1
|
Durer ZA, İnce Hİ, Düvenci ZŞ, Timuçin E, Gräwert T, Orun O, Kan B, Sayers Z. Insights into the solution structure of the actin-binding tail domain of metavinculin by small angle X-ray scattering and molecular dynamics simulations. Int J Biol Macromol 2025:144376. [PMID: 40409637 DOI: 10.1016/j.ijbiomac.2025.144376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 05/07/2025] [Accepted: 05/17/2025] [Indexed: 05/25/2025]
Abstract
Vinculin is a ubiquitously expressed focal adhesion protein that plays an important role in cell-matrix and cell-to-cell junctions. Metavinculin, a muscle-specific splice variant of vinculin, contains a 68-amino acid disordered insert region in its actin binding tail domain (MVt). Mutations in this insert are linked to cardiomyopathies. This study investigates the solution structures and structural ensembles of wild-type (WT) and two mutant MVts, ΔLeu954 and R975W, which have been associated with cardiomyopathies, using small-angle X-ray scattering (SAXS) and molecular dynamics (MD) simulations. SAXS analyses revealed subtle differences in the estimated maximum dimensions and corroborated the elongated shape of the MVts. Quantitative comparisons of SAXS profiles indicated similarity between the WT and ΔLeu954, whereas R975W exhibited differences in the small-angle region. MD simulations demonstrated reduced conformational flexibility and greater packing of the insert in WT compared to mutants. Notably, a salt-bridge observed between R975 and D928 in a WT simulation provides a structural basis for the destabilization caused by the R975W mutation. These findings provide insights into the structure and dynamics of WT and mutant MVt, reflecting the promise of SAXS combined with MD simulations to elucidate the structural properties of proteins with structural disorder.
Collapse
Affiliation(s)
- Zeynep Aslıhan Durer
- School of Medicine, Department of Biophysics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey; Faculty of Pharmacy, Department of Biochemistry, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.
| | - Hande İpek İnce
- Institute of Health Sciences, Department of Biophysics, Marmara University, Istanbul, Turkey
| | - Zeynep Şevval Düvenci
- Institute of Health Sciences, Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Emel Timuçin
- Institute of Health Sciences, Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey; School of Medicine, Department of Biostatistics and Medical Informatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Tobias Gräwert
- European Molecular Biology Laboratory - Hamburg Unit, Biological Small Angle Scattering Group, Hamburg, Germany
| | - Oya Orun
- Institute of Health Sciences, Department of Biophysics, Marmara University, Istanbul, Turkey; School of Medicine, Department of Biophysics, Marmara University, Istanbul, Turkey
| | - Beki Kan
- School of Medicine, Department of Biophysics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Zehra Sayers
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Sabanci University, İstanbul, Turkey
| |
Collapse
|
2
|
Han X, Hu Z, Surya W, Ma Q, Zhou F, Nordenskiöld L, Torres J, Lu L, Miao Y. The intrinsically disordered region of coronins fine-tunes oligomerization and actin polymerization. Cell Rep 2023; 42:112594. [PMID: 37269287 DOI: 10.1016/j.celrep.2023.112594] [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: 10/18/2022] [Revised: 04/21/2023] [Accepted: 05/16/2023] [Indexed: 06/05/2023] Open
Abstract
Coronins play critical roles in actin network formation. The diverse functions of coronins are regulated by the structured N-terminal β propeller and the C-terminal coiled coil (CC). However, less is known about a middle "unique region" (UR), which is an intrinsically disordered region (IDR). The UR/IDR is an evolutionarily conserved signature in the coronin family. By integrating biochemical and cell biology experiments, coarse-grained simulations, and protein engineering, we find that the IDR optimizes the biochemical activities of coronins in vivo and in vitro. The budding yeast coronin IDR plays essential roles in regulating Crn1 activity by fine-tuning CC oligomerization and maintaining Crn1 as a tetramer. The IDR-guided optimization of Crn1 oligomerization is critical for F-actin cross-linking and regulation of Arp2/3-mediated actin polymerization. The final oligomerization status and homogeneity of Crn1 are contributed by three examined factors: helix packing, the energy landscape of the CC, and the length and molecular grammar of the IDR.
Collapse
Affiliation(s)
- Xiao Han
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Zixin Hu
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Wahyu Surya
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Qianqian Ma
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Feng Zhou
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Lars Nordenskiöld
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Jaume Torres
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Lanyuan Lu
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Yansong Miao
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; Institute for Digital Molecular Analytics and Science, Nanyang Technological University, Singapore 636921, Singapore.
| |
Collapse
|
3
|
Shi B, Matsui T, Qian S, Weiss TM, Nicholl ID, Callaway DJE, Bu Z. An ensemble of cadherin-catenin-vinculin complex employs vinculin as the major F-actin binding mode. Biophys J 2023; 122:2456-2474. [PMID: 37147801 PMCID: PMC10323030 DOI: 10.1016/j.bpj.2023.04.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/14/2023] [Accepted: 04/27/2023] [Indexed: 05/07/2023] Open
Abstract
The cell-cell adhesion cadherin-catenin complexes recruit vinculin to the adherens junction (AJ) to modulate the mechanical couplings between neighboring cells. However, it is unclear how vinculin influences the AJ structure and function. Here, we identified two patches of salt bridges that lock vinculin in the head-tail autoinhibited conformation and reconstituted the full-length vinculin activation mimetics bound to the cadherin-catenin complex. The cadherin-catenin-vinculin complex contains multiple disordered linkers and is highly dynamic, which poses a challenge for structural studies. We determined the ensemble conformation of this complex using small-angle x-ray and selective deuteration/contrast variation small-angle neutron scattering. In the complex, both α-catenin and vinculin adopt an ensemble of flexible conformations, but vinculin has fully open conformations with the vinculin head and actin-binding tail domains well separated from each other. F-actin binding experiments show that the cadherin-catenin-vinculin complex binds and bundles F-actin. However, when the vinculin actin-binding domain is removed from the complex, only a minor fraction of the complex binds to F-actin. The results show that the dynamic cadherin-catenin-vinculin complex employs vinculin as the primary F-actin binding mode to strengthen AJ-cytoskeleton interactions.
Collapse
Affiliation(s)
- Bright Shi
- Department of Chemistry and Biochemistry, City College of New York, City University of New York (CUNY), New York; PhD Programs in Chemistry and Biochemistry, CUNY Graduate Center, New York
| | - Tsutomu Matsui
- Stanford Synchrotron Radiation Light Source, Menlo Park, California
| | - Shuo Qian
- Second Target Station Project, Oak Ridge National Laboratory, Oak Ridge, Tennessee
| | - Thomas M Weiss
- Stanford Synchrotron Radiation Light Source, Menlo Park, California
| | - Iain D Nicholl
- Department of Biomedical Science and Physiology, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom
| | - David J E Callaway
- Department of Chemistry and Biochemistry, City College of New York, City University of New York (CUNY), New York.
| | - Zimei Bu
- Department of Chemistry and Biochemistry, City College of New York, City University of New York (CUNY), New York; PhD Programs in Chemistry and Biochemistry, CUNY Graduate Center, New York.
| |
Collapse
|
4
|
Valencia-Gallardo C, Aguilar-Salvador DI, Khakzad H, Cocom-Chan B, Bou-Nader C, Velours C, Zarrouk Y, Le Clainche C, Malosse C, Lima DB, Quenech'Du N, Mazhar B, Essid S, Fontecave M, Asnacios A, Chamot-Rooke J, Malmström L, Tran Van Nhieu G. Shigella IpaA mediates actin bundling through diffusible vinculin oligomers with activation imprint. Cell Rep 2023; 42:112405. [PMID: 37071535 DOI: 10.1016/j.celrep.2023.112405] [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: 01/03/2022] [Revised: 02/22/2023] [Accepted: 04/03/2023] [Indexed: 04/19/2023] Open
Abstract
Upon activation, vinculin reinforces cytoskeletal anchorage during cell adhesion. Activating ligands classically disrupt intramolecular interactions between the vinculin head and tail domains that bind to actin filaments. Here, we show that Shigella IpaA triggers major allosteric changes in the head domain, leading to vinculin homo-oligomerization. Through the cooperative binding of its three vinculin-binding sites (VBSs), IpaA induces a striking reorientation of the D1 and D2 head subdomains associated with vinculin oligomerization. IpaA thus acts as a catalyst producing vinculin clusters that bundle actin at a distance from the activation site and trigger the formation of highly stable adhesions resisting the action of actin relaxing drugs. Unlike canonical activation, vinculin homo-oligomers induced by IpaA appear to keep a persistent imprint of the activated state in addition to their bundling activity, accounting for stable cell adhesion independent of force transduction and relevant to bacterial invasion.
Collapse
Affiliation(s)
- Cesar Valencia-Gallardo
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France
| | - Daniel-Isui Aguilar-Salvador
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France; Laboratoire de biologie et Pharmacie Appliquée (LBPA), CNRS UMR8113/INSERM U1282, Team "Ca(2+) Signaling and Microbial Infections," Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, 91190 Gif-sur-Yvette, France
| | - Hamed Khakzad
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France; Laboratoire de biologie et Pharmacie Appliquée (LBPA), CNRS UMR8113/INSERM U1282, Team "Ca(2+) Signaling and Microbial Infections," Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, 91190 Gif-sur-Yvette, France
| | - Benjamin Cocom-Chan
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France; Laboratoire de biologie et Pharmacie Appliquée (LBPA), CNRS UMR8113/INSERM U1282, Team "Ca(2+) Signaling and Microbial Infections," Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, 91190 Gif-sur-Yvette, France; Institute for Integrative Biology of the Cell (I2BC), CNRS UMR9198/INSERM U1280, Team "Ca(2+) Signaling and Microbial Infections," CEA, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Charles Bou-Nader
- Laboratoire de Chimie des Processus Biologiques, Collège De France, CNRS UMR8229, 75005 Paris, France
| | - Christophe Velours
- Fundamental Microbiology and Pathogenicity Laboratory, UMR 5234 CNRS-University of Bordeaux, SFR TransBioMed, 33076 Bordeaux, France
| | - Yosra Zarrouk
- Institute for Integrative Biology of the Cell (I2BC), CNRS UMR9198/INSERM U1280, Team "Ca(2+) Signaling and Microbial Infections," CEA, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Christophe Le Clainche
- Institute for Integrative Biology of the Cell (I2BC), CNRS UMR9198, Team "Cytoskeletal Dynamics and Motility", CEA, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Christian Malosse
- Institut Pasteur, Université Paris Cité, CNRS UAR 2024, Mass Spectrometry for Biology Unit, F-75015 Paris
| | - Diogo Borges Lima
- Institut Pasteur, Université Paris Cité, CNRS UAR 2024, Mass Spectrometry for Biology Unit, F-75015 Paris
| | - Nicole Quenech'Du
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France
| | - Bilal Mazhar
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France
| | - Sami Essid
- Laboratoire de biologie et Pharmacie Appliquée (LBPA), CNRS UMR8113/INSERM U1282, Team "Ca(2+) Signaling and Microbial Infections," Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, 91190 Gif-sur-Yvette, France
| | - Marc Fontecave
- Laboratoire de Chimie des Processus Biologiques, Collège De France, CNRS UMR8229, 75005 Paris, France
| | - Atef Asnacios
- Université Paris Cité, CNRS, Laboratoire Matière et Systèmes Complexes, UMR7057, F-75013 Paris, France
| | - Julia Chamot-Rooke
- Institut Pasteur, Université Paris Cité, CNRS UAR 2024, Mass Spectrometry for Biology Unit, F-75015 Paris
| | - Lars Malmström
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Guy Tran Van Nhieu
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France; Laboratoire de biologie et Pharmacie Appliquée (LBPA), CNRS UMR8113/INSERM U1282, Team "Ca(2+) Signaling and Microbial Infections," Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, 91190 Gif-sur-Yvette, France; Institute for Integrative Biology of the Cell (I2BC), CNRS UMR9198/INSERM U1280, Team "Ca(2+) Signaling and Microbial Infections," CEA, Université Paris-Saclay, 91190 Gif-sur-Yvette, France.
| |
Collapse
|
5
|
Complete Model of Vinculin Suggests the Mechanism of Activation by Helical Super-Bundle Unfurling. Protein J 2022; 41:55-70. [PMID: 35006498 DOI: 10.1007/s10930-022-10040-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2022] [Indexed: 12/24/2022]
Abstract
To shed light onto the activation mechanism of vinculin, we carried out a detailed refinement of chicken vinculin and compared it to the human protein which is greater than 95% identical. Refinement resulted in a complete and significantly improved model. This model includes important elements such as a pro-rich strap region (PRR) and C-terminus. The conformation of the PRR stabilized by its inter- and intra-molecular contacts shows a dynamic, but relatively stable motif that constitutes a docking platform for multiple molecules. The contact of the C-terminus with the PRR suggests that phosphorylation of Tyr1065 might control activation and membrane binding. Improved electron densities showed the presence of large solvent molecules such as phosphates/sulfates and a head-group of PIP2. The improved model allowed for a computational stability analysis to be performed by the program Corex/Best which located numerous hot-spots of increased and decreased stability. Proximity of the identified binding sites for regulatory partners involved in inducing or suppressing the activation of vinculin to the unstable elements sheds new light onto the activation pathway and differential activation. This stability analysis suggests that the activation pathway proceeds by unfurling of the super-bundle built from four bundles of helices without separation of the Vt region (840-1066) from the head. According to our mechanism, when activating proteins bind at the strap region a separation of N and C terminal bundles occurs, followed by unfurling of the super-bundle and flattening of the general shape of the molecule, which exposes the interaction sites for binding of auxiliary proteins.
Collapse
|
6
|
OUP accepted manuscript. Pathog Dis 2022; 80:6521441. [DOI: 10.1093/femspd/ftac003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/30/2021] [Accepted: 02/01/2022] [Indexed: 11/15/2022] Open
|
7
|
Vinje JB, Guadagno NA, Progida C, Sikorski P. Analysis of Actin and Focal Adhesion Organisation in U2OS Cells on Polymer Nanostructures. NANOSCALE RESEARCH LETTERS 2021; 16:143. [PMID: 34524556 PMCID: PMC8443752 DOI: 10.1186/s11671-021-03598-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/28/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND In this work, we explore how U2OS cells are affected by arrays of polymer nanopillars fabricated on flat glass surfaces. We focus on describing changes to the organisation of the actin cytoskeleton and in the location, number and shape of focal adhesions. From our findings we identify that the cells can be categorised into different regimes based on their spreading and adhesion behaviour on nanopillars. A quantitative analysis suggests that cells seeded on dense nanopillar arrays are suspended on top of the pillars with focal adhesions forming closer to the cell periphery compared to flat surfaces or sparse pillar arrays. This change is analogous to similar responses for cells seeded on soft substrates. RESULTS In this work, we explore how U2OS cells are affected by arrays of polymer nanopillars fabricated on flat glass surfaces. We focus on describing changes to the organisation of the actin cytoskeleton and in the location, number and shape of focal adhesions. From our findings we identify that the cells can be categorised into different regimes based on their spreading and adhesion behaviour on nanopillars. A quantitative analysis suggests that cells seeded on dense nanopillar arrays are suspended on top of the pillars with focal adhesions forming closer to the cell periphery compared to flat surfaces or sparse pillar arrays. This change is analogous to similar responses for cells seeded on soft substrates. CONCLUSION Overall, we show that the combination of high throughput nanofabrication, advanced optical microscopy, molecular biology tools to visualise cellular processes and data analysis can be used to investigate how cells interact with nanostructured surfaces and will in the future help to create culture substrates that induce particular cell function.
Collapse
Affiliation(s)
- Jakob B Vinje
- Department of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- Department of Electronic Systems, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | | | - Cinzia Progida
- Department of Biosciences, University of Oslo (UiO), Oslo, Norway
| | - Pawel Sikorski
- Department of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
8
|
Wang F, Fang M, Kong M, Wang C, Xu Y. Vinculin presents unfavorable prediction in ovarian cancer and prevents proliferation and migration of ovarian cancer cells. J Biochem Mol Toxicol 2020; 34:e22525. [PMID: 32369671 DOI: 10.1002/jbt.22525] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/21/2020] [Accepted: 04/27/2020] [Indexed: 12/24/2022]
Abstract
The influences of Vinculin on many cancers were blurry, including ovarian cancer. Thus, we concentrated on the efficient role of Vinculin in ovarian cancer and explored the potential mechanism(s). Expression of Vinculin in ovarian cancer tissues and cell lines was investigated by real-time polymerase chain reaction, immunohistochemistry, and Western blot. The Kaplan-Meier manner with the logrank was performed to assess overall survival. We further evaluated the relations between Vinculin expression and clinicopathological features of ovarian cancer. Moreover, Vinculin was overexpressed or silenced by respectively transfection with pcDNA-Vinculin or small interfering (si-Vinculin) into human ovarian cancer cell line Caov3 or human ovarian epithelial cell line (HOEpiC). Thereafter, cell viability, cell apoptosis, and migration were checked by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide, flow cytometer, and scratch assay, respectively. Likewise, the apoptosis- and migration-related proteins were distinguished by Western blot. Compared to the nontumor tissues or HOEpiC cells, Vinculin was significantly lower expressed in the ovarian cancer tissues and cells. Furthermore, we found out that Vinculin was primarily distributed at the cell membrane and cytoplasm. Moreover, Vinculin was negatively associated with International Federation of Gynecology and Obstetrics stage, grade, and distant metastasis. Overexpression of Vinculin dramatically weakened cell viability and migration and stimulated apoptosis. Conversely, suppression of Vinculin showed opposite results. Vinculin presents unfavorable prediction in ovarian cancer and inhibits ovarian cancer proliferation and migration.
Collapse
Affiliation(s)
- Fei Wang
- Department of Gynecology, Jining No.1 People's Hospital, Jining, Shandong, China
| | - Meixia Fang
- Department of Gynecology, Jining No.1 People's Hospital, Jining, Shandong, China
| | - Min Kong
- Department of Gynecology, Jining No.1 People's Hospital, Jining, Shandong, China
| | - Changhe Wang
- Department of Gynecology, Jining No.1 People's Hospital, Jining, Shandong, China
| | - Yuting Xu
- Department of Gynecology, Jining No.1 People's Hospital, Jining, Shandong, China
| |
Collapse
|
9
|
Kelley CF, Litschel T, Schumacher S, Dedden D, Schwille P, Mizuno N. Phosphoinositides regulate force-independent interactions between talin, vinculin, and actin. eLife 2020; 9:e56110. [PMID: 32657269 PMCID: PMC7384861 DOI: 10.7554/elife.56110] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/10/2020] [Indexed: 12/25/2022] Open
Abstract
Focal adhesions (FA) are large macromolecular assemblies which help transmit mechanical forces and regulatory signals between the extracellular matrix and an interacting cell. Two key proteins talin and vinculin connecting integrin to actomyosin networks in the cell. Both proteins bind to F-actin and each other, providing a foundation for network formation within FAs. However, the underlying mechanisms regulating their engagement remain unclear. Here, we report on the results of in vitro reconstitution of talin-vinculin-actin assemblies using synthetic membrane systems. We find that neither talin nor vinculin alone recruit actin filaments to the membrane. In contrast, phosphoinositide-rich membranes recruit and activate talin, and the membrane-bound talin then activates vinculin. Together, the two proteins then link actin to the membrane. Encapsulation of these components within vesicles reorganized actin into higher-order networks. Notably, these observations were made in the absence of applied force, whereby we infer that the initial assembly stage of FAs is force independent. Our findings demonstrate that the local membrane composition plays a key role in controlling the stepwise recruitment, activation, and engagement of proteins within FAs.
Collapse
Affiliation(s)
- Charlotte F Kelley
- Max Planck Institute of Biochemistry, Department of Structural Cell BiologyMartinsriedGermany
| | - Thomas Litschel
- Max Planck Institute of Biochemistry, Department of Cellular and Molecular BiophysicsMartinsriedGermany
| | - Stephanie Schumacher
- Max Planck Institute of Biochemistry, Department of Structural Cell BiologyMartinsriedGermany
| | - Dirk Dedden
- Max Planck Institute of Biochemistry, Department of Structural Cell BiologyMartinsriedGermany
| | - Petra Schwille
- Max Planck Institute of Biochemistry, Department of Cellular and Molecular BiophysicsMartinsriedGermany
| | - Naoko Mizuno
- Max Planck Institute of Biochemistry, Department of Structural Cell BiologyMartinsriedGermany
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaUnited States
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
10
|
Abstract
Irritable bowel syndrome (IBS) is an extremely common and often very debilitating chronic functional gastrointestinal disorder. Despite its prevalence, significant associated healthcare costs, and quality-of-life issues for affected individuals, our understanding of its etiology remained limited. However, it is now evident that microbial factors play key roles in IBS pathophysiology. Acute gastroenteritis following exposure to pathogens can precipitate the development of IBS, and studies have demonstrated changes in the gut microbiome in IBS patients. These changes may explain some of the symptoms of IBS, including visceral hypersensitivity, as gut microbes exert effects on the host immune system and gut barrier function, as well as the brain-gut axis. Microbial differences also appear to underlie the two main functional categories of IBS: diarrhea-predominant IBS (IBS-D) is associated with small intestinal bacterial overgrowth, which can be diagnosed by a positive hydrogen breath test, and constipation-predominant IBS (IBS-C) is associated with increased levels of methanogenic archaea, which can be diagnosed by a positive methane breath test. Mechanistically, the pathogens that cause gastroenteritis and trigger subsequent IBS development produce a common toxin, cytolethal distending toxin B (CdtB), and antibodies raised against CdtB cross-react with the cytoskeletal protein vinculin and impair gut motility, facilitating bacterial overgrowth. In contrast, methane gas slows intestinal contractility, which may facilitate the development of constipation. While antibiotics and dietary manipulations have been used to relieve IBS symptoms, with varying success, elucidating the specific mechanisms by which gut microbes exert their effects on the host may allow the development of targeted treatments that may successfully treat the underlying causes of IBS.
Collapse
Affiliation(s)
- Mark Pimentel
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Anthony Lembo
- Division of Gastroenterology, Beth Israel Deaconess Hospital, Boston, MA, USA
| |
Collapse
|
11
|
Morales W, Rezaie A, Barlow G, Pimentel M. Second-Generation Biomarker Testing for Irritable Bowel Syndrome Using Plasma Anti-CdtB and Anti-Vinculin Levels. Dig Dis Sci 2019; 64:3115-3121. [PMID: 31152332 DOI: 10.1007/s10620-019-05684-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/24/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND ELISA testing for anti-CdtB and anti-vinculin can discriminate patients with irritable bowel syndrome with diarrhea (IBS-D) from those with inflammatory bowel disease (IBD). However, recent findings suggest the antigens can suffer from epitope instability. AIM This study aimed to assess effects of incorporating epitope stabilization on test characteristics for distinguishing IBS-D from IBD subjects. METHODS Plasma samples from IBS-D subjects from a large-scale clinical trial and subjects with endoscopically active IBD without concurrent immunomodulator therapy were used. After epitope stabilization, CdtB and vinculin were used in ELISA testing. Optical density readings were compared between IBS-D and IBD subjects. RESULTS Samples from 100 IBS-D and 31 IBD (22 UC and 9 CD) subjects were tested. IBS-D subjects had higher anti-CdtB titers (P = 0.0001) and higher anti-vinculin titers (P = 0.004) than IBD subjects. The specificities of anti-CdtB and anti-vinculin to differentiate IBS-D from IBD were 93.5% and 90.9%, respectively, with sensitivities of 43.0% and 52.2%, respectively. The positive likelihood ratios of identifying IBS-D with anti-CdtB and anti-vinculin were 6.7 and 5.7, respectively. Assuming a pretest probability of 57% for diagnosis of IBS-D in patients with abdominal pain and change in bowel habits, testing positive for both antibodies resulted in a posttest probability of > 98%. CONCLUSIONS Performing epitope stabilization for CdtB and vinculin enhances the test characteristics of ELISAs for anti-CdtB and anti-vinculin in discriminating IBS-D from IBD. Measurement of anti-CdtB and anti-vinculin with this second-generation methodology may further advance our understanding of the role of immunity in functional bowel diseases.
Collapse
Affiliation(s)
- Walter Morales
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, 8730 Alden Drive, Suite 240E, Los Angeles, CA, 90048, USA
| | - Ali Rezaie
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, 8730 Alden Drive, Suite 240E, Los Angeles, CA, 90048, USA
| | - Gillian Barlow
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, 8730 Alden Drive, Suite 240E, Los Angeles, CA, 90048, USA
| | - Mark Pimentel
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, 8730 Alden Drive, Suite 240E, Los Angeles, CA, 90048, USA.
| |
Collapse
|
12
|
Krokhotin A, Sarker M, Sevilla EA, Costantini LM, Griffith JD, Campbell SL, Dokholyan NV. Distinct Binding Modes of Vinculin Isoforms Underlie Their Functional Differences. Structure 2019; 27:1527-1536.e3. [PMID: 31422909 PMCID: PMC6774862 DOI: 10.1016/j.str.2019.07.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 05/23/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022]
Abstract
Vinculin and its splice isoform metavinculin play key roles in regulating cellular morphology, motility, and force transduction. Vinculin is distinct from metavinculin in its ability to bundle filamentous actin (F-actin). To elucidate the molecular basis for these differences, we employed computational and experimental approaches. Results from these analyses suggest that the C terminus of both vinculin and metavinculin form stable interactions with the F-actin surface. However, the metavinculin tail (MVt) domain contains a 68 amino acid insert, with helix 1 (H1) sequestered into a globular subdomain, which protrudes from the F-actin surface and prevents actin bundling by sterically occluding actin filaments. Consistent with our model, deletion and selective point mutations within the MVt H1 disrupt this protruding structure, and facilitate actin bundling similar to vinculin tail (Vt) domain.
Collapse
Affiliation(s)
- Andrey Krokhotin
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Departments of Pathology, Genetics and Developmental Biology, Howard Hughes Medical Institute, Stanford Medical School, Palo Alto, CA 94305, USA
| | - Muzaddid Sarker
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Ernesto Alva Sevilla
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lindsey M Costantini
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jack D Griffith
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sharon L Campbell
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Nikolay V Dokholyan
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Departments of Pharmacology and Departments of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
13
|
Lee HT, Sharek L, O’Brien ET, Urbina FL, Gupton SL, Superfine R, Burridge K, Campbell SL. Vinculin and metavinculin exhibit distinct effects on focal adhesion properties, cell migration, and mechanotransduction. PLoS One 2019; 14:e0221962. [PMID: 31483833 PMCID: PMC6726196 DOI: 10.1371/journal.pone.0221962] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 08/19/2019] [Indexed: 12/04/2022] Open
Abstract
Vinculin (Vcn) is a ubiquitously expressed cytoskeletal protein that links transmembrane receptors to actin filaments, and plays a key role in regulating cell adhesion, motility, and force transmission. Metavinculin (MVcn) is a Vcn splice isoform that contains an additional exon encoding a 68-residue insert within the actin binding tail domain. MVcn is selectively expressed at sub-stoichiometic amounts relative to Vcn in smooth and cardiac muscle cells. Mutations in the MVcn insert are linked to various cardiomyopathies. In vitro analysis has previously shown that while both proteins can engage filamentous (F)-actin, only Vcn can promote F-actin bundling. Moreover, we and others have shown that MVcn can negatively regulate Vcn-mediated F-actin bundling in vitro. To investigate functional differences between MVcn and Vcn, we stably expressed either Vcn or MVcn in Vcn-null mouse embryonic fibroblasts. While both MVcn and Vcn were observed at FAs, MVcn-expressing cells had larger but fewer focal adhesions per cell compared to Vcn-expressing cells. MVcn-expressing cells migrated faster and exhibited greater persistence compared to Vcn-expressing cells, even though Vcn-containing FAs assembled and disassembled faster. Magnetic tweezer measurements on Vcn-expressing cells show a typical cell stiffening phenotype in response to externally applied force; however, this was absent in Vcn-null and MVcn-expressing cells. Our findings that MVcn expression leads to larger but fewer FAs per cell, in conjunction with the inability of MVcn to bundle F-actin in vitro and rescue the cell stiffening response, are consistent with our previous findings of actin bundling deficient Vcn variants, suggesting that deficient actin-bundling may account for some of the differences between Vcn and MVcn.
Collapse
Affiliation(s)
- Hyunna T. Lee
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Lisa Sharek
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - E. Timothy O’Brien
- Department of Physics and Astronomy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Fabio L. Urbina
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Stephanie L. Gupton
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Richard Superfine
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Keith Burridge
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Sharon L. Campbell
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
14
|
Sarker M, Lee HT, Mei L, Krokhotin A, de Los Reyes SE, Yen L, Costantini LM, Griffith J, Dokholyan NV, Alushin GM, Campbell SL. Cardiomyopathy Mutations in Metavinculin Disrupt Regulation of Vinculin-Induced F-Actin Assemblies. J Mol Biol 2019; 431:1604-1618. [PMID: 30844403 DOI: 10.1016/j.jmb.2019.02.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/22/2019] [Accepted: 02/22/2019] [Indexed: 10/27/2022]
Abstract
Debilitating heart conditions, notably dilated and hypertrophic cardiomyopathies (CMs), are associated with point mutations in metavinculin, a larger isoform of the essential cytoskeletal protein vinculin. Metavinculin is co-expressed with vinculin at sub-stoichiometric ratios in cardiac tissues. CM mutations in the metavinculin tail domain (MVt) occur within the extra 68-residue insert that differentiates it from the vinculin tail domain (Vt). Vt binds actin filaments (F-actin) and promotes vinculin dimerization to bundle F-actin into thick fibers. While MVt binds to F-actin in a similar manner to Vt, MVt is incapable of F-actin bundling and inhibits Vt-mediated F-actin bundling. We performed F-actin co-sedimentation and negative-stain EM experiments to dissect the coordinated roles of metavinculin and vinculin in actin fiber assembly and the effects of three known metavinculin CM mutations. These CM mutants were found to weakly induce the formation of disordered F-actin assemblies. Notably, they fail to inhibit Vt-mediated F-actin bundling and instead promote formation of large assemblies embedded with linear bundles. Computational models of MVt bound to F-actin suggest that MVt undergoes a conformational change licensing the formation of a protruding sub-domain incorporating the insert, which sterically prevents dimerization and bundling of F-actin by Vt. Sub-domain formation is destabilized by CM mutations, disrupting this inhibitory mechanism. These findings provide new mechanistic insights into the ability of metavinculin to tune actin organization by vinculin and suggest that dysregulation of this process by CM mutants could underlie their malfunction in disease.
Collapse
Affiliation(s)
- Muzaddid Sarker
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hyunna T Lee
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lin Mei
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA
| | - Andrey Krokhotin
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Laura Yen
- Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY 10025, USA
| | - Lindsey M Costantini
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jack Griffith
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nikolay V Dokholyan
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Gregory M Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA
| | - Sharon L Campbell
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
15
|
Xia S, Yim EKF, Kanchanawong P. Molecular Organization of Integrin-Based Adhesion Complexes in Mouse Embryonic Stem Cells. ACS Biomater Sci Eng 2019; 5:3828-3842. [PMID: 33438423 DOI: 10.1021/acsbiomaterials.8b01124] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The mechanical microenvironment serves as an important factor influencing stem cell differentiation. Mechanobiological responses depend strongly on actomyosin contractility and integrin-based cell-extracellular matrix (ECM) interactions mediated by adhesive structures such as focal adhesions (FAs). While the roles of FAs in mechanobiology have been intensively studied in many mesenchymal and migratory cell types, recently it has been recognized that certain pluripotent stem cells (PSCs) exhibited significantly attenuated FA-mediated mechanobiological responses. FAs in such PSCs are sparsely distributed and much less prominent in comparison to "classical" FAs of typical adherent cells. Despite these differences, insights into how FAs in PSCs are structurally organized to perform their functions are still elusive. Using mouse embryonic stem cells (mESCs) to study PSC-ECM interactions, here we surveyed the molecular composition and nanostructural organization of FAs. We found that, despite being small in size, mESC FAs appeared to be compositionally mature, containing markers such as vinculin, zyxin, and α-actinin, and dependent on myosin II contractility. Using super-resolution microscopy, we revealed that mESC FAs were organized into a conserved multilayer nanoscale architecture. However, the nanodomain organization was compressed in mESCs, with the force transduction layer spanning ∼10 nm, significantly more compact than in FAs of other cell types. Furthermore, we found that the position and orientation of vinculin, a key mechanotransduction protein, were modulated in an ECM-dependent manner. Our analysis also revealed that while most core FA genes were expressed, the expression of LIM domain proteins was comparatively lower in PSCs. Altogether our results suggest that while core structural and mechanosensitive elements are operational in mESC FAs, their structural organization and regulatory aspects may diverge significantly from "classical" FAs, which may account for the attenuated mechanobiological responses of these cell types.
Collapse
Affiliation(s)
- Shumin Xia
- Mechanobiology Institute, Singapore, Republic of Singapore, 117411
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Pakorn Kanchanawong
- Mechanobiology Institute, Singapore, Republic of Singapore, 117411.,Department of Biomedical Engineering, National University of Singapore, Singapore, Republic of Singapore, 117411
| |
Collapse
|
16
|
Yu H, Wang X, Kang F, Chen Z, Meng Y, Dai M. Propofol attenuates inflammatory damage on neurons following cerebral infarction by inhibiting excessive activation of microglia. Int J Mol Med 2018; 43:452-460. [PMID: 30431058 DOI: 10.3892/ijmm.2018.3974] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 02/02/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Hang Yu
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Xiaozhi Wang
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Fuxin Kang
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Zhile Chen
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Yunxia Meng
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Mingming Dai
- Department of Internal Neurology, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| |
Collapse
|
17
|
Han MKL, van der Krogt GNM, de Rooij J. Zygotic vinculin is not essential for embryonic development in zebrafish. PLoS One 2017; 12:e0182278. [PMID: 28767718 PMCID: PMC5540497 DOI: 10.1371/journal.pone.0182278] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
The formation of multicellular tissues during development is governed by mechanical forces that drive cell shape and tissue architecture. Protein complexes at sites of adhesion to the extracellular matrix (ECM) and cell neighbors, not only transmit these mechanical forces, but also allow cells to respond to changes in force by inducing biochemical feedback pathways. Such force-induced signaling processes are termed mechanotransduction. Vinculin is a central protein in mechanotransduction that in both integrin-mediated cell-ECM and cadherin-mediated cell-cell adhesions mediates force-induced cytoskeletal remodeling and adhesion strengthening. Vinculin was found to be important for the integrity and remodeling of epithelial tissues in cell culture models and could therefore be expected to be of broad importance in epithelial morphogenesis in vivo. Besides a function in mouse heart development, however, the importance of vinculin in morphogenesis of other vertebrate tissues has remained unclear. To investigate this further, we knocked out vinculin functioning in zebrafish, which contain two fully functional isoforms designated as vinculin A and vinculin B that both show high sequence conservation with higher vertebrates. Using TALEN and CRISPR-Cas gene editing technology we generated vinculin-deficient zebrafish. While single vinculin A mutants are viable and able to reproduce, additional loss of zygotic vinculin B was lethal after embryonic stages. Remarkably, vinculin-deficient embryos do not show major developmental defects, apart from mild pericardial edemas. These results lead to the conclusion that vinculin is not of broad importance for the development and morphogenesis of zebrafish tissues.
Collapse
Affiliation(s)
- Mitchell K. L. Han
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerard N. M. van der Krogt
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Johan de Rooij
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
18
|
Rezaie A, Park SC, Morales W, Marsh E, Lembo A, Kim JH, Weitsman S, Chua KS, Barlow GM, Pimentel M. Assessment of Anti-vinculin and Anti-cytolethal Distending Toxin B Antibodies in Subtypes of Irritable Bowel Syndrome. Dig Dis Sci 2017; 62:1480-1485. [PMID: 28451914 DOI: 10.1007/s10620-017-4585-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 04/18/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Antibodies to cytolethal distending toxin B (CdtB) and vinculin are novel biomarkers that rule-in and differentiate irritable bowel syndrome with diarrhea (IBS-D) from other causes of diarrhea and healthy controls. AIM To determine whether these antibodies can also diagnose and differentiate other IBS subtypes. METHODS Subjects with IBS-D based on Rome III criteria (n = 2375) were recruited from a large-scale multicenter clinical trial (TARGET 3). Healthy subjects without gastrointestinal (GI) diseases or symptoms (n = 43) and subjects with mixed IBS (IBS-M) (n = 25) or IBS with constipation (IBS-C) (n = 30) were recruited from two major medical centers. Plasma levels of anti-CdtB and anti-vinculin antibodies in all subjects were determined by enzyme-linked immunosorbent assay. Optical densities of ≥1.68 and ≥2.80 were considered positive for anti-vinculin and anti-CdtB, respectively. Plasma levels of anti-CdtB and anti-vinculin antibodies were highest in IBS-D and lowest in IBS-C and healthy controls (P < 0.001). Levels in IBS-C subjects were not statistically different from controls (P > 0.1). Positivity for anti-CdtB or anti-vinculin resulted in a statistically significant negative gradient from IBS-D (58.1%) to IBS-M (44.0%), IBS-C (26.7%), and controls (16.3%) (P < 0.001). CONCLUSIONS Anti-CdtB and anti-vinculin titers and positivity rates differ in IBS subtypes, with higher antibody levels and positivity rates in IBS-D and IBS-M, and lower levels in IBS-C subjects that are similar to those in healthy controls. These antibodies appear useful in the diagnosis of IBS-M and IBS-D, but not IBS-C. Furthermore, these findings suggest that IBS-C is pathophysiologically distinct from subtypes with diarrheal components (i.e., IBS-M and IBS-D).
Collapse
Affiliation(s)
- Ali Rezaie
- GI Motility Program, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, 8730 Alden Drive, Thalians Bldg, #E226, Los Angeles, CA, 90048, USA.
| | - Sung Chul Park
- GI Motility Program, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, 8730 Alden Drive, Thalians Bldg, #E226, Los Angeles, CA, 90048, USA.,Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Walter Morales
- GI Motility Program, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, 8730 Alden Drive, Thalians Bldg, #E226, Los Angeles, CA, 90048, USA
| | - Emily Marsh
- GI Motility Program, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, 8730 Alden Drive, Thalians Bldg, #E226, Los Angeles, CA, 90048, USA
| | - Anthony Lembo
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jae Hak Kim
- Department of Internal Medicine, Dongguk University Ilsan Hospital, The Graduate School of Dongguk University, Goyang, South Korea
| | - Stacy Weitsman
- GI Motility Program, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, 8730 Alden Drive, Thalians Bldg, #E226, Los Angeles, CA, 90048, USA
| | - Kathleen S Chua
- GI Motility Program, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, 8730 Alden Drive, Thalians Bldg, #E226, Los Angeles, CA, 90048, USA
| | - Gillian M Barlow
- GI Motility Program, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, 8730 Alden Drive, Thalians Bldg, #E226, Los Angeles, CA, 90048, USA
| | - Mark Pimentel
- GI Motility Program, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, 8730 Alden Drive, Thalians Bldg, #E226, Los Angeles, CA, 90048, USA
| |
Collapse
|
19
|
Bays JL, DeMali KA. Vinculin in cell-cell and cell-matrix adhesions. Cell Mol Life Sci 2017; 74:2999-3009. [PMID: 28401269 PMCID: PMC5501900 DOI: 10.1007/s00018-017-2511-3] [Citation(s) in RCA: 292] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 02/07/2023]
Abstract
Vinculin was identified as a component of focal adhesions and adherens junctions nearly 40 years ago. Since that time, remarkable progress has been made in understanding its activation, regulation and function. Here we discuss the current understanding of the roles of vinculin in cell–cell and cell–matrix adhesions. Emphasis is placed on the how vinculin is recruited, activated and regulated. We also highlight the recent understanding of how vinculin responds to and transmits force at integrin- and cadherin-containing adhesion complexes to the cytoskeleton. Furthermore, we discuss roles of vinculin in binding to and rearranging the actin cytoskeleton.
Collapse
Affiliation(s)
- Jennifer L Bays
- Department of Biochemistry, University of Iowa, Iowa City, IA, 52242, USA
| | - Kris A DeMali
- Department of Biochemistry, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
20
|
Rezaie A, Pimentel M, Cohen E. Autoimmunity as a Potential Cause of Post-Infectious Gut Dysmotility: A Longitudinal Observation. Am J Gastroenterol 2017; 112:656-657. [PMID: 28381845 DOI: 10.1038/ajg.2017.8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Ali Rezaie
- GI Motility Program, Division of Gastroenterology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Mark Pimentel
- GI Motility Program, Division of Gastroenterology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Erica Cohen
- GI Motility Program, Division of Gastroenterology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
21
|
Thompson PM, Ramachandran S, Case LB, Tolbert CE, Tandon A, Pershad M, Dokholyan NV, Waterman CM, Campbell SL. A Structural Model for Vinculin Insertion into PIP 2-Containing Membranes and the Effect of Insertion on Vinculin Activation and Localization. Structure 2017; 25:264-275. [PMID: 28089450 DOI: 10.1016/j.str.2016.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/10/2016] [Accepted: 12/12/2016] [Indexed: 12/20/2022]
Abstract
Vinculin, a scaffolding protein that localizes to focal adhesions (FAs) and adherens junctions, links the actin cytoskeleton to the adhesive super-structure. While vinculin binds to a number of cytoskeletal proteins, it can also associate with phosphatidylinositol 4,5-bisphosphate (PIP2) to drive membrane association. To generate a structural model for PIP2-dependent interaction of vinculin with the lipid bilayer, we conducted lipid-association, nuclear magnetic resonance, and computational modeling experiments. We find that two basic patches on the vinculin tail drive membrane association: the basic collar specifically recognizes PIP2, while the basic ladder drives association with the lipid bilayer. Vinculin mutants with defects in PIP2-dependent liposome association were then expressed in vinculin knockout murine embryonic fibroblasts. Results from these analyses indicate that PIP2 binding is not required for localization of vinculin to FAs or FA strengthening, but is required for vinculin activation and turnover at FAs to promote its association with the force transduction FA nanodomain.
Collapse
Affiliation(s)
- Peter M Thompson
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Srinivas Ramachandran
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lindsay B Case
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Caitlin E Tolbert
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Arpit Tandon
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mihir Pershad
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nikolay V Dokholyan
- Department of Biochemistry and Biophysics, 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
| | - Clare M Waterman
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sharon L Campbell
- Department of Biochemistry and Biophysics, 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.
| |
Collapse
|
22
|
Differential lipid binding of vinculin isoforms promotes quasi-equivalent dimerization. Proc Natl Acad Sci U S A 2016; 113:9539-44. [PMID: 27503891 DOI: 10.1073/pnas.1600702113] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The main cause of death globally remains debilitating heart conditions, such as dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM), which are often due to mutations of specific components of adhesion complexes. Vinculin regulates these complexes and plays essential roles in intercalated discs that are necessary for muscle cell function and coordinated movement and in the development and function of the heart. Humans bearing familial or sporadic mutations in vinculin suffer from chronic, progressively debilitating DCM that ultimately leads to cardiac failure and death, whereas autosomal dominant mutations in vinculin can also provoke HCM, causing acute cardiac failure. The DCM/HCM-associated mutants of vinculin occur in the 68-residue insert unique to the muscle-specific, alternatively spliced isoform of vinculin, termed metavinculin (MV). Contrary to studies that suggested that phosphoinositol-4,5-bisphosphate (PIP2) only induces vinculin homodimers, which are asymmetric, we show that phospholipid binding results in a domain-swapped symmetric MV dimer via a quasi-equivalent interface compared with vinculin involving R975. Although one of the two PIP2 binding sites is preserved, the symmetric MV dimer that bridges two PIP2 molecules differs from the asymmetric vinculin dimer that bridges only one PIP2 Unlike vinculin, wild-type MV and the DCM/HCM-associated R975W mutant bind PIP2 in their inactive conformations, and R975W MV fails to dimerize. Mutating selective vinculin residues to their corresponding MV residues, or vice versa, switches the isoform's dimeric constellation and lipid binding site. Collectively, our data suggest that MV homodimerization modulates microfilament attachment at muscular adhesion sites and furthers our understanding of MV-mediated cardiac remodeling.
Collapse
|
23
|
Atherton P, Stutchbury B, Wang DY, Jethwa D, Tsang R, Meiler-Rodriguez E, Wang P, Bate N, Zent R, Barsukov IL, Goult BT, Critchley DR, Ballestrem C. Vinculin controls talin engagement with the actomyosin machinery. Nat Commun 2015; 6:10038. [PMID: 26634421 PMCID: PMC4686655 DOI: 10.1038/ncomms10038] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 10/26/2015] [Indexed: 12/20/2022] Open
Abstract
The link between extracellular-matrix-bound integrins and intracellular F-actin is essential for cell spreading and migration. Here, we demonstrate how the actin-binding proteins talin and vinculin cooperate to provide this link. By expressing structure-based talin mutants in talin null cells, we show that while the C-terminal actin-binding site (ABS3) in talin is required for adhesion complex assembly, the central ABS2 is essential for focal adhesion (FA) maturation. Thus, although ABS2 mutants support cell spreading, the cells lack FAs, fail to polarize and exert reduced force on the surrounding matrix. ABS2 is inhibited by the preceding mechanosensitive vinculin-binding R3 domain, and deletion of R2R3 or expression of constitutively active vinculin generates stable force-independent FAs, although cell polarity is compromised. Our data suggest a model whereby force acting on integrin-talin complexes via ABS3 promotes R3 unfolding and vinculin binding, activating ABS2 and locking talin into an actin-binding configuration that stabilizes FAs. The mechanosensitive proteins talin and vinculin mediate the linkage between integrin-bound extracellular matrix and the actin cytoskeleton. Here the authors dissect distinct roles for two actin-binding sites within talin on adhesion complex assembly and maturation, which are regulated by vinculin binding to talin.
Collapse
Affiliation(s)
- Paul Atherton
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| | - Ben Stutchbury
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| | - De-Yao Wang
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| | - Devina Jethwa
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| | - Ricky Tsang
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| | | | - Pengbo Wang
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| | - Neil Bate
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Roy Zent
- Vanderbilt Centre for Kidney Disease, Vanderbilt Division of Nephrology, S-3223 Medical Centre, North Nashville, Tennessee, USA
| | - Igor L Barsukov
- Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Canterbury, KENT CT2 7NJ, UK
| | - David R Critchley
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
24
|
Kim LY, Thompson PM, Lee HT, Pershad M, Campbell SL, Alushin GM. The Structural Basis of Actin Organization by Vinculin and Metavinculin. J Mol Biol 2015; 428:10-25. [PMID: 26493222 DOI: 10.1016/j.jmb.2015.09.031] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/23/2015] [Accepted: 09/25/2015] [Indexed: 11/30/2022]
Abstract
Vinculin is an essential adhesion protein that links membrane-bound integrin and cadherin receptors through their intracellular binding partners to filamentous actin, facilitating mechanotransduction. Here we present an 8.5-Å-resolution cryo-electron microscopy reconstruction and pseudo-atomic model of the vinculin tail (Vt) domain bound to F-actin. Upon actin engagement, the N-terminal "strap" and helix 1 are displaced from the Vt helical bundle to mediate actin bundling. We find that an analogous conformational change also occurs in the H1' helix of the tail domain of metavinculin (MVt) upon actin binding, a muscle-specific splice isoform that suppresses actin bundling by Vt. These data support a model in which metavinculin tunes the actin bundling activity of vinculin in a tissue-specific manner, providing a mechanistic framework for understanding metavinculin mutations associated with hereditary cardiomyopathies.
Collapse
Affiliation(s)
- Laura Y Kim
- Laboratory of Macromolecular Interactions, Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter M Thompson
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hyunna T Lee
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mihir Pershad
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sharon L Campbell
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Gregory M Alushin
- Laboratory of Macromolecular Interactions, Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
25
|
Oztug Durer ZA, McGillivary RM, Kang H, Elam WA, Vizcarra CL, Hanein D, De La Cruz EM, Reisler E, Quinlan ME. Metavinculin Tunes the Flexibility and the Architecture of Vinculin-Induced Bundles of Actin Filaments. J Mol Biol 2015; 427:2782-98. [PMID: 26168869 DOI: 10.1016/j.jmb.2015.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/06/2015] [Accepted: 07/07/2015] [Indexed: 11/19/2022]
Abstract
Vinculin is an abundant protein found at cell-cell and cell-extracellular matrix junctions. In muscles, a longer splice isoform of vinculin, metavinculin, is also expressed. The metavinculin-specific insert is part of the C-terminal tail domain, the actin-binding site of both isoforms. Mutations in the metavinculin-specific insert are linked to heart disease such as dilated cardiomyopathies. Vinculin tail domain (VT) both binds and bundles actin filaments. Metavinculin tail domain (MVT) binds actin filaments in a similar orientation but does not bundle filaments. Recently, MVT was reported to sever actin filaments. In this work, we asked how MVT influences F-actin alone or in combination with VT. Cosedimentation and limited proteolysis experiments indicated a similar actin binding affinity and mode for both VT and MVT. In real-time total internal reflection fluorescence microscopy experiments, MVT's severing activity was negligible. Instead, we found that MVT binding caused a 2-fold reduction in F-actin's bending persistence length and increased susceptibility to breakage. Using mutagenesis and site-directed labeling with fluorescence probes, we determined that MVT alters actin interprotomer contacts and dynamics, which presumably reflect the observed changes in bending persistence length. Finally, we found that MVT decreases the density and thickness of actin filament bundles generated by VT. Altogether, our data suggest that MVT alters actin filament flexibility and tunes filament organization in the presence of VT. Both of these activities are potentially important for muscle cell function. Perhaps MVT allows the load of muscle contraction to act as a signal to reorganize actin filaments.
Collapse
Affiliation(s)
- Zeynep A Oztug Durer
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095-1569, USA
| | - Rebecca M McGillivary
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095-1569, USA
| | - Hyeran Kang
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520-8114, USA
| | - W Austin Elam
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520-8114, USA
| | - Christina L Vizcarra
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095-1569, USA
| | - Dorit Hanein
- Bioinformatics and Structural Biology Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Enrique M De La Cruz
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520-8114, USA
| | - Emil Reisler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095-1569, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095-1570, USA
| | - Margot E Quinlan
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095-1569, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095-1570, USA.
| |
Collapse
|
26
|
Dumbauld DW, García AJ. A helping hand: How vinculin contributes to cell-matrix and cell-cell force transfer. Cell Adh Migr 2015; 8:550-7. [PMID: 25482640 DOI: 10.4161/cam.29139] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Vinculin helps cells regulate and respond to mechanical forces. It is a scaffolding protein that tightly regulates its interactions with potential binding partners within adhesive structures-including focal adhesions that link the cell to the extracellular matrix and adherens junctions that link cells to each other-that physically connect the force-generating actin cytoskeleton (CSK) with the extracellular environment. This tight control of binding partner interaction-mediated by vinculin's autoinhibitory head-tail interaction-allows vinculin to rapidly interact and detach in response to changes in the dynamic forces applied through the cell. In doing so, vinculin modulates the structural composition of focal adhesions and the cell's ability to generate traction forces and adhesion strength. Recent evidence suggests that vinculin plays a similar role in regulating the fate and function of cell-cell junctions, further underscoring the importance of this protein. Using our lab's recent work as a starting point, this commentary explores several outstanding questions regarding the nature of vinculin activation and its function within focal adhesions and adherens junctions.
Collapse
Affiliation(s)
- David W Dumbauld
- a Woodruff School of Mechanical Engineering; Georgia Institute of Technology ; Atlanta , GA USA
| | | |
Collapse
|
27
|
Pimentel M, Morales W, Pokkunuri V, Brikos C, Kim SM, Kim SE, Triantafyllou K, Weitsman S, Marsh Z, Marsh E, Chua KS, Srinivasan S, Barlow GM, Chang C. Autoimmunity Links Vinculin to the Pathophysiology of Chronic Functional Bowel Changes Following Campylobacter jejuni Infection in a Rat Model. Dig Dis Sci 2015; 60:1195-1205. [PMID: 25424202 DOI: 10.1007/s10620-014-3435-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 11/11/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Acute gastroenteritis can precipitate irritable bowel syndrome (IBS) in humans. Cytolethal distending toxin is common to all pathogens causing gastroenteritis. Its active subunit, CdtB, is associated with post-infectious bowel changes in a rat model of Campylobacter jejuni infection, including small intestinal bacterial overgrowth (SIBO). AIM To evaluate the role of host antibodies to CdtB in contributing to post-infectious functional sequelae in this rat model. METHODS Ileal tissues from non-IBS human subjects, C. jejuni-infected and control rats were immunostained with antibodies to CdtB, c-Kit, S-100, PGP 9.5 and vinculin. Cytosolic and membrane proteins from mouse enteric neuronal cell lysates were immunoprecipitated with anti-CdtB and analyzed by mass spectrometry. ELISAs were performed on rat cardiac serum using CdtB or vinculin as antigens. RESULTS Anti-CdtB antibodies bound to a cytosolic protein in interstitial cells of Cajal (ICC) and myenteric ganglia in C. jejuni-infected and naïve rats and human subjects. Mass spectrometry identified vinculin, confirmed by co-localization and ELISAs. Anti-CdtB antibodies were higher in C. jejuni-infected rats (1.27 ± 0.15) than controls (1.76 ± 0.12) (P < 0.05), and rats that developed SIBO (2.01 ± 0.18) vs. rats that did not (1.44 ± 0.11) (P = 0.019). Vinculin expression levels were reduced in C. jejuni-infected rats (0.058 ± 0.053) versus controls (0.087 ± 0.023) (P = 0.0001), with greater reductions in rats with two C. jejuni infections (P = 0.0001) and rats that developed SIBO (P = 0.001). CONCLUSIONS Host anti-CdtB antibodies cross-react with vinculin in ICC and myenteric ganglia, required for normal gut motility. Circulating antibody levels and loss of vinculin expression correlate with number of C. jejuni exposures and SIBO, suggesting that effects on vinculin are important in the effects of C. jejuni infection on the host gut.
Collapse
Affiliation(s)
- Mark Pimentel
- GI Motility Program, Division of Gastroenterology, Cedars-Sinai Medical Center, 8730 Alden Drive, Suite 225E, Los Angeles, CA, 90048, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Chinthalapudi K, Patil DN, Rangarajan ES, Rader C, Izard T. Lipid-directed vinculin dimerization. Biochemistry 2015; 54:2758-68. [PMID: 25880222 DOI: 10.1021/acs.biochem.5b00015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Vinculin localizes to cellular adhesions where it regulates motility, migration, development, wound healing, and response to force. Importantly, vinculin loss results in cancer phenotypes, cardiovascular disease, and embryonic lethality. At the plasma cell membrane, the most abundant phosphoinositide, phosphatidylinositol 4,5-bisphosphate (PIP2), binds the vinculin tail domain, Vt, and triggers homotypic and heterotypic interactions that amplify binding of vinculin to the actin network. Binding of PIP2 to Vt is necessary for maintaining optimal focal adhesions, for organizing stress fibers, for cell migration and spreading, and for the control of vinculin dynamics and turnover of focal adhesions. While the recently determined Vt/PIP2 crystal structure revealed the conformational changes occurring upon lipid binding and oligomerization, characterization of PIP2-induced vinculin oligomerization has been challenging in the adhesion biology field. Here, via a series of novel biochemical assays not performed in previous studies that relied on chemical cross-linking, we characterize the PIP2-induced vinculin oligomerization. Our results show that Vt/PIP2 forms a tight dimer with Vt or with the muscle-specific vinculin isoform, metavinculin, at sites of adhesion at the cell membrane. Insight into how PIP2 regulates clustering and into mechanisms that regulate cell adhesion allows the development for a more definite sensor for PIP2, and our developed techniques can be applied generally and thus open the door for the characterization of many other protein/PIP2 complexes under physiological conditions.
Collapse
Affiliation(s)
- Krishna Chinthalapudi
- †Cell Adhesion Laboratory, ‡Department of Cancer Biology, and §Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Dipak N Patil
- †Cell Adhesion Laboratory, ‡Department of Cancer Biology, and §Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Erumbi S Rangarajan
- †Cell Adhesion Laboratory, ‡Department of Cancer Biology, and §Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Christoph Rader
- †Cell Adhesion Laboratory, ‡Department of Cancer Biology, and §Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Tina Izard
- †Cell Adhesion Laboratory, ‡Department of Cancer Biology, and §Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458, United States
| |
Collapse
|
29
|
Vinculin-dependent actin bundling regulates cell migration and traction forces. Biochem J 2015; 465:383-93. [PMID: 25358683 DOI: 10.1042/bj20140872] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Vinculin binding to actin filaments is thought to be critical for force transduction within a cell, but direct experimental evidence to support this conclusion has been limited. In the present study, we found mutation (R1049E) of the vinculin tail impairs its ability to bind F-actin, stimulate actin polymerization, and bundle F-actin in vitro. Further, mutant (R1049E) vinculin expressing cells are altered in cell migration, which is accompanied by changes in cell adhesion, cell spreading and cell generation of traction forces, providing direct evidence for the critical role of vinculin in mechanotransduction at adhesion sites. Lastly, we discuss the viability of models detailing the F-actin-binding surface on vinculin in the context of our mutational analysis.
Collapse
|
30
|
Chinthalapudi K, Rangarajan ES, Patil DN, George EM, Brown DT, Izard T. Lipid binding promotes oligomerization and focal adhesion activity of vinculin. ACTA ACUST UNITED AC 2015; 207:643-56. [PMID: 25488920 PMCID: PMC4259812 DOI: 10.1083/jcb.201404128] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PIP2 binds vinculin and directs its oligomerization, which promotes proper focal adhesion structure and function. Adherens junctions (AJs) and focal adhesion (FA) complexes are necessary for cell migration and morphogenesis, and for the development, growth, and survival of all metazoans. Vinculin is an essential regulator of both AJs and FAs, where it provides links to the actin cytoskeleton. Phosphatidylinositol 4,5-bisphosphate (PIP2) affects the functions of many targets, including vinculin. Here we report the crystal structure of vinculin in complex with PIP2, which revealed that PIP2 binding alters vinculin structure to direct higher-order oligomerization and suggests that PIP2 and F-actin binding to vinculin are mutually permissive. Forced expression of PIP2-binding–deficient mutants of vinculin in vinculin-null mouse embryonic fibroblasts revealed that PIP2 binding is necessary for maintaining optimal FAs, for organization of actin stress fibers, and for cell migration and spreading. Finally, photobleaching experiments indicated that PIP2 binding is required for the control of vinculin dynamics and turnover in FAs. Thus, through oligomerization, PIP2 directs a transient vinculin sequestration at FAs that is necessary for proper FA function.
Collapse
Affiliation(s)
- Krishna Chinthalapudi
- Cell Adhesion Laboratory, Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458
| | - Erumbi S Rangarajan
- Cell Adhesion Laboratory, Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458
| | - Dipak N Patil
- Cell Adhesion Laboratory, Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458
| | - Eric M George
- Department of Biochemistry and Department of Physiology, University of Mississippi Medical Center, Jackson, MS 39216 Department of Biochemistry and Department of Physiology, University of Mississippi Medical Center, Jackson, MS 39216
| | - David T Brown
- Department of Biochemistry and Department of Physiology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Tina Izard
- Cell Adhesion Laboratory, Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458
| |
Collapse
|
31
|
RhoGTPases - A novel link between cytoskeleton organization and cisplatin resistance. Drug Resist Updat 2015; 19:22-32. [PMID: 25660168 DOI: 10.1016/j.drup.2015.01.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 01/13/2015] [Accepted: 01/18/2015] [Indexed: 12/11/2022]
Abstract
For more than three decades, platinum compounds have been the first line treatment for a wide spectrum of solid tumors. Yet, cisplatin resistance is a major impediment in cancer therapy, and deciphering the mechanisms underlying chemoresistance is crucial for the development of novel therapies with enhanced efficacy. The Rho subfamily of small GTPases plays a significant role in cancer progression, and a growing body of evidence points toward the involvement of these proteins in anticancer drug resistance, including cisplatin resistance. The cycling between active and inactive states, governed by the balance between their GEFs, GAPs and GDIs, RhoGTPases, acts as molecular switches with a pivotal role in actin cytoskeleton organization. The Rho subfamily of proteins is involved in many key cellular processes including adhesion, vesicular trafficking, proliferation, survival, cell morphology and cell-matrix interactions. Although RhoA, RhoB and RhoC are highly homologous and share some upstream regulators and downstream effectors, they each have different roles in cancer progression and chemoresistance. While RhoA and RhoC are upregulated in many tumors and can stimulate transformation, RhoB appears to exhibit tumor suppressor characteristics with proapoptotic effects. In the current review, we discuss the role of Rho subfamily of proteins in cancer, and focus on their involvement in intrinsic and acquired drug resistance.
Collapse
|
32
|
Binding of the WASP/N-WASP-interacting protein WIP to actin regulates focal adhesion assembly and adhesion. Mol Cell Biol 2014; 34:2600-10. [PMID: 24797074 DOI: 10.1128/mcb.00017-14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The actin cytoskeleton is essential for cell adhesion and migration, functions important for tumor invasion. In addition to binding N-WASP/WASP, WIP binds and stabilizes F-actin. WIP(-/-) fibroblasts were used to test the role of WIP in F-actin function. WIP(-/-) cells had defective focal adhesion (FA), stress fiber assembly, and adherence to substrates, functions that were restored by transduction of wild-type WIP. Protein and mRNA levels of several FA constituents regulated by the myocardin-related transcription factor (MRTF)–serum response factor (SRF) transcription factor complex were reduced in WIP(-/-) fibroblasts. The level of G-actin, which sequesters MRTF in the cytoplasm, was increased, and nuclear localization of MRTF-A and SRF was reduced, in WIP(-/-) fibroblasts. Transfection of an MRTF-A mutant that constitutively translocates to the nucleus or transfection of constitutively active SRF restored FA and stress fiber assembly. Fibroblasts from knock-in mice expressing a WIP mutant that fails to bind actin phenocopied WIP(-/-) fibroblasts. Thus, WIP is a novel regulator of FA assembly and cell adhesion.
Collapse
|
33
|
Tolbert CE, Thompson PM, Superfine R, Burridge K, Campbell SL. Phosphorylation at Y1065 in vinculin mediates actin bundling, cell spreading, and mechanical responses to force. Biochemistry 2014; 53:5526-36. [PMID: 25115937 PMCID: PMC4151700 DOI: 10.1021/bi500678x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
![]()
Vinculin
is an essential structural adaptor protein that localizes
to sites of adhesion and is involved in a number of cell processes
including adhesion, spreading, motility, force transduction, and cell
survival. The C-terminal vinculin tail domain (Vt) contains the necessary
structural components to bind and cross-link actin filaments. Actin
binding to Vt induces a conformational change that promotes dimerization
through the C-terminal hairpin of Vt and enables actin filament cross-linking.
Here we show that Src phosphorylation of Y1065 within the C-terminal
hairpin regulates Vt-mediated actin bundling and provide a detailed
characterization of Y1065 mutations. Furthermore, we show that phosphorylation
at Y1065 plays a role in cell spreading and the response to the application
of mechanical force.
Collapse
Affiliation(s)
- Caitlin E Tolbert
- Department of Cell Biology and Physiology, ‡Department of Biochemistry and Biophysics, §Graduate Molecular and Cellular Biophysics Program, ∥Department of Physics and Astronomy, and ⊥the Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | | | | | | | | |
Collapse
|
34
|
Bachir AI, Zareno J, Moissoglu K, Plow EF, Gratton E, Horwitz AR. Integrin-associated complexes form hierarchically with variable stoichiometry in nascent adhesions. Curr Biol 2014; 24:1845-53. [PMID: 25088556 DOI: 10.1016/j.cub.2014.07.011] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 06/03/2014] [Accepted: 07/04/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND A complex network of putative molecular interactions underlies the architecture and function of cell-matrix adhesions. Most of these interactions are implicated from coimmunoprecipitation studies using expressed components, but few have been demonstrated or characterized functionally in living cells. RESULTS We introduce fluorescence fluctuation methods to determine, at high spatial and temporal resolution, "when" and "where" molecular complexes form and their stoichiometry in nascent adhesions (NAs). We focus on integrin-associated molecules implicated in integrin activation and in the integrin-actin linkage in NAs and show that these molecules form integrin-containing complexes hierarchically within the adhesion itself. Integrin and kindlin reside in a molecular complex as soon as adhesions are visible; talin, although also present early, associates with the integrin-kindlin complex only after NAs have formed and in response to myosin II activity. Furthermore, talin and vinculin association precedes the formation of the integrin-talin complex. Finally, α-actinin enters NAs periodically and in clusters that transiently associate with integrins. The absolute number and stoichiometry of these molecules varies among the molecules studied and changes as adhesions mature. CONCLUSIONS These observations suggest a working model for NA assembly whereby transient α-actinin-integrin complexes help nucleate NAs within the lamellipodium. Subsequently, integrin complexes containing kindlin, but not talin, emerge. Once NAs have formed, myosin II activity promotes talin association with the integrin-kindlin complex in a stoichiometry consistent with each talin molecule linking two integrin-kindlin complexes.
Collapse
Affiliation(s)
- Alexia I Bachir
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA.
| | - Jessica Zareno
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Konstadinos Moissoglu
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Edward F Plow
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Enrico Gratton
- Laboratory of Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Alan R Horwitz
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
35
|
Bays JL, Peng X, Tolbert CE, Guilluy C, Angell AE, Pan Y, Superfine R, Burridge K, DeMali KA. Vinculin phosphorylation differentially regulates mechanotransduction at cell-cell and cell-matrix adhesions. ACTA ACUST UNITED AC 2014; 205:251-63. [PMID: 24751539 PMCID: PMC4003237 DOI: 10.1083/jcb.201309092] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Vinculin phosphorylation on residue Y822 is necessary for cell stiffening in response to tension on cadherins but not integrins. Cells experience mechanical forces throughout their lifetimes. Vinculin is critical for transmitting these forces, yet how it achieves its distinct functions at cell–cell and cell–matrix adhesions remains unanswered. Here, we show vinculin is phosphorylated at Y822 in cell–cell, but not cell–matrix, adhesions. Phosphorylation at Y822 was elevated when forces were applied to E-cadherin and was required for vinculin to integrate into the cadherin complex. The mutation Y822F ablated these activities and prevented cells from stiffening in response to forces on E-cadherin. In contrast, Y822 phosphorylation was not required for vinculin functions in cell–matrix adhesions, including integrin-induced cell stiffening. Finally, forces applied to E-cadherin activated Abelson (Abl) tyrosine kinase to phosphorylate vinculin; Abl inhibition mimicked the loss of vinculin phosphorylation. These data reveal an unexpected regulatory mechanism in which vinculin Y822 phosphorylation determines whether cadherins transmit force and provides a paradigm for how a shared component of adhesions can produce biologically distinct functions.
Collapse
Affiliation(s)
- Jennifer L Bays
- Department of Biochemistry, University of Iowa Roy J. Carver College of Medicine, Iowa City, IA 52242
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Thompson PM, Tolbert CE, Shen K, Kota P, Palmer SM, Plevock KM, Orlova A, Galkin VE, Burridge K, Egelman EH, Dokholyan NV, Superfine R, Campbell SL. Identification of an actin binding surface on vinculin that mediates mechanical cell and focal adhesion properties. Structure 2014; 22:697-706. [PMID: 24685146 DOI: 10.1016/j.str.2014.03.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 02/04/2014] [Accepted: 03/04/2014] [Indexed: 11/30/2022]
Abstract
Vinculin, a cytoskeletal scaffold protein essential for embryogenesis and cardiovascular function, localizes to focal adhesions and adherens junctions, connecting cell surface receptors to the actin cytoskeleton. While vinculin interacts with many adhesion proteins, its interaction with filamentous actin regulates cell morphology, motility, and mechanotransduction. Disruption of this interaction lowers cell traction forces and enhances actin flow rates. Although a model for the vinculin:actin complex exists, we recently identified actin-binding deficient mutants of vinculin outside sites predicted to bind actin and developed an alternative model to better define this actin-binding surface, using negative-stain electron microscopy (EM), discrete molecular dynamics, and mutagenesis. Actin-binding deficient vinculin variants expressed in vinculin knockout fibroblasts fail to rescue cell-spreading defects and reduce cellular response to external force. These findings highlight the importance of this actin-binding surface and provide the molecular basis for elucidating additional roles of this interaction, including actin-induced conformational changes that promote actin bundling.
Collapse
Affiliation(s)
- Peter M Thompson
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Molecular and Cellular Biophysics Program, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Caitlin E Tolbert
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kai Shen
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Pradeep Kota
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Molecular and Cellular Biophysics Program, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sean M Palmer
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Molecular and Cellular Biophysics Program, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Karen M Plevock
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Molecular and Cellular Biophysics Program, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Albina Orlova
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Vitold E Galkin
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Keith Burridge
- 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
| | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Nikolay V Dokholyan
- Department of Biochemistry and Biophysics, 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
| | - Richard Superfine
- Department of Physics and Astronomy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sharon L Campbell
- Department of Biochemistry and Biophysics, 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.
| |
Collapse
|
37
|
Hagiwara M, Kokubu E, Sugiura S, Komatsu T, Tada H, Isoda R, Tanigawa N, Kato Y, Ishida N, Kobayashi K, Nakashima M, Ishihara K, Matsushita K. Vinculin and Rab5 complex is required [correction of requited]for uptake of Staphylococcus aureus and interleukin-6 expression. PLoS One 2014; 9:e87373. [PMID: 24466349 PMCID: PMC3900708 DOI: 10.1371/journal.pone.0087373] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/24/2013] [Indexed: 01/27/2023] Open
Abstract
Vinculin, a 116-kDa membrane cytoskeletal protein, is an important molecule for cell adhesion; however, little is known about its other cellular functions. Here, we demonstrated that vinculin binds to Rab5 and is required for Staphylococcus aureus (S. aureus) uptake in cells. Viunculin directly bound to Rab5 and enhanced the activation of S. aureus uptake. Over-expression of active vinculin mutants enhanced S. aureus uptake, whereas over-expression of an inactive vinculin mutant decreased S. aureus uptake. Vinculin bound to Rab5 at the N-terminal region (1-258) of vinculin. Vinculin and Rab5 were involved in the S. aureus-induced phosphorylation of MAP kinases (p38, Erk, and JNK) and IL-6 expression. Finally, vinculin and Rab5 knockdown reduced infection of S. aureus, phosphorylation of MAPKs and IL-6 expression in murine lungs. Our results suggest that vinculin binds to Rab5 and that these two molecules cooperatively enhance bacterial infection and the inflammatory response.
Collapse
Affiliation(s)
- Makoto Hagiwara
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Eitoyo Kokubu
- Department of Microbiology, Tokyo Dental College, Chiba, Japan
| | - Shinsuke Sugiura
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Toshinori Komatsu
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Hiroyuki Tada
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Ryutaro Isoda
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Naomi Tanigawa
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Yoshiko Kato
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Naoyuki Ishida
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Kaoru Kobayashi
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Misako Nakashima
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | | | - Kenji Matsushita
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
- * E-mail:
| |
Collapse
|
38
|
Wong MS, Sidik SM, Mahmud R, Stanslas J. Molecular targets in the discovery and development of novel antimetastatic agents: current progress and future prospects. Clin Exp Pharmacol Physiol 2013; 40:307-19. [PMID: 23534409 DOI: 10.1111/1440-1681.12083] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 03/18/2013] [Accepted: 03/21/2013] [Indexed: 01/08/2023]
Abstract
Tumour invasion and metastasis have been recognized as major causal factors in the morbidity and mortality among cancer patients. Many advances in the knowledge of cancer metastasis have yielded an impressive array of attractive drug targets, including enzymes, receptors and multiple signalling pathways. The present review summarizes the molecular pathogenesis of metastasis and the identification of novel molecular targets used in the discovery of antimetastatic agents. Several promising targets have been highlighted, including receptor tyrosine kinases, effector molecules involved in angiogenesis, matrix metalloproteinases (MMPs), urokinase plasminogen activator, adhesion molecules and their receptors, signalling pathways (e.g. phosphatidylinositol 3-kinase, phospholipase Cγ1, mitogen-activated protein kinases, c-Src kinase, c-Met kinases and heat shock protein. The discovery and development of potential novel therapeutics for each of the targets are also discussed in this review. Among these, the most promising agents that have shown remarkable clinical outcome are anti-angiogenic agents (e.g. bevacizumab). Newer agents, such as c-Met kinase inhibitors, are still undergoing preclinical studies and are yet to have their clinical efficacy proven. Some therapeutics, such as first-generation MMP inhibitors (MMPIs; e.g. marimastat) and more selective versions of them (e.g. prinomastat, tanomastat), have undergone clinical trials. Unfortunately, these drugs produced serious adverse effects that led to the premature termination of their development. In the future, third-generation MMPIs and inhibitors of signalling pathways and adhesion molecules could form valuable novel classes of drugs in the anticancer armamentarium to combat metastasis.
Collapse
Affiliation(s)
- Mei S Wong
- Pharmacotherapeutics Unit, Department of Medicine, University Putra Malaysia, Serdang, Selangor, Malaysia
| | | | | | | |
Collapse
|
39
|
Thievessen I, Thompson PM, Berlemont S, Plevock KM, Plotnikov SV, Zemljic-Harpf A, Ross RS, Davidson MW, Danuser G, Campbell SL, Waterman CM. Vinculin-actin interaction couples actin retrograde flow to focal adhesions, but is dispensable for focal adhesion growth. ACTA ACUST UNITED AC 2013; 202:163-77. [PMID: 23836933 PMCID: PMC3704983 DOI: 10.1083/jcb.201303129] [Citation(s) in RCA: 201] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Vinculin functions as a molecular clutch that organizes leading edge F-actin, generates traction, and promotes focal adhesion formation and turnover but not adhesion growth. In migrating cells, integrin-based focal adhesions (FAs) assemble in protruding lamellipodia in association with rapid filamentous actin (F-actin) assembly and retrograde flow. How dynamic F-actin is coupled to FA is not known. We analyzed the role of vinculin in integrating F-actin and FA dynamics by vinculin gene disruption in primary fibroblasts. Vinculin slowed F-actin flow in maturing FA to establish a lamellipodium–lamellum border and generate high extracellular matrix (ECM) traction forces. In addition, vinculin promoted nascent FA formation and turnover in lamellipodia and inhibited the frequency and rate of FA maturation. Characterization of a vinculin point mutant that specifically disrupts F-actin binding showed that vinculin–F-actin interaction is critical for these functions. However, FA growth rate correlated with F-actin flow speed independently of vinculin. Thus, vinculin functions as a molecular clutch, organizing leading edge F-actin, generating ECM traction, and promoting FA formation and turnover, but vinculin is dispensible for FA growth.
Collapse
Affiliation(s)
- Ingo Thievessen
- Laboratory of Cell and Tissue Morphodynamics, Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Morphological Cues for Regulation of Cell Adhesion and Motility with Tailored Electrospun Scaffolds of PCL and PCL/PVP Blends. Cell Mol Bioeng 2013. [DOI: 10.1007/s12195-013-0293-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
41
|
Saez de Guinoa J, Barrio L, Carrasco YR. Vinculin Arrests Motile B Cells by Stabilizing Integrin Clustering at the Immune Synapse. THE JOURNAL OF IMMUNOLOGY 2013; 191:2742-51. [DOI: 10.4049/jimmunol.1300684] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
42
|
The regulation and functional impact of actin assembly at cadherin cell–cell adhesions. Semin Cell Dev Biol 2013; 24:298-307. [DOI: 10.1016/j.semcdb.2012.12.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 10/25/2012] [Accepted: 12/14/2012] [Indexed: 11/17/2022]
|
43
|
Berginski ME, Gomez SM. The Focal Adhesion Analysis Server: a web tool for analyzing focal adhesion dynamics. F1000Res 2013; 2:68. [PMID: 24358855 PMCID: PMC3752736 DOI: 10.12688/f1000research.2-68.v1] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/01/2013] [Indexed: 12/02/2022] Open
Abstract
The Focal Adhesion Analysis Server (FAAS) is a web-based implementation of a set of computer vision algorithms designed to quantify the behavior of focal adhesions in cells imaged in 2D cultures. The input consists of one or more images of a labeled focal adhesion protein. The outputs of the system include a range of static and dynamic measurements for the adhesions present in each image as well as how these properties change over time. The user is able to adjust several parameters important for proper focal adhesion identification. This system provides a straightforward tool for the global, unbiased assessment of focal adhesion behavior common in optical microscopy studies. The webserver is available at:
http://faas.bme.unc.edu/.
Collapse
Affiliation(s)
- Matthew E Berginski
- UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7575, USA
| | - Shawn M Gomez
- UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7575, USA ; UNC Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7575, USA ; UNC Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7575, USA
| |
Collapse
|
44
|
Vinculin and metavinculin: Oligomerization and interactions with F-actin. FEBS Lett 2013; 587:1220-9. [DOI: 10.1016/j.febslet.2013.02.042] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 02/20/2013] [Accepted: 02/20/2013] [Indexed: 01/09/2023]
|
45
|
Prasad Gajula MNV, Vogel KP, Rai A, Dietrich F, Steinhoff HJ. How far in-silico computing meets real experiments. A study on the structure and dynamics of spin labeled vinculin tail protein by molecular dynamics simulations and EPR spectroscopy. BMC Genomics 2013; 14 Suppl 2:S4. [PMID: 23445506 PMCID: PMC3582443 DOI: 10.1186/1471-2164-14-s2-s4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Investigation of conformational changes in a protein is a prerequisite to understand its biological function. To explore these conformational changes in proteins we developed a strategy with the combination of molecular dynamics (MD) simulations and electron paramagnetic resonance (EPR) spectroscopy. The major goal of this work is to investigate how far computer simulations can meet the experiments. Methods Vinculin tail protein is chosen as a model system as conformational changes within the vinculin protein are believed to be important for its biological function at the sites of cell adhesion. MD simulations were performed on vinculin tail protein both in water and in vacuo environments. EPR experimental data is compared with those of the simulated data for corresponding spin label positions. Results The calculated EPR spectra from MD simulations trajectories of selected spin labelled positions are comparable to experimental EPR spectra. The results show that the information contained in the spin label mobility provides a powerful means of mapping protein folds and their conformational changes. Conclusions The results suggest the localization of dynamic and flexible regions of the vinculin tail protein. This study shows MD simulations can be used as a complementary tool to interpret experimental EPR data.
Collapse
Affiliation(s)
- M N V Prasad Gajula
- CABin division, DST Ramanujan Fellow, Indian Agricultural Statistics Research Institute, PUSA campus, New Delhi-110012, India.
| | | | | | | | | |
Collapse
|
46
|
Tolbert CE, Burridge K, Campbell SL. Vinculin regulation of F-actin bundle formation: what does it mean for the cell? Cell Adh Migr 2013; 7:219-25. [PMID: 23307141 DOI: 10.4161/cam.23184] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Vinculin is an essential cell adhesion protein, found at both focal adhesions and adherens junctions, where it couples transmembrane proteins to the actin cytoskeleton. Vinculin is involved in controlling cell shape, motility and cell survival, and has more recently been shown to play a role in force transduction. The tail domain of vinculin (Vt) has the ability to both bind and bundle actin filaments. Binding to actin induces a conformational change in Vt believed to promote formation of a Vt dimer that is able to crosslink actin filaments. We have recently provided additional evidence for the actin-induced Vt dimer and have shown that the vinculin carboxyl (C)-terminal hairpin is critical for both the formation of the Vt dimer and for bundling F-actin. We have also demonstrated the importance of the C-terminal hairpin in cells as deletion of this region impacts both adhesion properties and force transduction. Intriguingly, we have identified bundling deficient variants of vinculin that show different cellular phenotypes. These results suggest additional role(s) for the C-terminal hairpin, distinct from its bundling function. In this commentary, we will expand on our previous findings and further investigate these actin bundling deficient vinculin variants.
Collapse
Affiliation(s)
- Caitlin E Tolbert
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | |
Collapse
|
47
|
Actin cytoskeleton manipulation by effector proteins secreted by diarrheagenic Escherichia coli pathotypes. BIOMED RESEARCH INTERNATIONAL 2012; 2013:374395. [PMID: 23509714 PMCID: PMC3591105 DOI: 10.1155/2013/374395] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 10/22/2012] [Indexed: 11/18/2022]
Abstract
The actin cytoskeleton is a dynamic structure necessary for cell and tissue organization, including the maintenance of epithelial barriers. Disruption of the epithelial barrier coincides with alterations of the actin cytoskeleton in several disease states. These disruptions primarily affect the paracellular space, which is normally regulated by tight junctions. Thereby, the actin cytoskeleton is a common and recurring target of bacterial virulence factors. In order to manipulate the actin cytoskeleton, bacteria secrete and inject toxins and effectors to hijack the host cell machinery, which interferes with host-cell pathways and with a number of actin binding proteins. An interesting model to study actin manipulation by bacterial effectors is Escherichia coli since due to its genome plasticity it has acquired diverse genetic mobile elements, which allow having different E. coli varieties in one bacterial species. These E. coli pathotypes, including intracellular and extracellular bacteria, interact with epithelial cells, and their interactions depend on a specific combination of virulence factors. In this paper we focus on E. coli effectors that mimic host cell proteins to manipulate the actin cytoskeleton. The study of bacterial effector-cytoskeleton interaction will contribute not only to the comprehension of the molecular causes of infectious diseases but also to increase our knowledge of cell biology.
Collapse
|
48
|
Janssen MEW, Liu H, Volkmann N, Hanein D. The C-terminal tail domain of metavinculin, vinculin's splice variant, severs actin filaments. ACTA ACUST UNITED AC 2012; 197:585-93. [PMID: 22613835 PMCID: PMC3365496 DOI: 10.1083/jcb.201111046] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Relative to vinculin, a unique 68-residue insert in the C-terminal tail of metavinculin results in a loss of actin filament-bundling activity but gain of actin filament-severing activity. Vinculin and its splice variant, metavinculin (MV), are key elements of multiple protein assemblies linking the extracellular matrix to the actin cytoskeleton. Vinculin is expressed ubiquitously, whereas MV is mainly expressed in smooth and cardiac muscle tissue. The only difference in amino acid sequence between the isoforms is a 68-residue insert in the C-terminal tail domain of MV (MVt). Although the functional role of this insert remains elusive, its importance is exemplified by point mutations that are associated with dilated and hypertrophic cardiomyopathy. In vinculin, the actin binding site resides in the tail domain. In this paper, we show that MVt binds actin filaments similarly to the vinculin tail domain. Unlike its splice variant, MVt did not bundle actin filaments. Instead, MVt promoted severing of actin filaments, most efficiently at substoichiometric concentrations. This surprising and seemingly contradictory alteration of vinculin function by the 68-residue insert may be essential for modulating compliance of vinculin-induced actin bundles when exposed to rapidly increasing external forces.
Collapse
Affiliation(s)
- Mandy E W Janssen
- Bioinformatics and Systems Biology Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|