [PMID: 36932089 PMCID: PMC10023807 DOI: 10.1038/s41467-023-36925-y] [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: 01/20/2022] [Accepted: 02/23/2023] [Indexed: 03/19/2023] Open
Abstract
Multicellular filamentous fungi have septal pores that allow cytoplasmic exchange, and thus connectivity, between neighboring cells in the filament. Hyphal wounding and other stress conditions induce septal pore closure to minimize cytoplasmic loss. However, the composition of the septal pore and the mechanisms underlying its function are not well understood. Here, we set out to identify new septal components by determining the subcellular localization of 776 uncharacterized proteins in a multicellular ascomycete, Aspergillus oryzae. The set of 776 uncharacterized proteins was selected on the basis that their genes were present in the genomes of multicellular, septal pore-bearing ascomycetes (three Aspergillus species, in subdivision Pezizomycotina) and absent/divergent in the genomes of septal pore-lacking ascomycetes (yeasts). Upon determining their subcellular localization, 62 proteins were found to localize to the septum or septal pore. Deletion of the encoding genes revealed that 23 proteins are involved in regulating septal pore plugging upon hyphal wounding. Thus, this study determines the subcellular localization of many uncharacterized proteins in A. oryzae and, in particular, identifies a set of proteins involved in septal pore function.
Collapse
Affiliation(s)
| | - Wei Cao
- Research Center for Agricultural Information Technology, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Shugo Nakamura
- Department of Information Networking for Innovation and Design, Faculty of Information Networking for Innovation and Design, Toyo University, Tokyo, Japan
| | - Jun-Ichi Maruyama
- Department of Biotechnology, The University of Tokyo, Tokyo, Japan.
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
22
|
Mencel ML, Bittner GD. Repair of traumatic lesions to the plasmalemma of neurons and other cells: Commonalities, conflicts, and controversies. Front Physiol 2023; 14:1114779. [PMID: 37008019 PMCID: PMC10050709 DOI: 10.3389/fphys.2023.1114779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/14/2023] [Indexed: 03/17/2023] Open
Abstract
Neuroscientists and Cell Biologists have known for many decades that eukaryotic cells, including neurons, are surrounded by a plasmalemma/axolemma consisting of a phospholipid bilayer that regulates trans-membrane diffusion of ions (including calcium) and other substances. Cells often incur plasmalemmal damage via traumatic injury and various diseases. If the damaged plasmalemma is not rapidly repaired within minutes, activation of apoptotic pathways by calcium influx often results in cell death. We review publications reporting what is less-well known (and not yet covered in neuroscience or cell biology textbooks): that calcium influx at the lesion sites ranging from small nm-sized holes to complete axonal transection activates parallel biochemical pathways that induce vesicles/membrane-bound structures to migrate and interact to restore original barrier properties and eventual reestablishment of the plasmalemma. We assess the reliability of, and problems with, various measures (e.g., membrane voltage, input resistance, current flow, tracer dyes, confocal microscopy, transmission and scanning electron microscopy) used individually and in combination to assess plasmalemmal sealing in various cell types (e.g., invertebrate giant axons, oocytes, hippocampal and other mammalian neurons). We identify controversies such as plug versus patch hypotheses that attempt to account for currently available data on the subcellular mechanisms of plasmalemmal repair/sealing. We describe current research gaps and potential future developments, such as much more extensive correlations of biochemical/biophysical measures with sub-cellular micromorphology. We compare and contrast naturally occurring sealing with recently-discovered artificially-induced plasmalemmal sealing by polyethylene glycol (PEG) that bypasses all natural pathways for membrane repair. We assess other recent developments such as adaptive membrane responses in neighboring cells following injury to an adjacent cell. Finally, we speculate how a better understanding of the mechanisms involved in natural and artificial plasmalemmal sealing is needed to develop better clinical treatments for muscular dystrophies, stroke and other ischemic conditions, and various cancers.
Collapse
Affiliation(s)
- Marshal L. Mencel
- Institute of Cell and Molecular Biology, University of Texas at Austin, Austin, TX, United States
| | - George D. Bittner
- Department of Neuroscience, University of Texas at Austin, Austin, TX, United States
- *Correspondence: George D. Bittner,
| |
Collapse
|
23
|
Chumki SA, van den Goor LM, Hall BN, Miller AL. p115RhoGEF activates RhoA to support tight junction maintenance and remodeling. Mol Biol Cell 2022; 33:ar136. [PMID: 36200892 PMCID: PMC9727809 DOI: 10.1091/mbc.e22-06-0205] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
In vertebrates, epithelial cell-cell junctions must rapidly remodel to maintain barrier function as cells undergo dynamic shape-change events. Consequently, localized leaks sometimes arise within the tight junction (TJ) barrier, which are repaired by short-lived activations of RhoA, called "Rho flares." However, how RhoA is activated at leak sites remains unknown. Here we asked which guanine nucleotide exchange factor (GEF) localizes to TJs to initiate Rho activity at Rho flares. We find that p115RhoGEF locally activates Rho flares at sites of TJ loss. Knockdown of p115RhoGEF leads to diminished Rho flare intensity and impaired TJ remodeling. p115RhoGEF knockdown also decreases junctional active RhoA levels, thus compromising the apical actomyosin array and junctional complex. Furthermore, p115RhoGEF is necessary to promote local leak repair to maintain TJ barrier function. In all, our work demonstrates a central role for p115RhoGEF in activating junctional RhoA to preserve barrier function and direct local TJ remodeling.
Collapse
Affiliation(s)
- Shahana A. Chumki
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI 48109
| | - Lotte M. van den Goor
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Benjamin N. Hall
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Ann L. Miller
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI 48109,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109,*Address correspondence to: Ann L. Miller ()
| |
Collapse
|
24
|
Higashi T, Saito AC, Fukazawa Y, Furuse M, Higashi AY, Ono M, Chiba H. EpCAM proteolysis and release of complexed claudin-7 repair and maintain the tight junction barrier. J Cell Biol 2022; 222:213688. [PMID: 36378161 PMCID: PMC9671161 DOI: 10.1083/jcb.202204079] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/09/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
TJs maintain the epithelial barrier by regulating paracellular permeability. Since TJs are under dynamically fluctuating intercellular tension, cells must continuously survey and repair any damage. However, the underlying mechanisms allowing cells to sense TJ damage and repair the barrier are not yet fully understood. Here, we showed that proteinases play an important role in the maintenance of the epithelial barrier. At TJ break sites, EpCAM-claudin-7 complexes on the basolateral membrane become accessible to apical membrane-anchored serine proteinases (MASPs) and the MASPs cleave EpCAM. Biochemical data and imaging analysis suggest that claudin-7 released from EpCAM contributes to the rapid repair of damaged TJs. Knockout (KO) of MASPs drastically reduced barrier function and live-imaging of TJ permeability showed that MASPs-KO cells exhibited increased size, duration, and frequency of leaks. Together, our results reveal a novel mechanism of TJ maintenance through the localized proteolysis of EpCAM at TJ leaks, and provide a better understanding of the dynamic regulation of epithelial permeability.
Collapse
Affiliation(s)
- Tomohito Higashi
- Department of Basic Pathology, Fukushima Medical University, Fukushima, Japan,Correspondence to Tomohito Higashi:
| | - Akira C. Saito
- Department of Basic Pathology, Fukushima Medical University, Fukushima, Japan
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Faculty of Medical Science, Life Science Innovation Center, University of Fukui, Fukui, Japan
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan,Department of Physiological Sciences, School of Life Science, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Atsuko Y. Higashi
- Department of Basic Pathology, Fukushima Medical University, Fukushima, Japan
| | - Masahiro Ono
- Department of Basic Pathology, Fukushima Medical University, Fukushima, Japan
| | - Hideki Chiba
- Department of Basic Pathology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
25
|
Ashraf APK, Gerke V. The resealing factor S100A11 interacts with annexins and extended synaptotagmin-1 in the course of plasma membrane wound repair. Front Cell Dev Biol 2022; 10:968164. [PMID: 36200035 PMCID: PMC9527316 DOI: 10.3389/fcell.2022.968164] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
After damage, cells repair their plasma membrane in an active process that is driven by Ca2+ entering through the wound. This triggers a range of Ca2+-regulated events such as the translocation of different Ca2+-binding proteins to the wound site which likely function in the repair process. The translocated proteins include Ca2+/phospholipid binding proteins of the annexin (ANX) family and S100A11, an EF hand-type Ca2+-binding protein which can interact with ANX. The molecular mechanism by which S100A11 mediates PM wound repair remains poorly understood although it likely involves interactions with ANX. Here, using S100A11 knockout endothelial cells and expression of S100A11 mutants, we show that endothelial S100A11 is essential for efficient plasma membrane wound repair and engages in Ca2+-dependent interactions with ANXA1 and ANXA2 through its C-terminal extension (residues 93–105). ANXA2 but not ANXA1 translocation to the wound is substantially inhibited in the absence of S100A11; however, the repair defect in S100A11 knockout cells is rescued by ectopic expression of an ANX interaction-defective S100A11 mutant, suggesting an ANX-independent role of S100A11 in membrane wound repair. In search for other interaction partners that could mediate this action of S100A11 we identify extended synaptotagmin 1 (E-Syt1), a protein tether that regulates endoplasmic reticulum-plasma membrane contact sites. E-Syt1 binds to S100A11 in the presence of Ca2+ and depletion of E-Syt1 interferes with wound site recruitment of S100A11 and proper membrane resealing. Thus, the role of S100A11 in membrane wound repair does not exclusively dependent on ANX interactions and a Ca2+-regulated S100A11-E-Syt1 complex acts as a yet unrecognized component of the membrane resealing machinery.
Collapse
|
26
|
Hui J, Stjepić V, Nakamura M, Parkhurst SM. Wrangling Actin Assemblies: Actin Ring Dynamics during Cell Wound Repair. Cells 2022; 11:2777. [PMID: 36139352 PMCID: PMC9497110 DOI: 10.3390/cells11182777] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 12/18/2022] Open
Abstract
To cope with continuous physiological and environmental stresses, cells of all sizes require an effective wound repair process to seal breaches to their cortex. Once a wound is recognized, the cell must rapidly plug the injury site, reorganize the cytoskeleton and the membrane to pull the wound closed, and finally remodel the cortex to return to homeostasis. Complementary studies using various model organisms have demonstrated the importance and complexity behind the formation and translocation of an actin ring at the wound periphery during the repair process. Proteins such as actin nucleators, actin bundling factors, actin-plasma membrane anchors, and disassembly factors are needed to regulate actin ring dynamics spatially and temporally. Notably, Rho family GTPases have been implicated throughout the repair process, whereas other proteins are required during specific phases. Interestingly, although different models share a similar set of recruited proteins, the way in which they use them to pull the wound closed can differ. Here, we describe what is currently known about the formation, translocation, and remodeling of the actin ring during the cell wound repair process in model organisms, as well as the overall impact of cell wound repair on daily events and its importance to our understanding of certain diseases and the development of therapeutic delivery modalities.
Collapse
Affiliation(s)
| | | | | | - Susan M. Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| |
Collapse
|
27
|
Demonbreun AR, Bogdanovic E, Vaught LA, Reiser NL, Fallon KS, Long AM, Oosterbaan CC, Hadhazy M, Page PG, Joseph PRB, Cowen G, Telenson AM, Khatri A, Sadleir KR, Vassar R, McNally EM. A conserved annexin A6-mediated membrane repair mechanism in muscle, heart, and nerve. JCI Insight 2022; 7:158107. [PMID: 35866481 PMCID: PMC9431694 DOI: 10.1172/jci.insight.158107] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/08/2022] [Indexed: 11/21/2022] Open
Abstract
Membrane instability and disruption underlie myriad acute and chronic disorders. Anxa6 encodes the membrane-associated protein annexin A6 and was identified as a genetic modifier of muscle repair and muscular dystrophy. To evaluate annexin A6’s role in membrane repair in vivo, we inserted sequences encoding green fluorescent protein (GFP) into the last coding exon of Anxa6. Heterozygous Anxa6gfp mice expressed a normal pattern of annexin A6 with reduced annexin A6GFP mRNA and protein. High-resolution imaging of wounded muscle fibers showed annexin A6GFP rapidly formed a repair cap at the site of injury. Injured cardiomyocytes and neurons also displayed repair caps after wounding, highlighting annexin A6–mediated repair caps as a feature in multiple cell types. Using surface plasmon resonance, we showed recombinant annexin A6 bound phosphatidylserine-containing lipids in a Ca2+- and dose-dependent fashion with appreciable binding at approximately 50 μM Ca2+. Exogenously added recombinant annexin A6 localized to repair caps and improved muscle membrane repair capacity in a dose-dependent fashion without disrupting endogenous annexin A6 localization, indicating annexin A6 promotes repair from both intracellular and extracellular compartments. Thus, annexin A6 orchestrates repair in multiple cell types, and recombinant annexin A6 may be useful in additional chronic disorders beyond skeletal muscle myopathies.
Collapse
Affiliation(s)
| | - Elena Bogdanovic
- Center for Genetic Medicine.,Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Lauren A Vaught
- Center for Genetic Medicine.,Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Nina L Reiser
- Center for Genetic Medicine.,Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Katherine S Fallon
- Center for Genetic Medicine.,Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ashlee M Long
- Center for Genetic Medicine.,Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Claire C Oosterbaan
- Center for Genetic Medicine.,Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Michele Hadhazy
- Center for Genetic Medicine.,Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | | | - Gabrielle Cowen
- Center for Genetic Medicine.,Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Ammaarah Khatri
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Katherine R Sadleir
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Robert Vassar
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine.,Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
28
|
Mo G, Li R, Swider Z, Leblanc J, Bement WM, Liu XJ. A localized calcium transient and polar body abscission. Cell Cycle 2022; 21:2239-2254. [PMID: 35775922 DOI: 10.1080/15384101.2022.2092815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Polar body emission is a special form of cytokinesis in oocyte meiosis that ensures the correct number of chromosomes in reproduction-competent eggs. The molecular mechanism of the last step, polar body abscission, is poorly understood. While it has been proposed that Ca2+ signaling plays important roles in embryonic cytokinesis, to date transient increases in intracellular free Ca2+ have been difficult to document in oocyte meiosis except for the global Ca2+ wave induced by sperm at fertilization. Here, we find that microinjection of the calcium chelator dibromo-BAPTA inhibits polar body abscission in Xenopus laevis oocytes. Using a novel, microtubule-targeted ratio-metric calcium sensor, we detected a calcium transient that is focused at the contractile ring-associated plasma membrane and which occurred after anaphase and constriction of the contractile ring but prior to abscission. This calcium transient was confirmed by mobile calcium probes. Further, the Ca2+-sensitive protein kinase Cβ C2 domain transiently translocated to the contractile ring-associated membrane simultaneously with the calcium transient. Collectively, these results demonstrate that a calcium transient, apparently originating at the contractile ring-associated plasma membrane, promotes polar body abscission.
Collapse
Affiliation(s)
- Guolong Mo
- Ottawa Hospital Research Institute, Ottawa Hospital General Campus, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Ruizhen Li
- Ottawa Hospital Research Institute, Ottawa Hospital General Campus, Ottawa, Ontario, Canada
| | - Zachary Swider
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA.,Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA
| | - Julie Leblanc
- Ottawa Hospital Research Institute, Ottawa Hospital General Campus, Ottawa, Ontario, Canada
| | - William M Bement
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA.,Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA.,Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - X Johné Liu
- Ottawa Hospital Research Institute, Ottawa Hospital General Campus, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Department of Obstetrics and Gynaecology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
29
|
Assembly of Tetraspanin-enriched macrodomains contains membrane damage to facilitate repair. Nat Cell Biol 2022; 24:825-832. [PMID: 35654840 DOI: 10.1038/s41556-022-00920-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/22/2022] [Indexed: 11/08/2022]
Abstract
Various mechanisms contribute to membrane repair1-8 but the machinery that mediates the repair of large wounds on the plasma membrane is less clear. We found that shortly after membrane damage, Tetraspanin-enriched macrodomains are assembled around the damage site. Tetraspanin-enriched macrodomains are in the liquid-ordered phase and form a rigid ring around the damaged site. This restricts the spread of the damage and prevents membrane disintegration, thus facilitating membrane repair by other mechanisms. Functionally, Tetraspanin 4 helps cells mitigate damage caused by laser, detergent, pyroptosis and natural killer cells. We propose that assembly of Tetraspanin-enriched macrodomains creates a physical barrier to contain membrane damage.
Collapse
|
30
|
Kozlov AV, Grillari J. Pathogenesis of Multiple Organ Failure: The Impact of Systemic Damage to Plasma Membranes. Front Med (Lausanne) 2022; 9:806462. [PMID: 35372390 PMCID: PMC8964500 DOI: 10.3389/fmed.2022.806462] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/09/2022] [Indexed: 11/19/2022] Open
Abstract
Multiple organ failure (MOF) is the major cause of morbidity and mortality in intensive care patients, but the mechanisms causing this severe syndrome are still poorly understood. Inflammatory response, tissue hypoxia, immune and cellular metabolic dysregulations, and endothelial and microvascular dysfunction are the main features of MOF, but the exact mechanisms leading to MOF are still unclear. Recent progress in the membrane research suggests that cellular plasma membranes play an important role in key functions of diverse organs. Exploration of mechanisms contributing to plasma membrane damage and repair suggest that these processes can be the missing link in the development of MOF. Elevated levels of extracellular phospholipases, reactive oxygen and nitrogen species, pore-forming proteins (PFPs), and dysregulation of osmotic homeostasis occurring upon systemic inflammatory response are the major extracellular inducers of plasma membrane damage, which may simultaneously operate in different organs causing their profound dysfunction. Hypoxia activates similar processes, but they predominantly occur within the cells targeting intracellular membrane compartments and ultimately causing cell death. To combat the plasma membrane damage cells have developed several repair mechanisms, such as exocytosis, shedding, and protein-driven membrane remodeling. Analysis of knowledge on these mechanisms reveals that systemic damage to plasma membranes may be associated with potentially reversible MOF, which can be quickly recovered, if pathological stimuli are eliminated. Alternatively, it can be transformed in a non-resolving phase, if repair mechanisms are not sufficient to deal with a large damage or if the damage is extended to intracellular compartments essential for vital cellular functions.
Collapse
Affiliation(s)
- Andrey V Kozlov
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation With AUVA, LBG, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Medical University of Vienna, Vienna, Austria.,Laboratory of Navigational Redox Lipidomics and Department of Human Pathology, IM Sechenov Moscow State Medical University, Vienna, Austria
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation With AUVA, LBG, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Medical University of Vienna, Vienna, Austria.,Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
31
|
Varadarajan S, Chumki SA, Stephenson RE, Misterovich ER, Wu JL, Dudley CE, Erofeev IS, Goryachev AB, Miller AL. Mechanosensitive calcium flashes promote sustained RhoA activation during tight junction remodeling. J Cell Biol 2022; 221:213049. [PMID: 35254388 PMCID: PMC8906493 DOI: 10.1083/jcb.202105107] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 12/03/2021] [Accepted: 02/02/2022] [Indexed: 11/22/2022] Open
Abstract
Epithelial cell–cell junctions remodel in response to mechanical stimuli to maintain barrier function. Previously, we found that local leaks in tight junctions (TJs) are rapidly repaired by local, transient RhoA activation, termed “Rho flares,” but how Rho flares are regulated is unknown. Here, we discovered that intracellular calcium flashes and junction elongation are early events in the Rho flare pathway. Both laser-induced and naturally occurring TJ breaks lead to local calcium flashes at the site of leaks. Additionally, junction elongation induced by optogenetics increases Rho flare frequency, suggesting that Rho flares are mechanically triggered. Depletion of intracellular calcium or inhibition of mechanosensitive calcium channels (MSCs) reduces the amplitude of calcium flashes and diminishes the sustained activation of Rho flares. MSC-dependent calcium influx is necessary to maintain global barrier function by regulating reinforcement of local TJ proteins via junction contraction. In all, we uncovered a novel role for MSC-dependent calcium flashes in TJ remodeling, allowing epithelial cells to repair local leaks induced by mechanical stimuli.
Collapse
Affiliation(s)
| | - Shahana A Chumki
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI
| | - Rachel E Stephenson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI
| | - Eileen R Misterovich
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI
| | - Jessica L Wu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI
| | - Claire E Dudley
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI
| | - Ivan S Erofeev
- Centre for Synthetic and Systems Biology, University of Edinburgh, Edinburgh, Scotland
| | - Andrew B Goryachev
- Centre for Synthetic and Systems Biology, University of Edinburgh, Edinburgh, Scotland
| | - Ann L Miller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI.,Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI
| |
Collapse
|
32
|
Koh SP, Pham NP, Piekny A. Seeing is believing: tools to study the role of Rho GTPases during cytokinesis. Small GTPases 2022; 13:211-224. [PMID: 34405757 PMCID: PMC9707540 DOI: 10.1080/21541248.2021.1957384] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cytokinesis is required to cleave the daughter cells at the end of mitosis and relies on the spatiotemporal control of RhoA GTPase. Cytokinesis failure can lead to changes in cell fate or aneuploidy, which can be detrimental during development and/or can lead to cancer. However, our knowledge of the pathways that regulate RhoA during cytokinesis is limited, and the role of other Rho family GTPases is not clear. This is largely because the study of Rho GTPases presents unique challenges using traditional cell biological and biochemical methods, and they have pleiotropic functions making genetic studies difficult to interpret. The recent generation of optogenetic tools and biosensors that control and detect active Rho has overcome some of these challenges and is helping to elucidate the role of RhoA in cytokinesis. However, improvements are needed to reveal the role of other Rho GTPases in cytokinesis, and to identify the molecular mechanisms that control Rho activity. This review examines some of the outstanding questions in cytokinesis, and explores tools for the imaging and control of Rho GTPases.
Collapse
Affiliation(s)
- Su Pin Koh
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Nhat Phi Pham
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Alisa Piekny
- Department of Biology, Concordia University, Montreal, QC, Canada,CONTACT Alisa Piekny Department of Biology, Concordia University, 7141 Sherbrooke St. W, Montreal, QC, Canada
| |
Collapse
|
33
|
Lee D, Chan SSY, Aksic N, Bajalovic N, Loke DK. Ultralong-Time Recovery and Low-Voltage Electroporation for Biological Cell Monitoring Enabled by a Microsized Multipulse Framework. ACS OMEGA 2021; 6:35325-35333. [PMID: 34984264 PMCID: PMC8717367 DOI: 10.1021/acsomega.1c04257] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/20/2021] [Indexed: 05/05/2023]
Abstract
Long-term nondestructive monitoring of cells is of significant importance for understanding cell proliferation, cell signaling, cell death, and other processes. However, traditional monitoring methods are limited to a certain range of testing conditions and may reduce cell viability. Here, we present a microgap, multishot electroporation (M2E) system for monitoring cell recovery for up to ∼2 h using ∼5 V pulses and with excellent cell viability using a medium cell population. Electric field simulations reveal the bias-voltage- and gap-size-dependent electric field intensities in the M2E system. In addition to excellent transparency with low cell toxicity, the M2E system does not require specialized components, expensive materials, complicated fabrication processes, or cell manipulations; it just consists of a micrometer-sized pattern and a low-voltage square-wave generator. Ultimately, the M2E system can offer a long-term and nontoxic method of cell monitoring.
Collapse
Affiliation(s)
- Denise Lee
- Department
of Science, Mathematics and Technology, Singapore University of Technology and Design, Singapore 487372, Singapore
| | - Sophia S. Y. Chan
- Department
of Science, Mathematics and Technology, Singapore University of Technology and Design, Singapore 487372, Singapore
| | - Nemanja Aksic
- Department
of Science, Mathematics and Technology, Singapore University of Technology and Design, Singapore 487372, Singapore
| | - Natasa Bajalovic
- Department
of Science, Mathematics and Technology, Singapore University of Technology and Design, Singapore 487372, Singapore
| | - Desmond K. Loke
- Department
of Science, Mathematics and Technology, Singapore University of Technology and Design, Singapore 487372, Singapore
- Office
of Innovation, Changi General Hospital, Singapore 529889, Singapore
| |
Collapse
|
34
|
Therapeutic Benefit of Galectin-1: Beyond Membrane Repair, a Multifaceted Approach to LGMD2B. Cells 2021; 10:cells10113210. [PMID: 34831431 PMCID: PMC8621416 DOI: 10.3390/cells10113210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 11/21/2022] Open
Abstract
Two of the main pathologies characterizing dysferlinopathies are disrupted muscle membrane repair and chronic inflammation, which lead to symptoms of muscle weakness and wasting. Here, we used recombinant human Galectin-1 (rHsGal-1) as a therapeutic for LGMD2B mouse and human models. Various redox and multimerization states of Gal-1 show that rHsGal-1 is the most effective form in both increasing muscle repair and decreasing inflammation, due to its monomer-dimer equilibrium. Dose-response testing shows an effective 25-fold safety profile between 0.54 and 13.5 mg/kg rHsGal-1 in Bla/J mice. Mice treated weekly with rHsGal-1 showed downregulation of canonical NF-κB inflammation markers, decreased muscle fat deposition, upregulated anti-inflammatory cytokines, increased membrane repair, and increased functional movement compared to non-treated mice. Gal-1 treatment also resulted in a positive self-upregulation loop of increased endogenous Gal-1 expression independent of NF-κB activation. A similar reduction in disease pathologies in patient-derived human cells demonstrates the therapeutic potential of Gal-1 in LGMD2B patients.
Collapse
|
35
|
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
|
36
|
O’Connor J, Akbar FB, Hutson MS, Page-McCaw A. Zones of cellular damage around pulsed-laser wounds. PLoS One 2021; 16:e0253032. [PMID: 34570791 PMCID: PMC8476025 DOI: 10.1371/journal.pone.0253032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/07/2021] [Indexed: 11/19/2022] Open
Abstract
After a tissue is wounded, cells surrounding the wound adopt distinct wound-healing behaviors to repair the tissue. Considerable effort has been spent on understanding the signaling pathways that regulate immune and tissue-resident cells as they respond to wounds, but these signals must ultimately originate from the physical damage inflicted by the wound. Tissue wounds comprise several types of cellular damage, and recent work indicates that different types of cellular damage initiate different types of signaling. Hence to understand wound signaling, it is important to identify and localize the types of wound-induced cellular damage. Laser ablation is widely used by researchers to create reproducible, aseptic wounds in a tissue that can be live-imaged. Because laser wounding involves a combination of photochemical, photothermal and photomechanical mechanisms, each with distinct spatial dependencies, cells around a pulsed-laser wound will experience a gradient of damage. Here we exploit this gradient to create a map of wound-induced cellular damage. Using genetically-encoded fluorescent proteins, we monitor damaged cellular and sub-cellular components of epithelial cells in living Drosophila pupae in the seconds to minutes following wounding. We hypothesized that the regions of damage would be predictably arrayed around wounds of varying sizes, and subsequent analysis found that all damage radii are linearly related over a 3-fold range of wound size. Thus, around laser wounds, the distinct regions of damage can be estimated after measuring any one. This report identifies several different types of cellular damage within a wounded epithelial tissue in a living animal. By quantitatively mapping the size and placement of these different types of damage, we set the foundation for tracing wound-induced signaling back to the damage that initiates it.
Collapse
Affiliation(s)
- James O’Connor
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Program in Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Fabiha Bushra Akbar
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - M. Shane Hutson
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Program in Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
37
|
Kato T, Ghadban L, Boucher E, Mandato CA. Tension modulation of actomyosin ring assembly and RhoGTPases activity: Perspectives from the Xenopus oocyte wound healing model. Cytoskeleton (Hoboken) 2021; 78:349-360. [PMID: 34541818 DOI: 10.1002/cm.21688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/20/2021] [Accepted: 08/26/2021] [Indexed: 11/10/2022]
Abstract
Cells are remarkably resilient structures; they are able to recover from injuries to their plasma membrane (PM) and cytoskeleton that would normally constitute existential threats. This capacity is exemplified by Xenopus laevis oocytes which can recover from very large PM defects through exocytotic and endocytic events and can repair damaged cortical cytoskeleton structures through the formation of a contractile actomyosin ring (AMR). Formation of the AMR involves the localized Ca2+ -dependent activation of RhoA and Cdc42, and the pre-patterning of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). However, this model fails to account for observations that suggest a link between cytoskeletal dynamics, intracellular tension, and AMR formation. It also does not explain why the formation of an AMR is not involved in the cytoskeletal repair program of adherent cells. We show here evidence for the support of tension as an essential regulatory signal for the formation of AMR. Indeed, oocytes in which global tension has been experimentally reduced were unable to form a functional AMR following injury, showing severely diminished RhoA activity at the wound site. These new insights place the cytoskeleton at the center of events involving changes in cell shape such as cytokinesis which also involves the formation and closure of an AMR.
Collapse
Affiliation(s)
- Tatsuya Kato
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Logine Ghadban
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Eric Boucher
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Craig A Mandato
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
38
|
Dias C, Nita E, Faktor J, Tynan AC, Hernychova L, Vojtesek B, Nylandsted J, Hupp TR, Kunath T, Ball KL. CHIP-dependent regulation of the actin cytoskeleton is linked to neuronal cell membrane integrity. iScience 2021; 24:102878. [PMID: 34401662 PMCID: PMC8350547 DOI: 10.1016/j.isci.2021.102878] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/13/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
CHIP is an E3-ubiquitin ligase that contributes to healthy aging and has been characterized as neuroprotective. To elucidate dominant CHIP-dependent changes in protein steady-state levels in a patient-derived human neuronal model, CHIP function was ablated using gene-editing and an unbiased proteomic analysis conducted to compare knock-out and wild-type isogenic induced pluripotent stem cell (iPSC)-derived cortical neurons. Rather than a broad effect on protein homeostasis, loss of CHIP function impacted on a focused cohort of proteins from actin cytoskeleton signaling and membrane integrity networks. In support of the proteomics, CHIP knockout cells had enhanced sensitivity to induced membrane damage. We conclude that the major readout of CHIP function in cortical neurons derived from iPSC of a patient with elevate α-synuclein, Parkinson's disease and dementia, is the modulation of substrates involved in maintaining cellular "health". Thus, regulation of the actin cytoskeletal and membrane integrity likely contributes to the neuroprotective function(s) of CHIP.
Collapse
Affiliation(s)
- Catarina Dias
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Erisa Nita
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Jakub Faktor
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
- University of Gdansk, International Centre for Cancer Vaccine Science, 80-822 Gdansk, Poland
| | - Ailish C. Tynan
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Lenka Hernychova
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Borivoj Vojtesek
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Jesper Nylandsted
- Membrane Integrity Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Ted R. Hupp
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
- University of Gdansk, International Centre for Cancer Vaccine Science, 80-822 Gdansk, Poland
| | - Tilo Kunath
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Kathryn L. Ball
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| |
Collapse
|
39
|
Mahlandt EK, Arts JJG, van der Meer WJ, van der Linden FH, Tol S, van Buul JD, Gadella TWJ, Goedhart J. Visualizing endogenous Rho activity with an improved localization-based, genetically encoded biosensor. J Cell Sci 2021; 134:272101. [PMID: 34357388 PMCID: PMC8445605 DOI: 10.1242/jcs.258823] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/26/2021] [Indexed: 12/05/2022] Open
Abstract
Rho GTPases are regulatory proteins, which orchestrate cell features such as morphology, polarity and movement. Therefore, probing Rho GTPase activity is key to understanding processes such as development and cell migration. Localization-based reporters for active Rho GTPases are attractive probes to study Rho GTPase-mediated processes in real time with subcellular resolution in living cells and tissue. Until now, relocation Rho biosensors (sensors that relocalize to the native location of active Rho GTPase) seem to have been only useful in certain organisms and have not been characterized well. In this paper, we systematically examined the contribution of the fluorescent protein and Rho-binding peptides on the performance of localization-based sensors. To test the performance, we compared relocation efficiency and specificity in cell-based assays. We identified several improved localization-based, genetically encoded fluorescent biosensors for detecting endogenous Rho activity. This enables a broader application of Rho relocation biosensors, which was demonstrated by using the improved biosensor to visualize Rho activity during several cellular processes, such as cell division, migration and G protein-coupled receptor signaling. Owing to the improved avidity of the new biosensors for Rho activity, cellular processes regulated by Rho can be better understood. This article has an associated First Person interview with the first author of the paper. Summary: The dT-2xrGBD location-based Rho biosensor relocalizes more efficiently than other sensors of this type, and this sensor enables the observation of endogenous Rho activity in cultured cells.
Collapse
Affiliation(s)
- Eike K Mahlandt
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Janine J G Arts
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands.,Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Werner J van der Meer
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Franka H van der Linden
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Simon Tol
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Jaap D van Buul
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands.,Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Theodorus W J Gadella
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Joachim Goedhart
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Sommer A, Hoeftberger M, Foissner I. Fluid-phase and membrane markers reveal spatio-temporal dynamics of membrane traffic and repair in the green alga Chara australis. PROTOPLASMA 2021; 258:711-728. [PMID: 33704568 PMCID: PMC8211606 DOI: 10.1007/s00709-021-01627-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/05/2020] [Indexed: 06/12/2023]
Abstract
We investigated the mechanisms and the spatio-temporal dynamics of fluid-phase and membrane internalization in the green alga Chara australis using fluorescent hydrazides markers alone, or in conjunction with styryl dyes. Using live-cell imaging, immunofluorescence and inhibitor studies we revealed that both fluid-phase and membrane dyes were actively taken up into the cytoplasm by clathrin-mediated endocytosis and stained various classes of endosomes including brefeldin A- and wortmannin-sensitive organelles (trans-Golgi network and multivesicular bodies). Uptake of fluorescent hydrazides was poorly sensitive to cytochalasin D, suggesting that actin plays a minor role in constitutive endocytosis in Chara internodal cells. Sequential pulse-labelling experiments revealed novel aspects of the temporal progression of endosomes in Chara internodal cells. The internalized fluid-phase marker distributed to early compartments within 10 min from dye exposure and after about 30 min, it was found almost exclusively in late endocytic compartments. Notably, fluid cargo consecutively internalized at time intervals of more than 1h, was not targeted to the same vesicular structures, but was sorted into distinct late compartments. We further found that fluorescent hydrazide dyes distributed not only to rapidly recycling endosomes but also to long-lived compartments that participated in plasma membrane repair after local laser injury. Our approach highlights the benefits of combining different fluid-phase markers in conjunction with membrane dyes in simultaneous and sequential application modus for investigating vesicle traffic, especially in organisms, which are still refractory to genetic transformation like characean algae.
Collapse
Affiliation(s)
- Aniela Sommer
- Department of Biosciences, University of Salzburg, Hellbrunnerstr. 34, 5020, Salzburg, Austria.
| | - Margit Hoeftberger
- Department of Biosciences, University of Salzburg, Hellbrunnerstr. 34, 5020, Salzburg, Austria
| | - Ilse Foissner
- Department of Biosciences, University of Salzburg, Hellbrunnerstr. 34, 5020, Salzburg, Austria.
| |
Collapse
|
41
|
Moe A, Holmes W, Golding AE, Zola J, Swider ZT, Edelstein-Keshet L, Bement W. Cross-talk-dependent cortical patterning of Rho GTPases during cell repair. Mol Biol Cell 2021; 32:1417-1432. [PMID: 34133216 PMCID: PMC8351735 DOI: 10.1091/mbc.e20-07-0481] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Rho GTPases such as Rho, Rac, and Cdc42 are important regulators of the cortical cytoskeleton in processes including cell division, locomotion, and repair. In these processes, Rho GTPases assume characteristic patterns wherein the active GTPases occupy mutually exclusive "zones" in the cell cortex. During cell wound repair, for example, a Rho zone encircles the wound edge and is in turn encircled by a Cdc42 zone. Here we evaluated the contributions of cross-talk between Rho and Cdc42 to the patterning of their respective zones in wounded Xenopus oocytes using experimental manipulations in combination with mathematical modeling. The results show that the position of the Cdc42 zone relative to the Rho zone and relative to the wound edge is controlled by the level of Rho activity. In contrast, the outer boundary of the Rho zone is limited by the level of Cdc42 activity. Models based on positive feedback within zones and negative feedback from Rho to the GEF-GAP Abr to Cdc42 capture some, but not all, of the observed behaviors. We conclude that GTPase zone positioning is controlled at the level of Rho activity and we speculate that the Cdc42 zone or something associated with it limits the spread of Rho activity.
Collapse
Affiliation(s)
- Alison Moe
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706.,Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706
| | - William Holmes
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN 37212
| | - Adriana E Golding
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706.,Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706
| | - Jessica Zola
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706
| | - Zachary T Swider
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706.,Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706
| | - Leah Edelstein-Keshet
- Department of Mathematics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - William Bement
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706.,Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706.,Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
42
|
Ammendolia DA, Bement WM, Brumell JH. Plasma membrane integrity: implications for health and disease. BMC Biol 2021; 19:71. [PMID: 33849525 PMCID: PMC8042475 DOI: 10.1186/s12915-021-00972-y] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Plasma membrane integrity is essential for cellular homeostasis. In vivo, cells experience plasma membrane damage from a multitude of stressors in the extra- and intra-cellular environment. To avoid lethal consequences, cells are equipped with repair pathways to restore membrane integrity. Here, we assess plasma membrane damage and repair from a whole-body perspective. We highlight the role of tissue-specific stressors in health and disease and examine membrane repair pathways across diverse cell types. Furthermore, we outline the impact of genetic and environmental factors on plasma membrane integrity and how these contribute to disease pathogenesis in different tissues.
Collapse
Affiliation(s)
- Dustin A Ammendolia
- Cell Biology Program, Hospital for Sick Children, 686 Bay Street PGCRL, Toronto, ON, M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - William M Bement
- Center for Quantitative Cell Imaging and Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - John H Brumell
- Cell Biology Program, Hospital for Sick Children, 686 Bay Street PGCRL, Toronto, ON, M5G 0A4, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A1, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A1, Canada. .,SickKids IBD Centre, Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.
| |
Collapse
|
43
|
Plasma membrane integrity in health and disease: significance and therapeutic potential. Cell Discov 2021; 7:4. [PMID: 33462191 PMCID: PMC7813858 DOI: 10.1038/s41421-020-00233-2] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022] Open
Abstract
Maintenance of plasma membrane integrity is essential for normal cell viability and function. Thus, robust membrane repair mechanisms have evolved to counteract the eminent threat of a torn plasma membrane. Different repair mechanisms and the bio-physical parameters required for efficient repair are now emerging from different research groups. However, less is known about when these mechanisms come into play. This review focuses on the existence of membrane disruptions and repair mechanisms in both physiological and pathological conditions, and across multiple cell types, albeit to different degrees. Fundamentally, irrespective of the source of membrane disruption, aberrant calcium influx is the common stimulus that activates the membrane repair response. Inadequate repair responses can tip the balance between physiology and pathology, highlighting the significance of plasma membrane integrity. For example, an over-activated repair response can promote cancer invasion, while the inability to efficiently repair membrane can drive neurodegeneration and muscular dystrophies. The interdisciplinary view explored here emphasises the widespread potential of targeting plasma membrane repair mechanisms for therapeutic purposes.
Collapse
|
44
|
Carim SC, Kechad A, Hickson GRX. Animal Cell Cytokinesis: The Rho-Dependent Actomyosin-Anilloseptin Contractile Ring as a Membrane Microdomain Gathering, Compressing, and Sorting Machine. Front Cell Dev Biol 2020; 8:575226. [PMID: 33117802 PMCID: PMC7575755 DOI: 10.3389/fcell.2020.575226] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022] Open
Abstract
Cytokinesis is the last step of cell division that partitions the cellular organelles and cytoplasm of one cell into two. In animal cells, cytokinesis requires Rho-GTPase-dependent assembly of F-actin and myosin II (actomyosin) to form an equatorial contractile ring (CR) that bisects the cell. Despite 50 years of research, the precise mechanisms of CR assembly, tension generation and closure remain elusive. This hypothesis article considers a holistic view of the CR that, in addition to actomyosin, includes another Rho-dependent cytoskeletal sub-network containing the scaffold protein, Anillin, and septin filaments (collectively termed anillo-septin). We synthesize evidence from our prior work in Drosophila S2 cells that actomyosin and anillo-septin form separable networks that are independently anchored to the plasma membrane. This latter realization leads to a simple conceptual model in which CR assembly and closure depend upon the micro-management of the membrane microdomains to which actomyosin and anillo-septin sub-networks are attached. During CR assembly, actomyosin contractility gathers and compresses its underlying membrane microdomain attachment sites. These microdomains resist this compression, which builds tension. During CR closure, membrane microdomains are transferred from the actomyosin sub-network to the anillo-septin sub-network, with which they flow out of the CR as it advances. This relative outflow of membrane microdomains regulates tension, reduces the circumference of the CR and promotes actomyosin disassembly all at the same time. According to this hypothesis, the metazoan CR can be viewed as a membrane microdomain gathering, compressing and sorting machine that intrinsically buffers its own tension through coordination of actomyosin contractility and anillo-septin-membrane relative outflow, all controlled by Rho. Central to this model is the abandonment of the dogmatic view that the plasma membrane is always readily deformable by the underlying cytoskeleton. Rather, the membrane resists compression to build tension. The notion that the CR might generate tension through resistance to compression of its own membrane microdomain attachment sites, can account for numerous otherwise puzzling observations and warrants further investigation using multiple systems and methods.
Collapse
Affiliation(s)
- Sabrya C. Carim
- CHU Sainte-Justine Research Center, Université de Montréal, Montréal, QC, Canada
| | - Amel Kechad
- CHU Sainte-Justine Research Center, Université de Montréal, Montréal, QC, Canada
| | - Gilles R. X. Hickson
- CHU Sainte-Justine Research Center, Université de Montréal, Montréal, QC, Canada
- Département de Pathologie et Biologie Cellulaire, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
45
|
de Oliveira S, Houseright RA, Korte BG, Huttenlocher A. DnaJ-PKAc fusion induces liver inflammation in a zebrafish model of fibrolamellar carcinoma. Dis Model Mech 2020; 13:dmm042564. [PMID: 32102783 PMCID: PMC7197716 DOI: 10.1242/dmm.042564] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
Fibrolamellar carcinoma (FLC) is a rare liver cancer that affects adolescents and young adults. Genomic analysis of FLC has revealed a 400 kb deletion in chromosome 19 that leads to the chimeric transcript DNAJB1-PRKACA (DnaJ-PKAc), comprised of the first exon of heat shock protein 40 (DNAJB1) and exons 2-10 of the catalytic subunit of protein kinase A (PRKACA). Here, we report a new zebrafish model of FLC induced by ectopic expression of zebrafish Dnaja-Pkaca (zfDnaJa-Pkaca) in hepatocytes that is amenable to live imaging of early innate immune inflammation. Expression of zfDnaJa-Pkaca in hepatocytes induces hepatomegaly and increased hepatocyte size. In addition, FLC larvae exhibit early innate immune inflammation characterized by early infiltration of neutrophils and macrophages into the liver microenvironment. Increased Caspase-a (the zebrafish homolog for human caspase-1) activity was also found in the liver of FLC larvae, and pharmacological inhibition of Tnfα and caspase-a decreased liver size and inflammation. Overall, these findings show that innate immune inflammation is an early feature in a zebrafish model of FLC and that pharmacological inhibition of TNFα or caspase-1 activity might be targets to treat inflammation and progression in FLC patients.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sofia de Oliveira
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ruth A Houseright
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Benjamin G Korte
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53792, USA
| |
Collapse
|
46
|
Talukder MSU, Pervin MS, Tanvir MIO, Fujimoto K, Tanaka M, Itoh G, Yumura S. Ca 2+-Calmodulin Dependent Wound Repair in Dictyostelium Cell Membrane. Cells 2020; 9:cells9041058. [PMID: 32340342 PMCID: PMC7226253 DOI: 10.3390/cells9041058] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/18/2020] [Accepted: 04/21/2020] [Indexed: 12/20/2022] Open
Abstract
Wound repair of cell membrane is a vital physiological phenomenon. We examined wound repair in Dictyostelium cells by using a laserporation, which we recently invented. We examined the influx of fluorescent dyes from the external medium and monitored the cytosolic Ca2+ after wounding. The influx of Ca2+ through the wound pore was essential for wound repair. Annexin and ESCRT components accumulated at the wound site upon wounding as previously described in animal cells, but these were not essential for wound repair in Dictyostelium cells. We discovered that calmodulin accumulated at the wound site upon wounding, which was essential for wound repair. The membrane accumulated at the wound site to plug the wound pore by two-steps, depending on Ca2+ influx and calmodulin. From several lines of evidence, the membrane plug was derived from de novo generated vesicles at the wound site. Actin filaments also accumulated at the wound site, depending on Ca2+ influx and calmodulin. Actin accumulation was essential for wound repair, but microtubules were not essential. A molecular mechanism of wound repair will be discussed.
Collapse
Affiliation(s)
- Md. Shahabe Uddin Talukder
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8511, Japan; (M.S.U.T.); (M.S.P.); (M.I.O.T.); (K.F.); (M.T.)
- Institute of Food and Radiation Biology, AERE, Bangladesh Atomic Energy Commission, Savar, Dhaka 3787, Bangladesh
| | - Mst. Shaela Pervin
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8511, Japan; (M.S.U.T.); (M.S.P.); (M.I.O.T.); (K.F.); (M.T.)
- Rajshahi Diabetic Association General Hospital, Luxmipur, Jhautala, Rajshahi 6000, Bangladesh
| | - Md. Istiaq Obaidi Tanvir
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8511, Japan; (M.S.U.T.); (M.S.P.); (M.I.O.T.); (K.F.); (M.T.)
| | - Koushiro Fujimoto
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8511, Japan; (M.S.U.T.); (M.S.P.); (M.I.O.T.); (K.F.); (M.T.)
| | - Masahito Tanaka
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8511, Japan; (M.S.U.T.); (M.S.P.); (M.I.O.T.); (K.F.); (M.T.)
| | - Go Itoh
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Shigehiko Yumura
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8511, Japan; (M.S.U.T.); (M.S.P.); (M.I.O.T.); (K.F.); (M.T.)
- Correspondence: yumura@yamaguchi–u.ac.jp; Tel./Fax: +81-83-933-5717
| |
Collapse
|
47
|
Han PF, Che XD, Li HZ, Gao YY, Wei XC, Li PC. Annexin A1 involved in the regulation of inflammation and cell signaling pathways. Chin J Traumatol 2020; 23:96-101. [PMID: 32201231 PMCID: PMC7156956 DOI: 10.1016/j.cjtee.2020.02.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 11/25/2019] [Accepted: 01/04/2020] [Indexed: 02/08/2023] Open
Abstract
With the deepening of research, proteomics has developed into a science covering the study of all the structural and functional characteristics of proteins and the dynamic change rules. The essence of various biological activities is revealed from the perspectives of the biological structure, functional activity and corresponding regulatory mechanism of proteins by proteomics. Among them, phospholipid-binding protein is one of the hotspots of proteomics, especially annexin A1, which is widely present in various tissues and cells of the body. It has the capability of binding to phospholipid membranes reversibly in a calcium ion dependent manner. In order to provide possible research ideas for researchers, who are interested in this protein, the biological effects of annexin A1, such as inflammatory regulation, cell signal transduction, cell proliferation, differentiation and apoptosis are described in this paper.
Collapse
Affiliation(s)
- Peng-Fei Han
- Department of Orthopaedic Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi 046000, China
| | - Xian-Da Che
- Department of Orthopaedic Surgery, the Second Hospital of Shanxi Medical University, Taiyuan 030009, China
| | - Hong-Zhuo Li
- Department of Orthopaedic Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi 046000, China
| | - Yang-Yang Gao
- Department of Orthopaedic Surgery, the Second Hospital of Shanxi Medical University, Taiyuan 030009, China
| | - Xiao-Chun Wei
- Department of Orthopaedic Surgery, the Second Hospital of Shanxi Medical University, Taiyuan 030009, China
| | - Peng-Cui Li
- Department of Orthopaedic Surgery, the Second Hospital of Shanxi Medical University, Taiyuan 030009, China,Corresponding author.
| |
Collapse
|
48
|
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
|
49
|
Hagihara T, Kondo J, Endo H, Ohue M, Sakai Y, Inoue M. Hydrodynamic stress stimulates growth of cell clusters via the ANXA1/PI3K/AKT axis in colorectal cancer. Sci Rep 2019; 9:20027. [PMID: 31882967 PMCID: PMC6934682 DOI: 10.1038/s41598-019-56739-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/11/2019] [Indexed: 01/06/2023] Open
Abstract
Cancer cells are exposed to various stresses in vivo, including hydrodynamic stress (HDS). HDS on cancer cells in the blood stream can influence the metastatic potential. Recent studies revealed that circulating tumor cell clusters are more responsible for metastasis than circulating single cells. Nevertheless, most studies on HDS are based on single cells prepared from established cancer cell lines. Here, we used cancer tissue-originated spheroids (CTOS) as a patient-derived, 3D organoid model to investigate the effect of HDS on cancer cell clusters. We found that HDS induced the growth of cancer cell clusters in a population of colorectal CTOSs. Microarray analyses revealed that the multifunctional protein, Annexin 1 (ANXA1), was upregulated upon HDS exposure. Chemically-induced membrane damage also triggered the expression of ANXA1. A knockdown of ANXA1 revealed that ANXA1 regulated HDS-stimulated growth in colorectal CTOSs. Mechanistically, activating the PI3K/AKT pathway downstream of ANXA1 contributed to the phenotype. These findings demonstrate that HDS induces the growth of cancer cell clusters via ANXA1/PI3K/AKT axis, which helps to elucidate the pro-metastatic feature of circulating cancer cell clusters.
Collapse
Affiliation(s)
- Takeshi Hagihara
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Yoshida-honmachi, Sakyo-ku, Kyoto, 606-8501, Japan.,Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, Yoshida-honmachi, Sakyo-ku, Kyoto, 606-8501, Japan.,Department of Biochemistry, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka, 541-8567, Japan
| | - Jumpei Kondo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Yoshida-honmachi, Sakyo-ku, Kyoto, 606-8501, Japan. .,Department of Biochemistry, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka, 541-8567, Japan.
| | - Hiroko Endo
- Department of Biochemistry, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka, 541-8567, Japan
| | - Masayuki Ohue
- Department of Biochemistry, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka, 541-8567, Japan
| | - Yoshiharu Sakai
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, Yoshida-honmachi, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Yoshida-honmachi, Sakyo-ku, Kyoto, 606-8501, Japan.,Department of Biochemistry, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka, 541-8567, Japan
| |
Collapse
|
50
|
Abstract
Background The study of the mechanisms controlling wound healing is an attractive area within the field of biology, with it having a potentially significant impact on the health sector given the current medical burden associated with healing in the elderly population. Healing is a complex process and includes many steps that are regulated by coding and noncoding RNAs, proteins and other molecules. Nitric oxide (NO) is one of these small molecule regulators and its function has already been associated with inflammation and angiogenesis during adult healing. Results Our results showed that NO is also an essential component during embryonic scarless healing and acts via a previously unknown mechanism. NO is mainly produced during the early phase of healing and it is crucial for the expression of genes associated with healing. However, we also observed a late phase of healing, which occurs for several hours after wound closure and takes place under the epidermis and includes tissue remodelling that is dependent on NO. We also found that the NO is associated with multiple cellular metabolic pathways, in particularly the glucose metabolism pathway. This is particular noteworthy as the use of NO donors have already been found to be beneficial for the treatment of chronic healing defects (including those associated with diabetes) and it is possible that its mechanism of action follows those observed during embryonic wound healing. Conclusions Our study describes a new role of NO during healing, which may potentially translate to improved therapeutic treatments, especially for individual suffering with problematic healing.
Collapse
|