1
|
Tao D, Wang H, Chang S, Cheng J, Da N, Zhang L, Yang J, Wang W, Xu F, Li B. Matrix Viscoelasticity Orchestrates Osteogenesis via Mechanotransduction Mediated Metabolic Switch in Macrophages. Adv Healthc Mater 2025; 14:e2405097. [PMID: 40042258 PMCID: PMC12023826 DOI: 10.1002/adhm.202405097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/22/2025] [Indexed: 04/26/2025]
Abstract
Understanding the interplay between extracellular matrix (ECM) mechanics and macrophage cellular processes is crucial for bone regeneration. While ECM stiffness has been extensively studied, the role of ECM viscoelasticity (e.g., stress relaxation) in the bone marrow niche and its effects on macrophage function remain unclear. Here, this study reveals how matrix viscoelasticity orchestrates osteogenesis by modulating macrophage metabolism through vasodilator-stimulated phosphoprotein (VASP) / hypoxia-inducible factor 1 alpha (HIF1α) signaling. In the rapid maxillary expansion (RME) model, significant stress relaxation occurs in regenerated bone marrow during the initial 17 days, coinciding with increased transforming growth factor-beta 1 (TGF-β1+) F4/80+ macrophages. Fast stress relaxation enhances macrophage recruitment of mesenchymal stem cells (MSCs) by upregulating TGF-β1. Using a hydrogel-macrophage system mimicking bone marrow viscoelasticity, cranial defect regeneration is significantly improved. Moreover, fast stress relaxation shifts macrophage metabolism from glycolysis to oxidative phosphorylation (OXPHOS) via VASP/HIF1α signaling, facilitating a reparative phenotype. These findings elucidate the relationship between ECM viscoelasticity and macrophage metabolism, suggesting new therapeutic avenues for bone regeneration through mechanomedicine.
Collapse
Affiliation(s)
- Dihao Tao
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseaseShaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'anShaanxi710049P. R. China
| | - Hanzhe Wang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseaseShaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'anShaanxi710049P. R. China
| | - Shiping Chang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseaseShaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'anShaanxi710049P. R. China
| | - Jiayu Cheng
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseaseShaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Ningning Da
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseaseShaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Li Zhang
- Department of ProsthodonticsNanjing Stomatological HospitalAffiliated Hospital of Medical SchoolInstitute of StomatologyNanjing UniversityNanjingJiangsu210000P. R. China
| | - Jianhua Yang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseaseShaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Wenzhe Wang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseaseShaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'anShaanxi710049P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationXi'an Jiaotong UniversityXi'anShaanxi710049P. R. China
| | - Bei Li
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseaseShaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| |
Collapse
|
2
|
Jacobs T, Isasti Sanchez J, Reger S, Luschnig S. Rho/Rok-dependent regulation of actomyosin contractility at tricellular junctions restricts epithelial permeability in Drosophila. Curr Biol 2025; 35:1181-1196.e5. [PMID: 39965573 DOI: 10.1016/j.cub.2025.01.043] [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: 10/03/2024] [Revised: 01/02/2025] [Accepted: 01/21/2025] [Indexed: 02/20/2025]
Abstract
Cell contacts in epithelia are remodeled to regulate paracellular permeability and to control the passage of migrating cells, but how barrier function is modulated while preserving epithelial integrity is not clear. In the follicular epithelium of Drosophila ovaries, tricellular junctions (TCJs) open transiently in a process termed patency to allow passage of externally produced yolk proteins for uptake by the oocyte. Here, we show that modulation of actomyosin contractility at cell vertices controls TCJ permeability. Before patency, circumferential actomyosin bundles are anchored at apical follicle cell vertices, where tension-sensing junctional proteins, Rho-associated kinase (Rok), and active myosin II accumulate and maintain vertices closed. TCJ opening is initiated by redistribution of myosin II from circumferential bundles to the medial zone, accompanied by decreasing tension on vertices. This transition requires activation of Cofilin-dependent filamentous actin (F-actin) disassembly by the phosphatase Slingshot and myosin II inactivation by myosin light-chain phosphatase and is counteracted by Rok. Accordingly, constitutive activation of myosin or of Rho signaling prevents vertex opening, whereas reduced myosin II or Rok activity causes excessive vertex opening. Thus, the opening of intercellular gaps in the follicular epithelium relies on relaxation of actomyosin contractility rather than active actomyosin-based pulling forces. Conversely, F-actin assembly is required for closing intercellular gaps after patency. Our findings are consistent with a force transduction model in which TCJ integrity is maintained by vertex-anchored contractile actomyosin. We propose that the cell-type-specific organization of actomyosin at cell vertices determines the mode of contractility-dependent regulation of epithelial permeability.
Collapse
Affiliation(s)
- Thea Jacobs
- Institute of Integrative Cell Biology and Physiology, Cells in Motion (CiM) Interfaculty Center, University of Münster, Röntgenstrasse 16, 48149 Münster, Germany
| | - Jone Isasti Sanchez
- Institute of Integrative Cell Biology and Physiology, Cells in Motion (CiM) Interfaculty Center, University of Münster, Röntgenstrasse 16, 48149 Münster, Germany
| | - Steven Reger
- Institute of Integrative Cell Biology and Physiology, Cells in Motion (CiM) Interfaculty Center, University of Münster, Röntgenstrasse 16, 48149 Münster, Germany
| | - Stefan Luschnig
- Institute of Integrative Cell Biology and Physiology, Cells in Motion (CiM) Interfaculty Center, University of Münster, Röntgenstrasse 16, 48149 Münster, Germany.
| |
Collapse
|
3
|
Ligunas GD, Paniagua GF, LaBelle J, Ramos-Martinez A, Shen K, Gerlt EH, Aguilar K, Nguyen N, Materna SC, Woo S. Tissue-specific and endogenous protein labeling with split fluorescent proteins. Dev Biol 2024; 514:109-116. [PMID: 38908500 PMCID: PMC11463824 DOI: 10.1016/j.ydbio.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
The ability to label proteins by fusion with genetically encoded fluorescent proteins is a powerful tool for understanding dynamic biological processes. However, current approaches for expressing fluorescent protein fusions possess drawbacks, especially at the whole organism level. Expression by transgenesis risks potential overexpression artifacts while fluorescent protein insertion at endogenous loci is technically difficult and, more importantly, does not allow for tissue-specific study of broadly expressed proteins. To overcome these limitations, we have adopted the split fluorescent protein system mNeonGreen21-10/11 (split-mNG2) to achieve tissue-specific and endogenous protein labeling in zebrafish. In our approach, mNG21-10 is expressed under a tissue-specific promoter using standard transgenesis while mNG211 is inserted into protein-coding genes of interest using CRISPR/Cas-directed gene editing. Each mNG2 fragment on its own is not fluorescent, but when co-expressed the fragments self-assemble into a fluorescent complex. Here, we report successful use of split-mNG2 to achieve differential labeling of the cytoskeleton genes tubb4b and krt8 in various tissues. We also demonstrate that by anchoring the mNG21-10 component to specific cellular compartments, the split-mNG2 system can be used to manipulate protein localization. Our approach should be broadly useful for a wide range of applications.
Collapse
Affiliation(s)
- Gloria D Ligunas
- Department of Molecular and Cell Biology, University of California, Merced, CA, USA; Quantitative and Systems Biology Graduate Group, University of California, Merced, CA, USA
| | - German F Paniagua
- Department of Molecular and Cell Biology, University of California, Merced, CA, USA
| | - Jesselynn LaBelle
- Department of Molecular and Cell Biology, University of California, Merced, CA, USA; Quantitative and Systems Biology Graduate Group, University of California, Merced, CA, USA
| | - Adela Ramos-Martinez
- Department of Molecular and Cell Biology, University of California, Merced, CA, USA
| | - Kyle Shen
- Department of Molecular and Cell Biology, University of California, Merced, CA, USA
| | - Emma H Gerlt
- Department of Molecular and Cell Biology, University of California, Merced, CA, USA
| | - Kaddy Aguilar
- Department of Molecular and Cell Biology, University of California, Merced, CA, USA
| | - Ngoc Nguyen
- Department of Molecular and Cell Biology, University of California, Merced, CA, USA
| | - Stefan C Materna
- Department of Molecular and Cell Biology, University of California, Merced, CA, USA; Quantitative and Systems Biology Graduate Group, University of California, Merced, CA, USA; Health Sciences Research Institute, University of California, Merced, CA, USA
| | - Stephanie Woo
- Department of Molecular and Cell Biology, University of California, Merced, CA, USA; Quantitative and Systems Biology Graduate Group, University of California, Merced, CA, USA; Health Sciences Research Institute, University of California, Merced, CA, USA.
| |
Collapse
|
4
|
Aydin F, Katkar HH, Morganthaler A, Harker AJ, Kovar DR, Voth GA. Prediction of the essential intermolecular contacts for side-binding of VASP on F-actin. Cytoskeleton (Hoboken) 2024; 81:382-392. [PMID: 38647032 PMCID: PMC11333183 DOI: 10.1002/cm.21864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Vasodilator-stimulated phosphoprotein (VASP) family proteins play a crucial role in mediating the actin network architecture in the cytoskeleton. The Ena/VASP homology 2 (EVH2) domain in each of the four identical arms of the tetrameric VASP consists of a loading poly-Pro region, a G-actin-binding domain (GAB), and an F-actin-binding domain (FAB). Together, the poly-Pro, GAB, and FAB domains allow VASP to bind to sides of actin filaments in a bundle, and recruit profilin-G-actin to processively elongate the filaments. The atomic resolution structure of the ternary complex, consisting of the loading poly-Pro region and GAB domain of VASP with profilin-actin, has been solved over a decade ago; however, a detailed structure of the FAB-F-actin complex has not been resolved to date. Experimental insights, based on homology of the FAB domain with the C region of WASP, have been used to hypothesize that the FAB domain binds to the cleft between subdomains 1 and 3 of F-actin. Here, in order to develop our understanding of the VASP-actin complex, we first augment known structural information about the GAB domain binding to actin with the missing FAB domain-actin structure, which we predict using homology modeling and docking simulations. In earlier work, we used mutagenesis and kinetic modeling to study the role of domain-level binding-unbinding kinetics of Ena/VASP on actin filaments in a bundle, specifically on the side of actin filaments. We further look at the nature of the side-binding of the FAB domain of VASP at the atomistic level using our predicted structure, and tabulate effective mutation sites on the FAB domain that would disrupt the VASP-actin complex. We test the binding affinity of Ena with mutated FAB domain using total internal reflection fluorescence microscopy experiments. The binding affinity of VASP is affected significantly for the mutant, providing additional support for our predicted structure.
Collapse
Affiliation(s)
- Fikret Aydin
- Department of Chemistry, The James Franck Institute, and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
| | - Harshwardhan H. Katkar
- Department of Chemistry, The James Franck Institute, and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
| | - Alisha Morganthaler
- Department of Biochemistry and Molecular Biology and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Alyssa J. Harker
- Department of Biochemistry and Molecular Biology and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - David R. Kovar
- Department of Biochemistry and Molecular Biology and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Gregory A. Voth
- Department of Chemistry, The James Franck Institute, and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
| |
Collapse
|
5
|
Engelhardt PM, Strippel J, Albat D, Chiha S, Rojas Pión J, Plein L, Kühne R, Müller M, Schmalz HG. C-Terminal Decarboxylation of Proline-Derived Building Blocks for Protein-Binding Peptides. Chemistry 2024; 30:e202401678. [PMID: 38770931 DOI: 10.1002/chem.202401678] [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: 04/29/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 05/22/2024]
Abstract
Using a set of conformationally restricted Proline-derived Modules (ProMs), our group has recently succeeded in developing inhibitors for the enabled/vasodilator-stimulated phosphoprotein homology 1 (EVH1) domain, which is a key mediator of cell migration and plays an important role in tumor metastasis. While these (formally) pentapeptidic compounds show nanomolecular binding affinities towards EVH1, their drug-like properties and cell permeability need to be further optimized before they can be clinically tested as therapeutic agents against metastasis. In this study, we sought to improve these properties by removing the C-terminal carboxylic acid function of our peptoids, either by late-stage decarboxylation or by direct synthesis. For late-stage decarboxylation of ProM-like systems, a method for reductive halo decarboxylation was optimized and applied to several proline-derived substrates. In this way, a series of new decarboxy ProMs suitable as building blocks for decarboxy EVH1 inhibitors were obtained. In addition, we incorporated decarboxy-ProM-1 into the pentapeptide-like compound Ac[2ClF][ProM-2][Decarb-ProM-1], which showed similar affinity towards EVH1 as the methyl ester derivative (Ac[2Cl-F][ProM-2][ProM1]OMe). However, despite better calculated drug-like properties, this compound did not inhibit chemotaxis in a cellular assay.
Collapse
Affiliation(s)
- Pascal M Engelhardt
- University of Cologne, Department of Chemistry, Greinstraße 4, 50939, Cologne, Germany
| | - Julian Strippel
- University of Cologne, Department of Chemistry, Greinstraße 4, 50939, Cologne, Germany
| | - Dominik Albat
- University of Cologne, Department of Chemistry, Greinstraße 4, 50939, Cologne, Germany
- Prosion Therapeutics GmbH, Luxemburger Str. 90, 50939, Köln, Germany
| | - Slim Chiha
- Prosion Therapeutics GmbH, Luxemburger Str. 90, 50939, Köln, Germany
| | | | - Laura Plein
- University of Cologne, Department of Chemistry, Greinstraße 4, 50939, Cologne, Germany
| | - Ronald Kühne
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Matthias Müller
- Prosion Therapeutics GmbH, Luxemburger Str. 90, 50939, Köln, Germany
| | - Hans-Günther Schmalz
- University of Cologne, Department of Chemistry, Greinstraße 4, 50939, Cologne, Germany
| |
Collapse
|
6
|
Phua DY, Sun X, Alushin GM. Force-activated zyxin assemblies coordinate actin nucleation and crosslinking to orchestrate stress fiber repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.594765. [PMID: 38798419 PMCID: PMC11118565 DOI: 10.1101/2024.05.17.594765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
As the cytoskeleton sustains cell and tissue forces, it incurs physical damage that must be repaired to maintain mechanical homeostasis. The LIM-domain protein zyxin detects force-induced ruptures in actin-myosin stress fibers, coordinating downstream repair factors to restore stress fiber integrity through unclear mechanisms. Here, we reconstitute stress fiber repair with purified proteins, uncovering detailed links between zyxin's force-regulated binding interactions and cytoskeletal dynamics. In addition to binding individual tensed actin filaments (F-actin), zyxin's LIM domains form force-dependent assemblies that bridge broken filament fragments. Zyxin assemblies engage repair factors through multi-valent interactions, coordinating nucleation of new F-actin by VASP and its crosslinking into aligned bundles by ɑ-actinin. Through these combined activities, stress fiber repair initiates within the cores of micron-scale damage sites in cells, explaining how these F-actin depleted regions are rapidly restored. Thus, zyxin's force-dependent organization of actin repair machinery inherently operates at the network scale to maintain cytoskeletal integrity.
Collapse
Affiliation(s)
- Donovan Y.Z. Phua
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Xiaoyu Sun
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Gregory M. Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
7
|
Chandrasekaran A, Graham K, Stachowiak JC, Rangamani P. Kinetic trapping organizes actin filaments within liquid-like protein droplets. Nat Commun 2024; 15:3139. [PMID: 38605007 PMCID: PMC11009352 DOI: 10.1038/s41467-024-46726-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 03/07/2024] [Indexed: 04/13/2024] Open
Abstract
Several actin-binding proteins (ABPs) phase separate to form condensates capable of curating the actin network shapes. Here, we use computational modeling to understand the principles of actin network organization within VASP condensate droplets. Our simulations reveal that the different actin shapes, namely shells, rings, and mixture states are highly dependent on the kinetics of VASP-actin interactions, suggesting that they arise from kinetic trapping. Specifically, we show that reducing the residence time of VASP on actin filaments reduces degree of bundling, thereby promoting assembly of shells rather than rings. We validate the model predictions experimentally using a VASP-mutant with decreased bundling capability. Finally, we investigate the ring opening within deformed droplets and found that the sphere-to-ellipsoid transition is favored under a wide range of filament lengths while the ellipsoid-to-rod transition is only permitted when filaments have a specific range of lengths. Our findings highlight key mechanisms of actin organization within phase-separated ABPs.
Collapse
Affiliation(s)
- Aravind Chandrasekaran
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, 92093-0411, USA
| | - Kristin Graham
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Jeanne C Stachowiak
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA.
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA.
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, 92093-0411, USA.
| |
Collapse
|
8
|
Ligunas GD, Paniagua G, LaBelle J, Ramos-Martinez A, Shen K, Gerlt EH, Aguilar K, Nguyen A, Materna SC, Woo S. Tissue-specific and endogenous protein labeling with split fluorescent proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.581822. [PMID: 38464062 PMCID: PMC10925240 DOI: 10.1101/2024.02.28.581822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The ability to label proteins by fusion with genetically encoded fluorescent proteins is a powerful tool for understanding dynamic biological processes. However, current approaches for expressing fluorescent protein fusions possess drawbacks, especially at the whole organism level. Expression by transgenesis risks potential overexpression artifacts while fluorescent protein insertion at endogenous loci is technically difficult and, more importantly, does not allow for tissue-specific study of broadly expressed proteins. To overcome these limitations, we have adopted the split fluorescent protein system mNeonGreen21-10/11 (split-mNG2) to achieve tissue-specific and endogenous protein labeling in zebrafish. In our approach, mNG21-10 is expressed under a tissue-specific promoter using standard transgenesis while mNG211 is inserted into protein-coding genes of interest using CRISPR/Cas-directed gene editing. Each mNG2 fragment on its own is not fluorescent, but when co-expressed the fragments self-assemble into a fluorescent complex. Here, we report successful use of split-mNG2 to achieve differential labeling of the cytoskeleton genes tubb4b and krt8 in various tissues. We also demonstrate that by anchoring the mNG21-10 component to specific cellular compartments, the split-mNG2 system can be used to manipulate protein function. Our approach should be broadly useful for a wide range of applications.
Collapse
Affiliation(s)
- Gloria D. Ligunas
- Department of Molecular Cell Biology, University of California, Merced, CA USA
- Quantitative and Systems Biology Graduate Group, University of California, Merced, CA USA
| | - German Paniagua
- Department of Molecular Cell Biology, University of California, Merced, CA USA
| | - Jesselynn LaBelle
- Department of Molecular Cell Biology, University of California, Merced, CA USA
- Quantitative and Systems Biology Graduate Group, University of California, Merced, CA USA
| | | | - Kyle Shen
- Department of Molecular Cell Biology, University of California, Merced, CA USA
| | - Emma H. Gerlt
- Department of Molecular Cell Biology, University of California, Merced, CA USA
| | - Kaddy Aguilar
- Department of Molecular Cell Biology, University of California, Merced, CA USA
| | - Alice Nguyen
- Department of Molecular Cell Biology, University of California, Merced, CA USA
| | - Stefan C. Materna
- Department of Molecular Cell Biology, University of California, Merced, CA USA
- Quantitative and Systems Biology Graduate Group, University of California, Merced, CA USA
- Health Sciences Research Institute, University of California, Merced, CA USA
| | - Stephanie Woo
- Department of Molecular Cell Biology, University of California, Merced, CA USA
- Quantitative and Systems Biology Graduate Group, University of California, Merced, CA USA
- Health Sciences Research Institute, University of California, Merced, CA USA
| |
Collapse
|
9
|
McCormick LE, Suarez C, Herring LE, Cannon KS, Kovar DR, Brown NG, Gupton SL. Multi-monoubiquitylation controls VASP-mediated actin dynamics. J Cell Sci 2024; 137:jcs261527. [PMID: 38277158 PMCID: PMC10917064 DOI: 10.1242/jcs.261527] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/13/2023] [Indexed: 01/13/2024] Open
Abstract
The actin cytoskeleton performs multiple cellular functions, and as such, actin polymerization must be tightly regulated. We previously demonstrated that reversible, non-degradative ubiquitylation regulates the function of the actin polymerase VASP in developing neurons. However, the underlying mechanism of how ubiquitylation impacts VASP activity was unknown. Here, we show that mimicking multi-monoubiquitylation of VASP at K240 and K286 negatively regulates VASP interactions with actin. Using in vitro biochemical assays, we demonstrate the reduced ability of multi-monoubiquitylated VASP to bind, bundle, and elongate actin filaments. However, multi-monoubiquitylated VASP maintained the ability to bind and protect barbed ends from capping protein. Finally, we demonstrate the electroporation of recombinant multi-monoubiquitylated VASP protein altered cell spreading morphology. Collectively, these results suggest a mechanism in which ubiquitylation controls VASP-mediated actin dynamics.
Collapse
Affiliation(s)
- Laura E. McCormick
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Cristian Suarez
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Laura E. Herring
- Michael Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kevin S. Cannon
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David R. Kovar
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Nicholas G. Brown
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Stephanie L. Gupton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
10
|
Gui J, Zhou H, Wan H, Yang D, Liu Q, Zhu L, Mi Y. The Role of Vasodilator-stimulated Phosphoproteins in the Development of Malignant Tumors. Curr Cancer Drug Targets 2024; 24:477-489. [PMID: 37962042 PMCID: PMC11092557 DOI: 10.2174/0115680096262439231023110106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/06/2023] [Accepted: 09/06/2023] [Indexed: 11/15/2023]
Abstract
Vasodilator-stimulated phosphoprotein (VASP) is an actin-binding protein that includes three structural domains: Enabled/VASP homolog1 (EVH1), EVH2, and proline-rich (PRR). VASP plays an important role in various cellular behaviors related to cytoskeletal regulation. More importantly, VASP plays a key role in the progression of several malignant tumors and is associated with malignant cell proliferation, invasion, and metastasis. Here, we have summarized current studies on the impact of VASP on the development of several malignant tumors and their mechanisms. This study provides a new theoretical basis for clinical molecular diagnosis and molecular targeted therapy.
Collapse
Affiliation(s)
- Jiandong Gui
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Hangsheng Zhou
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Hongyuan Wan
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Dongjie Yang
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Qing Liu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
- Huadong Sanatorium, 67 Dajishan, Wuxi 214122, Jiangsu Province, China
| | - Lijie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| |
Collapse
|
11
|
McCormick LE, Suarez C, Herring LE, Cannon KS, Kovar DR, Brown NG, Gupton SL. Multi-monoubiquitination controls VASP-mediated actin dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.16.549237. [PMID: 37503134 PMCID: PMC10370145 DOI: 10.1101/2023.07.16.549237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The actin cytoskeleton performs multiple cellular functions, and as such, actin polymerization must be tightly regulated. We previously demonstrated that reversible, non-degradative ubiquitination regulates the function of the actin polymerase VASP in developing neurons. However, the underlying mechanism of how ubiquitination impacts VASP activity was unknown. Here we show that mimicking multi-monoubiquitination of VASP at K240 and K286 negatively regulates VASP interactions with actin. Using in vitro biochemical assays, we demonstrate the reduced ability of multi-monoubiquitinated VASP to bind, bundle, and elongate actin filaments. However, multi-monoubiquitinated VASP maintained the ability to bind and protect barbed ends from capping protein. Lastly, we demonstrate the introduction of recombinant multi-monoubiquitinated VASP protein altered cell spreading morphology. Collectively, these results suggest a mechanism in which ubiquitination controls VASP-mediated actin dynamics.
Collapse
Affiliation(s)
- Laura E McCormick
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Cristian Suarez
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637
| | - Laura E Herring
- Michael Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Kevin S Cannon
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637
| | - Nicholas G Brown
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
12
|
Fang HY, Forghani R, Clarke A, McQueen PG, Chandrasekaran A, O’Neill KM, Losert W, Papoian GA, Giniger E. Enabled primarily controls filopodial morphology, not actin organization, in the TSM1 growth cone in Drosophila. Mol Biol Cell 2023; 34:ar83. [PMID: 37223966 PMCID: PMC10398877 DOI: 10.1091/mbc.e23-01-0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 05/25/2023] Open
Abstract
Ena/VASP proteins are processive actin polymerases that are required throughout animal phylogeny for many morphogenetic processes, including axon growth and guidance. Here we use in vivo live imaging of morphology and actin distribution to determine the role of Ena in promoting the growth of the TSM1 axon of the Drosophila wing. Altering Ena activity causes stalling and misrouting of TSM1. Our data show that Ena has a substantial impact on filopodial morphology in this growth cone but exerts only modest effects on actin distribution. This is in contrast to the main regulator of Ena, Abl tyrosine kinase, which was shown previously to have profound effects on actin and only mild effects on TSM1 growth cone morphology. We interpret these data as suggesting that the primary role of Ena in this axon may be to link actin to the morphogenetic processes of the plasma membrane, rather than to regulate actin organization itself. These data also suggest that a key role of Ena, acting downstream of Abl, may be to maintain consistent organization and reliable evolution of growth cone structure, even as Abl activity varies in response to guidance cues in the environment.
Collapse
Affiliation(s)
- Hsiao Yu Fang
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Rameen Forghani
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Akanni Clarke
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Philip G. McQueen
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Aravind Chandrasekaran
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20752
| | - Kate M. O’Neill
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
- Institute for Physical Sciences and Department of Physics, University of Maryland, College Park, MD 20752
| | - Wolfgang Losert
- Institute for Physical Sciences and Department of Physics, University of Maryland, College Park, MD 20752
| | - Garegin A. Papoian
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20752
| | - Edward Giniger
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
13
|
Benz PM, Frömel T, Laban H, Zink J, Ulrich L, Groneberg D, Boon RA, Poley P, Renne T, de Wit C, Fleming I. Cardiovascular Functions of Ena/VASP Proteins: Past, Present and Beyond. Cells 2023; 12:1740. [PMID: 37443774 PMCID: PMC10340426 DOI: 10.3390/cells12131740] [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: 04/26/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Actin binding proteins are of crucial importance for the spatiotemporal regulation of actin cytoskeletal dynamics, thereby mediating a tremendous range of cellular processes. Since their initial discovery more than 30 years ago, the enabled/vasodilator-stimulated phosphoprotein (Ena/VASP) family has evolved as one of the most fascinating and versatile family of actin regulating proteins. The proteins directly enhance actin filament assembly, but they also organize higher order actin networks and link kinase signaling pathways to actin filament assembly. Thereby, Ena/VASP proteins regulate dynamic cellular processes ranging from membrane protrusions and trafficking, and cell-cell and cell-matrix adhesions, to the generation of mechanical tension and contractile force. Important insights have been gained into the physiological functions of Ena/VASP proteins in platelets, leukocytes, endothelial cells, smooth muscle cells and cardiomyocytes. In this review, we summarize the unique and redundant functions of Ena/VASP proteins in cardiovascular cells and discuss the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Peter M. Benz
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
| | - Timo Frömel
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Hebatullah Laban
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Joana Zink
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Lea Ulrich
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Dieter Groneberg
- Institute of Physiology I, University of Würzburg, 97070 Würzburg, Germany
| | - Reinier A. Boon
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
- Cardiopulmonary Institute, 60596 Frankfurt am Main, Germany
- Centre of Molecular Medicine, Institute of Cardiovascular Regeneration, Goethe-University, 60596 Frankfurt am Main, Germany
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Centre, 1081 HZ Amsterdam, The Netherlands
| | - Philip Poley
- Institut für Physiologie, Universität zu Lübeck, 23562 Lübeck, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany
| | - Thomas Renne
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 VN51 Dublin, Ireland
| | - Cor de Wit
- Institut für Physiologie, Universität zu Lübeck, 23562 Lübeck, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
- Cardiopulmonary Institute, 60596 Frankfurt am Main, Germany
| |
Collapse
|
14
|
Parker SS, Ly KT, Grant AD, Sweetland J, Wang AM, Parker JD, Roman MR, Saboda K, Roe DJ, Padi M, Wolgemuth CW, Langlais P, Mouneimne G. EVL and MIM/MTSS1 regulate actin cytoskeletal remodeling to promote dendritic filopodia in neurons. J Cell Biol 2023; 222:e202106081. [PMID: 36828364 PMCID: PMC9998662 DOI: 10.1083/jcb.202106081] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/22/2022] [Accepted: 01/20/2023] [Indexed: 02/25/2023] Open
Abstract
Dendritic spines are the postsynaptic compartment of a neuronal synapse and are critical for synaptic connectivity and plasticity. A developmental precursor to dendritic spines, dendritic filopodia (DF), facilitate synapse formation by sampling the environment for suitable axon partners during neurodevelopment and learning. Despite the significance of the actin cytoskeleton in driving these dynamic protrusions, the actin elongation factors involved are not well characterized. We identified the Ena/VASP protein EVL as uniquely required for the morphogenesis and dynamics of DF. Using a combination of genetic and optogenetic manipulations, we demonstrated that EVL promotes protrusive motility through membrane-direct actin polymerization at DF tips. EVL forms a complex at nascent protrusions and DF tips with MIM/MTSS1, an I-BAR protein important for the initiation of DF. We proposed a model in which EVL cooperates with MIM to coalesce and elongate branched actin filaments, establishing the dynamic lamellipodia-like architecture of DF.
Collapse
Affiliation(s)
- Sara S. Parker
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Kenneth Tran Ly
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Adam D. Grant
- Cancer Biology Program, University of Arizona Cancer Center, Tucson, AZ, USA
| | - Jillian Sweetland
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Ashley M. Wang
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - James D. Parker
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Mackenzie R. Roman
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Kathylynn Saboda
- University of Arizona Cancer Center and Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Denise J. Roe
- University of Arizona Cancer Center and Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Megha Padi
- Cancer Biology Program, University of Arizona Cancer Center, Tucson, AZ, USA
- Department of Molecular and Cellular Biology, College of Science, University of Arizona, Tucson, AZ, USA
| | - Charles W. Wolgemuth
- University of Arizona Cancer Center and Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
- Department of Molecular and Cellular Biology, College of Science, University of Arizona, Tucson, AZ, USA
- Department of Physics, College of Science, University of Arizona, Tucson, AZ, USA
- Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, USA
| | - Paul Langlais
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Ghassan Mouneimne
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
- Cancer Biology Program, University of Arizona Cancer Center, Tucson, AZ, USA
| |
Collapse
|
15
|
Mukherjee D, Previs RA, Haines CN, Abo MA, Juras PK, Strickland KC, Chakraborty B, Artham S, Whitaker R, Hebert KL, Fontenot J, Patierno SR, Freedman JA, Lau FH, Burow M, Chang CY, McDonnell DP. Ca 2+ /Calmodulin Dependent Protein Kinase Kinase-2 (CaMKK2) promotes Protein Kinase G (PKG)-dependent actin cytoskeletal assembly to increase tumor metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.536051. [PMID: 37131673 PMCID: PMC10153149 DOI: 10.1101/2023.04.17.536051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Triple-negative breast cancers (TNBCs) tend to become highly invasive early during cancer development. Despite some successes in the initial treatment of patients diagnosed with early-stage localized TNBC, the rate of metastatic recurrence remains high with poor long-term survival outcomes. Here we show that elevated expression of the serine/threonine-kinase, Calcium/Calmodulin (CaM)-dependent protein kinase kinase-2 (CaMKK2), is highly correlated with tumor invasiveness. We determined that genetic disruption of CaMKK2 expression, or inhibition of its activity, disrupted spontaneous metastatic outgrowth from primary tumors in murine xenograft models of TNBC. High-grade serous ovarian cancer (HGSOC), a high-risk, poor-prognosis ovarian cancer subtype, shares many genetic features with TNBC, and importantly, CaMKK2 inhibition effectively blocked metastatic progression in a validated xenograft model of this disease. Probing the mechanistic links between CaMKK2 and metastasis we defined the elements of a new signaling pathway that impacts actin cytoskeletal dynamics in a manner which increases cell migration/invasion and metastasis. Notably, CaMKK2 increases the expression of the phosphodiesterase PDE1A which decreases the cGMP-dependent activity of protein kinase G1 (PKG1). This inhibition of PKG1 results in decreased phosphorylation of Vasodilator-Stimulated Phosphoprotein (VASP), which in its hypophosphorylated state binds to and regulates F-actin assembly to facilitate contraction/cell movement. Together, these data establish a targetable CaMKK2-PDE1A-PKG1-VASP signaling pathway that controls cancer cell motility and metastasis. Further, it credentials CaMKK2 as a therapeutic target that can be exploited in the discovery of agents for use in the neoadjuvant/adjuvant setting to restrict tumor invasiveness in patients diagnosed with early-stage TNBC or localized HGSOC.
Collapse
|
16
|
Pokrant T, Hein JI, Körber S, Disanza A, Pich A, Scita G, Rottner K, Faix J. Ena/VASP clustering at microspike tips involves lamellipodin but not I-BAR proteins, and absolutely requires unconventional myosin-X. Proc Natl Acad Sci U S A 2023. [PMID: 36598940 DOI: 10.1101/2022.05.12.491613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
Sheet-like membrane protrusions at the leading edge, termed lamellipodia, drive 2D-cell migration using active actin polymerization. Microspikes comprise actin-filament bundles embedded within lamellipodia, but the molecular mechanisms driving their formation and their potential functional relevance have remained elusive. Microspike formation requires the specific activity of clustered Ena/VASP proteins at their tips to enable processive actin assembly in the presence of capping protein, but the factors and mechanisms mediating Ena/VASP clustering are poorly understood. Systematic analyses of B16-F1 melanoma mutants lacking potential candidate proteins revealed that neither inverse BAR-domain proteins, nor lamellipodin or Abi is essential for clustering, although they differentially contribute to lamellipodial VASP accumulation. In contrast, unconventional myosin-X (MyoX) identified here as proximal to VASP was obligatory for Ena/VASP clustering and microspike formation. Interestingly, and despite the invariable distribution of other relevant marker proteins, the width of lamellipodia in MyoX-KO mutants was significantly reduced as compared with B16-F1 control, suggesting that microspikes contribute to lamellipodium stability. Consistently, MyoX removal caused marked defects in protrusion and random 2D-cell migration. Strikingly, Ena/VASP-deficiency also uncoupled MyoX cluster dynamics from actin assembly in lamellipodia, establishing their tight functional association in microspike formation.
Collapse
Affiliation(s)
- Thomas Pokrant
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Jens Ingo Hein
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Sarah Körber
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Andrea Disanza
- IFOM ETS (Istituto Fondazione di Oncologia Molecolare ETS), - The AIRC (Italian Association for Cancer Research) Institute of Molecular Oncology, 20139 Milan, Italy
| | - Andreas Pich
- Research Core Unit Proteomics, Hannover Medical School, 30625 Hannover, Germany
| | - Giorgio Scita
- IFOM ETS (Istituto Fondazione di Oncologia Molecolare ETS), - The AIRC (Italian Association for Cancer Research) Institute of Molecular Oncology, 20139 Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, 20139 Milan, Italy
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany
- Molecular Cell Biology Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
17
|
Yun S, Cha SS, Kim JH. DJ-1 promotes cell migration by interacting with Mena, the mammalian homolog of Drosophila enabled. Adv Biol Regul 2022; 88:100943. [PMID: 36542983 DOI: 10.1016/j.jbior.2022.100943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/26/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
DJ-1 has gained extensive attention after being identified in 2003 as a protein implicated in the pathogenesis of early-onset Parkinson's disease. Since then, efforts have revealed versatile DJ-1 functions in reactive oxygen species (ROS) control, transcriptional regulation, chaperone function, fertility, and cell transformation. Herein, we report a novel function of DJ-1 in actin cytoskeletal rearrangements. DJ-1 was identified as a new binding partner of Mena, a protein of the Enah/VASP family, and it promoted cancer cell migration by Mena-dependent actin polymerization and filopodia formation. These results suggest a novel molecular mechanism for DJ-1-dependent cancer cell invasion and metastasis.
Collapse
Affiliation(s)
- Sanguk Yun
- Department of Biotechnology, Inje University, Gimhae, 50834, Republic of Korea.
| | - Sun-Shin Cha
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, 50612, Yangsan, Republic of Korea.
| |
Collapse
|
18
|
Montaño-Rendón F, Walpole GF, Krause M, Hammond GR, Grinstein S, Fairn GD. PtdIns(3,4)P2, Lamellipodin, and VASP coordinate actin dynamics during phagocytosis in macrophages. J Cell Biol 2022; 221:e202207042. [PMID: 36165850 PMCID: PMC9521245 DOI: 10.1083/jcb.202207042] [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: 07/20/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/22/2022] Open
Abstract
Phosphoinositides are pivotal regulators of vesicular traffic and signaling during phagocytosis. Phagosome formation, the initial step of the process, is characterized by local membrane remodeling and reorganization of the actin cytoskeleton that leads to formation of the pseudopods that drive particle engulfment. Using genetically encoded fluorescent probes, we found that upon particle engagement a localized pool of PtdIns(3,4)P2 is generated by the sequential activities of class I phosphoinositide 3-kinases and phosphoinositide 5-phosphatases. Depletion of this locally generated pool of PtdIns(3,4)P2 blocks pseudopod progression and ultimately phagocytosis. We show that the PtdIns(3,4)P2 effector Lamellipodin (Lpd) is recruited to nascent phagosomes by PtdIns(3,4)P2. Furthermore, we show that silencing of Lpd inhibits phagocytosis and produces aberrant pseudopodia with disorganized actin filaments. Finally, vasodilator-stimulated phosphoprotein (VASP) was identified as a key actin-regulatory protein mediating phagosome formation downstream of Lpd. Mechanistically, our findings imply that a pathway involving PtdIns(3,4)P2, Lpd, and VASP mediates phagocytosis at the stage of particle engulfment.
Collapse
Affiliation(s)
- Fernando Montaño-Rendón
- Division of Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Glenn F.W. Walpole
- Division of Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Matthias Krause
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, UK
| | - Gerald R.V. Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Sergio Grinstein
- Division of Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Gregory D. Fairn
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
19
|
Visweshwaran SP, Nayab H, Hoffmann L, Gil M, Liu F, Kühne R, Maritzen T. Ena/VASP proteins at the crossroads of actin nucleation pathways in dendritic cell migration. Front Cell Dev Biol 2022; 10:1008898. [PMID: 36274843 PMCID: PMC9581539 DOI: 10.3389/fcell.2022.1008898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/12/2022] [Indexed: 12/05/2022] Open
Abstract
As sentinels of our immune system dendritic cells (DCs) rely on efficient cell migration for patrolling peripheral tissues and delivering sampled antigens to secondary lymphoid organs for the activation of T-cells. Dynamic actin polymerization is key to their macropinocytic and migratory properties. Both major actin nucleation machineries, formins and the Arp2/3 complex, are critical for different aspects of DC functionality, by driving the generation of linear and branched actin filaments, respectively. However, the importance of a third group of actin nucleators, the Ena/VASP family, has not been addressed yet. Here, we show that the two family members Evl and VASP are expressed in murine DCs and that their loss negatively affects DC macropinocytosis, spreading, and migration. Our interactome analysis reveals Ena/VASP proteins to be ideally positioned for orchestrating the different actin nucleation pathways by binding to the formin mDia1 as well as to the WAVE regulatory complex, a stimulator of Arp2/3. In fact, Evl/VASP deficient murine DCs are more vulnerable to inhibition of Arp2/3 demonstrating that Ena/VASP proteins contribute to the robustness and efficiency of DC migration.
Collapse
Affiliation(s)
| | - Hafiza Nayab
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Lennart Hoffmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Marine Gil
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Fan Liu
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Ronald Kühne
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Tanja Maritzen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
- Department of Nanophysiology, Technische Universität Kaiserslautern, Kaiserslautern, Germany
- *Correspondence: Tanja Maritzen,
| |
Collapse
|
20
|
Chen TH, Garnir K, Chen CY, Jian CB, Gao HD, Cheng B, Tseng MC, Moucheron C, Kirsch-De Mesmaeker A, Lee HM. A Toolkit for Engineering Proteins in Living Cells: Peptide with a Tryptophan-Selective Ru-TAP Complex to Regioselectively Photolabel Specific Proteins. J Am Chem Soc 2022; 144:18117-18125. [PMID: 36135325 DOI: 10.1021/jacs.2c08342] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Using a chemical approach to crosslink functionally versatile bioeffectors (such as peptides) to native proteins of interest (POI) directly inside a living cell is a useful toolbox for chemical biologists. However, this goal has not been reached due to unsatisfactory chemoselectivity, regioselectivity, and protein selectivity in protein labeling within living cells. Herein, we report the proof of concept of a cytocompatible and highly selective photolabeling strategy using a tryptophan-specific Ru-TAP complex as a photocrosslinker. Aside from the high selectivity, the photolabeling is blue light-driven by a photoinduced electron transfer (PeT) and allows the bioeffector to bear an additional UV-responsive unit. The two different photosensitivities are demonstrated by blue light-photocrosslinking a UV-sensitive peptide to POI. Our visible light photolabeling can generate photocaged proteins for subsequent activity manipulation by UV light. Cytoskeletal dynamics regulation is demonstrated in living cells via the unprecedented POI photomanipulation and proves that our methodology opens a new avenue to endogenous protein modification.
Collapse
Affiliation(s)
- Tzu-Ho Chen
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei 11529, Taiwan.,Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.,Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Kevin Garnir
- Laboratoire de Chimie Organique et Photochimie CP160/08, Université libre de Bruxelles, 50 Av. Franklin D. Roosevelt, 1050 Brussels, Belgium
| | - Chong-Yan Chen
- Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Cheng-Bang Jian
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.,Nano Science and Technology Program, Taiwan International Graduate Program, Academia Sinica, Taipei 11529, Taiwan.,Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Hua-De Gao
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.,Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Bill Cheng
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung 40227, Taiwan
| | - Mei-Chun Tseng
- Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Cécile Moucheron
- Laboratoire de Chimie Organique et Photochimie CP160/08, Université libre de Bruxelles, 50 Av. Franklin D. Roosevelt, 1050 Brussels, Belgium
| | - Andrée Kirsch-De Mesmaeker
- Laboratoire de Chimie Organique et Photochimie CP160/08, Université libre de Bruxelles, 50 Av. Franklin D. Roosevelt, 1050 Brussels, Belgium
| | - Hsien-Ming Lee
- Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
21
|
Faix J, Rottner K. Ena/VASP proteins in cell edge protrusion, migration and adhesion. J Cell Sci 2022; 135:274697. [DOI: 10.1242/jcs.259226] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
ABSTRACT
The tightly coordinated, spatiotemporal control of actin filament remodeling provides the basis of fundamental cellular processes, such as cell migration and adhesion. Specific protein assemblies, composed of various actin-binding proteins, are thought to operate in these processes to nucleate and elongate new filaments, arrange them into complex three-dimensional (3D) arrays and recycle them to replenish the actin monomer pool. Actin filament assembly is not only necessary to generate pushing forces against the leading edge membrane or to propel pathogens through the cytoplasm, but also coincides with the generation of stress fibers (SFs) and focal adhesions (FAs) that generate, transmit and sense mechanical tension. The only protein families known to date that directly enhance the elongation of actin filaments are formins and the family of Ena/VASP proteins. Their mechanisms of action, however, in enhancing processive filament elongation are distinct. The aim of this Review is to summarize our current knowledge on the molecular mechanisms of Ena/VASP-mediated actin filament assembly, and to discuss recent insights into the cell biological functions of Ena/VASP proteins in cell edge protrusion, migration and adhesion.
Collapse
Affiliation(s)
- Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technical University Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
- Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| |
Collapse
|
22
|
Borriello L, Coste A, Traub B, Sharma VP, Karagiannis GS, Lin Y, Wang Y, Ye X, Duran CL, Chen X, Friedman M, Sosa MS, Sun D, Dalla E, Singh DK, Oktay MH, Aguirre-Ghiso JA, Condeelis JS, Entenberg D. Primary tumor associated macrophages activate programs of invasion and dormancy in disseminating tumor cells. Nat Commun 2022; 13:626. [PMID: 35110548 PMCID: PMC8811052 DOI: 10.1038/s41467-022-28076-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 01/07/2022] [Indexed: 02/07/2023] Open
Abstract
Metastases are initiated by disseminated tumor cells (DTCs) that colonize distant organs. Growing evidence suggests that the microenvironment of the primary tumor primes DTCs for dormant or proliferative fates. However, the manner in which this occurs remains poorly understood. Here, using the Window for High-Resolution Intravital Imaging of the Lung (WHRIL), we study the live lung longitudinally and follow the fate of individual DTCs that spontaneously disseminate from orthotopic breast tumors. We find that spontaneously DTCs have increased levels of retention, increased speed of extravasation, and greater survival after extravasation, compared to experimentally metastasized tumor cells. Detailed analysis reveals that a subset of macrophages within the primary tumor induces a pro-dissemination and pro-dormancy DTC phenotype. Our work provides insight into how specific primary tumor microenvironments prime a subpopulation of cells for expression of proteins associated with dissemination and dormancy.
Collapse
Affiliation(s)
- Lucia Borriello
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Anouchka Coste
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Brian Traub
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Ved P Sharma
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - George S Karagiannis
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Department of Microbiology and Immunology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Cancer Dormancy and Tumor Microenvironment Institute and, Einstein Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Yu Lin
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Yarong Wang
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Xianjun Ye
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Camille L Duran
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Xiaoming Chen
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Madeline Friedman
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Maria Soledad Sosa
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dan Sun
- Cancer Dormancy and Tumor Microenvironment Institute and, Einstein Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine/Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Erica Dalla
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deepak K Singh
- Cancer Dormancy and Tumor Microenvironment Institute and, Einstein Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine/Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Maja H Oktay
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Cancer Dormancy and Tumor Microenvironment Institute and, Einstein Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
- Department of Pathology, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Julio A Aguirre-Ghiso
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Cancer Dormancy and Tumor Microenvironment Institute and, Einstein Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
- Department of Cell Biology, Albert Einstein College of Medicine/Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Cancer Dormancy and Tumor Microenvironment Institute and, Einstein Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| | - David Entenberg
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Cancer Dormancy and Tumor Microenvironment Institute and, Einstein Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
- Department of Pathology, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
23
|
Yu-Kemp HC, Szymanski RA, Cortes DB, Gadda NC, Lillich ML, Maddox AS, Peifer M. Micron-scale supramolecular myosin arrays help mediate cytoskeletal assembly at mature adherens junctions. J Cell Biol 2022; 221:212872. [PMID: 34812842 PMCID: PMC8614156 DOI: 10.1083/jcb.202103074] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 09/28/2021] [Accepted: 10/14/2021] [Indexed: 01/19/2023] Open
Abstract
Epithelial cells assemble specialized actomyosin structures at E-Cadherin–based cell–cell junctions, and the force exerted drives cell shape change during morphogenesis. The mechanisms that build this supramolecular actomyosin structure remain unclear. We used ZO-knockdown MDCK cells, which assemble a robust, polarized, and highly organized actomyosin cytoskeleton at the zonula adherens, combining genetic and pharmacologic approaches with superresolution microscopy to define molecular machines required. To our surprise, inhibiting individual actin assembly pathways (Arp2/3, formins, or Ena/VASP) did not prevent or delay assembly of this polarized actomyosin structure. Instead, as junctions matured, micron-scale supramolecular myosin arrays assembled, with aligned stacks of myosin filaments adjacent to the apical membrane, overlying disorganized actin filaments. This suggested that myosin arrays might bundle actin at mature junctions. Consistent with this idea, inhibiting ROCK or myosin ATPase disrupted myosin localization/organization and prevented actin bundling and polarization. We obtained similar results in Caco-2 cells. These results suggest a novel role for myosin self-assembly, helping drive actin organization to facilitate cell shape change.
Collapse
Affiliation(s)
- Hui-Chia Yu-Kemp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Rachel A Szymanski
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Daniel B Cortes
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Nicole C Gadda
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Madeline L Lillich
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Amy S Maddox
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC.,Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Mark Peifer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC.,Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
24
|
Hwang T, Parker SS, Hill SM, Ilunga MW, Grant RA, Mouneimne G, Keating AE. A distributed residue network permits conformational binding specificity in a conserved family of actin remodelers. eLife 2021; 10:e70601. [PMID: 34854809 PMCID: PMC8639148 DOI: 10.7554/elife.70601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/08/2021] [Indexed: 11/23/2022] Open
Abstract
Metazoan proteomes contain many paralogous proteins that have evolved distinct functions. The Ena/VASP family of actin regulators consists of three members that share an EVH1 interaction domain with a 100 % conserved binding site. A proteome-wide screen revealed photoreceptor cilium actin regulator (PCARE) as a high-affinity ligand for ENAH EVH1. Here, we report the surprising observation that PCARE is ~100-fold specific for ENAH over paralogs VASP and EVL and can selectively bind ENAH and inhibit ENAH-dependent adhesion in cells. Specificity arises from a mechanism whereby PCARE stabilizes a conformation of the ENAH EVH1 domain that is inaccessible to family members VASP and EVL. Structure-based modeling rapidly identified seven residues distributed throughout EVL that are sufficient to differentiate binding by ENAH vs. EVL. By exploiting the ENAH-specific conformation, we rationally designed the tightest and most selective ENAH binder to date. Our work uncovers a conformational mechanism of interaction specificity that distinguishes highly similar paralogs and establishes tools for dissecting specific Ena/VASP functions in processes including cancer cell invasion.
Collapse
Affiliation(s)
- Theresa Hwang
- Department of Biology, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Sara S Parker
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of ArizonaTucsonUnited States
| | - Samantha M Hill
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of ArizonaTucsonUnited States
| | - Meucci W Ilunga
- Department of Biology, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Robert A Grant
- Department of Biology, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Ghassan Mouneimne
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of ArizonaTucsonUnited States
| | - Amy E Keating
- Department of Biology, Massachusetts Institute of TechnologyCambridgeUnited States
- Department of Biological Engineering and Koch Institue for Integrative Cancer Research, Massachusetts Institute of TechnologyCambridgeUnited States
| |
Collapse
|
25
|
Mascarenhas JB, Gaber AA, Larrinaga TM, Mayfield R, Novak S, Camp SM, Gregorio C, Jacobson JR, Cress AE, Dudek SM, Garcia JGN. EVL is a novel focal adhesion protein involved in the regulation of cytoskeletal dynamics and vascular permeability. Pulm Circ 2021; 11:20458940211049002. [PMID: 34631011 PMCID: PMC8493322 DOI: 10.1177/20458940211049002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022] Open
Abstract
Increases in lung vascular permeability is a cardinal feature of inflammatory disease and represents an imbalance in vascular contractile forces and barrier-restorative forces, with both forces highly dependent upon the actin cytoskeleton. The current study investigates the role of Ena-VASP-like (EVL), a member of the Ena-VASP family known to regulate the actin cytoskeleton, in regulating vascular permeability responses and lung endothelial cell barrier integrity. Utilizing changes in transendothelial electricial resistance (TEER) to measure endothelial cell barrier responses, we demonstrate that EVL expression regulates endothelial cell responses to both sphingosine-1-phospate (S1P), a vascular barrier-enhancing agonist, and to thrombin, a barrier-disrupting stimulus. Total internal reflection fluorescence demonstrates that EVL is present in endothelial cell focal adhesions and impacts focal adhesion size, distribution, and the number of focal adhesions generated in response to S1P and thrombin challenge, with the focal adhesion kinase (FAK) a key contributor in S1P-stimulated EVL-transduced endothelial cell but a limited role in thrombin-induced focal adhesion rearrangements. In summary, these data indicate that EVL is a focal adhesion protein intimately involved in regulation of cytoskeletal responses to endothelial cell barrier-altering stimuli. Keywords: cytoskeleton, vascular barrier, sphingosine-1-phosphate, thrombin, focal adhesion kinase (FAK), Ena-VASP like protein (EVL), cytoskeletal regulatory protein
Collapse
Affiliation(s)
| | - Amir A Gaber
- Department of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Tania M Larrinaga
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Rachel Mayfield
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Stefanie Novak
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Sara M Camp
- Department of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Carol Gregorio
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Jeffrey R Jacobson
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Anne E Cress
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Steven M Dudek
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Joe G N Garcia
- Department of Medicine, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
26
|
Augustin V, Kins S. Fe65: A Scaffolding Protein of Actin Regulators. Cells 2021; 10:cells10071599. [PMID: 34202290 PMCID: PMC8304848 DOI: 10.3390/cells10071599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 01/19/2023] Open
Abstract
The scaffolding protein family Fe65, composed of Fe65, Fe65L1, and Fe65L2, was identified as an interaction partner of the amyloid precursor protein (APP), which plays a key function in Alzheimer’s disease. All three Fe65 family members possess three highly conserved interaction domains, forming complexes with diverse binding partners that can be assigned to different cellular functions, such as transactivation of genes in the nucleus, modulation of calcium homeostasis and lipid metabolism, and regulation of the actin cytoskeleton. In this article, we rule out putative new intracellular signaling mechanisms of the APP-interacting protein Fe65 in the regulation of actin cytoskeleton dynamics in the context of various neuronal functions, such as cell migration, neurite outgrowth, and synaptic plasticity.
Collapse
|
27
|
Davidson AJ, Wood W. Macrophages Use Distinct Actin Regulators to Switch Engulfment Strategies and Ensure Phagocytic Plasticity In Vivo. Cell Rep 2021; 31:107692. [PMID: 32460022 PMCID: PMC7262594 DOI: 10.1016/j.celrep.2020.107692] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 04/16/2020] [Accepted: 05/05/2020] [Indexed: 01/02/2023] Open
Abstract
Macrophages must not only be responsive to an array of different stimuli, such as infection and cellular damage, but also perform phagocytosis within the diverse and complex tissue environments found in vivo. This requires a high degree of morphological and therefore cytoskeletal plasticity. Here, we use the exceptional genetics and in vivo imaging of Drosophila embryos to study macrophage phagocytic versatility during apoptotic corpse clearance. We find that macrophage phagocytosis is highly robust, arising from their possession of two distinct modes of engulfment that utilize exclusive suites of actin-regulatory proteins. “Lamellipodial phagocytosis” is Arp2/3-complex-dependent and allows cells to migrate toward and envelop apoptotic corpses. Alternatively, Diaphanous and Ena drive filopodial phagocytosis to reach out and draw in debris. Macrophages switch to “filopodial phagocytosis” to overcome spatial constraint, providing the robust plasticity necessary to ensure that whatever obstacle they encounter in vivo, they fulfil their critical clearance function. Macrophages use two distinct modes of engulfment: lamellipodial and filopodial phagocytosis Arp2/3-complex-dependent lamellipodial phagocytosis involves envelopment via a lamellipod Filopodial phagocytosis involves phagocytic filopods extended by Dia and/or Ena Macrophages switch to filopodial phagocytosis to overcome spatial constraint in vivo
Collapse
Affiliation(s)
- Andrew J Davidson
- Centre for Inflammation Research, University of Edinburgh, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Will Wood
- Centre for Inflammation Research, University of Edinburgh, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
28
|
Pearce S, Tucker CL. Dual Systems for Enhancing Control of Protein Activity through Induced Dimerization Approaches. Adv Biol (Weinh) 2021; 5:e2000234. [PMID: 34028215 PMCID: PMC8144547 DOI: 10.1002/adbi.202000234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/29/2020] [Indexed: 12/25/2022]
Abstract
To reveal the underpinnings of complex biological systems, a variety of approaches have been developed that allow switchable control of protein function. One powerful approach for switchable control is the use of inducible dimerization systems, which can be configured to control activity of a target protein upon induced dimerization triggered by chemicals or light. Individually, many inducible dimerization systems suffer from pre-defined dynamic ranges and overwhelming sensitivity to expression level and cellular context. Such systems often require extensive engineering efforts to overcome issues of background leakiness and restricted dynamic range. To address these limitations, recent tool development efforts have explored overlaying dimerizer systems with a second layer of regulation. Albeit more complex, the resulting layered systems have enhanced functionality, such as tighter control that can improve portability of these tools across platforms.
Collapse
Affiliation(s)
- Sarah Pearce
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, 80045, Colorado, USA
| | - Chandra L. Tucker
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, 80045, Colorado, USA
| |
Collapse
|
29
|
Brabec JL, Lara MK, Tyler AL, Mahoney JM. System-Level Analysis of Alzheimer's Disease Prioritizes Candidate Genes for Neurodegeneration. Front Genet 2021; 12:625246. [PMID: 33889174 PMCID: PMC8056044 DOI: 10.3389/fgene.2021.625246] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is a debilitating neurodegenerative disorder. Since the advent of the genome-wide association study (GWAS) we have come to understand much about the genes involved in AD heritability and pathophysiology. Large case-control meta-GWAS studies have increased our ability to prioritize weaker effect alleles, while the recent development of network-based functional prediction has provided a mechanism by which we can use machine learning to reprioritize GWAS hits in the functional context of relevant brain tissues like the hippocampus and amygdala. In parallel with these developments, groups like the Alzheimer’s Disease Neuroimaging Initiative (ADNI) have compiled rich compendia of AD patient data including genotype and biomarker information, including derived volume measures for relevant structures like the hippocampus and the amygdala. In this study we wanted to identify genes involved in AD-related atrophy of these two structures, which are often critically impaired over the course of the disease. To do this we developed a combined score prioritization method which uses the cumulative distribution function of a gene’s functional and positional score, to prioritize top genes that not only segregate with disease status, but also with hippocampal and amygdalar atrophy. Our method identified a mix of genes that had previously been identified in AD GWAS including APOE, TOMM40, and NECTIN2(PVRL2) and several others that have not been identified in AD genetic studies, but play integral roles in AD-effected functional pathways including IQSEC1, PFN1, and PAK2. Our findings support the viability of our novel combined score as a method for prioritizing region- and even cell-specific AD risk genes.
Collapse
Affiliation(s)
- Jeffrey L Brabec
- Department of Neurological Sciences, University of Vermont, Burlington, VT, United States
| | - Montana Kay Lara
- Department of Neurological Sciences, University of Vermont, Burlington, VT, United States
| | - Anna L Tyler
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - J Matthew Mahoney
- Department of Neurological Sciences, University of Vermont, Burlington, VT, United States.,The Jackson Laboratory, Bar Harbor, ME, United States
| |
Collapse
|
30
|
Higuchi M, Ishiyama K, Maruoka M, Kanamori R, Takaori-Kondo A, Watanabe N. Paradoxical activation of c-Src as a drug-resistant mechanism. Cell Rep 2021; 34:108876. [PMID: 33761359 DOI: 10.1016/j.celrep.2021.108876] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/29/2020] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
ATP-competitive inhibitors have been developed as promising anti-cancer agents. However, drug-resistance frequently occurs, and the underlying mechanisms are not fully understood. Here, we show that the activation of c-Src and its downstream phosphorylation cascade can be paradoxically induced by Src-targeted and RTK-targeted kinase inhibitors. We reveal that inhibitor binding induces a conformational change in c-Src, leading to the association of the active form c-Src with focal adhesion kinase (FAK). Reduction of the inhibitor concentration results in the dissociation of inhibitors from the c-Src-FAK complex, which allows c-Src to phosphorylate FAK and initiate FAK-Grb2-mediated Erk signaling. Furthermore, a drug-resistant mutation in c-Src, which reduces the affinity of inhibitors for c-Src, converts Src inhibitors into facilitators of cell proliferation by enhancing the phosphorylation of FAK and Erk in c-Src-mutated cells. Our data thus reveal paradoxical enhancement of cell growth evoked by target-based kinase inhibitors, providing potentially important clues for the future development of effective and safe cancer treatment.
Collapse
Affiliation(s)
- Makio Higuchi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenichi Ishiyama
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Hematology and Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masahiro Maruoka
- Laboratory of Single-Molecule Cell Biology, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Ryosuke Kanamori
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Naoki Watanabe
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Kyoto, Japan.
| |
Collapse
|
31
|
King TR, Kramer J, Cheng YS, Swope D, Kramer SG. Enabled/VASP is required to mediate proper sealing of opposing cardioblasts during Drosophila dorsal vessel formation. Dev Dyn 2021; 250:1173-1190. [PMID: 33587326 DOI: 10.1002/dvdy.317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 01/16/2021] [Accepted: 02/02/2021] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION The Drosophila dorsal vessel (DV) is comprised of two opposing rows of cardioblasts (CBs) that migrate toward the dorsal midline during development. While approaching the midline, CBs change shape, enabling dorsal and ventral attachments with their contralateral partners to create a linear tube with a central lumen. We previously demonstrated DV closure occurs via a "buttoning" mechanism where specific CBs advance ahead of their lateral neighbors, and attach creating transient holes, which eventually seal. RESULTS Here, we investigate the role of the actin-regulatory protein enabled (Ena) in DV closure. Loss of Ena results in DV cell shape and alignment defects. Live analysis of DV formation in ena mutants shows a reduction in CB leading edge protrusion length and gaps in the DV between contralateral CB pairs. These gaps occur primarily between a specific genetic subtype of CBs, which express the transcription factor seven-up (Svp) and form the ostia inflow tracts of the heart. In WT embryos these gaps between Svp+ CBs are observed transiently during the final stages of DV closure. CONCLUSIONS Our data suggest that Ena modulates the actin cytoskeleton in order to facilitate the complete sealing of the DV during the final stages of cardiac tube formation.
Collapse
Affiliation(s)
- Tiffany R King
- Graduate Program in Cell and Developmental Biology, Rutgers Graduate School of Biomedical Sciences at Robert Wood Johnson Medical School, Department of Pathology and Laboratory Medicine, Piscataway, New Jersey, USA.,Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joseph Kramer
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Yi-Shan Cheng
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - David Swope
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, USA.,Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, New Jersey, USA
| | - Sunita G Kramer
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| |
Collapse
|
32
|
Melchionna R, Trono P, Tocci A, Nisticò P. Actin Cytoskeleton and Regulation of TGFβ Signaling: Exploring Their Links. Biomolecules 2021; 11:biom11020336. [PMID: 33672325 PMCID: PMC7926735 DOI: 10.3390/biom11020336] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/15/2021] [Accepted: 02/20/2021] [Indexed: 12/14/2022] Open
Abstract
Human tissues, to maintain their architecture and function, respond to injuries by activating intricate biochemical and physical mechanisms that regulates intercellular communication crucial in maintaining tissue homeostasis. Coordination of the communication occurs through the activity of different actin cytoskeletal regulators, physically connected to extracellular matrix through integrins, generating a platform of biochemical and biomechanical signaling that is deregulated in cancer. Among the major pathways, a controller of cellular functions is the cytokine transforming growth factor β (TGFβ), which remains a complex and central signaling network still to be interpreted and explained in cancer progression. Here, we discuss the link between actin dynamics and TGFβ signaling with the aim of exploring their aberrant interaction in cancer.
Collapse
Affiliation(s)
- Roberta Melchionna
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, via Chianesi 53, 00144 Rome, Italy; (R.M.); (P.T.); (A.T.)
| | - Paola Trono
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, via Chianesi 53, 00144 Rome, Italy; (R.M.); (P.T.); (A.T.)
- Institute of Biochemistry and Cell Biology, National Research Council, via Ramarini 32, 00015 Monterotondo Scalo, Rome, Italy
| | - Annalisa Tocci
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, via Chianesi 53, 00144 Rome, Italy; (R.M.); (P.T.); (A.T.)
| | - Paola Nisticò
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, via Chianesi 53, 00144 Rome, Italy; (R.M.); (P.T.); (A.T.)
- Correspondence: ; Tel.: +39-0652662539
| |
Collapse
|
33
|
Bleicher P, Nast-Kolb T, Sciortino A, de la Trobe YA, Pokrant T, Faix J, Bausch AR. Intra-bundle contractions enable extensile properties of active actin networks. Sci Rep 2021; 11:2677. [PMID: 33514794 PMCID: PMC7846802 DOI: 10.1038/s41598-021-81601-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/05/2021] [Indexed: 12/27/2022] Open
Abstract
The cellular cortex is a dynamic and contractile actomyosin network modulated by actin-binding proteins. We reconstituted a minimal cortex adhered to a model cell membrane mimicking two processes mediated by the motor protein myosin: contractility and high turnover of actin monomers. Myosin reorganized these networks by extensile intra‑bundle contractions leading to an altered growth mechanism. Hereby, stress within tethered bundles induced nicking of filaments followed by repair via incorporation of free monomers. This mechanism was able to break the symmetry of the previously disordered network resulting in the generation of extensile clusters, reminiscent of structures found within cells.
Collapse
Affiliation(s)
- P Bleicher
- Physik-Department, Lehrstuhl für Biophysik E27, Technische Universität München, Garching, Germany.
- Center for Protein Assemblies (CPA), Ernst-Otto-Fischer Str. 8, 85747, Garching, Germany.
| | - T Nast-Kolb
- Physik-Department, Lehrstuhl für Biophysik E27, Technische Universität München, Garching, Germany
- Center for Protein Assemblies (CPA), Ernst-Otto-Fischer Str. 8, 85747, Garching, Germany
| | - A Sciortino
- Physik-Department, Lehrstuhl für Biophysik E27, Technische Universität München, Garching, Germany
- Center for Protein Assemblies (CPA), Ernst-Otto-Fischer Str. 8, 85747, Garching, Germany
| | - Y A de la Trobe
- Physik-Department, Lehrstuhl für Biophysik E27, Technische Universität München, Garching, Germany
- Center for Protein Assemblies (CPA), Ernst-Otto-Fischer Str. 8, 85747, Garching, Germany
| | - T Pokrant
- Institut für Biophysikalische Chemie, Medizinische Hochschule Hannover, Hannover, Germany
| | - J Faix
- Institut für Biophysikalische Chemie, Medizinische Hochschule Hannover, Hannover, Germany
| | - A R Bausch
- Physik-Department, Lehrstuhl für Biophysik E27, Technische Universität München, Garching, Germany.
- Center for Protein Assemblies (CPA), Ernst-Otto-Fischer Str. 8, 85747, Garching, Germany.
| |
Collapse
|
34
|
Designed nanomolar small-molecule inhibitors of Ena/VASP EVH1 interaction impair invasion and extravasation of breast cancer cells. Proc Natl Acad Sci U S A 2020; 117:29684-29690. [PMID: 33184177 PMCID: PMC7703624 DOI: 10.1073/pnas.2007213117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Protein–protein interactions mediated by proline-rich motifs are involved in regulation of many important signaling cascades. These motifs belong to the most abundant recognition motifs in the eukaryotic genome and preferentially adopt a left-handed polyproline helix II, a secondary structure element that has been notoriously difficult to mimic with small molecules. Here, we present a structure-guided design effort yielding a toolkit of chemical entities that enables rational construction of selective small molecule inhibitors for these protein domains. We succeeded in developing an inhibitor for the Ena/VASP protein family that is active in vivo and reduces extravasation of invasive breast cancer cells in a zebrafish model. Battling metastasis through inhibition of cell motility is considered a promising approach to support cancer therapies. In this context, Ena/VASP-depending signaling pathways, in particular interactions with their EVH1 domains, are promising targets for pharmaceutical intervention. However, protein–protein interactions involving proline-rich segments are notoriously difficult to address by small molecules. Hence, structure-based design efforts in combination with the chemical synthesis of additional molecular entities are required. Building on a previously developed nonpeptidic micromolar inhibitor, we determined 22 crystal structures of ENAH EVH1 in complex with inhibitors and rationally extended our library of conformationally defined proline-derived modules (ProMs) to succeed in developing a nanomolar inhibitor (Kd=120 nM,MW=734 Da). In contrast to the previous inhibitor, the optimized compounds reduced extravasation of invasive breast cancer cells in a zebrafish model. This study represents an example of successful, structure-guided development of low molecular weight inhibitors specifically and selectively addressing a proline-rich sequence-recognizing domain that is characterized by a shallow epitope lacking defined binding pockets. The evolved high-affinity inhibitor may now serve as a tool in validating the basic therapeutic concept, i.e., the suppression of cancer metastasis by inhibiting a crucial protein–protein interaction involved in actin filament processing and cell migration.
Collapse
|
35
|
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: 66] [Impact Index Per Article: 13.2] [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
|
36
|
Cheng KW, Mullins RD. Initiation and disassembly of filopodia tip complexes containing VASP and lamellipodin. Mol Biol Cell 2020; 31:2021-2034. [PMID: 32579429 PMCID: PMC7543071 DOI: 10.1091/mbc.e20-04-0270] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The shapes of many eukaryotic cells depends on the actin cytoskeleton, and changes in actin assembly dynamics underlie many changes in cell shape. Ena/VASP-family actin polymerases, for example, modulate cell shape by accelerating actin filament assembly locally and slowing filament capping. When concentrated into discrete foci at the leading edge, VASP promotes filopodia assembly and forms part of a poorly understood molecular complex that remains associated with growing filopodia tips. Here we identify precursors of this filopodia tip complex in migrating B16F1 cells: small leading-edge clusters of the adaptor protein lamellipodin (Lpd) that subsequently recruit VASP and initiate filopodia formation. Dimerization, membrane association, and VASP binding are all required for lamellipodin to incorporate into filopodia tip complexes, and overexpression of monomeric, membrane-targeted lamellipodin mutants disrupts tip complex assembly. Once formed, tip complexes containing VASP and lamellipodin grow by fusing with each other, but their growth is limited by a size-dependent dynamic instability. Our results demonstrate that assembly and disassembly dynamics of filopodia tip complexes are determined, in part, by a network of multivalent interactions between Ena/VASP proteins, EVH1 ligands, and actin filaments.
Collapse
Affiliation(s)
- Karen W Cheng
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94143
| | - R Dyche Mullins
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94143.,Howard Hughes Medical Institute, University of California, San Francisco, CA 94143
| |
Collapse
|
37
|
Lamb MC, Anliker KK, Tootle TL. Fascin regulates protrusions and delamination to mediate invasive, collective cell migration in vivo. Dev Dyn 2020; 249:961-982. [PMID: 32352613 DOI: 10.1002/dvdy.186] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/26/2020] [Accepted: 04/23/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The actin bundling protein Fascin is essential for developmental cell migrations and promotes cancer metastasis. In addition to bundling actin, Fascin has several actin-independent roles; how these other functions contribute to cell migration remains unclear. Border cell migration during Drosophila oogenesis provides an excellent model to study Fascin's various roles during invasive, collective cell migration. RESULTS On-time border cell migration during Stage 9 requires Fascin (Drosophila Singed). Fascin functions not only within the migrating border cells, but also within the nurse cells, the substrate for this migration. Fascin genetically interacts with the actin elongation factor Enabled to promote on-time Stage 9 migration and overexpression of Enabled suppresses the defects seen with loss of Fascin. Loss of Fascin results in increased, shorter and mislocalized protrusions during migration. Additionally, loss of Fascin inhibits border cell delamination and increases E-Cadherin (Drosophila Shotgun) adhesions on both the border cells and nurse cells. CONCLUSIONS Overall, Fascin promotes on-time border cell migration during Stage 9 and contributes to multiple aspects of this invasive, collective cell migration, including both protrusion dynamics and delamination. These findings have implications beyond Drosophila, as border cell migration has emerged as a model to study mechanisms mediating cancer metastasis.
Collapse
Affiliation(s)
- Maureen C Lamb
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Kelsey K Anliker
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Tina L Tootle
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
38
|
Skruber K, Warp PV, Shklyarov R, Thomas JD, Swanson MS, Henty-Ridilla JL, Read TA, Vitriol EA. Arp2/3 and Mena/VASP Require Profilin 1 for Actin Network Assembly at the Leading Edge. Curr Biol 2020; 30:2651-2664.e5. [PMID: 32470361 DOI: 10.1016/j.cub.2020.04.085] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/09/2020] [Accepted: 04/29/2020] [Indexed: 12/27/2022]
Abstract
Cells have many types of actin structures, which must assemble from a common monomer pool. Yet, it remains poorly understood how monomers are distributed to and shared between different filament networks. Simplified model systems suggest that monomers are limited and heterogeneous, which alters actin network assembly through biased polymerization and internetwork competition. However, less is known about how monomers influence complex actin structures, where different networks competing for monomers overlap and are functionally interdependent. One example is the leading edge of migrating cells, which contains filament networks generated by multiple assembly factors. The leading edge dynamically switches between the formation of different actin structures, such as lamellipodia or filopodia, by altering the balance of these assembly factors' activities. Here, we sought to determine how the monomer-binding protein profilin 1 (PFN1) controls the assembly and organization of actin in mammalian cells. Actin polymerization in PFN1 knockout cells was severely disrupted, particularly at the leading edge, where both Arp2/3 and Mena/VASP-based filament assembly was inhibited. Further studies showed that in the absence of PFN1, Arp2/3 no longer localizes to the leading edge and Mena/VASP is non-functional. Additionally, we discovered that discrete stages of internetwork competition and collaboration between Arp2/3 and Mena/VASP networks exist at different PFN1 concentrations. Low levels of PFN1 caused filopodia to form exclusively at the leading edge, while higher concentrations inhibited filopodia and favored lamellipodia and pre-filopodia bundles. These results demonstrate that dramatic changes to actin architecture can be made simply by modifying PFN1 availability.
Collapse
Affiliation(s)
- Kristen Skruber
- Department of Anatomy and Cell Biology, University of Florida, College of Medicine, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Peyton V Warp
- Department of Anatomy and Cell Biology, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Rachael Shklyarov
- Department of Anatomy and Cell Biology, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - James D Thomas
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics, and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics, and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Jessica L Henty-Ridilla
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, NY 13210, USA
| | - Tracy-Ann Read
- Department of Anatomy and Cell Biology, University of Florida, College of Medicine, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Eric A Vitriol
- Department of Anatomy and Cell Biology, University of Florida, College of Medicine, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, Gainesville, FL 32610, USA.
| |
Collapse
|
39
|
Damiano-Guercio J, Kurzawa L, Mueller J, Dimchev G, Schaks M, Nemethova M, Pokrant T, Brühmann S, Linkner J, Blanchoin L, Sixt M, Rottner K, Faix J. Loss of Ena/VASP interferes with lamellipodium architecture, motility and integrin-dependent adhesion. eLife 2020; 9:55351. [PMID: 32391788 PMCID: PMC7239657 DOI: 10.7554/elife.55351] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/08/2020] [Indexed: 01/21/2023] Open
Abstract
Cell migration entails networks and bundles of actin filaments termed lamellipodia and microspikes or filopodia, respectively, as well as focal adhesions, all of which recruit Ena/VASP family members hitherto thought to antagonize efficient cell motility. However, we find these proteins to act as positive regulators of migration in different murine cell lines. CRISPR/Cas9-mediated loss of Ena/VASP proteins reduced lamellipodial actin assembly and perturbed lamellipodial architecture, as evidenced by changed network geometry as well as reduction of filament length and number that was accompanied by abnormal Arp2/3 complex and heterodimeric capping protein accumulation. Loss of Ena/VASP function also abolished the formation of microspikes normally embedded in lamellipodia, but not of filopodia capable of emanating without lamellipodia. Ena/VASP-deficiency also impaired integrin-mediated adhesion accompanied by reduced traction forces exerted through these structures. Our data thus uncover novel Ena/VASP functions of these actin polymerases that are fully consistent with their promotion of cell migration.
Collapse
Affiliation(s)
| | - Laëtitia Kurzawa
- CytoMorphoLab, Laboratoire de Physiologie cellulaire et Végétale, Interdisciplinary ResearchInstitute of Grenoble, CEA, CNRS, INRA, Grenoble-Alpes University, Grenoble, France.,CytomorphoLab, Hôpital Saint-Louis, Institut Universitaire d'Hematologie, UMRS1160, INSERM/AP-HP/UniversitéParis Diderot, Paris, France
| | - Jan Mueller
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Georgi Dimchev
- Division of Molecular Cell Biology, Zoological Institute, Technical University Braunschweig, Braunschweig, Germany
| | - Matthias Schaks
- Division of Molecular Cell Biology, Zoological Institute, Technical University Braunschweig, Braunschweig, Germany.,Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Maria Nemethova
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Thomas Pokrant
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Stefan Brühmann
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Joern Linkner
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Laurent Blanchoin
- CytoMorphoLab, Laboratoire de Physiologie cellulaire et Végétale, Interdisciplinary ResearchInstitute of Grenoble, CEA, CNRS, INRA, Grenoble-Alpes University, Grenoble, France.,CytomorphoLab, Hôpital Saint-Louis, Institut Universitaire d'Hematologie, UMRS1160, INSERM/AP-HP/UniversitéParis Diderot, Paris, France
| | - Michael Sixt
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technical University Braunschweig, Braunschweig, Germany.,Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
40
|
RIAM-VASP Module Relays Integrin Complement Receptors in Outside-In Signaling Driving Particle Engulfment. Cells 2020; 9:cells9051166. [PMID: 32397169 PMCID: PMC7291270 DOI: 10.3390/cells9051166] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/27/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
The phagocytic integrins and complement receptors αMβ2/CR3 and αXβ2/CR4 are classically associated with the phagocytosis of iC3b-opsonized particles. The activation of this receptor is dependent on signals derived from other receptors (inside-out signaling) with the crucial involvement of the Rap1-RIAM-Talin-1 pathway. Here, we analyze the implication of RIAM and its binding partner VASP in the signaling events occurring downstream of β2 integrins (outside-in) during complement-mediated phagocytosis. To this end, we used HL-60 promyelocytic cell lines deficient in RIAM or VASP or overexpressing EGFP-tagged VASP to determine VASP dynamics at phagocytic cups. Our results indicate that RIAM-deficient HL-60 cells presented impaired particle internalization and altered integrin downstream signaling during complement-dependent phagocytosis. Similarly, VASP deficiency completely blocked phagocytosis, while VASP overexpression increased the random movement of phagocytic particles at the cell surface, with reduced internalization. Moreover, the recruitment of VASP to particle contact sites, amount of pSer157-VASP and formation of actin-rich phagocytic cups were dependent on RIAM expression. Our results suggested that RIAM worked as a relay for integrin complement receptors in outside-in signaling, coordinating integrin activation and cytoskeletal rearrangements via its interaction with VASP.
Collapse
|
41
|
McNeill EM, Thompson C, Berke B, Chou VT, Rusch J, Duckworth A, DeProto J, Taylor A, Gates J, Gertler F, Keshishian H, Van Vactor D. Drosophila enabled promotes synapse morphogenesis and regulates active zone form and function. Neural Dev 2020; 15:4. [PMID: 32183907 PMCID: PMC7076993 DOI: 10.1186/s13064-020-00141-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 02/25/2020] [Indexed: 11/10/2022] Open
Abstract
Background Recent studies of synapse form and function highlight the importance of the actin cytoskeleton in regulating multiple aspects of morphogenesis, neurotransmission, and neural plasticity. The conserved actin-associated protein Enabled (Ena) is known to regulate development of the Drosophila larval neuromuscular junction through a postsynaptic mechanism. However, the functions and regulation of Ena within the presynaptic terminal has not been determined. Methods Here, we use a conditional genetic approach to address a presynaptic role for Ena on presynaptic morphology and ultrastructure, and also examine the pathway in which Ena functions through epistasis experiments. Results We find that Ena is required to promote the morphogenesis of presynaptic boutons and branches, in contrast to its inhibitory role in muscle. Moreover, while postsynaptic Ena is regulated by microRNA-mediated mechanisms, presynaptic Ena relays the output of the highly conserved receptor protein tyrosine phosphatase Dlar and associated proteins including the heparan sulfate proteoglycan Syndecan, and the non-receptor Abelson tyrosine kinase to regulate addition of presynaptic varicosities. Interestingly, Ena also influences active zones, where it restricts active zone size, regulates the recruitment of synaptic vesicles, and controls the amplitude and frequency of spontaneous glutamate release. Conclusion We thus show that Ena, under control of the Dlar pathway, is required for presynaptic terminal morphogenesis and bouton addition and that Ena has active zone and neurotransmission phenotypes. Notably, in contrast to Dlar, Ena appears to integrate multiple pathways that regulate synapse form and function.
Collapse
Affiliation(s)
- Elizabeth M McNeill
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA.
| | - Cheryl Thompson
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Brett Berke
- Department of Biology, Yale University, New Haven, CT, USA
| | - Vivian T Chou
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| | - Jannette Rusch
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - April Duckworth
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Jamin DeProto
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Alicia Taylor
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA.,Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Julie Gates
- Department of Biology, Bucknell University, Lewisburg, PA, USA
| | - Frank Gertler
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, England
| | | | - David Van Vactor
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
42
|
Koseki K, Taniguchi D, Yamashiro S, Mizuno H, Vavylonis D, Watanabe N. Lamellipodium tip actin barbed ends serve as a force sensor. Genes Cells 2019; 24:705-718. [PMID: 31514256 DOI: 10.1111/gtc.12720] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 09/04/2019] [Accepted: 09/04/2019] [Indexed: 01/31/2023]
Abstract
Cells change direction of migration by sensing rigidity of environment and traction force, yet its underlying mechanism is unclear. Here, we show that tip actin barbed ends serve as an active "force sensor" at the leading edge. We established a method to visualize intracellular single-molecule fluorescent actin through an elastic culture substrate. We found that immediately after cell edge stretch, actin assembly increased specifically at the lamellipodium tip. The rate of actin assembly increased with increasing stretch speed. Furthermore, tip actin polymerization remained elevated at the subsequent hold step, which was accompanied by a decrease in the load on the tip barbed ends. Stretch-induced tip actin polymerization was still observed without either the WAVE complex or Ena/VASP proteins. The observed relationships between forces and tip actin polymerization are consistent with a force-velocity relationship as predicted by the Brownian ratchet mechanism. Stretch caused extra membrane protrusion with respect to the stretched substrate and increased local tip polymerization by >5% of total cellular actin in 30 s. Our data reveal that augmentation of lamellipodium tip actin assembly is directly coupled to the load decrease, which may serve as a force sensor for directed cell protrusion.
Collapse
Affiliation(s)
- Kazuma Koseki
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Taniguchi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sawako Yamashiro
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Kyoto, Japan
| | - Hiroaki Mizuno
- Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Kyoto, Japan
| | | | - Naoki Watanabe
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Kyoto, Japan
| |
Collapse
|
43
|
Puleo JI, Parker SS, Roman MR, Watson AW, Eliato KR, Peng L, Saboda K, Roe DJ, Ros R, Gertler FB, Mouneimne G. Mechanosensing during directed cell migration requires dynamic actin polymerization at focal adhesions. J Cell Biol 2019; 218:4215-4235. [PMID: 31594807 PMCID: PMC6891092 DOI: 10.1083/jcb.201902101] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 06/16/2019] [Accepted: 09/06/2019] [Indexed: 11/22/2022] Open
Abstract
The mechanical properties of a cell's microenvironment influence many aspects of cellular behavior, including cell migration. Durotaxis, the migration toward increasing matrix stiffness, has been implicated in processes ranging from development to cancer. During durotaxis, mechanical stimulation by matrix rigidity leads to directed migration. Studies suggest that cells sense mechanical stimuli, or mechanosense, through the acto-myosin cytoskeleton at focal adhesions (FAs); however, FA actin cytoskeletal remodeling and its role in mechanosensing are not fully understood. Here, we show that the Ena/VASP family member, Ena/VASP-like (EVL), polymerizes actin at FAs, which promotes cell-matrix adhesion and mechanosensing. Importantly, we show that EVL regulates mechanically directed motility, and that suppression of EVL expression impedes 3D durotactic invasion. We propose a model in which EVL-mediated actin polymerization at FAs promotes mechanosensing and durotaxis by maturing, and thus reinforcing, FAs. These findings establish dynamic FA actin polymerization as a central aspect of mechanosensing and identify EVL as a crucial regulator of this process.
Collapse
Affiliation(s)
- Julieann I Puleo
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| | - Sara S Parker
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| | - Mackenzie R Roman
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| | - Adam W Watson
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| | - Kiarash Rahmani Eliato
- Department of Physics, Center for Biological Physics, and Biodesign Institute, Arizona State University, Tempe, AZ
| | - Leilei Peng
- College of Optical Sciences, University of Arizona, Tucson, AZ
| | - Kathylynn Saboda
- University of Arizona Cancer Center and Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ
| | - Denise J Roe
- University of Arizona Cancer Center and Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ
| | - Robert Ros
- Department of Physics, Center for Biological Physics, and Biodesign Institute, Arizona State University, Tempe, AZ
| | - Frank B Gertler
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
| | - Ghassan Mouneimne
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| |
Collapse
|
44
|
Gao Y, Hisey E, Bradshaw TWA, Erata E, Brown WE, Courtland JL, Uezu A, Xiang Y, Diao Y, Soderling SH. Plug-and-Play Protein Modification Using Homology-Independent Universal Genome Engineering. Neuron 2019; 103:583-597.e8. [PMID: 31272828 PMCID: PMC7200071 DOI: 10.1016/j.neuron.2019.05.047] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/13/2019] [Accepted: 05/29/2019] [Indexed: 12/20/2022]
Abstract
Analysis of endogenous protein localization, function, and dynamics is fundamental to the study of all cells, including the diversity of cell types in the brain. However, current approaches are often low throughput and resource intensive. Here, we describe a CRISPR-Cas9-based homology-independent universal genome engineering (HiUGE) method for endogenous protein manipulation that is straightforward, scalable, and highly flexible in terms of genomic target and application. HiUGE employs adeno-associated virus (AAV) vectors of autonomous insertional sequences (payloads) encoding diverse functional modifications that can integrate into virtually any genomic target loci specified by easily assembled gene-specific guide-RNA (GS-gRNA) vectors. We demonstrate that universal HiUGE donors enable rapid alterations of proteins in vitro or in vivo for protein labeling and dynamic visualization, neural-circuit-specific protein modification, subcellular rerouting and sequestration, and truncation-based structure-function analysis. Thus, the "plug-and-play" nature of HiUGE enables high-throughput and modular analysis of mechanisms driving protein functions in cellular neurobiology.
Collapse
Affiliation(s)
- Yudong Gao
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | - Erin Hisey
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | - Tyler W A Bradshaw
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical School, Durham, NC 27710, USA
| | - Eda Erata
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | - Walter E Brown
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | - Jamie L Courtland
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical School, Durham, NC 27710, USA
| | - Akiyoshi Uezu
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | - Yu Xiang
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | - Yarui Diao
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | - Scott H Soderling
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical School, Durham, NC 27710, USA.
| |
Collapse
|
45
|
O'Banion CP, Vickerman BM, Haar L, Lawrence DS. Compartmentalized cAMP Generation by Engineered Photoactivated Adenylyl Cyclases. Cell Chem Biol 2019; 26:1393-1406.e7. [PMID: 31353320 DOI: 10.1016/j.chembiol.2019.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 04/15/2019] [Accepted: 07/07/2019] [Indexed: 12/22/2022]
Abstract
Because small-molecule activators of adenylyl cyclases (AC) affect ACs cell-wide, it is challenging to explore the signaling consequences of AC activity emanating from specific intracellular compartments. We explored this issue using a series of engineered, optogenetic, spatially restricted, photoactivable adenylyl cyclases (PACs) positioned at the plasma membrane (PM), the outer mitochondrial membrane (OMM), and the nucleus (Nu). The biochemical consequences of brief photostimulation of PAC is primarily limited to the intracellular site occupied by the PAC. By contrast, sustained photostimulation results in distal cAMP signaling. Prolonged cAMP generation at the OMM profoundly stimulates nuclear protein kinase (PKA) activity. We have found that phosphodiesterases 3 (OMM and PM) and 4 (PM) modulate proximal (local) cAMP-triggered activity, whereas phosphodiesterase 4 regulates distal cAMP activity as well as the migration of PKA's catalytic subunit into the nucleus.
Collapse
Affiliation(s)
- Colin P O'Banion
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brianna M Vickerman
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Lauren Haar
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - David S Lawrence
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
46
|
Gau D, Veon W, Shroff SG, Roy P. The VASP-profilin1 (Pfn1) interaction is critical for efficient cell migration and is regulated by cell-substrate adhesion in a PKA-dependent manner. J Biol Chem 2019; 294:6972-6985. [PMID: 30814249 DOI: 10.1074/jbc.ra118.005255] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 02/22/2019] [Indexed: 12/20/2022] Open
Abstract
Dynamic regulation of the actin cytoskeleton is an essential feature of cell motility. Action of Enabled (Ena)/vasodilator-stimulated phosphoprotein (VASP), a family of conserved actin-elongating proteins, is an important aspect of regulation of the actin cytoskeletal architecture at the leading edge that controls membrane protrusion and cell motility. In this study, we performed mutagenesis experiments in overexpression and knockdown-rescue settings to provide, for the first time, direct evidence of the role of the actin-binding protein profilin1 (Pfn1) in VASP-mediated regulation of cell motility. We found that VASP's interaction with Pfn1 is promoted by cell-substrate adhesion and requires down-regulation of PKA activity. Our experimental data further suggest that PKA-mediated Ser137 phosphorylation of Pfn1 potentially negatively regulates the Pfn1-VASP interaction. Finally, Pfn1's ability to be phosphorylated on Ser137 was partly responsible for the anti-migratory action elicited by exposing cells to a cAMP/PKA agonist. On the basis of these findings, we propose a mechanism of adhesion-protrusion coupling in cell motility that involves dynamic regulation of Pfn1 by PKA activity.
Collapse
Affiliation(s)
- David Gau
- From the Department of Bioengineering, University of Pittsburgh and
| | - William Veon
- From the Department of Bioengineering, University of Pittsburgh and
| | - Sanjeev G Shroff
- From the Department of Bioengineering, University of Pittsburgh and
| | - Partha Roy
- From the Department of Bioengineering, University of Pittsburgh and .,the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| |
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: 57] [Impact Index Per Article: 8.1] [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
|
|
49
|
Banerjee A, Roy JK. Bantam regulates the axonal geometry of Drosophila larval brain by modulating actin regulator enabled. INVERTEBRATE NEUROSCIENCE 2018; 18:7. [DOI: 10.1007/s10158-018-0212-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 05/10/2018] [Indexed: 01/01/2023]
|
50
|
Laban H, Weigert A, Zink J, Elgheznawy A, Schürmann C, Günther L, Abdel Malik R, Bothur S, Wingert S, Bremer R, Rieger MA, Brüne B, Brandes RP, Fleming I, Benz PM. VASP regulates leukocyte infiltration, polarization, and vascular repair after ischemia. J Cell Biol 2018; 217:1503-1519. [PMID: 29507126 PMCID: PMC5881493 DOI: 10.1083/jcb.201702048] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 07/06/2017] [Accepted: 01/26/2018] [Indexed: 01/14/2023] Open
Abstract
In ischemic vascular diseases, leukocyte recruitment and polarization are crucial for revascularization and tissue repair. The study of Laban et al. provides evidence that VASP is a major regulator of leukocyte recruitment and polarization and vascular repair after ischemia. Mechanistically, the study supports a novel role of VASP in chemokine receptor trafficking. In ischemic vascular diseases, leukocyte recruitment and polarization are crucial for revascularization and tissue repair. We investigated the role of vasodilator-stimulated phosphoprotein (VASP) in vascular repair. After hindlimb ischemia induction, blood flow recovery, angiogenesis, arteriogenesis, and leukocyte infiltration into ischemic muscles in VASP−/− mice were accelerated. VASP deficiency also elevated the polarization of the macrophages through increased signal transducer and activator of transcription (STAT) signaling, which augmented the release of chemokines, cytokines, and growth factors to promote leukocyte recruitment and vascular repair. Importantly, VASP deletion in bone marrow–derived cells was sufficient to mimic the increased blood flow recovery of global VASP−/− mice. In chemotaxis experiments, VASP−/− neutrophils/monocytes were significantly more responsive to M1-related chemokines than wild-type controls. Mechanistically, VASP formed complexes with the chemokine receptor CCR2 and β-arrestin-2, and CCR2 receptor internalization was significantly reduced in VASP−/− leukocytes. Our data indicate that VASP is a major regulator of leukocyte recruitment and polarization in postischemic revascularization and support a novel role of VASP in chemokine receptor trafficking.
Collapse
Affiliation(s)
- Hebatullah Laban
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Andreas Weigert
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Joana Zink
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Amro Elgheznawy
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Christoph Schürmann
- German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Lea Günther
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Randa Abdel Malik
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Sabrina Bothur
- LOEWE Center for Cell and Gene Therapy and Department for Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Susanne Wingert
- LOEWE Center for Cell and Gene Therapy and Department for Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Rolf Bremer
- HBB Datenkommunikation and Abrechnungssysteme, Hannover, Germany
| | - Michael A Rieger
- LOEWE Center for Cell and Gene Therapy and Department for Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Ralf P Brandes
- German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Peter M Benz
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany .,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| |
Collapse
|