1
|
Driscoll MK, Welf ES, Weems A, Sapoznik E, Zhou F, Murali VS, García-Arcos JM, Roh-Johnson M, Piel M, Dean KM, Fiolka R, Danuser G. Proteolysis-free amoeboid migration of melanoma cells through crowded environments via bleb-driven worrying. Dev Cell 2024; 59:2414-2428.e8. [PMID: 38870943 PMCID: PMC11421976 DOI: 10.1016/j.devcel.2024.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/27/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024]
Abstract
In crowded microenvironments, migrating cells must find or make a path. Amoeboid cells are thought to find a path by deforming their bodies to squeeze through tight spaces. Yet, some amoeboid cells seem to maintain a near-spherical morphology as they move. To examine how they do so, we visualized amoeboid human melanoma cells in dense environments and found that they carve tunnels via bleb-driven degradation of extracellular matrix components without the need for proteolytic degradation. Interactions between adhesions and collagen at the cell front induce a signaling cascade that promotes bleb enlargement via branched actin polymerization. Large blebs abrade collagen, creating feedback between extracellular matrix structure, cell morphology, and polarization that enables both path generation and persistent movement.
Collapse
Affiliation(s)
- Meghan K Driscoll
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Erik S Welf
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Andrew Weems
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Etai Sapoznik
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Felix Zhou
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vasanth S Murali
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Minna Roh-Johnson
- Department of Biochemistry, School of Medicine, University of Utah, Salt Lake City, UT 84113, USA
| | - Matthieu Piel
- Institut Curie, UMR144, CNRS, PSL University, Paris, France
| | - Kevin M Dean
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Reto Fiolka
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
2
|
Nguyen KC, Jameson CD, Baldwin SA, Nardini JT, Smith RC, Haugh JM, Flores KB. Quantifying collective motion patterns in mesenchymal cell populations using topological data analysis and agent-based modeling. Math Biosci 2024; 370:109158. [PMID: 38373479 DOI: 10.1016/j.mbs.2024.109158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 02/06/2024] [Accepted: 02/11/2024] [Indexed: 02/21/2024]
Abstract
Fibroblasts in a confluent monolayer are known to adopt elongated morphologies in which cells are oriented parallel to their neighbors. We collected and analyzed new microscopy movies to show that confluent fibroblasts are motile and that neighboring cells often move in anti-parallel directions in a collective motion phenomenon we refer to as "fluidization" of the cell population. We used machine learning to perform cell tracking for each movie and then leveraged topological data analysis (TDA) to show that time-varying point-clouds generated by the tracks contain significant topological information content that is driven by fluidization, i.e., the anti-parallel movement of individual neighboring cells and neighboring groups of cells over long distances. We then utilized the TDA summaries extracted from each movie to perform Bayesian parameter estimation for the D'Orsgona model, an agent-based model (ABM) known to produce a wide array of different patterns, including patterns that are qualitatively similar to fluidization. Although the D'Orsgona ABM is a phenomenological model that only describes inter-cellular attraction and repulsion, the estimated region of D'Orsogna model parameter space was consistent across all movies, suggesting that a specific level of inter-cellular repulsion force at close range may be a mechanism that helps drive fluidization patterns in confluent mesenchymal cell populations.
Collapse
Affiliation(s)
- Kyle C Nguyen
- Biomathematics Graduate Program, North Carolina State University, Raleigh, NC 27607, USA; Center for Research in Scientific Computation, North Carolina State University, Raleigh, NC 27607, USA.
| | | | - Scott A Baldwin
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, USA
| | - John T Nardini
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ 08628, USA
| | - Ralph C Smith
- Department of Mathematics, North Carolina State University, Raleigh, NC 27607, USA
| | - Jason M Haugh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, USA
| | - Kevin B Flores
- Center for Research in Scientific Computation, North Carolina State University, Raleigh, NC 27607, USA; Department of Mathematics, North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
3
|
Matsubayashi HT, Mountain J, Takahashi N, Deb Roy A, Yao T, Peterson AF, Saez Gonzalez C, Kawamata I, Inoue T. Non-catalytic role of phosphoinositide 3-kinase in mesenchymal cell migration through non-canonical induction of p85β/AP2-mediated endocytosis. Nat Commun 2024; 15:2612. [PMID: 38521786 PMCID: PMC10960865 DOI: 10.1038/s41467-024-46855-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: 01/11/2023] [Accepted: 03/13/2024] [Indexed: 03/25/2024] Open
Abstract
Class IA phosphoinositide 3-kinase (PI3K) galvanizes fundamental cellular processes such as migration, proliferation, and differentiation. To enable these multifaceted roles, the catalytic subunit p110 utilizes the multi-domain, regulatory subunit p85 through its inter SH2 domain (iSH2). In cell migration, its product PI(3,4,5)P3 generates locomotive activity. While non-catalytic roles are also implicated, underlying mechanisms and their relationship to PI(3,4,5)P3 signaling remain elusive. Here, we report that a disordered region of iSH2 contains AP2 binding motifs which can trigger clathrin and dynamin-mediated endocytosis independent of PI3K catalytic activity. The AP2 binding motif mutants of p85 aberrantly accumulate at focal adhesions and increase both velocity and persistency in fibroblast migration. We thus propose the dual functionality of PI3K in the control of cell motility, catalytic and non-catalytic, arising distinctly from juxtaposed regions within iSH2.
Collapse
Affiliation(s)
- Hideaki T Matsubayashi
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
- Center for Cell Dynamics, Institute of Basic Biomedical Sciences, Johns Hopkins University, Baltimore, MD, USA.
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Tohoku, Japan.
| | - Jack Mountain
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Center for Cell Dynamics, Institute of Basic Biomedical Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Nozomi Takahashi
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Tohoku, Japan
| | - Abhijit Deb Roy
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Center for Cell Dynamics, Institute of Basic Biomedical Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Tony Yao
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Center for Cell Dynamics, Institute of Basic Biomedical Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Amy F Peterson
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Center for Cell Dynamics, Institute of Basic Biomedical Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Cristian Saez Gonzalez
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Center for Cell Dynamics, Institute of Basic Biomedical Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Ibuki Kawamata
- Department of Robotics, Tohoku University, Tohoku, Japan
- Natural Science Division, Ochanomizu University, Kyoto, Japan
- Graduate School of Science, Kyoto University, Kyoto, Japan
| | - Takanari Inoue
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
- Center for Cell Dynamics, Institute of Basic Biomedical Sciences, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
4
|
Burgess SG, Paul NR, Richards MW, Ault JR, Askenatzis L, Claydon SG, Corbyn R, Machesky LM, Bayliss R. A nanobody inhibitor of Fascin-1 actin-bundling activity and filopodia formation. Open Biol 2024; 14:230376. [PMID: 38503329 PMCID: PMC10960945 DOI: 10.1098/rsob.230376] [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: 10/13/2023] [Accepted: 01/16/2024] [Indexed: 03/21/2024] Open
Abstract
Fascin-1-mediated actin-bundling activity is central to the generation of plasma membrane protrusions required for cell migration. Dysregulated formation of cellular protrusions is observed in metastatic cancers, where they are required for increased invasiveness, and is often correlated with increased Fascin-1 abundance. Therefore, there is interest in generating therapeutic Fascin-1 inhibitors. We present the identification of Nb 3E11, a nanobody inhibitor of Fascin-1 actin-bundling activity and filopodia formation. The crystal structure of the Fascin-1/Nb 3E11 complex reveals the structural mechanism of inhibition. Nb 3E11 occludes an actin-binding site on the third β-trefoil domain of Fascin-1 that is currently not targeted by chemical inhibitors. Binding of Nb 3E11 to Fascin-1 induces a conformational change in the adjacent domains to stabilize Fascin-1 in an inhibitory state similar to that adopted in the presence of small-molecule inhibitors. Nb 3E11 could be used as a tool inhibitor molecule to aid in the development of Fascin-1 targeted therapeutics.
Collapse
Affiliation(s)
- Selena G. Burgess
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Nikki R. Paul
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Mark W. Richards
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - James R. Ault
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Laurie Askenatzis
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Sophie G. Claydon
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Ryan Corbyn
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Laura M. Machesky
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Richard Bayliss
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
5
|
Yim YI, Pedrosa A, Wu X, Chinthalapudi K, Cheney RE, Hammer JA. Mechanisms underlying Myosin 10's contribution to the maintenance of mitotic spindle bipolarity. Mol Biol Cell 2024; 35:ar14. [PMID: 38019611 PMCID: PMC10881153 DOI: 10.1091/mbc.e23-07-0282] [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: 07/24/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
Myosin 10 (Myo10) couples microtubules and integrin-based adhesions to movement along actin filaments via its microtubule-binding MyTH4 domain and integrin-binding FERM domain, respectively. Here we show that Myo10-depleted HeLa cells and mouse embryo fibroblasts (MEFs) both exhibit a pronounced increase in the frequency of multipolar spindles. Staining of unsynchronized metaphase cells showed that the primary driver of spindle multipolarity in Myo10-depleted MEFs and in Myo10-depleted HeLa cells lacking supernumerary centrosomes is pericentriolar material (PCM) fragmentation, which creates y-tubulin-positive acentriolar foci that serve as extra spindle poles. For HeLa cells possessing supernumerary centrosomes, Myo10 depletion further accentuates spindle multipolarity by impairing the clustering of the extra spindle poles. Complementation experiments show that Myo10 must interact with both microtubules and integrins to promote PCM/pole integrity. Conversely, Myo10 only needs interact with integrins to promote supernumerary centrosome clustering. Importantly, images of metaphase Halo-Myo10 knockin cells show that the myosin localizes exclusively to the spindle and the tips of adhesive retraction fibers. We conclude that Myo10 promotes PCM/pole integrity in part by interacting with spindle microtubules, and that it promotes supernumerary centrosome clustering by supporting retraction fiber-based cell adhesion, which likely serves to anchor the microtubule-based forces driving pole focusing.
Collapse
Affiliation(s)
- Yang-In Yim
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Antonio Pedrosa
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xufeng Wu
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Richard E. Cheney
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
6
|
Mittal N, Michels EB, Massey AE, Qiu Y, Royer-Weeden SP, Smith BR, Cartagena-Rivera AX, Han SJ. Myosin-independent stiffness sensing by fibroblasts is regulated by the viscoelasticity of flowing actin. COMMUNICATIONS MATERIALS 2024; 5:6. [PMID: 38741699 PMCID: PMC11090405 DOI: 10.1038/s43246-024-00444-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 01/02/2024] [Indexed: 05/16/2024]
Abstract
The stiffness of the extracellular matrix induces differential tension within integrin-based adhesions, triggering differential mechanoresponses. However, it has been unclear if the stiffness-dependent differential tension is induced solely by myosin activity. Here, we report that in the absence of myosin contractility, 3T3 fibroblasts still transmit stiffness-dependent differential levels of traction. This myosin-independent differential traction is regulated by polymerizing actin assisted by actin nucleators Arp2/3 and formin where formin has a stronger contribution than Arp2/3 to both traction and actin flow. Intriguingly, despite only slight changes in F-actin flow speed observed in cells with the combined inhibition of Arp2/3 and myosin compared to cells with sole myosin inhibition, they show a 4-times reduction in traction than cells with myosin-only inhibition. Our analyses indicate that traditional models based on rigid F-actin are inadequate for capturing such dramatic force reduction with similar actin flow. Instead, incorporating the F-actin network's viscoelastic properties is crucial. Our new model including the F-actin viscoelasticity reveals that Arp2/3 and formin enhance stiffness sensitivity by mechanically reinforcing the F-actin network, thereby facilitating more effective transmission of flow-induced forces. This model is validated by cell stiffness measurement with atomic force microscopy and experimental observation of model-predicted stiffness-dependent actin flow fluctuation.
Collapse
Affiliation(s)
- Nikhil Mittal
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA
- Health Research Institute, Michigan Technological University, Houghton, MI, USA
| | - Etienne B. Michels
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA
| | - Andrew E. Massey
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Yunxiu Qiu
- Department of Biomedical Engineering, Michigan State University, Lansing, MI, USA
| | - Shaina P. Royer-Weeden
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA
| | - Bryan R. Smith
- Department of Biomedical Engineering, Michigan State University, Lansing, MI, USA
| | - Alexander X. Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Sangyoon J. Han
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA
- Health Research Institute, Michigan Technological University, Houghton, MI, USA
- Department of Mechanical Engineering and Engineering Mechanics, Michigan Technological University, Houghton, MI, USA
| |
Collapse
|
7
|
Liu Y, Niu R, Deng R, Wang Y, Song S, Zhang H. Multi-Enzyme Co-Expressed Nanomedicine for Anti-Metastasis Tumor Therapy by Up-Regulating Cellular Oxidative Stress and Depleting Cholesterol. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307752. [PMID: 37734072 DOI: 10.1002/adma.202307752] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/07/2023] [Indexed: 09/23/2023]
Abstract
Tumor cells movement and migration are inseparable from the integrity of lipid rafts and the formation of lamellipodia, and lipid rafts are also a prerequisite for the formation of lamellipodia. Therefore, destroying the lipid rafts is an effective strategy to inhibit tumor metastasis. Herein, a multi-enzyme co-expressed nanomedicine: cholesterol oxidase (CHO) loaded Co─PN3 single-atom nanozyme (Co─PN3 SA/CHO) that can up-regulate cellular oxidative stress, disrupt the integrity of lipid rafts, and inhibit lamellipodia formation to induce anti-metastasis tumor therapy, is developed. In this process, Co─PN3 SA can catalyze oxygen (O2 ) and hydrogen peroxide (H2 O2 ) to generate reactive oxygen species (ROS) via oxidase-like and Fenton-like properties. The doping of P atoms optimizes the adsorption process of the intermediate at the active site and enhances the ROS generation properties of nanomedicine. Meantime, O2 produced by catalase-like catalysis can combine with excess cholesterol to generate more H2 O2 under CHO catalysis, achieving enhanced oxidative damage to tumor cells. Most importantly, cholesterol depletion in tumor cells also disrupts the integrity of lipid rafts and inhibits the formation of lamellipodia, greatly inhibiting the proliferation and metastasis of tumor cells. This strategy by up-regulating cellular oxidative stress and depleting cellular cholesterol constructs a new idea for anti-metastasis-oriented cancer therapy strategies.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| | - Rui Niu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Ruiping Deng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| | - Yinghui Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Shuyan Song
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Department of Chemistry, Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
8
|
Heilala M, Lehtonen A, Arasalo O, Peura A, Pokki J, Ikkala O, Nonappa, Klefström J, Munne PM. Fibrin Stiffness Regulates Phenotypic Plasticity of Metastatic Breast Cancer Cells. Adv Healthc Mater 2023; 12:e2301137. [PMID: 37671812 PMCID: PMC11469292 DOI: 10.1002/adhm.202301137] [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: 04/11/2023] [Revised: 08/18/2023] [Indexed: 09/07/2023]
Abstract
The extracellular matrix (ECM)-regulated phenotypic plasticity is crucial for metastatic progression of triple negative breast cancer (TNBC). While ECM faithful cell-based models are available for in situ and invasive tumors, such as cell aggregate cultures in reconstituted basement membrane and in collagenous gels, there are no ECM faithful models for metastatic circulating tumor cells (CTCs). Such models are essential to represent the stage of metastasis where clinical relevance and therapeutic opportunities are significant. Here, CTC-like DU4475 TNBC cells are cultured in mechanically tunable 3D fibrin hydrogels. This is motivated, as in circulation fibrin aids CTC survival by forming a protective coating reducing shear stress and immune cell-mediated cytotoxicity and promotes several stages of late metastatic processes at the interface between circulation and tissue. This work shows that fibrin hydrogels support DU4475 cell growth, resulting in spheroid formation. Furthermore, increasing fibrin stiffness from 57 to 175 Pa leads to highly motile, actin and tubulin containing cellular protrusions, which are associated with specific cell morphology and gene expression patterns that markedly differ from basement membrane or suspension cultures. Thus, mechanically tunable fibrin gels reveal specific matrix-based regulation of TNBC cell phenotype and offer scaffolds for CTC-like cells with better mechano-biological properties than liquid.
Collapse
Affiliation(s)
- Maria Heilala
- Department of Applied PhysicsAalto UniversityP.O. Box 15100AaltoEspooFI‐00076Finland
| | - Arttu Lehtonen
- Department of Electrical Engineering and AutomationAalto UniversityP.O. Box 12200AaltoEspooFI‐00076Finland
| | - Ossi Arasalo
- Department of Electrical Engineering and AutomationAalto UniversityP.O. Box 12200AaltoEspooFI‐00076Finland
| | - Aino Peura
- Finnish Cancer Institute and FICAN SouthHelsinki University Hospital & Cancer Cell Circuitry LaboratoryTranslational Cancer MedicineMedical FacultyUniversity of HelsinkiP.O. Box 63 (Haartmaninkatu 8)Helsinki00014Finland
| | - Juho Pokki
- Department of Electrical Engineering and AutomationAalto UniversityP.O. Box 12200AaltoEspooFI‐00076Finland
| | - Olli Ikkala
- Department of Applied PhysicsAalto UniversityP.O. Box 15100AaltoEspooFI‐00076Finland
| | - Nonappa
- Faculty of Engineering and Natural SciencesTampere UniversityP.O. Box 541TampereFI‐33720Finland
| | - Juha Klefström
- Finnish Cancer Institute and FICAN SouthHelsinki University Hospital & Cancer Cell Circuitry LaboratoryTranslational Cancer MedicineMedical FacultyUniversity of HelsinkiP.O. Box 63 (Haartmaninkatu 8)Helsinki00014Finland
| | - Pauliina M. Munne
- Finnish Cancer Institute and FICAN SouthHelsinki University Hospital & Cancer Cell Circuitry LaboratoryTranslational Cancer MedicineMedical FacultyUniversity of HelsinkiP.O. Box 63 (Haartmaninkatu 8)Helsinki00014Finland
| |
Collapse
|
9
|
Ji C, Huang Y. Durotaxis and negative durotaxis: where should cells go? Commun Biol 2023; 6:1169. [PMID: 37973823 PMCID: PMC10654570 DOI: 10.1038/s42003-023-05554-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023] Open
Abstract
Durotaxis and negative durotaxis are processes in which cell migration is directed by extracellular stiffness. Durotaxis is the tendency of cells to migrate toward stiffer areas, while negative durotaxis occurs when cells migrate toward regions with lower stiffness. The mechanisms of both processes are not yet fully understood. Additionally, the connection between durotaxis and negative durotaxis remains unclear. In this review, we compare the mechanisms underlying durotaxis and negative durotaxis, summarize the basic principles of both, discuss the possible reasons why some cell types exhibit durotaxis while others exhibit negative durotaxis, propose mechanisms of switching between these processes, and emphasize the challenges in the investigation of durotaxis and negative durotaxis.
Collapse
Affiliation(s)
- Congcong Ji
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yuxing Huang
- Center for Precision Medicine Multi-Omics Research, Peking University Health Science Center, Peking University, Beijing, 100191, China.
| |
Collapse
|
10
|
Mogilner A, Savinov M. Crawling, waving, inch worming, dilating, and pivoting mechanics of migrating cells: Lessons from Ken Jacobson. Biophys J 2023; 122:3551-3559. [PMID: 36934300 PMCID: PMC10541468 DOI: 10.1016/j.bpj.2023.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/07/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Research on the locomotion of single cells on hard, flat surfaces brought insight into the mechanisms of leading-edge protrusion, spatially graded adhesion, front-rear coordination, and how intracellular and traction forces are harnessed to execute various maneuvers. Here, we highlight how, by studying a variety of cell types, shapes, and movements, Ken Jacobson and his collaborators made several discoveries that triggered the mechanistic understanding of cell motility. We then review the recent advancements and current perspectives in this field.
Collapse
Affiliation(s)
- Alex Mogilner
- Courant Institute of Mathematical Sciences, New York University, New York, New York; Department of Biology, New York University, New York, New York.
| | - Mariya Savinov
- Courant Institute of Mathematical Sciences, New York University, New York, New York
| |
Collapse
|
11
|
Ruggiero C, Tamburello M, Rossini E, Zini S, Durand N, Cantini G, Cioppi F, Hantel C, Kiseljak-Vassiliades K, Wierman ME, Landwehr LS, Weigand I, Kurlbaum M, Zizioli D, Turtoi A, Yang S, Berruti A, Luconi M, Sigala S, Lalli E. FSCN1 as a new druggable target in adrenocortical carcinoma. Int J Cancer 2023; 153:210-223. [PMID: 36971100 DOI: 10.1002/ijc.34526] [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: 11/21/2022] [Revised: 03/02/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023]
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy with a high risk of relapse and metastatic spread. The actin-bundling protein fascin (FSCN1) is overexpressed in aggressive ACC and represents a reliable prognostic indicator. FSCN1 has been shown to synergize with VAV2, a guanine nucleotide exchange factor for the Rho/Rac GTPase family, to enhance the invasion properties of ACC cancer cells. Based on those results, we investigated the effects of FSCN1 inactivation by CRISPR/Cas9 or pharmacological blockade on the invasive properties of ACC cells, both in vitro and in an in vivo metastatic ACC zebrafish model. Here, we showed that FSCN1 is a transcriptional target for β-catenin in H295R ACC cells and that its inactivation resulted in defects in cell attachment and proliferation. FSCN1 knock-out modulated the expression of genes involved in cytoskeleton dynamics and cell adhesion. When Steroidogenic Factor-1 (SF-1) dosage was upregulated in H295R cells, activating their invasive capacities, FSCN1 knock-out reduced the number of filopodia, lamellipodia/ruffles and focal adhesions, while decreasing cell invasion in Matrigel. Similar effects were produced by the FSCN1 inhibitor G2-044, which also diminished the invasion of other ACC cell lines expressing lower levels of FSCN1 than H295R. In the zebrafish model, metastases formation was significantly reduced in FSCN1 knock-out cells and G2-044 significantly reduced the number of metastases formed by ACC cells. Our results indicate that FSCN1 is a new druggable target for ACC and provide the rationale for future clinical trials with FSCN1 inhibitors in patients with ACC.
Collapse
Affiliation(s)
- Carmen Ruggiero
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, 06560, Valbonne, France
- Université Côte d'Azur, 06560, Valbonne, France
| | - Mariangela Tamburello
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Elisa Rossini
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Silvia Zini
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Nelly Durand
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, 06560, Valbonne, France
- Université Côte d'Azur, 06560, Valbonne, France
| | - Giulia Cantini
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50134, Florence, Italy
| | - Francesca Cioppi
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50134, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134, Florence, Italy
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), 8091, Zürich, Switzerland
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, 01307, Dresden, Germany
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, 80045, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Affairs Medical Center, 80045, Aurora, Colorado, USA
| | - Margaret E Wierman
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, 80045, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Affairs Medical Center, 80045, Aurora, Colorado, USA
| | - Laura-Sophie Landwehr
- Division of Endocrinology and Diabetology-Department of Internal Medicine I, University Hospital, University of Würzburg, 97080, Würzburg, Germany
| | - Isabel Weigand
- Division of Endocrinology and Diabetology-Department of Internal Medicine I, University Hospital, University of Würzburg, 97080, Würzburg, Germany
- Department of Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, 81377, Munich, Germany
| | - Max Kurlbaum
- Division of Endocrinology and Diabetology-Department of Internal Medicine I, University Hospital, University of Würzburg, 97080, Würzburg, Germany
| | - Daniela Zizioli
- Section of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Andrei Turtoi
- Tumor Microenvironment and Resistance to Therapy Laboratory, Institut de Recherche en Cancérologie de Montpellier, Université de Montpellier-INSERM U1194, 34090, Montpellier, France
- Platform for Translational Oncometabolomics, Biocampus, CNRS-INSERM-Université de Montpellier, 34090, Montpellier, France
| | - Shengyu Yang
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, 17033, Hershey, Pennsylvania, USA
| | - Alfredo Berruti
- Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia and ASST Spedali Civili di Brescia, 25123, Brescia, Italy
| | - Michaela Luconi
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50134, Florence, Italy
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, 06560, Valbonne, France
- Université Côte d'Azur, 06560, Valbonne, France
- Inserm, 06560, Valbonne, France
| |
Collapse
|
12
|
Yim YI, Pedrosa A, Wu X, Chinthalapudi K, Cheney RE, Hammer JA. Myosin 10 uses its MyTH4 and FERM domains differentially to support two aspects of spindle pole biology required for mitotic spindle bipolarity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.15.545002. [PMID: 37398378 PMCID: PMC10312724 DOI: 10.1101/2023.06.15.545002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Myosin 10 (Myo10) has the ability to link actin filaments to integrin-based adhesions and to microtubules by virtue of its integrin-binding FERM domain and microtubule-binding MyTH4 domain, respectively. Here we used Myo10 knockout cells to define Myo10's contribution to the maintenance of spindle bipolarity, and complementation to quantitate the relative contributions of its MyTH4 and FERM domains. Myo10 knockout HeLa cells and mouse embryo fibroblasts (MEFs) both exhibit a pronounced increase in the frequency of multipolar spindles. Staining of unsynchronized metaphase cells showed that the primary driver of spindle multipolarity in knockout MEFs and knockout HeLa cells lacking supernumerary centrosomes is pericentriolar material (PCM) fragmentation, which creates γ-tubulin-positive acentriolar foci that serve as additional spindle poles. For HeLa cells possessing supernumerary centrosomes, Myo10 depletion further accentuates spindle multipolarity by impairing the clustering of the extra spindle poles. Complementation experiments show that Myo10 must interact with both integrins and microtubules to promote PCM/pole integrity. Conversely, Myo10's ability to promote the clustering of supernumerary centrosomes only requires that it interact with integrins. Importantly, images of Halo-Myo10 knock-in cells show that the myosin localizes exclusively within adhesive retraction fibers during mitosis. Based on these and other results, we conclude that Myo10 promotes PCM/pole integrity at a distance, and that it facilitates supernumerary centrosome clustering by promoting retraction fiber-based cell adhesion, which likely provides an anchor for the microtubule-based forces driving pole focusing.
Collapse
Affiliation(s)
- Yang-In Yim
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Antonio Pedrosa
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Xufeng Wu
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH
| | - Richard E. Cheney
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
13
|
Abu Bakar N, Wan Ibrahim WN, Zulkiflli AR, Saleh Hodin NA, Kim TY, Ling YS, Md Ajat MM, Shaari K, Shohaimi S, Nasruddin NS, Mohd Faudzi SM, Kim CH. Embryonic mercury exposure in zebrafish: Alteration of metabolites and gene expression, related to visual and behavioral impairments. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 256:114862. [PMID: 37004432 DOI: 10.1016/j.ecoenv.2023.114862] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/05/2023] [Accepted: 03/30/2023] [Indexed: 06/19/2023]
Abstract
The widespread presence of mercury, a heavy metal found in the environment and used in numerous industries and domestic, raises concerns about its potential impact on human health. Nevertheless, the adverse effects of this environmental toxicant at low concentrations are often underestimated. There are emerging studies showing that accumulation of mercury in the eye may contribute to visual impairment and a comorbidity between autism spectrum disorders (ASD) trait and visual impairment. However, the underlying mechanism of visual impairment in humans and rodents is challenging. In response to this issue, zebrafish larvae with a cone-dominated retinal visual system were exposed to 100 nM mercury chloride (HgCl2), according to our previous study, followed by light-dark stimulation, a social assay, and color preference to examine the functionality of the visual system in relation to ASD-like behavior. Exposure of embryos to HgCl2 from gastrulation to hatching increased locomotor activity in the dark, reduced shoaling and exploratory behavior, and impaired color preference. Defects in microridges as the first barrier may serve as primary tools for HgCl2 toxicity affecting vision. Depletion of polyunsaturated fatty acids (PUFAs), linoleic acid, arachidonic acid (ARA), alpha-linoleic acid, docosahexaenoic acid (DHA), stearic acid, L-phenylalanine, isoleucine, L-lysine, and N-acetylputrescine, along with the increase of gamma-aminobutyric acid (GABA), sphingosine-1-phosphate, and citrulline assayed by liquid chromatography-mass spectrometry (LC-MS) suggest that these metabolites serve as biomarkers of retinal impairments that affect vision and behavior. Although suppression of adsl, shank3a, tsc1b, and nrxn1a gene expression was observed, among these tsc1b showed more positive correlation with ASD. Collectively, these results contribute new insights into the possible mechanism of mercury toxicity give rise to visual, cognitive, and social deficits in zebrafish.
Collapse
Affiliation(s)
- Noraini Abu Bakar
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Wan Norhamidah Wan Ibrahim
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Natural Medicines and Product Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Abdul Rahman Zulkiflli
- Natural Medicines and Product Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Nur Atikah Saleh Hodin
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Tae-Yoon Kim
- Department of Biology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Yee Soon Ling
- Biotechnology Research Institute, Universiti Malaysia Sabah, Jalan UMS, 88400 Kota Kinabalu, Sabah, Malaysia
| | - Mohd Mokrish Md Ajat
- Natural Medicines and Product Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Malaysia
| | - Khozirah Shaari
- Natural Medicines and Product Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Shamarina Shohaimi
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Nurrul Shaqinah Nasruddin
- Centre for Craniofacial Diagnostics, Faculty of Dentistry, Universiti Kebangsaan Malaysia (UKM), 50300 Kuala Lumpur, Malaysia
| | - Siti Munirah Mohd Faudzi
- Natural Medicines and Product Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
14
|
Matsubayashi H, Mountain J, Yao T, Peterson A, Roy AD, Inoue T. Non-catalytic role of phosphoinositide 3-kinase in mesenchymal cell migration through non-canonical induction of p85β/AP-2-mediated endocytosis. RESEARCH SQUARE 2023:rs.3.rs-2432041. [PMID: 36712095 PMCID: PMC9882665 DOI: 10.21203/rs.3.rs-2432041/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Class IA phosphoinositide 3-kinase (PI3K) galvanizes fundamental cellular processes such as migration, proliferation, and differentiation. To enable multifaceted roles, the catalytic subunit p110 utilizes a multi-domain, regulatory subunit p85 through its inter SH2 domain (iSH2). In cell migration, their product PI(3,4,5)P3 generates locomotive activity. While non-catalytic roles are also implicated, underlying mechanisms and its relationship to PI(3,4,5)P3 signaling remain elusive. Here, we report that a disordered region of iSH2 contains previously uncharacterized AP-2 binding motifs which can trigger clathrin and dynamin-mediated endocytosis independent of PI3K catalytic activity. The AP-2 binding motif mutants of p85 aberrantly accumulate at focal adhesions and upregulate both velocity and persistency in fibroblast migration. We thus propose the dual functionality of PI3K in the control of cell motility, catalytic and non-catalytic, arising distinctly from juxtaposed regions within iSH2.
Collapse
|
15
|
Matsubayashi HT, Mountain J, Yao T, Peterson AF, Deb Roy A, Inoue T. Non-catalytic role of phosphoinositide 3-kinase in mesenchymal cell migration through non-canonical induction of p85β/AP-2-mediated endocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2022.12.31.522383. [PMID: 36712134 PMCID: PMC9881872 DOI: 10.1101/2022.12.31.522383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Class IA phosphoinositide 3-kinase (PI3K) galvanizes fundamental cellular processes such as migration, proliferation, and differentiation. To enable multifaceted roles, the catalytic subunit p110 utilizes a multidomain, regulatory subunit p85 through its inter SH2 domain (iSH2). In cell migration, their product PI(3,4,5)P3 generates locomotive activity. While non-catalytic roles are also implicated, underlying mechanisms and its relationship to PI(3,4,5)P3 signaling remain elusive. Here, we report that a disordered region of iSH2 contains previously uncharacterized AP-2 binding motifs which can trigger clathrin and dynamin-mediated endocytosis independent of PI3K catalytic activity. The AP-2 binding motif mutants of p85 aberrantly accumulate at focal adhesions and upregulate both velocity and persistency in fibroblast migration. We thus propose the dual functionality of PI3K in the control of cell motility, catalytic and non-catalytic, arising distinctly from juxtaposed regions within iSH2.
Collapse
Affiliation(s)
- Hideaki T. Matsubayashi
- Department of Cell Biology, School of Medicine, Johns Hopkins University
- Center for Cell Dynamics, Institute of Basic Biomedical Sciences, Johns Hopkins University
| | - Jack Mountain
- Department of Cell Biology, School of Medicine, Johns Hopkins University
- Center for Cell Dynamics, Institute of Basic Biomedical Sciences, Johns Hopkins University
| | - Tony Yao
- Department of Cell Biology, School of Medicine, Johns Hopkins University
- Center for Cell Dynamics, Institute of Basic Biomedical Sciences, Johns Hopkins University
| | - Amy F. Peterson
- Department of Cell Biology, School of Medicine, Johns Hopkins University
- Center for Cell Dynamics, Institute of Basic Biomedical Sciences, Johns Hopkins University
| | - Abhijit Deb Roy
- Department of Cell Biology, School of Medicine, Johns Hopkins University
- Center for Cell Dynamics, Institute of Basic Biomedical Sciences, Johns Hopkins University
| | - Takanari Inoue
- Department of Cell Biology, School of Medicine, Johns Hopkins University
- Center for Cell Dynamics, Institute of Basic Biomedical Sciences, Johns Hopkins University
| |
Collapse
|
16
|
A network of mixed actin polarity in the leading edge of spreading cells. Commun Biol 2022; 5:1338. [PMID: 36473943 PMCID: PMC9727120 DOI: 10.1038/s42003-022-04288-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Physical interactions of cells with the underlying extracellular matrix (ECM) play key roles in multiple cellular processes. The actin cytoskeleton is a central driver and regulator of cellular dynamics, that produces membrane-protrusions such as lamellipodia and filopodia. Here, we examined actin organization in expanding lamellipodia during early stages of cell spreading. To gain insight into the 3D actin organization, we plated fibroblasts on galectin-8 coated EM grids, an ECM protein presents in disease states. We then combined cryo-electron tomography with advanced image processing tools for reconstructing the structure of F-actin in the lamellipodia. This approach enabled us to resolve the polarity and orientation of filaments, and the structure of the Arp2/3 complexes associated with F-actin branches. We show that F-actin in lamellipodial protrusions forms a dense network with three distinct sub-domains. One consists primarily of radial filaments, with their barbed ends pointing towards the membrane, the other is enriched with parallel filaments that run between the radial fibers, in addition to an intermediate sub-domain. Surprisingly, a minor, yet significant (~10%) population of actin filaments, are oriented with their barbed-ends towards the cell center. Our results provide structural insights into F-actin assembly and dynamic reorganization in the leading edge of spreading cells.
Collapse
|
17
|
Lappalainen P, Kotila T, Jégou A, Romet-Lemonne G. Biochemical and mechanical regulation of actin dynamics. Nat Rev Mol Cell Biol 2022; 23:836-852. [PMID: 35918536 DOI: 10.1038/s41580-022-00508-4] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2022] [Indexed: 12/30/2022]
Abstract
Polymerization of actin filaments against membranes produces force for numerous cellular processes, such as migration, morphogenesis, endocytosis, phagocytosis and organelle dynamics. Consequently, aberrant actin cytoskeleton dynamics are linked to various diseases, including cancer, as well as immunological and neurological disorders. Understanding how actin filaments generate forces in cells, how force production is regulated by the interplay between actin-binding proteins and how the actin-regulatory machinery responds to mechanical load are at the heart of many cellular, developmental and pathological processes. During the past few years, our understanding of the mechanisms controlling actin filament assembly and disassembly has evolved substantially. It has also become evident that the activities of key actin-binding proteins are not regulated solely by biochemical signalling pathways, as mechanical regulation is critical for these proteins. Indeed, the architecture and dynamics of the actin cytoskeleton are directly tuned by mechanical load. Here we discuss the general mechanisms by which key actin regulators, often in synergy with each other, control actin filament assembly, disassembly, and monomer recycling. By using an updated view of actin dynamics as a framework, we discuss how the mechanics and geometry of actin networks control actin-binding proteins, and how this translates into force production in endocytosis and mesenchymal cell migration.
Collapse
Affiliation(s)
- Pekka Lappalainen
- Institute of Biotechnology and Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland.
| | - Tommi Kotila
- Institute of Biotechnology and Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Antoine Jégou
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | | |
Collapse
|
18
|
Jang HJ, Yoon YJ, Choi J, Lee YJ, Lee S, Cho W, Byun WG, Park SB, Han DC, Kwon BM. S-Benproperine, an Active Stereoisomer of Benproperine, Suppresses Cancer Migration and Tumor Metastasis by Targeting ARPC2. Pharmaceuticals (Basel) 2022; 15:ph15121462. [PMID: 36558913 PMCID: PMC9785746 DOI: 10.3390/ph15121462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/19/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Metastasis, in which cancer cells migrate to other tissues and form new tumors, is a major cause of both cancer death and treatment failure. In a previous study, benproperine (Benp) was identified as a cancer cell migration inhibitor and an inhibitor of actin-related protein 2/3 complex subunit 2 (ARPC2). However, Benp is a racemic mixture, and which stereoisomer is the active isomer remains unclear. In this study, we found that S-Benp is an active isomer and inhibits the migration and invasion of cancer cells much more strongly than R-Benp, with no effect on normal cells. The metastasis inhibitory effect of S-Benp was also verified in an animal model. Validating that inhibitors bind to their targets in cells and tissues has been a very challenging task in drug discovery. The direct interactions between ARPC2 and S-Benp were verified by surface plasmon resonance analysis (SPR), a cellular thermal shift assay (CETSA), and drug affinity responsive target stability (DARTS). In the mutant study with ARPC2F225A cells, S-Benp did not bind to ARPC2F225A according to CETSA and DARTS. Furthermore, we validated that S-Benp colocalized with ARPC2 in cancer cells and directly bound to ARPC2 in tumor tissues using Cy3-conjugated S-Benp according to CETSA. Finally, actin polymerization assays and immunocytochemistry showed that S-Benp suppressed actin remodeling such as lamellipodium formation. Taken together, our data suggest that S-Benp is an active stereoisomer of Benp and a potential metastasis inhibitor via ARPC2 binding.
Collapse
Affiliation(s)
- Hyun-Jin Jang
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahakro, Daejeon 34141, Republic of Korea
| | - Yae Jin Yoon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahakro, Daejeon 34141, Republic of Korea
| | - Jiyeon Choi
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahakro, Daejeon 34141, Republic of Korea
| | - Yu-Jin Lee
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahakro, Daejeon 34141, Republic of Korea
| | - Sangku Lee
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahakro, Daejeon 34141, Republic of Korea
| | - Wansang Cho
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Wan Gi Byun
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Seung Bum Park
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Dong Cho Han
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahakro, Daejeon 34141, Republic of Korea
- KRIBB School of Bioscience, University of Science and Technology in Korea, 217 Gajeongro, Daejeon 34113, Republic of Korea
- Correspondence: (D.C.H.); (B.-M.K.)
| | - Byoung-Mog Kwon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahakro, Daejeon 34141, Republic of Korea
- Central Research Institute, VS Pharm Tech Co., Ltd., Daejeon 35209, Republic of Korea
- Correspondence: (D.C.H.); (B.-M.K.)
| |
Collapse
|
19
|
Molina E, Cataldo VF, Eggers C, Muñoz-Madrid V, Glavic Á. p53 Related Protein Kinase is Required for Arp2/3-Dependent Actin Dynamics of Hemocytes in Drosophila melanogaster. Front Cell Dev Biol 2022; 10:859105. [PMID: 35721516 PMCID: PMC9201722 DOI: 10.3389/fcell.2022.859105] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/22/2022] [Indexed: 11/21/2022] Open
Abstract
Cells extend membrane protrusions like lamellipodia and filopodia from the leading edge to sense, to move and to form new contacts. The Arp2/3 complex sustains lamellipodia formation, and in conjunction with the actomyosin contractile system, provides mechanical strength to the cell. Drosophila p53-related protein kinase (Prpk), a Tsc5p ortholog, has been described as essential for cell growth and proliferation. In addition, Prpk interacts with proteins associated to actin filament dynamics such as α-spectrin and the Arp2/3 complex subunit Arpc4. Here, we investigated the role of Prpk in cell shape changes, specifically regarding actin filament dynamics and membrane protrusion formation. We found that reductions in Prpk alter cell shape and the structure of lamellipodia, mimicking the phenotypes evoked by Arp2/3 complex deficiencies. Prpk co-localize and co-immunoprecipitates with the Arp2/3 complex subunit Arpc1 and with the small GTPase Rab35. Importantly, expression of Rab35, known by its ability to recruit upstream regulators of the Arp2/3 complex, could rescue the Prpk knockdown phenotypes. Finally, we evaluated the requirement of Prpk in different developmental contexts, where it was shown to be essential for correct Arp2/3 complex distribution and actin dynamics required for hemocytes migration, recruitment, and phagocytosis during immune response.
Collapse
Affiliation(s)
- Emiliano Molina
- FONDAP Center for Genome Regulation, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Vicente F. Cataldo
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristián Eggers
- Department for Chemistry and Biochemistry and Pharmaceutical Sciences, Faculty of Science, University of Bern, Bern, Switzerland
| | - Valentina Muñoz-Madrid
- FONDAP Center for Genome Regulation, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Álvaro Glavic
- FONDAP Center for Genome Regulation, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- *Correspondence: Álvaro Glavic,
| |
Collapse
|
20
|
Korkmazhan E, Kennard AS, Garzon-Coral C, Vasquez CG, Dunn AR. Tether-guided lamellipodia enable rapid wound healing. Biophys J 2022; 121:1029-1037. [PMID: 35167863 PMCID: PMC8943750 DOI: 10.1016/j.bpj.2022.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/15/2021] [Accepted: 02/03/2022] [Indexed: 11/20/2022] Open
Abstract
Adhesion between animal cells and the underlying extracellular matrix is challenged during wounding, cell division, and a variety of pathological processes. How cells recover adhesion in the immediate aftermath of detachment from the extracellular matrix remains incompletely understood, due in part to technical limitations. Here, we used acute chemical and mechanical perturbations to examine how epithelial cells respond to partial delamination events. In both cases, we found that cells extended lamellipodia to establish readhesion within seconds of detachment. These lamellipodia were guided by sparse membrane tethers whose tips remained attached to their original points of adhesion, yielding lamellipodia that appear to be qualitatively distinct from those observed during cell migration. In vivo measurements in the context of a zebrafish wound assay showed a similar behavior, in which membrane tethers guided rapidly extending lamellipodia. In the case of mechanical wounding events, cells selectively extended tether-guided lamellipodia in the direction opposite of the pulling force, resulting in the rapid reestablishment of contact with the substrate. We suggest that membrane tether-guided lamellipodial respreading may represent a general mechanism to reestablish tissue integrity in the face of acute disruption.
Collapse
Affiliation(s)
- Elgin Korkmazhan
- Graduate Program in Biophysics, Stanford University, Stanford, California; Department of Chemical Engineering, Stanford University, Stanford, California
| | - Andrew S Kennard
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, Washington
| | - Carlos Garzon-Coral
- Department of Chemical Engineering, Stanford University, Stanford, California
| | - Claudia G Vasquez
- Department of Chemical Engineering, Stanford University, Stanford, California
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, California.
| |
Collapse
|
21
|
Lu Y, Zhang P, Chen H, Tong Q, Wang J, Li Q, Tian C, Yang J, Li S, Zhang Z, Yuan H, Xiang M. Cytochalasin Q exerts anti-melanoma effect by inhibiting creatine kinase B. Toxicol Appl Pharmacol 2022; 441:115971. [PMID: 35276125 DOI: 10.1016/j.taap.2022.115971] [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: 01/03/2022] [Revised: 02/24/2022] [Accepted: 03/03/2022] [Indexed: 10/18/2022]
Abstract
Due to the pivotal role of microfilament in cancer cells, targeting microfilaments with cytochalasins is considered a promising anticancer strategy. Here, we obtained cytochalasin Q (CQ) from Xylaria sp. DO1801, the endophytic fungi from the root of plant Damnacanthus officinarum, and discovered its anti-melanoma activity in vivo and in vitro attributing to microfilament depolymerization. Mechanistically, CQ directly bound to and inactivated creatine kinase B (CKB), an enzyme phosphorylating creatine to phosphocreatine (PCr) and regenerating ATP to cope with high energy demand, and then inhibited the creatine metabolism as well as cytosolic glycolysis in melanoma cells. Preloading PCr recovered ATP generation, reversed microfilament depolymerization and blunted anti-melanoma efficacy of CQ. Knockdown of CKB resulted in reduced ATP level, perturbed microfilament, inhibited proliferation and induced apoptosis, and manifested lower sensitivity to CQ. Further, we found that either CQ or CKB depletion suppressed the PI3K/AKT/FoxO1 pathway, whereas 740Y-P, a PI3K agonist, elevated protein expression of CKB suppressed by CQ. Taken together, our study highlights the significant anti-melanoma effect and proposes a PI3K/AKT/FoxO1/ CKB feedback circuit for the activity of CQ, opening new opportunities for current chemotherapy.
Collapse
Affiliation(s)
- Yi Lu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Peng Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongdan Chen
- Breast and Thyroid Surgical Department, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400014, China; Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qingyi Tong
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jia Wang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qing Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cheng Tian
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian Yang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Senlin Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zijun Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huimin Yuan
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
22
|
Simvastatin Downregulates Cofilin and Stathmin to Inhibit Skeletal Muscle Cells Migration. Int J Mol Sci 2022; 23:ijms23052848. [PMID: 35269994 PMCID: PMC8911248 DOI: 10.3390/ijms23052848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/27/2022] [Accepted: 03/02/2022] [Indexed: 01/27/2023] Open
Abstract
Statins are the most effective therapeutic agents for reducing cholesterol synthesis. Given their widespread use, many adverse effects from statins have been reported; of these, musculoskeletal complications occurred in 15% of patients after receiving statins for 6 months, and simvastatin was the most commonly administered statin among these cases. This study investigated the negative effects of simvastatin on skeletal muscle cells. We performed RNA sequencing analysis to determine gene expression in simvastatin-treated cells. Cell proliferation and migration were examined through cell cycle analysis and the transwell filter migration assay, respectively. Cytoskeleton rearrangement was examined through F-actin and tubulin staining. Western blot analysis was performed to determine the expression of cell cycle-regulated and cytoskeleton-related proteins. Transfection of small interfering RNAs (siRNAs) was performed to validate the role of cofilin and stathmin in the simvastatin-mediated inhibition of cell migration. The results revealed that simvastatin inhibited the proliferation and migration of skeletal muscle cells and affected the rearrangement of F-actin and tubulin. Simvastatin reduced the expression of cofilin and stathmin. The knockdown of both cofilin and stathmin by specific siRNA synergistically impaired cell migration. In conclusion, our results indicated that simvastatin inhibited skeletal muscle cell migration by reducing the expressions of cofilin and stathmin.
Collapse
|
23
|
Yang W, Wei X, Jiao Y, Bai Y, Sam WN, Yan Q, Sun X, Li G, Ma J, Wei W, Tian D, Zheng F. STAT3/HIF-1α/fascin-1 axis promotes RA FLSs migration and invasion ability under hypoxia. Mol Immunol 2021; 142:83-94. [PMID: 34971867 DOI: 10.1016/j.molimm.2021.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/15/2021] [Accepted: 12/01/2021] [Indexed: 12/24/2022]
Abstract
Rheumatoid arthritis (RA) synovium was identified as "tumor-like" tissues because of the hypoxic microenvironment, significant cell proliferation, and invasion phenotypes. It was reported that hypoxia promoted tumor aggressiveness via up-regulated expression of fascin-1 in cancer. However, the role of fascin-1 in RA synovial hyperplasia and joint injury progression remains unknown. In the current study, we first identified that both fascin-1 and HIF-1α were highly expressed in the RA synovium, in which they were widely colocalized, compared to osteoarthritis(OA). As well, levels of fascin-1 in RA fibroblast-like synoviocytes(FLSs) were found significantly higher than those in OA FLSs. Further, it was demonstrated that the mRNA and protein levels of fascin-1 in RA FLSs were up-regulated in hypoxia (3 % O2) and experimental hypoxia induced by cobalt chloride. Mechanistically, the HIF-1α-mediated hypoxia environment activated the gene expression of the fascin-1 protein, which in turn promoted the migration and invasion of RA FLSs. Accordingly, the restoration of FLSs migration and invasion was observed following siRNA-mediated silencing of fascin-1 and HIF-1α expression. Notably, under the experimental hypoxia, we found that the expression levels of fascin-1, HIF-1α, and p-STAT3 were increased in a time-dependent manner, and fascin-1and HIF-1α expressions were dependent on p-STAT3. Our results indicated that hypoxia-induced fascin-1 up-regulation promoted RA FLSs migration and invasion through the STAT3/HIF-1α/fascin-1 axis, which might represent a novel therapeutic target for the treatment of RA.
Collapse
Affiliation(s)
- Wang Yang
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Xinyue Wei
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Yachong Jiao
- Department of Clinical Laboratory, The Third Hospital of Hebei Medical University, Hebei, China
| | - Yingyu Bai
- Laboratory for Mechanisms and Therapies of Heart Diseases, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wilfried Noel Sam
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Qiushuang Yan
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Xuguo Sun
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jun Ma
- Department of Health Statistics, College of Public Health, Tianjin Medical University, Tianjin, China.
| | - Wei Wei
- Department of Rheumatology, General Hospital, Tianjin Medical University, Tianjin, China.
| | - Derun Tian
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin, China.
| | - Fang Zheng
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
24
|
Kashgari G, Venkatesh S, Refuerzo S, Pham B, Bayat A, Klein RH, Ramos R, Ta AP, Plikus MV, Wang PH, Andersen B. GRHL3 activates FSCN1 to relax cell-cell adhesions between migrating keratinocytes during wound reepithelialization. JCI Insight 2021; 6:e142577. [PMID: 34494554 PMCID: PMC8492311 DOI: 10.1172/jci.insight.142577] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 07/21/2021] [Indexed: 01/23/2023] Open
Abstract
The migrating keratinocyte wound front is required for skin wound closure. Despite significant advances in wound healing research, we do not fully understand the molecular mechanisms that orchestrate collective keratinocyte migration. Here, we show that, in the wound front, the epidermal transcription factor Grainyhead like-3 (GRHL3) mediates decreased expression of the adherens junction protein E-cadherin; this results in relaxed adhesions between suprabasal keratinocytes, thus promoting collective cell migration and wound closure. Wound fronts from mice lacking GRHL3 in epithelial cells (Grhl3-cKO) have lower expression of Fascin-1 (FSCN1), a known negative regulator of E-cadherin. Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) on wounded keratinocytes shows decreased wound-induced chromatin accessibility near the Fscn1 gene in Grhl3-cKO mice, a region enriched for GRHL3 motifs. These data reveal a wound-induced GRHL3/FSCN1/E-cadherin pathway that regulates keratinocyte-keratinocyte adhesion during wound-front migration; this pathway is activated in acute human wounds and is altered in diabetic wounds in mice, suggesting translational relevance.
Collapse
Affiliation(s)
| | | | | | - Brandon Pham
- Department of Biological Chemistry, School of Medicine
| | - Anita Bayat
- Department of Biological Chemistry, School of Medicine
| | | | - Raul Ramos
- Department of Developmental & Cell Biology, School of Biological Sciences, and
| | - Albert Paul Ta
- Department of Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine (UCI), California, USA
| | - Maksim V Plikus
- Department of Developmental & Cell Biology, School of Biological Sciences, and
| | - Ping H Wang
- Department of Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine (UCI), California, USA
| | - Bogi Andersen
- Department of Biological Chemistry, School of Medicine.,Department of Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine (UCI), California, USA
| |
Collapse
|
25
|
SenGupta S, Parent CA, Bear JE. The principles of directed cell migration. Nat Rev Mol Cell Biol 2021; 22:529-547. [PMID: 33990789 PMCID: PMC8663916 DOI: 10.1038/s41580-021-00366-6] [Citation(s) in RCA: 258] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 02/03/2023]
Abstract
Cells have the ability to respond to various types of environmental cues, and in many cases these cues induce directed cell migration towards or away from these signals. How cells sense these cues and how they transmit that information to the cytoskeletal machinery governing cell translocation is one of the oldest and most challenging problems in biology. Chemotaxis, or migration towards diffusible chemical cues, has been studied for more than a century, but information is just now beginning to emerge about how cells respond to other cues, such as substrate-associated cues during haptotaxis (chemical cues on the surface), durotaxis (mechanical substrate compliance) and topotaxis (geometric features of substrate). Here we propose four common principles, or pillars, that underlie all forms of directed migration. First, a signal must be generated, a process that in physiological environments is much more nuanced than early studies suggested. Second, the signal must be sensed, sometimes by cell surface receptors, but also in ways that are not entirely clear, such as in the case of mechanical cues. Third, the signal has to be transmitted from the sensing modules to the machinery that executes the actual movement, a step that often requires amplification. Fourth, the signal has to be converted into the application of asymmetric force relative to the substrate, which involves mostly the cytoskeleton, but perhaps other players as well. Use of these four pillars has allowed us to compare some of the similarities between different types of directed migration, but also to highlight the remarkable diversity in the mechanisms that cells use to respond to different cues provided by their environment.
Collapse
Affiliation(s)
- Shuvasree SenGupta
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carole A Parent
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - James E Bear
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
26
|
Baldwin SA, Van Bruggen SM, Koelbl JM, Appalabhotla R, Bear JE, Haugh JM. Microfluidic devices fitted with "flowver" paper pumps generate steady, tunable gradients for extended observation of chemotactic cell migration. BIOMICROFLUIDICS 2021; 15:044101. [PMID: 34290842 PMCID: PMC8282348 DOI: 10.1063/5.0054764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/19/2021] [Indexed: 05/11/2023]
Abstract
Microfluidics approaches have gained popularity in the field of directed cell migration, enabling control of the extracellular environment and integration with live-cell microscopy; however, technical hurdles remain. Among the challenges are the stability and predictability of the environment, which are especially critical for the observation of fibroblasts and other slow-moving cells. Such experiments require several hours and are typically plagued by the introduction of bubbles and other disturbances that naturally arise in standard microfluidics protocols. Here, we report on the development of a passive pumping strategy, driven by the high capillary pressure and evaporative capacity of paper, and its application to study fibroblast chemotaxis. The paper pumps-flowvers (flow + clover)-are inexpensive, compact, and scalable, and they allow nearly bubble-free operation, with a predictable volumetric flow rate on the order of μl/min, for several hours. To demonstrate the utility of this approach, we combined the flowver pumping strategy with a Y-junction microfluidic device to generate a chemoattractant gradient landscape that is both stable (6+ h) and predictable (by finite-element modeling calculations). Integrated with fluorescence microscopy, we were able to recapitulate previous, live-cell imaging studies of fibroblast chemotaxis to platelet derived growth factor (PDGF), with an order-of-magnitude gain in throughput. The increased throughput of single-cell analysis allowed us to more precisely define PDGF gradient conditions conducive for chemotaxis; we were also able to interpret how the orientation of signaling through the phosphoinositide 3-kinase pathway affects the cells' sensing of and response to conducive gradients.
Collapse
Affiliation(s)
- Scott A. Baldwin
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, North Carolina 27695, USA
| | - Shawn M. Van Bruggen
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, North Carolina 27695, USA
| | - Joseph M. Koelbl
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, North Carolina 27695, USA
| | - Ravikanth Appalabhotla
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, North Carolina 27695, USA
| | - James E. Bear
- Department of Cell Biology and Physiology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Jason M. Haugh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, North Carolina 27695, USA
| |
Collapse
|
27
|
van Steen AC, van der Meer WJ, Hoefer IE, van Buul JD. Actin remodelling of the endothelium during transendothelial migration of leukocytes. Atherosclerosis 2020; 315:102-110. [DOI: 10.1016/j.atherosclerosis.2020.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/30/2022]
|
28
|
Predictive assembling model reveals the self-adaptive elastic properties of lamellipodial actin networks for cell migration. Commun Biol 2020; 3:616. [PMID: 33106551 PMCID: PMC7588425 DOI: 10.1038/s42003-020-01335-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 09/30/2020] [Indexed: 12/14/2022] Open
Abstract
Branched actin network supports cell migration through extracellular microenvironments. However, it is unknown how intracellular proteins adapt the elastic properties of the network to the highly varying extracellular resistance. Here we develop a three-dimensional assembling model to simulate the realistic self-assembling process of the network by encompassing intracellular proteins and their dynamic interactions. Combining this multiscale model with finite element method, we reveal that the network can not only sense the variation of extracellular resistance but also self-adapt its elastic properties through remodeling with intracellular proteins. Such resistance-adaptive elastic behaviours are versatile and essential in supporting cell migration through varying extracellular microenvironments. The bending deformation mechanism and anisotropic Poisson's ratios determine why lamellipodia persistently evolve into sheet-like structures. Our predictions are confirmed by published experiments. The revealed self-adaptive elastic properties of the networks are also applicable to the endocytosis, phagocytosis, vesicle trafficking, intracellular pathogen transport and dendritic spine formation.
Collapse
|
29
|
Guak H, Krawczyk CM. Implications of cellular metabolism for immune cell migration. Immunology 2020; 161:200-208. [PMID: 32920838 DOI: 10.1111/imm.13260] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/25/2020] [Accepted: 09/02/2020] [Indexed: 12/15/2022] Open
Abstract
Cell migration is an essential, energetically demanding process in immunity. Immune cells navigate the body via chemokines and other immune mediators, which are altered under inflammatory conditions of injury or infection. Several factors determine the migratory abilities of different types of immune cells in diverse contexts, including the precise co-ordination of cytoskeletal remodelling, the expression of specific chemokine receptors and integrins, and environmental conditions. In this review, we present an overview of recent advances in our understanding of the relationship of each of these factors with cellular metabolism, with a focus on the spatial organization of glycolysis and mitochondria, reciprocal regulation of chemokine receptors and the influence of environmental changes.
Collapse
Affiliation(s)
- Hannah Guak
- Department of Physiology, McGill University, Montreal, QC, Canada.,Metabolic and Nutritional Programming Group, Van Andel Institute, Grand Rapids, MI, USA
| | - Connie M Krawczyk
- Metabolic and Nutritional Programming Group, Van Andel Institute, Grand Rapids, MI, USA
| |
Collapse
|
30
|
Garbett D, Bisaria A, Yang C, McCarthy DG, Hayer A, Moerner WE, Svitkina TM, Meyer T. T-Plastin reinforces membrane protrusions to bridge matrix gaps during cell migration. Nat Commun 2020; 11:4818. [PMID: 32968060 PMCID: PMC7511357 DOI: 10.1038/s41467-020-18586-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022] Open
Abstract
Migrating cells move across diverse assemblies of extracellular matrix (ECM) that can be separated by micron-scale gaps. For membranes to protrude and reattach across a gap, actin filaments, which are relatively weak as single filaments, must polymerize outward from adhesion sites to push membranes towards distant sites of new adhesion. Here, using micropatterned ECMs, we identify T-Plastin, one of the most ancient actin bundling proteins, as an actin stabilizer that promotes membrane protrusions and enables bridging of ECM gaps. We show that T-Plastin widens and lengthens protrusions and is specifically enriched in active protrusions where F-actin is devoid of non-muscle myosin II activity. Together, our study uncovers critical roles of the actin bundler T-Plastin to promote protrusions and migration when adhesion is spatially-gapped. In vivo, cells migrate across a diverse landscape of extracellular matrix containing gaps which present a challenge for cells to protrude across. Here, the authors show that T-Plastin strengthens protrusive actin networks to promote protrusion, extracellular matrix gap-bridging, and cell migration.
Collapse
Affiliation(s)
- Damien Garbett
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA.
| | - Anjali Bisaria
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Changsong Yang
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Arnold Hayer
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA.,Department of Biology, McGill University, Montréal, Canada
| | - W E Moerner
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Tatyana M Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Tobias Meyer
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA. .,Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
31
|
Xia T, Zhao R, Feng F, Yang L. The Effect of Matrix Stiffness on Human Hepatocyte Migration and Function-An In Vitro Research. Polymers (Basel) 2020; 12:polym12091903. [PMID: 32846973 PMCID: PMC7564768 DOI: 10.3390/polym12091903] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/08/2020] [Accepted: 08/18/2020] [Indexed: 01/30/2023] Open
Abstract
The extracellular matrix (ECM) regulates cellular function through the dynamic biomechanical and biochemical interplay between the resident cells and their microenvironment. Pathologically stiff ECM promotes phenotype changes in hepatocytes during liver fibrosis. To investigate the effect of ECM stiffness on hepatocyte migration and function, we designed an easy fabricated polyvinyl alcohol (PVA) hydrogel in which stiffness can be controlled by changing the concentration of glutaraldehyde. Three stiffnesses of hydrogels corresponding to the health of liver tissue, early stage, and end stage of fibrosis were selected. These were 4.8 kPa (soft), 21 kPa (moderate), and 45 kPa (stiff). For hepatocytes attachment, the hydrogel was coated with fibronectin. To evaluate the optimal concentration of fibronectin, hydrogel was coated with 0.1 mg/mL, 0.01 mg/mL, 0.005 mg/mL, or 0.003 mg/mL fibronectin, and the migratory behavior of single hepatocyte cultured on different concentrations of fibronectin was analyzed. To further explore the effect of substrate stiffness on hepatocyte migration, we used a stiffness controllable commercial 3D collagen gel, which has similar substrate stiffness to that of PVA hydrogel. Our result confirmed the PVA hydrogel biocompatibility with high hepatocytes survival. Fibronectin (0.01 mg/mL) promoted optimal migratory behavior for single hepatocytes. However, for confluent hepatocytes, a stiff substrate promoted hepatocellular migration compared with the soft and moderate groups via enhancing the formation of actin- and tubulin-rich structures. The gene expression analysis and protein expression analysis showed that the stiff substrate altered the phenotype of hepatocytes and induced apoptosis. Hepatocytes in stiff 3D hydrogel showed a higher proportion of cell death and expression of filopodia.
Collapse
Affiliation(s)
| | | | | | - Li Yang
- Correspondence: (T.X.); (L.Y.)
| |
Collapse
|
32
|
Actin polymerization downstream of integrins: signaling pathways and mechanotransduction. Biochem J 2020; 477:1-21. [PMID: 31913455 DOI: 10.1042/bcj20170719] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 11/17/2019] [Accepted: 12/16/2019] [Indexed: 01/04/2023]
Abstract
A cell constantly adapts to its environment. Cell decisions to survive, to proliferate or to migrate are dictated not only by soluble growth factors, but also through the direct interaction of the cell with the surrounding extracellular matrix (ECM). Integrins and their connections to the actin cytoskeleton are crucial for monitoring cell attachment and the physical properties of the substratum. Cell adhesion dynamics are modulated in complex ways by the polymerization of branched and linear actin arrays, which in turn reinforce ECM-cytoskeleton connection. This review describes the major actin regulators, Ena/VASP proteins, formins and Arp2/3 complexes, in the context of signaling pathways downstream of integrins. We focus on the specific signaling pathways that transduce the rigidity of the substrate and which control durotaxis, i.e. directed migration of cells towards increased ECM rigidity. By doing so, we highlight several recent findings on mechanotransduction and put them into a broad integrative perspective that is the result of decades of intense research on the actin cytoskeleton and its regulation.
Collapse
|
33
|
Topographic cues reveal filopodia-mediated cell locomotion in 3D microenvironment. Biointerphases 2020; 15:031001. [PMID: 32366106 DOI: 10.1116/1.5141051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
In cell-material interactions, the formation and functioning of filopodia have been demonstrated to be very sensitive to topographic cues. However, substrate-exploring functions of filopodia in a 3D microenvironment remain elusive. In this study, the silk fibroin film with a micropillar structure was prepared to reveal a filopodial-mediated cell response to 3D topographic cues. The micropillars provided a confined space for cell spreading by a simplified 3D structure, allowing initial cells to settle on the bottom of substrates rather than on the top of micropillars. Shortly after cell adhesion, the authors describe how cells transform from a filopodia-rich spherical cell state to a lamellipodia-dominated state that enables cell to climb along micropillars and spread on the top of the micropillars. The authors found that filopodia not only served as sensors for pathfinding but also provided nucleation scaffolds for the formation and orientation of minilamellipodia on the micropillar substrate. On the route of long filopodial extension following micropillars, all three functional filopodial adhesions have the ability to form veil-like minilamellipodium, simply by tethering the filopodium to the micropillars. Stable filopodia contacts consistently stimulated the local protrusion of a lamellipodium, which ultimately steered cell migration. Their results suggest the filopodia-mediated cell locomotion in the 3D microenvironment using a filopodia-to-minilamellipodium transformation mechanism.
Collapse
|
34
|
Matrix-transmitted paratensile signaling enables myofibroblast-fibroblast cross talk in fibrosis expansion. Proc Natl Acad Sci U S A 2020; 117:10832-10838. [PMID: 32358190 DOI: 10.1073/pnas.1910650117] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
While the concept of intercellular mechanical communication has been revealed, the mechanistic insights have been poorly evidenced in the context of myofibroblast-fibroblast interaction during fibrosis expansion. Here we report and systematically investigate the mechanical force-mediated myofibroblast-fibroblast cross talk via the fibrous matrix, which we termed paratensile signaling. Paratensile signaling enables instantaneous and long-range mechanotransduction via collagen fibers (less than 1 s over 70 μm) to activate a single fibroblast, which is intracellularly mediated by DDR2 and integrin signaling pathways in a calcium-dependent manner through the mechanosensitive Piezo1 ion channel. By correlating in vitro fibroblast foci growth models with mathematical modeling, we demonstrate that the single-cell-level spatiotemporal feature of paratensile signaling can be applied to elucidate the tissue-level fibrosis expansion and that blocking paratensile signaling can effectively attenuate the fibroblast to myofibroblast transition at the border of fibrotic and normal tissue. Our comprehensive investigation of paratensile signaling in fibrosis expansion broadens the understanding of cellular dynamics during fibrogenesis and inspires antifibrotic intervention strategies targeting paratensile signaling.
Collapse
|
35
|
How Actin Tracks Affect Myosin Motors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1239:183-197. [DOI: 10.1007/978-3-030-38062-5_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
|
36
|
Electromigration of cell surface macromolecules in DC electric fields during cell polarization and galvanotaxis. J Theor Biol 2019; 478:58-73. [DOI: 10.1016/j.jtbi.2019.06.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022]
|
37
|
Miao Q, Hill MC, Chen F, Mo Q, Ku AT, Ramos C, Sock E, Lefebvre V, Nguyen H. SOX11 and SOX4 drive the reactivation of an embryonic gene program during murine wound repair. Nat Commun 2019; 10:4042. [PMID: 31492871 PMCID: PMC6731344 DOI: 10.1038/s41467-019-11880-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 08/08/2019] [Indexed: 01/08/2023] Open
Abstract
Tissue injury induces changes in cellular identity, but the underlying molecular mechanisms remain obscure. Here, we show that upon damage in a mouse model, epidermal cells at the wound edge convert to an embryonic-like state, altering particularly the cytoskeletal/extracellular matrix (ECM) components and differentiation program. We show that SOX11 and its closest relative SOX4 dictate embryonic epidermal state, regulating genes involved in epidermal development as well as cytoskeletal/ECM organization. Correspondingly, postnatal induction of SOX11 represses epidermal terminal differentiation while deficiency of Sox11 and Sox4 accelerates differentiation and dramatically impairs cell motility and re-epithelialization. Amongst the embryonic genes reactivated at the wound edge, we identify fascin actin-bundling protein 1 (FSCN1) as a critical direct target of SOX11 and SOX4 regulating cell migration. Our study identifies the reactivated embryonic gene program during wound repair and demonstrates that SOX11 and SOX4 play a central role in this process.
Collapse
Affiliation(s)
- Qi Miao
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA.
- Center for Cell and Gene Therapy, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA.
| | - Matthew C Hill
- Program in Developmental Biology, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA
| | - Fengju Chen
- Dan L. Duncan Cancer Center, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA
| | - Qianxing Mo
- Dan L. Duncan Cancer Center, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA
- Department of Biostatistics & Bioinformatics, H. Lee Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA
| | - Amy T Ku
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA
| | - Carlos Ramos
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA
| | - Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, 91054, Erlangen, Germany
| | - Véronique Lefebvre
- Department of Surgery/Division of Orthopedic Surgery, Children's Hospital of Philadelphia, 3615 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Hoang Nguyen
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA.
- Center for Cell and Gene Therapy, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA.
- Program in Developmental Biology, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA.
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, BCM 505, Houston, TX, 77030, USA.
| |
Collapse
|
38
|
Alieva NO, Efremov AK, Hu S, Oh D, Chen Z, Natarajan M, Ong HT, Jégou A, Romet-Lemonne G, Groves JT, Sheetz MP, Yan J, Bershadsky AD. Myosin IIA and formin dependent mechanosensitivity of filopodia adhesion. Nat Commun 2019; 10:3593. [PMID: 31399564 PMCID: PMC6689027 DOI: 10.1038/s41467-019-10964-w] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 06/07/2019] [Indexed: 12/21/2022] Open
Abstract
Filopodia, dynamic membrane protrusions driven by polymerization of an actin filament core, can adhere to the extracellular matrix and experience both external and cell-generated pulling forces. The role of such forces in filopodia adhesion is however insufficiently understood. Here, we study filopodia induced by overexpression of myosin X, typical for cancer cells. The lifetime of such filopodia positively correlates with the presence of myosin IIA filaments at the filopodia bases. Application of pulling forces to the filopodia tips through attached fibronectin-coated laser-trapped beads results in sustained growth of the filopodia. Pharmacological inhibition or knockdown of myosin IIA abolishes the filopodia adhesion to the beads. Formin inhibitor SMIFH2, which causes detachment of actin filaments from formin molecules, produces similar effect. Thus, centripetal force generated by myosin IIA filaments at the base of filopodium and transmitted to the tip through actin core in a formin-dependent fashion is required for filopodia adhesion.
Collapse
Affiliation(s)
- N O Alieva
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - A K Efremov
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore.,Center for BioImaging Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117557, Singapore
| | - S Hu
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - D Oh
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Z Chen
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore.,Department of Chemistry, University of California, Berkeley, CA, 94720, USA
| | - M Natarajan
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - H T Ong
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - A Jégou
- Institut Jacques Monod, CNRS, Université de Paris, 15 rue Helene Brion, F-75013, Paris, France
| | - G Romet-Lemonne
- Institut Jacques Monod, CNRS, Université de Paris, 15 rue Helene Brion, F-75013, Paris, France
| | - J T Groves
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore.,Department of Chemistry, University of California, Berkeley, CA, 94720, USA
| | - M P Sheetz
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore.,Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | - J Yan
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore.,Center for BioImaging Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117557, Singapore.,Department of Physics, National University of Singapore, Singapore, 117542, Singapore
| | - A D Bershadsky
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore. .,Weizmann Institute of Science, Herzl St 234, Rehovot, 7610001, Israel.
| |
Collapse
|
39
|
Poli G, Ruggiero C, Cantini G, Canu L, Baroni G, Armignacco R, Jouinot A, Santi R, Ercolino T, Ragazzon B, Assie G, Mannelli M, Nesi G, Lalli E, Luconi M. Fascin-1 Is a Novel Prognostic Biomarker Associated With Tumor Invasiveness in Adrenocortical Carcinoma. J Clin Endocrinol Metab 2019; 104:1712-1724. [PMID: 30476173 DOI: 10.1210/jc.2018-01717] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/20/2018] [Indexed: 01/04/2023]
Abstract
CONTEXT Novel tumor markers are urgently needed to better stratify adrenocortical cancer (ACC) patients and improve therapies for this aggressive neoplasm. OBJECTIVE To assess the diagnostic and prognostic value of the actin-bundling protein fascin-1 (FSCN1) in adrenocortical tumors. DESIGN, SETTING AND PARTICIPANTS A local series of 37 malignant/37 benign adrenocortical tumors at Careggi University Hospital and two independent validation ACC cohorts (Cochin, TCGA) from the European Network for the Study of Adrenal Tumors were studied. MAIN OUTCOME MEASURES FSCN1 expression was quantified by immunohistochemistry, Western blot and quantitative RT-PCR in ACC specimens; overall and disease-free survival associated with FSCN1 expression were assessed by Kaplan-Meier analysis and compared with that of Ki67 labeling index and tumor stage. RESULTS Despite the low diagnostic power, in the Florence ACC series, FSCN1 immunohistochemical detection appeared as an independent prognostic factor, also refining results obtained with staging and Ki67 labeling index. The robust prognostic power of FSCN1 levels was further confirmed in two independent ACC cohorts. A positive correlation was found between FSCN1 and steroidogenic factor-1 (SF-1), with a substantially higher expression of both factors in ACCs at advanced stages and with at least one of the three Weiss score parameters associated with invasiveness. Moreover, we demonstrated FSCN1 role in promoting cell invasion in a human ACC cell line only in the case of increased SF-1 dosage. CONCLUSIONS These findings show that FSCN1 is a novel independent prognostic marker in ACC and may serve as a potential therapeutic target to block tumor spread.
Collapse
Affiliation(s)
- Giada Poli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Florence, Italy
| | - Carmen Ruggiero
- Université Côte d'Azur, Sophia Antipolis, Valbonne, France
- CNRS UMR7275, Sophia Antipolis, Valbonne, France
- NEOGENEX CNRS International Associated Laboratory, Sophia Antipolis, Valbonne, France
- Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Valbonne, France
| | - Giulia Cantini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Florence, Italy
| | - Letizia Canu
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Florence, Italy
- Careggi University Hospital (AOUC), Florence, Italy
| | - Gianna Baroni
- Careggi University Hospital (AOUC), Florence, Italy
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Roberta Armignacco
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Florence, Italy
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne Jouinot
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Department of Endocrinology Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Raffaella Santi
- Careggi University Hospital (AOUC), Florence, Italy
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | | | - Bruno Ragazzon
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Guillaume Assie
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Department of Endocrinology Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Reference Center for Rare Adrenal Diseases Reference Center for Rare Adrenal Cancer Network COMETE, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Massimo Mannelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Florence, Italy
- Careggi University Hospital (AOUC), Florence, Italy
| | - Gabriella Nesi
- Careggi University Hospital (AOUC), Florence, Italy
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Enzo Lalli
- Université Côte d'Azur, Sophia Antipolis, Valbonne, France
- CNRS UMR7275, Sophia Antipolis, Valbonne, France
- NEOGENEX CNRS International Associated Laboratory, Sophia Antipolis, Valbonne, France
- Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Valbonne, France
| | - Michaela Luconi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Florence, Italy
- Careggi University Hospital (AOUC), Florence, Italy
| |
Collapse
|
40
|
Tanaka M, Fujii Y, Hirano K, Higaki T, Nagasaki A, Ishikawa R, Okajima T, Katoh K. Fascin in lamellipodia contributes to cell elasticity by controlling the orientation of filamentous actin. Genes Cells 2019; 24:202-213. [PMID: 30664308 DOI: 10.1111/gtc.12671] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 12/27/2018] [Accepted: 12/28/2018] [Indexed: 11/30/2022]
Abstract
Fascin, an actin-bundling protein, is present in the filopodia and lamellipodia of growth cones. However, few studies have examined lamellipodial fascin because it is difficult to observe. In this study, we evaluated lamellipodial fascin. We visualized the actin meshwork of lamellipodia in live growth cones by super-resolution microscopy. Fascin was colocalized with the actin meshwork in lamellipodia. Ser39 of fascin is a well-known phosphorylation site that controls the binding of fascin to actin filaments. Fluorescence recovery after photobleaching experiments with confocal microscopy showed that binding of fascin was controlled by phosphorylation of Ser39 in lamellipodia. Moreover, TPA, an agonist of protein kinase C, induced phosphorylation of fascin and dissociation from actin filaments in lamellipodia. Time series images showed that dissociation of fascin from the actin meshwork was induced by TPA. As fascin dissociated from actin filaments, the orientation of the actin filaments became parallel to the leading edge. The angle of actin filaments against the leading edge was changed from 73° to 15°. This decreased the elasticity of the lamellipodia by 40%, as measured by atomic force microscopy. These data suggest that actin bundles made by fascin contribute to elasticity of the growth cone.
Collapse
Affiliation(s)
- Minami Tanaka
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Yuki Fujii
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Kazumi Hirano
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Takumi Higaki
- International Research Organization for Advanced Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Akira Nagasaki
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Ryoki Ishikawa
- School of Nursing, Gunma Prefectural College of Health Sciences, Maebashi, Japan
| | - Takaharu Okajima
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Kaoru Katoh
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| |
Collapse
|
41
|
McGuire S, Kara B, Hart PC, Montag A, Wroblewski K, Fazal S, Huang XY, Lengyel E, Kenny HA. Inhibition of fascin in cancer and stromal cells blocks ovarian cancer metastasis. Gynecol Oncol 2019; 153:405-415. [PMID: 30797592 DOI: 10.1016/j.ygyno.2019.01.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/15/2019] [Accepted: 01/20/2019] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Ovarian cancer (OvCa) metastasis requires the coordinated motility of both cancer and stromal cells. Cellular movement is a dynamic process that involves the synchronized assembly of f-actin bundles into cytoskeletal protrusions by fascin. Fascin directly binds f-actin and is an integral component of filopodia, lamellapodia and stress fibers. Here, we examine the expression pattern and function of fascin in the cancer and stromal cells of OvCa tumors. METHODS Fascin expression was evaluated in human cells and tissues using immunohistochemistry and immunofluorescence. The functional role of fascin in cancer and stromal cells was assessed with in vitro functional assays, an ex vivo colonization assay and in vivo metastasis assays using siRNA/shRNA and an inhibitor. The effect of fascin inhibition on Cdc42 and Rac1 activity was evaluated using GTPase activity assays and immunofluorescence. RESULTS Fascin expression was found to be higher in the stromal cell, when compared to the cancer cell, compartment of ovarian tumors. The low expression of fascin in the cancer cells of the primary tumor indicated a favorable prognosis for non-serous OvCa patients. In vitro, both knockdown and pharmacologic inhibition of fascin decreased the migration of cancer and stromal cells. The inhibition of fascin impaired Cdc42 and Rac1 activity in cancer cells, and cytoskeletal reorganization in the cancer and stromal cells. Inhibition of fascin ex vivo blocked OvCa cell colonization of human omental tissue and in vivo prevented and reduced OvCa metastases in mice. Likewise, knockdown of fascin specifically in the OvCa cells using a fascin-specific lentiviral-shRNA also blocked metastasis in vivo. CONCLUSION This study reveals the therapeutic potential of pharmacologically inhibiting fascin in both cancer and stromal cells of the OvCa tumor microenvironment.
Collapse
Affiliation(s)
- Sean McGuire
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, United States of America
| | - Betul Kara
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, United States of America
| | - Peter C Hart
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, United States of America
| | - Anthony Montag
- Department of Pathology, University of Chicago, Chicago, IL 60637, United States of America
| | - Kristen Wroblewski
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, United States of America
| | - Sarah Fazal
- Cellular Screening Center, University of Chicago, Chicago, IL 60637, United States of America
| | - Xin-Yun Huang
- Department of Physiology, Cornell University Weill Medical College, New York, NY 10065, United States of America
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, United States of America.
| | - Hilary A Kenny
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, United States of America.
| |
Collapse
|
42
|
Jacquemet G, Stubb A, Saup R, Miihkinen M, Kremneva E, Hamidi H, Ivaska J. Filopodome Mapping Identifies p130Cas as a Mechanosensitive Regulator of Filopodia Stability. Curr Biol 2019; 29:202-216.e7. [PMID: 30639111 PMCID: PMC6345628 DOI: 10.1016/j.cub.2018.11.053] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/25/2018] [Accepted: 11/20/2018] [Indexed: 01/09/2023]
Abstract
Filopodia are adhesive cellular protrusions specialized in the detection of extracellular matrix (ECM)-derived cues. Although ECM engagement at focal adhesions is known to trigger the recruitment of hundreds of proteins ("adhesome") to fine-tune cellular behavior, the components of the filopodia adhesions remain undefined. Here, we performed a structured-illumination-microscopy-based screen to map the localization of 80 target proteins, linked to cell adhesion and migration, within myosin-X-induced filopodia. We demonstrate preferential enrichment of several adhesion proteins to either filopodia tips, filopodia shafts, or shaft subdomains, suggesting divergent, spatially restricted functions for these proteins. Moreover, proteins with phosphoinositide (PI) binding sites are particularly enriched in filopodia. This, together with the strong localization of PI(3,4)P2 in filopodia tips, predicts critical roles for PIs in regulating filopodia ultra-structure and function. Our mapping further reveals that filopodia adhesions consist of a unique set of proteins, the filopodome, that are distinct from classical nascent adhesions, focal adhesions, and fibrillar adhesions. Using live imaging, we observe that filopodia adhesions can give rise to nascent adhesions, which, in turn, form focal adhesions. We demonstrate that p130Cas (BCAR1) is recruited to filopodia tips via its C-terminal Cas family homology domain (CCHD) and acts as a mechanosensitive regulator of filopodia stability. Finally, we demonstrate that our map based on myosin-X-induced filopodia can be translated to endogenous filopodia and fascin- and IRSp53-mediated filopodia.
Collapse
Affiliation(s)
- Guillaume Jacquemet
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.
| | - Aki Stubb
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Rafael Saup
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Mitro Miihkinen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Elena Kremneva
- Institute of Biotechnology, University of Helsinki, PO Box 56, 00014 Helsinki, Finland
| | - Hellyeh Hamidi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland; Department of Biochemistry, University of Turku, Turku, Finland.
| |
Collapse
|
43
|
Tvorogova A, Saidova A, Smirnova T, Vorobjev I. Dynamic microtubules drive fibroblast spreading. Biol Open 2018; 7:7/12/bio038968. [PMID: 30545950 PMCID: PMC6310885 DOI: 10.1242/bio.038968] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
When cells with a mesenchymal type of motility come into contact with an adhesive substrate they adhere and start spreading by the formation of lamellipodia. Using a label-free approach and virtual synchronization approach we analyzed spreading in fibroblasts and cancer cells. In all cell lines spreading is a non-linear process undergoing isotropic or anisotropic modes with first fast (5–20 min) and then slow (30–120 min) phases. In the first 10 min cell area increases 2–4 times, while the absolute rate of initial spreading decreases 2–8 times. Fast spreading depends on actin polymerization and dynamic microtubules. Inhibition of microtubule growth was sufficient for a slowdown of initial spreading. Inhibition of myosin II in the presence of stable microtubules restored fast spreading. Inhibition of actin polymerization or complete depolymerization of microtubules slowed down fast spreading. However, in these cases inhibition of myosin II only partially restored spreading kinetics. We conclude that rapid growth of microtubules towards cell margins at the first stage of cell spreading temporarily inhibits phosphorylation of myosin II and is essential for the fast isotropic spreading. Comparison of the fibroblasts with cancer cells shows that fast spreading in different cell types shares similar kinetics and mechanisms, and strongly depends on dynamic microtubules. Summary: Cell spreading is a non-linear process. The fast spreading phase depends on dynamic microtubules (MTs). Rapid growth of MTs towards the cell membrane promotes the temporal relaxation of acto-myosin contractility.
Collapse
Affiliation(s)
- Anna Tvorogova
- Department of Electron Microscopy, A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov State University, 1-40 Leninskie Gory, Moscow 119991, Russia
| | - Aleena Saidova
- Biological Faculty, M.V. Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow 119991, Russia
| | - Tatiana Smirnova
- Biological Faculty, M.V. Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow 119991, Russia
| | - Ivan Vorobjev
- Department of Electron Microscopy, A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov State University, 1-40 Leninskie Gory, Moscow 119991, Russia .,Biological Faculty, M.V. Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow 119991, Russia.,Department of Biology, School of Science and Technology, Nazarbayev University, Kabanbay Batyr ave. 53, Astana 010000, Kazakhstan
| |
Collapse
|
44
|
Saw TB, Xi W, Ladoux B, Lim CT. Biological Tissues as Active Nematic Liquid Crystals. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1802579. [PMID: 30156334 DOI: 10.1002/adma.201802579] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/11/2018] [Indexed: 05/27/2023]
Abstract
Live tissues can self-organize and be described as active materials composed of cells that generate active stresses through continuous injection of energy. In vitro reconstituted molecular networks, as well as single-cell cytoskeletons show that their filamentous structures can portray nematic liquid crystalline properties and can promote nonequilibrium processes induced by active processes at the microscale. The appearance of collective patterns, the formation of topological singularities, and spontaneous phase transition within the cell cytoskeleton are emergent properties that drive cellular functions. More integrated systems such as tissues have cells that can be seen as coarse-grained active nematic particles and their interaction can dictate many important tissue processes such as epithelial cell extrusion and migration as observed in vitro and in vivo. Here, a brief introduction to the concept of active nematics is provided, and the main focus is on the use of this framework in the systematic study of predominantly 2D tissue architectures and dynamics in vitro. In addition how the nematic state is important in tissue behavior, such as epithelial expansion, tissue homeostasis, and the atherosclerosis disease state, is discussed. Finally, how the nematic organization of cells can be controlled in vitro for tissue engineering purposes is briefly discussed.
Collapse
Affiliation(s)
- Thuan Beng Saw
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore, 117583, Singapore
| | - Wang Xi
- Institut Jacques Monod (IJM), CNRS UMR 7592 and Université Paris Diderot, Paris, France
| | - Benoit Ladoux
- Institut Jacques Monod (IJM), CNRS UMR 7592 and Université Paris Diderot, Paris, France
- Mechanobiology Institute (MBI), National University of Singapore, Singapore, 117411, Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore, 117583, Singapore
- Mechanobiology Institute (MBI), National University of Singapore, Singapore, 117411, Singapore
- Biomedical Institute for Global Health, Research and Technology (BIGHEART), National University of Singapore, MD6, 14 Medical Drive, #14-01, Singapore, 117599, Singapore
| |
Collapse
|
45
|
Young LE, Latario CJ, Higgs HN. Roles for Ena/VASP proteins in FMNL3-mediated filopodial assembly. J Cell Sci 2018; 131:131/21/jcs220814. [PMID: 30373894 DOI: 10.1242/jcs.220814] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/25/2018] [Indexed: 02/01/2023] Open
Abstract
Filopodia are actin-dependent finger-like structures that protrude from the plasma membrane. Actin filament barbed-end-binding proteins localized to filopodial tips are key to filopodial assembly. Two classes of barbed-end-binding proteins are formins and Ena/VASP proteins, and both classes have been localized to filopodial tips in specific cellular contexts. Here, we examine the filopodial roles of the FMNL formins and Ena/VASP proteins in U2OS cells. FMNL3 suppression reduces filopodial assembly by 90%, and FMNL3 is enriched at >95% of filopodial tips. Suppression of VASP or Mena (also known as ENAH) reduces filopodial assembly by >75%. However, VASP and Mena do not display consistent filopodial tip localization, but are enriched in focal adhesions (FAs). Interestingly, >85% of FMNL3-containing filopodia are associated with FAs. Two situations increase Ena/VASP filopodial localization: (1) expression of myosin-X, and (2) actively spreading cells. In spreading cells, filopodia often mark sites of nascent adhesions. Interestingly, VASP suppression in spreading cells causes a significant increase in adhesion assembly at filopodial tips. This work demonstrates that, in U2OS cells, Ena/VASP proteins play roles in filopodia beyond those at filopodial tips.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Lorna E Young
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| | - Casey J Latario
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| | - Henry N Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| |
Collapse
|
46
|
Gao W, Zhang C, Li W, Li H, Sang J, Zhao Q, Bo Y, Luo H, Zheng X, Lu Y, Shi Y, Yang D, Zhang R, Li Z, Cui J, Zhang Y, Niu M, Li J, Wu Z, Guo H, Xiang C, Wang J, Hou J, Zhang L, Thorne RF, Cui Y, Wu Y, Wen S, Wang B. Promoter Methylation-Regulated miR-145-5p Inhibits Laryngeal Squamous Cell Carcinoma Progression by Targeting FSCN1. Mol Ther 2018; 27:365-379. [PMID: 30341010 PMCID: PMC6369713 DOI: 10.1016/j.ymthe.2018.09.018] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 01/16/2023] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a common form of head and neck cancer with poor prognosis. However, the mechanism underlying the pathogenesis of LSCC remains unclear. Here, we demonstrated increased expression of fascin actin-bundling protein 1 (FSCN1) and decreased expression of microRNA-145-5p (miR-145-5p) in a clinical cohort of LSCC. Luciferase assay revealed that miR-145-5p is a negative regulator of FSCN1. Importantly, low miR-145-5p expression was correlated with TNM (tumor, node, metastasis) status and metastasis. Moreover, cases with low miR-145-5p/high FSCN1 expression showed poor prognosis, and these characteristics together served as independent prognostic indicators of survival. Gain- and loss-of-function studies showed that miR-145-5p overexpression or FSCN1 knockdown inhibited LSCC migration, invasion, and growth by suppressing the epithelial-mesenchymal transition along with inducing cell-cycle arrest and apoptosis. Additionally, hypermethylation of the miR-145-5p promoter suggested that repression of miR-145-5p arises through epigenetic inactivation. LSCC tumor growth in vivo could be inhibited by using miR-145-5p agomir or FSCN1 small interfering RNA (siRNA), which highlights the potential for clinical translation. Collectively, our findings indicate that miR-145-5p plays critical roles in inhibiting the progression of LSCC by suppressing FSCN1. Both miR-145-5p and FSCN1 are important potential prognostic markers and therapeutic targets for LSCC.
Collapse
Affiliation(s)
- Wei Gao
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Chunming Zhang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Wenqi Li
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Huizheng Li
- Department of Otolaryngology Head & Neck Surgery, Dalian Municipal Friendship Hospital, Dalian 116100, Liaoning, China
| | - Jiangwei Sang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Qinli Zhao
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Yunfeng Bo
- Department of Pathology, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Hongjie Luo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Xiwang Zheng
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Yan Lu
- Department of Otolaryngology Head & Neck Surgery, The First Hospital, Jinzhou Medical University, Jinzhou 121001, Liaoning, China
| | - Yong Shi
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Dongli Yang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Ruiping Zhang
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China; Department of MRI & CT, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Zhenyu Li
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China; Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030001, Shanxi, China
| | - Jiajia Cui
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Yuliang Zhang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Min Niu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Jun Li
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Zhongqiang Wu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Huina Guo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Caixia Xiang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Juan Wang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Juan Hou
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Lu Zhang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, School of Medicine, Henan University, Zhengzhou 450053, Henan, China; School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Yongping Cui
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Yongyan Wu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China.
| | - Shuxin Wen
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China.
| | - Binquan Wang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan 030001, Shanxi, China; Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan 030001, Shanxi, China; The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi Province, Taiyuan 030001, Shanxi, China.
| |
Collapse
|
47
|
Fischer RS, Lam PY, Huttenlocher A, Waterman CM. Filopodia and focal adhesions: An integrated system driving branching morphogenesis in neuronal pathfinding and angiogenesis. Dev Biol 2018; 451:86-95. [PMID: 30193787 DOI: 10.1016/j.ydbio.2018.08.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/08/2018] [Accepted: 08/29/2018] [Indexed: 12/31/2022]
Abstract
Single cell branching during development in vertebrates is typified by neuronal branching to form neurites and vascular branches formed by sprouting angiogenesis. Neurons and endothelial tip cells possess subcellular protrusions that share many common features from the morphological to the molecular level. Both systems utilize filopodia as their cellular protrusion organelles and depend on specific integrin-mediated adhesions to the local extracellular matrix for guidance in their pathfinding. We discuss the similar molecular machineries involved in these two types of cell branch formation and use their analogy to propose a new mechanism for angiogenic filopodia function, namely as adhesion assembly sites. In support of this model we provide primary data of angiogenesis in zebrafish in vivo showing that the actin assembly factor VASP participates in both filopodia formation and adhesion assembly at the base of the filopodia, enabling forward progress of the tip cell. The use of filopodia and their associated adhesions provide a common mechanism for neuronal and endothelial pathfinding during development in response to extracellular matrix cues.
Collapse
Affiliation(s)
- Robert S Fischer
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, United States
| | - Pui-Ying Lam
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, United States
| | - Anna Huttenlocher
- Departments of Pediatrics and Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, United States
| | - Clare M Waterman
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, United States.
| |
Collapse
|
48
|
Pakshir P, Hinz B. The big five in fibrosis: Macrophages, myofibroblasts, matrix, mechanics, and miscommunication. Matrix Biol 2018; 68-69:81-93. [DOI: 10.1016/j.matbio.2018.01.019] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 01/25/2018] [Accepted: 01/28/2018] [Indexed: 02/07/2023]
|
49
|
Wang C, Choi HJ, Kim SJ, Desai A, Lee N, Kim D, Bae Y, Lee K. Deconvolution of subcellular protrusion heterogeneity and the underlying actin regulator dynamics from live cell imaging. Nat Commun 2018; 9:1688. [PMID: 29703977 PMCID: PMC5923236 DOI: 10.1038/s41467-018-04030-0] [Citation(s) in RCA: 11] [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: 03/04/2017] [Accepted: 03/29/2018] [Indexed: 12/25/2022] Open
Abstract
Cell protrusion is morphodynamically heterogeneous at the subcellular level. However, the mechanism of cell protrusion has been understood based on the ensemble average of actin regulator dynamics. Here, we establish a computational framework called HACKS (deconvolution of heterogeneous activity in coordination of cytoskeleton at the subcellular level) to deconvolve the subcellular heterogeneity of lamellipodial protrusion from live cell imaging. HACKS identifies distinct subcellular protrusion phenotypes based on machine-learning algorithms and reveals their underlying actin regulator dynamics at the leading edge. Using our method, we discover "accelerating protrusion", which is driven by the temporally ordered coordination of Arp2/3 and VASP activities. We validate our finding by pharmacological perturbations and further identify the fine regulation of Arp2/3 and VASP recruitment associated with accelerating protrusion. Our study suggests HACKS can identify specific subcellular protrusion phenotypes susceptible to pharmacological perturbation and reveal how actin regulator dynamics are changed by the perturbation.
Collapse
Affiliation(s)
- Chuangqi Wang
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609, USA
| | - Hee June Choi
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609, USA
| | - Sung-Jin Kim
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609, USA
| | - Aesha Desai
- Department of Pathology and Anatomical Sciences, Computational Cell Biology, Anatomy and Pathology Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14203, USA
| | - Namgyu Lee
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Dohoon Kim
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Yongho Bae
- Department of Pathology and Anatomical Sciences, Computational Cell Biology, Anatomy and Pathology Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14203, USA
| | - Kwonmoo Lee
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609, USA.
| |
Collapse
|
50
|
Abstract
Cell migration directed by substrate-bound chemical cues is called haptotaxis. This study shows that grip and slip between the cell adhesion molecule (CAM) L1-CAM and the adhesive substrates, which occur asymmetrically under the growth cone, direct growth cone migration mediated by laminin. This mechanism is disrupted in a human patient of L1-CAM syndrome, suffering corpus callosum agenesis and corticospinal tract hypoplasia. These findings provide a conceptual framework for understanding the regulation and dysregulation of cell migration on the bases of force generation. Chemical cues presented on the adhesive substrate direct cell migration, a process termed haptotaxis. To migrate, cells must generate traction forces upon the substrate. However, how cells probe substrate-bound cues and generate directional forces for migration remains unclear. Here, we show that the cell adhesion molecule (CAM) L1-CAM is involved in laminin-induced haptotaxis of axonal growth cones. L1-CAM underwent grip and slip on the substrate. The ratio of the grip state was higher on laminin than on the control substrate polylysine; this was accompanied by an increase in the traction force upon laminin. Our data suggest that the directional force for laminin-induced growth cone haptotaxis is generated by the grip and slip of L1-CAM on the substrates, which occur asymmetrically under the growth cone. This mechanism is distinct from the conventional cell signaling models for directional cell migration. We further show that this mechanism is disrupted in a human patient with L1-CAM syndrome, suffering corpus callosum agenesis and corticospinal tract hypoplasia.
Collapse
|