1
|
Almashwali A, Lal B, Khor SF. Methane Hydrate-in-Oil Systems in the Presence of Natural Amino Acid-Equilibrium Phase Condition Measurements. ACS OMEGA 2024; 9:47442-47452. [PMID: 39651083 PMCID: PMC11618404 DOI: 10.1021/acsomega.4c05430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/26/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024]
Abstract
This experimental study reports the thermodynamic influence of three different amino acids on methane hydrate in oil-dominated systems, namely, glycine, proline, and alanine. To thoroughly examine the effect of selected amino acids on methane (CH4) hydrate formation compared to the commercial inhibitor monoethylene glycol (MEG) in the presence of oil, the hydrate liquid-vapor equilibrium (H-Lw-Lo-V) curve is used to measure amino acid aqueous solutions. All experiments are performed at a concentration of 10 wt % by using the isochoric T-cycle technique in a high-pressure reactor cell at the selected range of pressures with temperatures of 4.0-9.0 MPa and 276.5-286.0 K, respectively. Results show that all studied amino acids inhibit hydrate formation of methane; the inhibition trend shows as glycine > alanine > proline in both systems; in the brine water system, the inhibition performance was higher than in the pure water system due to the presence of NaCl. Glycine showed the highest inhibition strength in both systems with an average reduced temperature in pure and brine water of 0.92 and 1.75 K, respectively, at 10 wt %, making the inhibition performance of glycine comparable to the commercial inhibitor MEG. The inhibition effect is attributed to the amino acid's hydrogen bonding energies and side group alkyl chain. Calculating the dissociation enthalpies of methane hydrates in the presence of amino acids using the Clausius-Clapeyron equation implies that the amino acids do not occupy the cage structures during methane hydrate formation.
Collapse
Affiliation(s)
- Abdulrab
Abdulwahab Almashwali
- Chemical
Engineering Department, Universiti Teknologi
PETRONAS, Bandar
Seri Iskandar, Perak 32610, Malaysia
- ISER
Institute of Sustainable Energy & Resources, Universiti Teknologi PETRONAS, Bandar Seri Iskandar, Perak 32610, Malaysia
| | - Bhajan Lal
- Chemical
Engineering Department, Universiti Teknologi
PETRONAS, Bandar
Seri Iskandar, Perak 32610, Malaysia
- ISER
Institute of Sustainable Energy & Resources, Universiti Teknologi PETRONAS, Bandar Seri Iskandar, Perak 32610, Malaysia
| | - Siak Foo Khor
- Chemical
Engineering Department, Universiti Teknologi
PETRONAS, Bandar
Seri Iskandar, Perak 32610, Malaysia
- ISER
Institute of Sustainable Energy & Resources, Universiti Teknologi PETRONAS, Bandar Seri Iskandar, Perak 32610, Malaysia
| |
Collapse
|
2
|
Franz F, Tapia-Rojo R, Winograd-Katz S, Boujemaa-Paterski R, Li W, Unger T, Albeck S, Aponte-Santamaria C, Garcia-Manyes S, Medalia O, Geiger B, Gräter F. Allosteric activation of vinculin by talin. Nat Commun 2023; 14:4311. [PMID: 37463895 DOI: 10.1038/s41467-023-39646-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 06/22/2023] [Indexed: 07/20/2023] Open
Abstract
The talin-vinculin axis is a key mechanosensing component of cellular focal adhesions. How talin and vinculin respond to forces and regulate one another remains unclear. By combining single-molecule magnetic tweezers experiments, Molecular Dynamics simulations, actin-bundling assays, and adhesion assembly experiments in live cells, we here describe a two-ways allosteric network within vinculin as a regulator of the talin-vinculin interaction. We directly observe a maturation process of vinculin upon talin binding, which reinforces the binding to talin at a rate of 0.03 s-1. This allosteric transition can compete with force-induced dissociation of vinculin from talin only at forces up to 10 pN. Mimicking the allosteric activation by mutation yields a vinculin molecule that bundles actin and localizes to focal adhesions in a force-independent manner. Hence, the allosteric switch confines talin-vinculin interactions and focal adhesion build-up to intermediate force levels. The 'allosteric vinculin mutant' is a valuable molecular tool to further dissect the mechanical and biochemical signalling circuits at focal adhesions and elsewhere.
Collapse
Affiliation(s)
- Florian Franz
- Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Mathematikon, INF 205, 69120, Heidelberg, Germany
| | - Rafael Tapia-Rojo
- Department of Physics, Randall Centre for Cell and Molecular Biophysics, Centre for the Physical Science of Life and London Centre for Nanotechnology, King's College London, Strand, WC2R 2LS London, UK.
- Single Molecule Mechanobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, London, UK.
| | - Sabina Winograd-Katz
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Wenhong Li
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Unger
- The Dana and Yossie Hollander Center for Structural Proteomics, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Albeck
- The Dana and Yossie Hollander Center for Structural Proteomics, Weizmann Institute of Science, Rehovot, Israel
| | - Camilo Aponte-Santamaria
- Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Mathematikon, INF 205, 69120, Heidelberg, Germany
| | - Sergi Garcia-Manyes
- Department of Physics, Randall Centre for Cell and Molecular Biophysics, Centre for the Physical Science of Life and London Centre for Nanotechnology, King's College London, Strand, WC2R 2LS London, UK
- Single Molecule Mechanobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, London, UK
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, 8057, Zurich, Switzerland.
| | - Benjamin Geiger
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Frauke Gräter
- Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany.
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Mathematikon, INF 205, 69120, Heidelberg, Germany.
- IMSEAM, Heidelberg University, INF 225, 69120, Heidelberg, Germany.
| |
Collapse
|
3
|
Bowers DT, McCulloch ME, Brown JL. Evaluation of focal adhesion mediated subcellular curvature sensing in response to engineered extracellular matrix. Biointerphases 2023; 18:021004. [PMID: 37019799 PMCID: PMC10079328 DOI: 10.1116/6.0002440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/24/2023] [Accepted: 03/14/2023] [Indexed: 04/07/2023] Open
Abstract
Fibril curvature is bioinstructive to attached cells. Similar to natural healthy tissues, an engineered extracellular matrix can be designed to stimulate cells to adopt desired phenotypes. To take full advantage of the curvature control in biomaterial fabrication methodologies, an understanding of the response to fibril subcellular curvature is required. In this work, we examined morphology, signaling, and function of human cells attached to electrospun nanofibers. We controlled curvature across an order of magnitude using nondegradable poly(methyl methacrylate) (PMMA) attached to a stiff substrate with flat PMMA as a control. Focal adhesion length and the distance of maximum intensity from the geographic center of the vinculin positive focal adhesion both peaked at a fiber curvature of 2.5 μm-1 (both ∼2× the flat surface control). Vinculin experienced slightly less tension when attached to nanofiber substrates. Vinculin expression was also more affected by a subcellular curvature than structural proteins α-tubulin or α-actinin. Among the phosphorylation sites we examined (FAK397, 576/577, 925, and Src416), FAK925 exhibited the most dependance on the nanofiber curvature. A RhoA/ROCK dependance of migration velocity across curvatures combined with an observation of cell membrane wrapping around nanofibers suggested a hybrid of migration modes for cells attached to fibers as has been observed in 3D matrices. Careful selection of nanofiber curvature for regenerative engineering scaffolds and substrates used to study cell biology is required to maximize the potential of these techniques for scientific exploration and ultimately improvement of human health.
Collapse
Affiliation(s)
- Daniel T. Bowers
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Mary Elizabeth McCulloch
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Justin L. Brown
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802
| |
Collapse
|
4
|
Pang Y, Thomas P. mPRα and PR co-operate in progesterone inhibition of endothelial cell focal adhesion. J Mol Endocrinol 2023; 70:JME-22-0073. [PMID: 36099054 DOI: 10.1530/jme-22-0073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 01/19/2023]
Abstract
Progesterone causes vascular smooth muscle cell relaxation through membrane progesterone receptors (mPRs), which are members of the progestin and adipoQ receptor (PAQR) family, and nuclear PRs (nPRs). However, beneficial vascular effects of progesterone in preventing pre-atherosclerosis and the involvement of mPRs and nPRs remain unclear. The results show short- to long-term treatments with 100 nM progesterone (P4) and specific agonists for mPRs, OD 02-0, and nPRs, R5020, inhibited pre-atherosclerotic events in human umbilical vein endothelial cells (HUVECs), decreasing focal adhesion (FA) by monocytes, FA signaling, HUVEC migration and invasion, and vinculin expression. Progesterone and OD 02-0, but not R5020, inhibited phosphorylation of Src and focal adhesion kinase, critical kinases of FA signaling, within 20 min and migration and invasion of HUVECs and monocyte adhesion after 3 h. These inhibitory P4 and 02-0 effects were attenuated with MAP kinase and Pi3k inhibitors, indicating involvement of these kinases in this mPR-mediated action. However, after 16 h, OD 02-0 was no longer effective in inhibiting FA signaling, while both progesterone and R5020 decreased the activity of the two kinases. Knockdown of receptor expression with siRNA confirmed that mPRα mediates short-term and nPR long-term inhibitory effects of progesterone on FA signaling. Thus, progesterone inhibition of FA signaling and pre-atherosclerosis is coordinated through mPRα and nPRs.
Collapse
Affiliation(s)
- Yefei Pang
- Marine Science Institute, University of Texas at Austin, Port Aransas, Texas, USA
| | - Peter Thomas
- Marine Science Institute, University of Texas at Austin, Port Aransas, Texas, USA
| |
Collapse
|
5
|
Akashi Y, Shimoo Y, Hashiguchi H, Nakajima K, Kokubun K, Matsuzaka K. Effects of Excimer Laser Treatment of Zirconia Disks on the Adhesion of L929 Fibroblasts. MATERIALS (BASEL, SWITZERLAND) 2022; 16:115. [PMID: 36614454 PMCID: PMC9821666 DOI: 10.3390/ma16010115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/07/2022] [Accepted: 12/19/2022] [Indexed: 06/17/2023]
Abstract
The adhesion of zirconia and soft tissue is very important for the success of zirconia implants. The aim of this study was to characterize the effects of excimer laser treatment of zirconia on the adhesion of L929 fibroblasts. In this study, polished zirconia disks treated with an excimer laser were the experimental group and untreated zirconia disks were the control group. Surface roughness and contact angles of zirconia disks were measured. mRNA expression levels of integrin β1 and collagen type I α1 in L929 fibroblasts cultured on zirconia disks were measured using qRT-PCR. Cell morphology was evaluated using 3D laser microscopy and the expression of vinculin was characterized using confocal microscopy. There was no significant difference in the surface roughness of zirconia disks, but contact angles were significantly lower. mRNA expression of integrin β1 was significantly higher at 3, 6 and 24 h and of collagen type I α1 was significantly higher at 6 and 24 h. L929 fibroblasts tended to form elongated microspikes and vinculin colocalization in those microspikes. Furthermore, vinculin was strongly expressed in filopodia of L929 fibroblasts at 24 h. These results suggest that excimer laser treatment improves adhesion between zirconia disks and L929 fibroblasts.
Collapse
Affiliation(s)
- Yoshihiko Akashi
- Department of Pathology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Yoshiaki Shimoo
- Department of Pathology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
- MALO DENTAL and MEDICAL TOKYO, FUKUHARA GINZA 8F, 7-8-10 Ginza, Chuo-ku, Tokyo 104-0061, Japan
| | - Hayato Hashiguchi
- Department of Pathology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Kei Nakajima
- Department of Pathology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Katsutoshi Kokubun
- Department of Pathology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Kenichi Matsuzaka
- Department of Pathology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| |
Collapse
|
6
|
Zhang H, Xie L, Chen S, Qiu Y, Sun Y, Kong W. Thyroxine Regulates the Opening of the Organ of Corti through Affecting P-Cadherin and Acetylated Microtubule. Int J Mol Sci 2022; 23:13339. [PMID: 36362134 PMCID: PMC9656988 DOI: 10.3390/ijms232113339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 10/10/2023] Open
Abstract
Different serum thyroxine levels may influence the morphology of the inner ear during development. A well-developed organ of Corti (OC) is considered to be critical to the function of hearing. In our study, we treated mice with triiodothyronine (T3) and found that the opening of the OC occurred sooner than in control mice. We also observed an increased formation of acetylated microtubules and a decrease in the adhesion junction molecule P-cadherin the during opening of the OC. Our investigation indicates that thyroxin affects P-cadherin expression and microtubule acetylation to influence the opening of the OC.
Collapse
Affiliation(s)
- Huimin Zhang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Le Xie
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Sen Chen
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yue Qiu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weijia Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
7
|
Costantini A, Mäkitie RE, Hartmann MA, Fratzl-Zelman N, Zillikens MC, Kornak U, Søe K, Mäkitie O. Early-Onset Osteoporosis: Rare Monogenic Forms Elucidate the Complexity of Disease Pathogenesis Beyond Type I Collagen. J Bone Miner Res 2022; 37:1623-1641. [PMID: 35949115 PMCID: PMC9542053 DOI: 10.1002/jbmr.4668] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/22/2022] [Accepted: 08/01/2022] [Indexed: 12/05/2022]
Abstract
Early-onset osteoporosis (EOOP), characterized by low bone mineral density (BMD) and fractures, affects children, premenopausal women and men aged <50 years. EOOP may be secondary to a chronic illness, long-term medication, nutritional deficiencies, etc. If no such cause is identified, EOOP is regarded primary and may then be related to rare variants in genes playing a pivotal role in bone homeostasis. If the cause remains unknown, EOOP is considered idiopathic. The scope of this review is to guide through clinical and genetic diagnostics of EOOP, summarize the present knowledge on rare monogenic forms of EOOP, and describe how analysis of bone biopsy samples can lead to a better understanding of the disease pathogenesis. The diagnostic pathway of EOOP is often complicated and extensive assessments may be needed to reliably exclude secondary causes. Due to the genetic heterogeneity and overlapping features in the various genetic forms of EOOP and other bone fragility disorders, the genetic diagnosis usually requires the use of next-generation sequencing to investigate several genes simultaneously. Recent discoveries have elucidated the complexity of disease pathogenesis both regarding genetic architecture and bone tissue-level pathology. Two rare monogenic forms of EOOP are due to defects in genes partaking in the canonical WNT pathway: LRP5 and WNT1. Variants in the genes encoding plastin-3 (PLS3) and sphingomyelin synthase 2 (SGMS2) have also been found in children and young adults with skeletal fragility. The molecular mechanisms leading from gene defects to clinical manifestations are often not fully understood. Detailed analysis of patient-derived transiliac bone biopsies gives valuable information to understand disease pathogenesis, distinguishes EOOP from other bone fragility disorders, and guides in patient management, but is not widely available in clinical settings. Despite the great advances in this field, EOOP remains an insufficiently explored entity and further research is needed to optimize diagnostic and therapeutic approaches. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Alice Costantini
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Paris Cité University, INSERM UMR1163, Institut Imagine, Paris, France
| | - Riikka E Mäkitie
- Folkhälsan Institute of Genetics, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Otorhinolaryngology-Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Markus A Hartmann
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria.,Vienna Bone and Growth Center, Vienna, Austria
| | - Nadja Fratzl-Zelman
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria.,Vienna Bone and Growth Center, Vienna, Austria
| | - M Carola Zillikens
- Bone Center, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Uwe Kornak
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Kent Søe
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense, Denmark.,Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Outi Mäkitie
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Folkhälsan Institute of Genetics, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Children's Hospital and Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
8
|
Fierro Morales JC, Xue Q, Roh-Johnson M. An evolutionary and physiological perspective on cell-substrate adhesion machinery for cell migration. Front Cell Dev Biol 2022; 10:943606. [PMID: 36092727 PMCID: PMC9453864 DOI: 10.3389/fcell.2022.943606] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Cell-substrate adhesion is a critical aspect of many forms of cell migration. Cell adhesion to an extracellular matrix (ECM) generates traction forces necessary for efficient migration. One of the most well-studied structures cells use to adhere to the ECM is focal adhesions, which are composed of a multilayered protein complex physically linking the ECM to the intracellular actin cytoskeleton. Much of our understanding of focal adhesions, however, is primarily derived from in vitro studies in Metazoan systems. Though these studies provide a valuable foundation to the cell-substrate adhesion field, the evolution of cell-substrate adhesion machinery across evolutionary space and the role of focal adhesions in vivo are largely understudied within the field. Furthering investigation in these areas is necessary to bolster our understanding of the role cell-substrate adhesion machinery across Eukaryotes plays during cell migration in physiological contexts such as cancer and pathogenesis. In this review, we review studies of cell-substrate adhesion machinery in organisms evolutionary distant from Metazoa and cover the current understanding and ongoing work on how focal adhesions function in single and collective cell migration in an in vivo environment, with an emphasis on work that directly visualizes cell-substrate adhesions. Finally, we discuss nuances that ought to be considered moving forward and the importance of future investigation in these emerging fields for application in other fields pertinent to adhesion-based processes.
Collapse
Affiliation(s)
| | | | - Minna Roh-Johnson
- Department of Biochemistry, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
9
|
Braun L, Schoen I, Vogel V. PIP 2-induced membrane binding of the vinculin tail competes with its other binding partners. Biophys J 2021; 120:4608-4622. [PMID: 34411575 DOI: 10.1016/j.bpj.2021.08.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/14/2021] [Accepted: 08/11/2021] [Indexed: 01/09/2023] Open
Abstract
Vinculin plays a key role during the first phase of focal adhesion formation and interacts with the plasma membrane through specific binding of its tail domain to the lipid phosphatidylinositol 4,5-bisphosphate (PIP2). Our understanding of the PIP2-vinculin interaction has been hampered by contradictory biochemical and structural data. Here, we used a multiscale molecular dynamics simulation approach, in which unbiased coarse-grained molecular dynamics were used to generate starting structures for subsequent microsecond-long all-atom simulations. This allowed us to map the interaction of the vinculin tail with PIP2-enriched membranes in atomistic detail. In agreement with experimental data, we have shown that membrane binding is sterically incompatible with the intramolecular interaction between vinculin's head and tail domain. Our simulations further confirmed biochemical and structural results, which identified two positively charged surfaces, the basic collar and the basic ladder, as the main PIP2 interaction sites. By introducing a valency-disaggregated binding network analysis, we were able to map the protein-lipid interactions in unprecedented detail. In contrast to the basic collar, in which PIP2 is specifically recognized by an up to hexavalent binding pocket, the basic ladder forms a series of low-valency binding sites. Importantly, many of these PIP2 binding residues are also involved in maintaining vinculin in a closed, autoinhibited conformation. These findings led us to propose a molecular mechanism for the coupling between vinculin activation and membrane binding. Finally, our refined binding site suggests an allosteric relationship between PIP2 and F-actin binding that disfavors simultaneous interaction with both ligands, despite nonoverlapping binding sites.
Collapse
Affiliation(s)
- Lukas Braun
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
| | - Ingmar Schoen
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Nishimura Y, Shi S, Li Q, Bershadsky AD, Viasnoff V. Crosstalk between myosin II and formin functions in the regulation of force generation and actomyosin dynamics in stress fibers. Cells Dev 2021; 168:203736. [PMID: 34455135 DOI: 10.1016/j.cdev.2021.203736] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/23/2021] [Accepted: 08/16/2021] [Indexed: 10/20/2022]
Abstract
REF52 fibroblasts have a well-developed contractile machinery, the most prominent elements of which are actomyosin stress fibers with highly ordered organization of actin and myosin IIA filaments. The relationship between contractile activity and turnover dynamics of stress fibers is not sufficiently understood. Here, we simultaneously measured the forces exerted by stress fibers (using traction force microscopy or micropillar array sensors) and the dynamics of actin and myosin (using photoconversion-based monitoring of actin incorporation and high-resolution fluorescence microscopy of myosin II light chain). Our data revealed new features of the crosstalk between myosin II-driven contractility and stress fiber dynamics. During normal stress fiber turnover, actin incorporated all along the stress fibers and not only at focal adhesions. Incorporation of actin into stress fibers/focal adhesions, as well as actin and myosin II filaments flow along stress fibers, strongly depends on myosin II activity. Myosin II-dependent generation of traction forces does not depend on incorporation of actin into stress fibers per se, but still requires formin activity. This previously overlooked function of formins in maintenance of the actin cytoskeleton connectivity could be the main mechanism of formin involvement in traction force generation.
Collapse
Affiliation(s)
- Yukako Nishimura
- Mechanobiology Institute, National University of Singapore, T-Lab, 5A Engineering Drive 1, 117411, Singapore; Division of Developmental Physiology, Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-0815, Japan
| | - Shidong Shi
- Mechanobiology Institute, National University of Singapore, T-Lab, 5A Engineering Drive 1, 117411, Singapore
| | - Qingsen Li
- IFOM-FIRC Institute of Molecular Oncology, Via Adamello, 16, 20139 Milan, Italy
| | - Alexander D Bershadsky
- Mechanobiology Institute, National University of Singapore, T-Lab, 5A Engineering Drive 1, 117411, Singapore; Department of Molecular Cell Biology, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot 7610001, Israel.
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, T-Lab, 5A Engineering Drive 1, 117411, Singapore; CNRS UMI 3639, Singapore; Department of Biological Sciences, National university of Singapore, S3 #05-01, 16 Science Drive 4, 117558, Singapore.
| |
Collapse
|
11
|
Hyaluronic Acid Treatment Improves Healing of the Tenorrhaphy Site by Suppressing Adhesions through Extracellular Matrix Remodeling in a Rat Model. Polymers (Basel) 2021; 13:polym13060928. [PMID: 33802991 PMCID: PMC8002636 DOI: 10.3390/polym13060928] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 11/17/2022] Open
Abstract
Due to the limited supply of vessels and nerves, acute or chronic tendon injuries often result in significant and persistent complications, such as pain and sprains, as well as the loss of joint functions. Among these complications, tendon adhesions within the surrounding soft tissue have been shown to significantly impair the range of motion. In this study, to elucidate the effects of a hyaluronic acid (HA) injection at the site of tenorrhaphy on tendon adhesion formation, we used a full transection model of a rat’s Achilles tendon to investigate the anti-adhesive function of HA. Our initial findings showed that significantly lower adhesion scores were observed in the HA-treated experimental group than in the normal saline-treated control group, as determined by macroscopic and histological evaluations. Hematoxylin and eosin, as well as picrosirius red staining, showed denser and irregular collagen fibers, with the larger number of infiltrating inflammatory cells in the control group indicating severe adhesion formation. Furthermore, we observed that the expression of tendon adhesion markers in operated tendon tissue, such as collagen type I, transforming growth factor-β1, and plasminogen activator inhibitor-1, was suppressed at both the gene and protein levels following HA treatment. These results suggest that HA injections could reduce tendon adhesion formation by significantly ameliorating inflammatory-associated reactions.
Collapse
|
12
|
Panda PP, Bohot M, Chaturvedi MM, Purohit JS. Purification and partial characterization of vinculin from chicken liver nuclear extract. Biologia (Bratisl) 2021. [DOI: 10.1007/s11756-021-00691-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
13
|
Chen H, Wang T, Yang J, Huang S, Zeng P. Improved Detection of Potentially Pleiotropic Genes in Coronary Artery Disease and Chronic Kidney Disease Using GWAS Summary Statistics. Front Genet 2020; 11:592461. [PMID: 33343632 PMCID: PMC7744760 DOI: 10.3389/fgene.2020.592461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022] Open
Abstract
The coexistence of coronary artery disease (CAD) and chronic kidney disease (CKD) implies overlapped genetic foundation. However, the common genetic determination between the two diseases remains largely unknown. Relying on summary statistics publicly available from large scale genome-wide association studies (n = 184,305 for CAD and n = 567,460 for CKD), we observed significant positive genetic correlation between CAD and CKD (rg = 0.173, p = 0.024) via the linkage disequilibrium score regression. Next, we implemented gene-based association analysis for each disease through MAGMA (Multi-marker Analysis of GenoMic Annotation) and detected 763 and 827 genes associated with CAD or CKD (FDR < 0.05). Among those 72 genes were shared between the two diseases. Furthermore, by integrating the overlapped genetic information between CAD and CKD, we implemented two pleiotropy-informed informatics approaches including cFDR (conditional false discovery rate) and GPA (Genetic analysis incorporating Pleiotropy and Annotation), and identified 169 and 504 shared genes (FDR < 0.05), of which 121 genes were simultaneously discovered by cFDR and GPA. Importantly, we found 11 potentially new pleiotropic genes related to both CAD and CKD (i.e., ARHGEF19, RSG1, NDST2, CAMK2G, VCL, LRP10, RBM23, USP10, WNT9B, GOSR2, and RPRML). Five of the newly identified pleiotropic genes were further repeated via an additional dataset CAD available from UK Biobank. Our functional enrichment analysis showed that those pleiotropic genes were enriched in diverse relevant pathway processes including quaternary ammonium group transmembrane transporter, dopamine transport. Overall, this study identifies common genetic architectures overlapped between CAD and CKD and will help to advance understanding of the molecular mechanisms underlying the comorbidity of the two diseases.
Collapse
Affiliation(s)
- Haimiao Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Ting Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Jinna Yang
- Department of Infectious Diseases, People's Hospital of Zhuji, Shaoxing, China
| | - Shuiping Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China.,Center for Medical Statistics and Data Analysis, School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Ping Zeng
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China.,Center for Medical Statistics and Data Analysis, School of Public Health, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
14
|
Boujemaa-Paterski R, Martins B, Eibauer M, Beales CT, Geiger B, Medalia O. Talin-activated vinculin interacts with branched actin networks to initiate bundles. eLife 2020; 9:e53990. [PMID: 33185186 PMCID: PMC7682986 DOI: 10.7554/elife.53990] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 11/12/2020] [Indexed: 12/18/2022] Open
Abstract
Vinculin plays a fundamental role in integrin-mediated cell adhesion. Activated by talin, it interacts with diverse adhesome components, enabling mechanical coupling between the actin cytoskeleton and the extracellular matrix. Here we studied the interactions of activated full-length vinculin with actin and the way it regulates the organization and dynamics of the Arp2/3 complex-mediated branched actin network. Through a combination of surface patterning and light microscopy experiments we show that vinculin can bundle dendritic actin networks through rapid binding and filament crosslinking. We show that vinculin promotes stable but flexible actin bundles having a mixed-polarity organization, as confirmed by cryo-electron tomography. Adhesion-like synthetic design of vinculin activation by surface-bound talin revealed that clustered vinculin can initiate and immobilize bundles from mobile Arp2/3-branched networks. Our results provide a molecular basis for coordinate actin bundle formation at nascent adhesions.
Collapse
Affiliation(s)
- Rajaa Boujemaa-Paterski
- Department of Biochemistry, University of ZurichZurichSwitzerland
- Université Grenoble AlpesGrenobleFrance
| | - Bruno Martins
- Department of Biochemistry, University of ZurichZurichSwitzerland
| | - Matthias Eibauer
- Department of Biochemistry, University of ZurichZurichSwitzerland
| | - Charlie T Beales
- Department of Biochemistry, University of ZurichZurichSwitzerland
| | - Benjamin Geiger
- Department of Immunology, Weizmann Institute of ScienceRehovotIsrael
| | - Ohad Medalia
- Department of Biochemistry, University of ZurichZurichSwitzerland
| |
Collapse
|
15
|
Microinjection of Fluorescently Labeled 130K Protein into Living Fibroblasts: Localization at the Ends of Actin Microfilament Bundles and in Close Relation to Fibronectin. ACTA ACUST UNITED AC 2020. [DOI: 10.1017/s143192760000310x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Understanding the interactions between cell surface components and the underlying cytoskeleton continues to be a major goal in cell biology. In particular, little is known about the molecules involved in linking actin filaments to the plasma membrane in cells such as fibroblasts. A protein with a molecular weight of 130,000 (the 130K protein) was recently purified from smooth muscle by Geiger and localized to the ends of microfilament bundles in a variety of cell types. Independently we had purified this protein and were studying its possible interactions with other structural proteins when we learned of its interesting location, one that would be consistent with some role in the attachment of actin filaments to the plasma membrane. We were prompted to investigate this interesting protein further by microinjection of the f1uorescent1y labeled protein into living cells, a technique which had recently been successfully applied to the study of α-actinin.
Collapse
|
16
|
Miller AE, Hu P, Barker TH. Feeling Things Out: Bidirectional Signaling of the Cell-ECM Interface, Implications in the Mechanobiology of Cell Spreading, Migration, Proliferation, and Differentiation. Adv Healthc Mater 2020; 9:e1901445. [PMID: 32037719 PMCID: PMC7274903 DOI: 10.1002/adhm.201901445] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/10/2020] [Indexed: 12/16/2022]
Abstract
Biophysical cues stemming from the extracellular environment are rapidly transduced into discernible chemical messages (mechanotransduction) that direct cellular activities-placing the extracellular matrix (ECM) as a potent regulator of cell behavior. Dynamic reciprocity between the cell and its associated matrix is essential to the maintenance of tissue homeostasis and dysregulation of both ECM mechanical signaling, via pathological ECM turnover, and internal mechanotransduction pathways contribute to disease progression. This review covers the current understandings of the key modes of signaling used by both the cell and ECM to coregulate one another. By taking an outside-in approach, the inherent complexities and regulatory processes at each level of signaling (ECM, plasma membrane, focal adhesion, and cytoplasm) are captured to give a comprehensive picture of the internal and external mechanoregulatory environment. Specific emphasis is placed on the focal adhesion complex which acts as a central hub of mechanical signaling, regulating cell spreading, migration, proliferation, and differentiation. In addition, a wealth of available knowledge on mechanotransduction is curated to generate an integrated signaling network encompassing the central components of the focal adhesion, cytoplasm and nucleus that act in concert to promote durotaxis, proliferation, and differentiation in a stiffness-dependent manner.
Collapse
Affiliation(s)
- Andrew E Miller
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| | - Ping Hu
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| |
Collapse
|
17
|
Kluger C, Braun L, Sedlak SM, Pippig DA, Bauer MS, Miller K, Milles LF, Gaub HE, Vogel V. Different Vinculin Binding Sites Use the Same Mechanism to Regulate Directional Force Transduction. Biophys J 2020; 118:1344-1356. [PMID: 32109366 PMCID: PMC7091509 DOI: 10.1016/j.bpj.2019.12.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/17/2019] [Accepted: 12/30/2019] [Indexed: 12/18/2022] Open
Abstract
Vinculin is a universal adaptor protein that transiently reinforces the mechanical stability of adhesion complexes. It stabilizes mechanical connections that cells establish between the actomyosin cytoskeleton and the extracellular matrix via integrins or to neighboring cells via cadherins, yet little is known regarding its mechanical design. Vinculin binding sites (VBSs) from different nonhomologous actin-binding proteins use conserved helical motifs to associate with the vinculin head domain. We studied the mechanical stability of such complexes by pulling VBS peptides derived from talin, α-actinin, and Shigella IpaA out of the vinculin head domain. Experimental data from atomic force microscopy single-molecule force spectroscopy and steered molecular dynamics (SMD) simulations both revealed greater mechanical stability of the complex for shear-like than for zipper-like pulling configurations. This suggests that reinforcement occurs along preferential force directions, thus stabilizing those cytoskeletal filament architectures that result in shear-like pulling geometries. Large force-induced conformational changes in the vinculin head domain, as well as protein-specific fine-tuning of the VBS sequence, including sequence inversion, allow for an even more nuanced force response.
Collapse
Affiliation(s)
- Carleen Kluger
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lukas Braun
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Steffen M Sedlak
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Diana A Pippig
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Magnus S Bauer
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ken Miller
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lukas F Milles
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hermann E Gaub
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
18
|
Khan RB, Goult BT. Adhesions Assemble!-Autoinhibition as a Major Regulatory Mechanism of Integrin-Mediated Adhesion. Front Mol Biosci 2019; 6:144. [PMID: 31921890 PMCID: PMC6927945 DOI: 10.3389/fmolb.2019.00144] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/26/2019] [Indexed: 01/14/2023] Open
Abstract
The advent of cell-cell and cell-extracellular adhesion enabled cells to interact in a coherent manner, forming larger structures and giving rise to the development of tissues, organs and complex multicellular life forms. The development of such organisms required tight regulation of dynamic adhesive structures by signaling pathways that coordinate cell attachment. Integrin-mediated adhesion to the extracellular matrix provides cells with support, survival signals and context-dependent cues that enable cells to run different cellular programs. One mysterious aspect of the process is how hundreds of proteins assemble seemingly spontaneously onto the activated integrin. An emerging concept is that adhesion assembly is regulated by autoinhibition of key proteins, a highly dynamic event that is modulated by a variety of signaling events. By enabling precise control of the activation state of proteins, autoinhibition enables localization of inactive proteins and the formation of pre-complexes. In response to the correct signals, these proteins become active and interact with other proteins, ultimately leading to development of cell-matrix junctions. Autoinhibition of key components of such adhesion complexes—including core components integrin, talin, vinculin, and FAK and important peripheral regulators such as RIAM, Src, and DLC1—leads to a view that the majority of proteins involved in complex assembly might be regulated by intramolecular interactions. Autoinhibition is relieved via multiple different signals including post-translation modification and proteolysis. More recently, mechanical forces have been shown to stabilize and increase the lifetimes of active conformations, identifying autoinhibition as a means of encoding mechanosensitivity. The complexity and scope for nuanced adhesion dynamics facilitated via autoinhibition provides numerous points of regulation. In this review, we discuss what is known about this mode of regulation and how it leads to rapid and tightly controlled assembly and disassembly of cell-matrix adhesion.
Collapse
Affiliation(s)
- Rejina B Khan
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
19
|
Krüger-Genge A, Blocki A, Franke RP, Jung F. Vascular Endothelial Cell Biology: An Update. Int J Mol Sci 2019; 20:ijms20184411. [PMID: 31500313 PMCID: PMC6769656 DOI: 10.3390/ijms20184411] [Citation(s) in RCA: 641] [Impact Index Per Article: 106.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022] Open
Abstract
The vascular endothelium, a monolayer of endothelial cells (EC), constitutes the inner cellular lining of arteries, veins and capillaries and therefore is in direct contact with the components and cells of blood. The endothelium is not only a mere barrier between blood and tissues but also an endocrine organ. It actively controls the degree of vascular relaxation and constriction, and the extravasation of solutes, fluid, macromolecules and hormones, as well as that of platelets and blood cells. Through control of vascular tone, EC regulate the regional blood flow. They also direct inflammatory cells to foreign materials, areas in need of repair or defense against infections. In addition, EC are important in controlling blood fluidity, platelet adhesion and aggregation, leukocyte activation, adhesion, and transmigration. They also tightly keep the balance between coagulation and fibrinolysis and play a major role in the regulation of immune responses, inflammation and angiogenesis. To fulfill these different tasks, EC are heterogeneous and perform distinctly in the various organs and along the vascular tree. Important morphological, physiological and phenotypic differences between EC in the different parts of the arterial tree as well as between arteries and veins optimally support their specified functions in these vascular areas. This review updates the current knowledge about the morphology and function of endothelial cells, particularly their differences in different localizations around the body paying attention specifically to their different responses to physical, biochemical and environmental stimuli considering the different origins of the EC.
Collapse
Affiliation(s)
- Anne Krüger-Genge
- Department of Biomaterials and Healthcare, Division of Life Science and Bioprocesses, Fraunhofer Institute for Applied Polymer Research (IAP), Potsdam-Golm 14476, Germany.
- Department of Anesthesia, Pain Management and Perioperative Medicine, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 2Y9, Canada.
| | - Anna Blocki
- Institute for Tissue Engineering and Regenerative Medicine (ITERM), School of Biomedical Sciences (SBS), Chinese University of Hong Kong (CUHK), New Territories, Hong Kong, China
| | - Ralf-Peter Franke
- Central Institute for Biomedical Technology, Dep. Biomaterials, University of Ulm, Albert-Einstein-Allee 47, 89081 Ulm, Germany
| | - Friedrich Jung
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology, 01968 Senftenberg, Germany
| |
Collapse
|
20
|
Konishi S, Yano T, Tanaka H, Mizuno T, Kanoh H, Tsukita K, Namba T, Tamura A, Yonemura S, Gotoh S, Matsumoto H, Hirai T, Tsukita S. Vinculin is critical for the robustness of the epithelial cell sheet paracellular barrier for ions. Life Sci Alliance 2019; 2:2/4/e201900414. [PMID: 31399484 PMCID: PMC6689668 DOI: 10.26508/lsa.201900414] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022] Open
Abstract
Vinculin in the apical junctional complex maintains the paracellular barrier function specifically for ions, but not for large solutes, by buffering mechanical fluctuations. The paracellular barrier function of tight junctions (TJs) in epithelial cell sheets is robustly maintained against mechanical fluctuations, by molecular mechanisms that are poorly understood. Vinculin is an adaptor of a mechanosensory complex at the adherens junction. Here, we generated vinculin KO Eph4 epithelial cells and analyzed their confluent cell-sheet properties. We found that vinculin is dispensable for the basic TJ structural integrity and the paracellular barrier function for larger solutes. However, vinculin is indispensable for the paracellular barrier function for ions. In addition, TJs stochastically showed dynamically distorted patterns in vinculin KO cell sheets. These KO phenotypes were rescued by transfecting full-length vinculin and by relaxing the actomyosin tension with blebbistatin, a myosin II ATPase activity inhibitor. Our findings indicate that vinculin resists mechanical fluctuations to maintain the TJ paracellular barrier function for ions in epithelial cell sheets.
Collapse
Affiliation(s)
- Satoshi Konishi
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoki Yano
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroo Tanaka
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Pharmacology, Teikyo University, Tokyo, Japan.,Strategic Innovation and Research Center, Teikyo University, Tokyo, Japan
| | - Tomoaki Mizuno
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hatsuho Kanoh
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Kazuto Tsukita
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshinori Namba
- Graduate School of Arts and Sciences, Tokyo University, Tokyo, Japan
| | - Atsushi Tamura
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Pharmacology, Teikyo University, Tokyo, Japan.,Strategic Innovation and Research Center, Teikyo University, Tokyo, Japan
| | - Shigenobu Yonemura
- Department of Cell Biology, Tokushima University Graduate School of Medical Science, Tokushima, Japan.,Laboratory for Ultrastructural Research, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Shimpei Gotoh
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hisako Matsumoto
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sachiko Tsukita
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka, Japan .,Strategic Innovation and Research Center, Teikyo University, Tokyo, Japan
| |
Collapse
|
21
|
ROCK Inhibitor-Induced Promotion of Retinal Pigment Epithelial Cell Motility during Wound Healing. J Ophthalmol 2019; 2019:9428738. [PMID: 31316826 PMCID: PMC6607728 DOI: 10.1155/2019/9428738] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 12/12/2022] Open
Abstract
Purpose No standard therapy for RPE tear, a complication of neovascular age-related macular degeneration, exists even though RPE tears cause severe vision loss, and promotion of cell proliferation and/or migration could be a candidate RPE tear therapy. The aim of this study is to evaluate the effect of Rho-associated coiled-coil containing kinase (ROCK) inhibitor Y27632 on retinal pigment epithelial (RPE) cell motility during wound healing. Methods Human RPE cells were cultured in media with and without 10 μM Y27632. A luminescent cell viability assay and vinculin immunocytochemistry were used to test the Y27632 effect on RPE cell adhesion. The mean size of vinculin puncta was quantified from immunofluorescence images. RPE cell motility during wound healing was evaluated using time-lapse imaging and measuring cell migration distances and cell coverage rate in wound fields. Results The number of adhered RPE and mean size of vinculin puncta were, respectively, 20519 cells and 3.65 μm2 under nontreatment and 23569 cells and 0.66 μm2 under Y27632 treatment. Cell migration distance and cell coverage percentage for untreated and Y27632-treated cells were 98.9 and 59.4% and 203.4 and 92.5%, respectively. Conclusions Inhibition of ROCK signaling by using 10 μM Y27632 promoted RPE cell motility during wound healing by reducing RPE cell adhesion strength.
Collapse
|
22
|
Sit B, Gutmann D, Iskratsch T. Costameres, dense plaques and podosomes: the cell matrix adhesions in cardiovascular mechanosensing. J Muscle Res Cell Motil 2019; 40:197-209. [PMID: 31214894 PMCID: PMC6726830 DOI: 10.1007/s10974-019-09529-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/15/2019] [Indexed: 12/12/2022]
Abstract
The stiffness of the cardiovascular environment changes during ageing and in disease and contributes to disease incidence and progression. For instance, increased arterial stiffness can lead to atherosclerosis, while stiffening of the heart due to fibrosis can increase the chances of heart failure. Cells can sense the stiffness of the extracellular matrix through integrin adhesions and other mechanosensitive structures and in response to this initiate mechanosignalling pathways that ultimately change the cellular behaviour. Over the past decades, interest in mechanobiology has steadily increased and with this also our understanding of the molecular basis of mechanosensing and transduction. However, much of our knowledge about the mechanisms is derived from studies investigating focal adhesions in non-muscle cells, which are distinct in several regards from the cell-matrix adhesions in cardiomyocytes (costameres) or vascular smooth muscle cells (dense plaques or podosomes). Therefore, we will look here first at the evidence for mechanical sensing in the cardiovascular system, before comparing the different cytoskeletal arrangements and adhesion sites in cardiomyocytes and vascular smooth muscle cells and what is known about mechanical sensing through the various structures.
Collapse
Affiliation(s)
- Brian Sit
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, London, UK
| | - Daniel Gutmann
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, London, UK
| | - Thomas Iskratsch
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, London, UK.
| |
Collapse
|
23
|
Jalal S, Shi S, Acharya V, Huang RYJ, Viasnoff V, Bershadsky AD, Tee YH. Actin cytoskeleton self-organization in single epithelial cells and fibroblasts under isotropic confinement. J Cell Sci 2019; 132:jcs.220780. [PMID: 30787030 PMCID: PMC6432717 DOI: 10.1242/jcs.220780] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 01/24/2019] [Indexed: 12/23/2022] Open
Abstract
Actin cytoskeleton self-organization in two cell types, fibroblasts and epitheliocytes, was studied in cells confined to isotropic adhesive islands. In fibroblasts plated onto islands of optimal size, an initially circular actin pattern evolves into a radial pattern of actin bundles that undergo asymmetric chiral swirling before finally producing parallel linear stress fibers. Epitheliocytes, however, did not exhibit succession through all the actin patterns described above. Upon confinement, the actin cytoskeleton in non-keratinocyte epitheliocytes was arrested at the circular stage, while in keratinocytes it progressed as far as the radial pattern but still could not break symmetry. Epithelial–mesenchymal transition pushed actin cytoskeleton development from circular towards radial patterns but remained insufficient to cause chirality. Knockout of cytokeratins also did not promote actin chirality development in keratinocytes. Left–right asymmetric cytoskeleton swirling could, however, be induced in keratinocytes by treatment with small doses of the G-actin sequestering drug, latrunculin A in a transcription-independent manner. Both the nucleus and the cytokeratin network followed the induced chiral swirling. Development of chirality in keratinocytes was controlled by DIAPH1 (mDia1) and VASP, proteins involved in regulation of actin polymerization. This article has an associated First Person interview with the first author of the paper. Summary: Epitheliocytes cannot develop the F-actin patterns typically observed in fibroblasts, but can do so after treatments affecting actin polymerization. Regulators of actin polymerization, DIAPH1 and VASP, control this process.
Collapse
Affiliation(s)
- Salma Jalal
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Shidong Shi
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | | | - Ruby Yun-Ju Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599.,Department of Obstetrics & Gynaecology, National University Hospital, Singapore 119228.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, Singapore 117411.,Centre National Pour la Recherche Scientifique, Singapore 117411.,Department of Biological Sciences, National University of Singapore, Singapore 117558
| | - Alexander D Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore 117411 .,Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yee Han Tee
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| |
Collapse
|
24
|
Krüger-Genge A, Dietze S, Yan W, Liu Y, Fang L, Kratz K, Lendlein A, Jung F. Endothelial cell migration, adhesion and proliferation on different polymeric substrates. Clin Hemorheol Microcirc 2019; 70:511-529. [DOI: 10.3233/ch-189317] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Anne Krüger-Genge
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Stefanie Dietze
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Wan Yan
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Yue Liu
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Liang Fang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Karl Kratz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Friedrich Jung
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| |
Collapse
|
25
|
Lynch AM, Wagner BD, Weiss SJ, Wall KM, Palestine AG, Mathias MT, Siringo FS, Cathcart JN, Patnaik JL, Drolet DW, Janjic N, Mandava N. Proteomic Profiles in Advanced Age-Related Macular Degeneration Using an Aptamer-Based Proteomic Technology. Transl Vis Sci Technol 2019; 8:14. [PMID: 30697465 PMCID: PMC6348995 DOI: 10.1167/tvst.8.1.14] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 12/02/2018] [Indexed: 12/12/2022] Open
Abstract
Purpose To explore top-ranked plasma proteins related to neovascular age-related macular degeneration (AMD) and geographic atrophy (GA), and explore pathways related to neovascular AMD and GA. Methods We conducted a pilot study of patients with neovascular AMD (n = 10), GA (n = 10), and age-matched cataract controls (n = 10) who were recruited into an AMD registry. We measured 4001 proteins in ethylenediaminetetraacetic acid plasma samples using an aptamer-based proteomic technology. Relative concentrations of each of 4001 proteins were log (base 2) transformed and compared between cases of neovascular AMD and GA versus controls using linear regression. Pathway analysis was conducted using pathways downloaded from Reactome. Results In this pilot study, higher levels of vinculin and lower levels of CD177 were found in patients with neovascular AMD compared with controls. Neuregulin-4 was higher and soluble intercellular adhesion molecule-1 was lower in patients with GA compared with controls. For neovascular AMD, cargo trafficking to the periciliary membrane, fibroblast growth factor receptor 3b ligand binding and activation, and vascular endothelial growth factor–related pathways were in the top ranked pathways. The top-ranked pathways for GA included several related to ErbB4 signaling. Conclusions We found different proteins and different pathways associated with neovascular AMD and GA. Vinculin and some of the top-ranked pathways have been previously associated with AMD, whereas others have not been described. Translational Relevance Biomarkers identified in plasma likely reflect systemic alterations in protein expression and may improve our understanding of the mechanisms leading to AMD.
Collapse
Affiliation(s)
- Anne M Lynch
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Brandie D Wagner
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | | | | | - Alan G Palestine
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marc T Mathias
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Frank S Siringo
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jennifer N Cathcart
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jennifer L Patnaik
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA
| | | | | | - Naresh Mandava
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
26
|
Boulter E, Tissot FS, Dilly J, Pisano S, Féral CC. Cyclic uniaxial mechanical stretching of cells using a LEGO® parts-based mechanical stretcher system. J Cell Sci 2019; 133:jcs.234666. [DOI: 10.1242/jcs.234666] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 12/01/2019] [Indexed: 12/27/2022] Open
Abstract
Mechanical cues are essential to the regulation of cell and tissue physiology. Henceforth, it has become an utmost necessity for cell biologists to account for those mechanical parameters when investigating biological processes and they need devices to manipulate cells accordingly. Here, we report a simple mechanical cell stretching system that can generate uniaxial cyclic mechanical stretch on cells in tissue culture. This system is based upon a low-cost battery-powered uniaxial cyclic mechanical stretcher exclusively built out of LEGO® parts combined to a stretchable PDMS tissue culture plate in order to grow and stretch cells. We characterize the system and show that it can be used with a wide variety of downstream applications including immunofluorescence, Western blotting and biochemical assays. We also illustrate how this system can be useful in a study as we investigated the behavior of integrin adhesion complexes upon cell stretching. We therefore present a cost-effective, multipurpose cell stretching system that should help understand mechanical signaling.
Collapse
Affiliation(s)
- Etienne Boulter
- Université Côte d'Azur, INSERM, CNRS, IRCAN, 06107 Nice France
| | - Floriane S. Tissot
- Université Côte d'Azur, INSERM, CNRS, IRCAN, 06107 Nice France
- Imperial College London, London UK
- The Francis Crick Institute, London UK
| | - Julien Dilly
- Université Côte d'Azur, INSERM, CNRS, IRCAN, 06107 Nice France
| | - Sabrina Pisano
- Université Côte d'Azur, INSERM, CNRS, IRCAN, 06107 Nice France
| | - Chloé C. Féral
- Université Côte d'Azur, INSERM, CNRS, IRCAN, 06107 Nice France
| |
Collapse
|
27
|
Gough RE, Goult BT. The tale of two talins - two isoforms to fine-tune integrin signalling. FEBS Lett 2018; 592:2108-2125. [PMID: 29723415 PMCID: PMC6032930 DOI: 10.1002/1873-3468.13081] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/12/2018] [Accepted: 04/26/2018] [Indexed: 11/08/2022]
Abstract
Talins are cytoplasmic adapter proteins essential for integrin-mediated cell adhesion to the extracellular matrix. Talins control the activation state of integrins, link integrins to cytoskeletal actin, recruit numerous signalling molecules that mediate integrin signalling and coordinate recruitment of microtubules to adhesion sites via interaction with KANK (kidney ankyrin repeat-containing) proteins. Vertebrates have two talin genes, TLN1 and TLN2. Although talin1 and talin2 share 76% protein sequence identity (88% similarity), they are not functionally redundant, and the differences between the two isoforms are not fully understood. In this Review, we focus on the similarities and differences between the two talins in terms of structure, biochemistry and function, which hint at subtle differences in fine-tuning adhesion signalling.
Collapse
|
28
|
Fish Scale-Derived Scaffolds for Culturing Human Corneal Endothelial Cells. Stem Cells Int 2018; 2018:8146834. [PMID: 29853917 PMCID: PMC5949177 DOI: 10.1155/2018/8146834] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/22/2018] [Accepted: 03/19/2018] [Indexed: 11/18/2022] Open
Abstract
Purpose To investigate the biocompatibility of fish scale-derived scaffolds (FSS) with primary human corneal endothelial cells (HCEnCs). Methods HCEnCs were isolated from 30 donor corneas in a donor-matched study and plated in precoated Lab-Tek slides (n = 15) and FSS (n = 15). Cell morphology, proliferation/migration, and glucose uptake were studied (n = 30). Hoechst, ethidium homodimer, and calcein AM (HEC) staining was performed to determine viability and toxicity (n = 6). The cell surface area was calculated based on calcein AM staining. HCEnCs were stained for ZO-1 (n = 6) to detect tight junctions and to measure cell morphology; Ki-67 (n = 6) to measure proliferating cells; and vinculin to quantify focal adhesions (n = 6). The formation of de novo extracellular matrix was analyzed using histology (n = 6). Results HCEnCs attach and grow faster on Lab-Tek slides compared to the undulating topography of the FSS. At day 11, HCEnCs on Lab-Tek slide grew 100% confluent, while FSS was only 65% confluent (p = 0.0883), with no significant difference in glucose uptake between the two (p = 0.5181) (2.2 μg/mL in Lab-Tek versus 2.05 μg/mL in FSS). HEC staining showed no toxicity. The surface area of the cells in Lab-Tek was 409.1 μm2 compared to 452.2 μm2 on FSS, which was not significant (p = 0.5325). ZO-1 showed the presence of tight junctions in both conditions; however, hexagonality was higher (74% in Lab-Tek versus 45% in FSS; p = 0.0006) with significantly less polymorphic cells on Lab-Tek slides (8% in Lab-Tek versus 16% in FSS; p = 0.0041). Proliferative cells were detected in both conditions (4.6% in Lab-Tek versus 4.2% in FSS; p = 0.5922). Vinculin expression was marginally higher in HCEnCs cultured on Lab-Tek (234 versus 199 focal adhesions; p = 0.0507). Histological analysis did not show the formation of a basement membrane. Conclusions HCEnCs cultured on precoated FSS form a monolayer, displaying correct morphology, cytocompatibility, and absence of toxicity. FSS needs further modification in terms of structure and surface chemistry before considering it as a potential carrier for cultured HCEnCs.
Collapse
|
29
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
30
|
Burridge K. Focal adhesions: a personal perspective on a half century of progress. FEBS J 2017; 284:3355-3361. [PMID: 28796323 DOI: 10.1111/febs.14195] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/07/2017] [Indexed: 12/28/2022]
Abstract
Focal adhesions (FAs) are specialized sites within the cell where clustered integrin receptors interact with the extracellular matrix on the outside of cells and with the actin cytoskeleton on the inside. They provide strong adhesion to the matrix and transmit mechanical tension generated within cells across the plasma membrane to the external environment. Additionally, they act as scaffolds for many signaling pathways triggered by integrin engagement or mechanical force exerted on cells. Here I describe my personal perspective on FA research which I have witnessed since the initial discovery and description of FAs as electron dense regions of the ventral plasma nearly half a century ago.
Collapse
Affiliation(s)
- Keith Burridge
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
31
|
Taskin MB, Xia D, Besenbacher F, Dong M, Chen M. Nanotopography featured polycaprolactone/polyethyleneoxide microfibers modulate endothelial cell response. NANOSCALE 2017; 9:9218-9229. [PMID: 28654129 DOI: 10.1039/c7nr03326e] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Among many physical properties, surface nanotopography has been found to strongly affect cell adhesion, migration and other functions. Accurate biological interpretation requires the nanotopography to be presented in a three-dimensional (3D) micro-environment. Herein, immiscible blends of polycaprolactone (PCL)/polyethyleneoxide (PEO) were electrospun into a grounded coagulation bath, resulting in macroporous microfibers with nanotopography featured surfaces. Variations in PCL/PEO ratios enabled tunable surface nanotopographic structures, from longitudinal submicron grooves to transverse nano-lamellae. Chemical composition, crystallinity and quantitative nanomechanical analysis confirmed that the interplay of the two semi-crystalline immiscible polymers and the pairing of miscible solvents/non-solvents in both the electrospinning solution and the bath solution were critical for the formation of the secondary structure. It was found that the nanotopography features promoted the proliferation of human umbilical vein endothelial cells (HUVECs) compared with their smooth film counterparts. An analysis of the cell adhesion related markers, vinculin and phosphorylated focal adhesion kinase (pFAK), further revealed that the nanotopographies enhanced the nascent adhesion complex formation compared with smooth PCL fibers, even in the scaffolds with a high PEO content, which is often considered as a non-adhesive material.
Collapse
Affiliation(s)
- Mehmet Berat Taskin
- Interdisiplinary Nanoscience Center (iNANO), Aarhus University, DK-8000 Aarhus C, Denmark.
| | | | | | | | | |
Collapse
|
32
|
Bachir AI, Horwitz AR, Nelson WJ, Bianchini JM. Actin-Based Adhesion Modules Mediate Cell Interactions with the Extracellular Matrix and Neighboring Cells. Cold Spring Harb Perspect Biol 2017; 9:9/7/a023234. [PMID: 28679638 DOI: 10.1101/cshperspect.a023234] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell adhesions link cells to the extracellular matrix (ECM) and to each other and depend on interactions with the actin cytoskeleton. Both cell-ECM and cell-cell adhesion sites contain discrete, yet overlapping, functional modules. These modules establish physical associations with the actin cytoskeleton, locally modulate actin organization and dynamics, and trigger intracellular signaling pathways. Interplay between these modules generates distinct actin architectures that underlie different stages, types, and functions of cell-ECM and cell-cell adhesions. Actomyosin contractility is required to generate mature, stable adhesions, as well as to sense and translate the mechanical properties of the cellular environment into changes in cell organization and behavior. Here, we review the organization and function of different adhesion modules and how they interact with the actin cytoskeleton. We highlight the molecular mechanisms of mechanotransduction in adhesions and how adhesion molecules mediate cross talk between cell-ECM and cell-cell adhesion sites.
Collapse
Affiliation(s)
- Alexia I Bachir
- Protein and Cell Analysis, Biosciences Division, Thermo Fisher Scientific, Eugene, Oregon 97402
| | - Alan Rick Horwitz
- Protein and Cell Analysis, Biosciences Division, Thermo Fisher Scientific, Eugene, Oregon 97402
| | - W James Nelson
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22903
| | - Julie M Bianchini
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22903
| |
Collapse
|
33
|
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
|
34
|
Parekh M, Ahmad S, Ruzza A, Ferrari S. Human Corneal Endothelial Cell Cultivation From Old Donor Corneas With Forced Attachment. Sci Rep 2017; 7:142. [PMID: 28273942 PMCID: PMC5428054 DOI: 10.1038/s41598-017-00209-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/14/2017] [Indexed: 02/05/2023] Open
Abstract
Human corneal endothelial cells (HCEnCs) are responsible for maintaining the transparency of the cornea. Damaged or diseased HCEnCs may cause blindness. Replacement of the diseased cells with a healthy donor endothelium is the only currently available treatment. Tissue-engineering can serve as an alternative to conventional donor corneal transplantation. Due to the global shortage of donor corneas, a wide interest in the development of cultured graft substitutes and artificial corneas has increased. Availability of the old donor corneas is higher especially for research. Although it can be proposed as a valuable source for cell culture, its less proliferative capability emerges a challenge for the researchers. This article describes the use of hyaluronic acid (HA) in combination with Rho-kinase inhibitor (ROCK) Y-27632 for the cultivation of HCEnCs from older donor corneas (age > 60 years). Four conditions including and excluding HA + ROCK and its effect on early attachment rates and proliferation was studied on forty-eight corneas. It was observed that HCEnCs reach confluence within 10–15 days when cultured with HA + ROCK. This approach improves the efficiency of cell adhesion due to force attachment. HCEnCs from old donor corneas can be cultured using this method which may further lead to cell-based therapy for treating corneal endothelial dysfunction.
Collapse
Affiliation(s)
- Mohit Parekh
- International Center for Ocular Physiopathology, The Veneto Eye Bank Foundation, Venice, Italy. .,Department of Molecular Medicine, School of Biomedicine, University of Padova, Padova, Italy.
| | - Sajjad Ahmad
- Moorfields eye hospital, London, UK.,Institute of Ophthalmology, University College London, London, UK
| | - Alessandro Ruzza
- International Center for Ocular Physiopathology, The Veneto Eye Bank Foundation, Venice, Italy
| | - Stefano Ferrari
- International Center for Ocular Physiopathology, The Veneto Eye Bank Foundation, Venice, Italy
| |
Collapse
|
35
|
Herath SCB, Sharghi-Namini S, Du Y, Wang D, Ge R, Wang QG, Asada H, Chen PCY. A Magneto-Microfluidic System for Investigating the Influence of an Externally Induced Force Gradient in a Collagen Type I ECM on HMVEC Sprouting. SLAS Technol 2016; 22:413-424. [DOI: 10.1177/2211068216680078] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Advances in mechanobiology have suggested that physiological and pathological angiogenesis may be differentiated based on the ways in which the cells interact with the extracellular matrix (ECM) that exhibits partially different mechanical properties. This warrants investigating the regulation of ECM stiffness on cell behavior using angiogenesis assays. In this article, we report the application of the technique of active manipulation of ECM stiffness to study in vitro angiogenic sprouting of human microvascular endothelial cells (HMVECs) in a microfluidic device. Magnetic beads were embedded in the ECM through bioconjugation (between the streptavidin-coated beads and collagen fibers) in order to create a pretension in the ECM when under the influence of an external magnetic field. The advantage of using this magneto-microfluidic system is that the resulting change in the local deformability of the collagen fibers is only apparent to a cell at the pericellular level near the site of an embedded bead, while the global intrinsic material properties of the ECM remain unchanged. The results demonstrate that this system represents an effective tool for inducing noninvasively an external force on cells through the ECM, and suggest the possibility of creating desired stiffness gradients in the ECM for manipulating cell behavior in vitro.
Collapse
Affiliation(s)
- Sahan C. B. Herath
- Department of Mechanical Engineering, National University of Singapore, Singapore
- Biosystem and Micromechanics Interdisciplinary Research Group, Singapore–MIT Alliance for Research and Technology Program, Singapore
| | - Soheila Sharghi-Namini
- Biosystem and Micromechanics Interdisciplinary Research Group, Singapore–MIT Alliance for Research and Technology Program, Singapore
| | - Yue Du
- Department of Mechanical Engineering, National University of Singapore, Singapore
- Biosystem and Micromechanics Interdisciplinary Research Group, Singapore–MIT Alliance for Research and Technology Program, Singapore
| | - Dongan Wang
- Division of Bioengineering, Nanyang Technological University, Singapore
| | - Ruowen Ge
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Qing-Guo Wang
- Institute for Intelligent Systems, University of Johannesburg, Johannesburg, South Africa
| | - Harry Asada
- Biosystem and Micromechanics Interdisciplinary Research Group, Singapore–MIT Alliance for Research and Technology Program, Singapore
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Peter C. Y. Chen
- Department of Mechanical Engineering, National University of Singapore, Singapore
- Biosystem and Micromechanics Interdisciplinary Research Group, Singapore–MIT Alliance for Research and Technology Program, Singapore
| |
Collapse
|
36
|
Freikamp A, Cost AL, Grashoff C. The Piconewton Force Awakens: Quantifying Mechanics in Cells. Trends Cell Biol 2016; 26:838-847. [DOI: 10.1016/j.tcb.2016.07.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/20/2016] [Accepted: 07/26/2016] [Indexed: 02/07/2023]
|
37
|
Li Z, Lee H, Zhu C. Molecular mechanisms of mechanotransduction in integrin-mediated cell-matrix adhesion. Exp Cell Res 2016; 349:85-94. [PMID: 27720950 DOI: 10.1016/j.yexcr.2016.10.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/30/2016] [Accepted: 10/03/2016] [Indexed: 01/09/2023]
Abstract
Cell-matrix adhesion complexes are multi-protein structures linking the extracellular matrix (ECM) to the cytoskeleton. They are essential to both cell motility and function by bidirectionally sensing and transmitting mechanical and biochemical stimulations. Several types of cell-matrix adhesions have been identified and they share many key molecular components, such as integrins and actin-integrin linkers. Mechanochemical coupling between ECM molecules and the actin cytoskeleton has been observed from the single cell to the single molecule level and from immune cells to neuronal cells. However, the mechanisms underlying force regulation of integrin-mediated mechanotransduction still need to be elucidated. In this review article, we focus on integrin-mediated adhesions and discuss force regulation of cell-matrix adhesions and key adaptor molecules, three different force-dependent behaviors, and molecular mechanisms for mechanochemical coupling in force regulation.
Collapse
Affiliation(s)
- Zhenhai Li
- Molecular Modeling and Simulation Group, National Institutes for Quantum and Radiological Science and Technology, 8-1-7 Umemidai, Kizugawa, Kyoto 619-0215, Japan
| | - Hyunjung Lee
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Cheng Zhu
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; George W Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| |
Collapse
|
38
|
Cheng F, Miao L, Wu Q, Gong X, Xiong J, Zhang J. Vinculin b deficiency causes epicardial hyperplasia and coronary vessel disorganization in zebrafish. Development 2016; 143:3522-3531. [PMID: 27578788 DOI: 10.1242/dev.132936] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 07/26/2016] [Indexed: 12/29/2022]
Abstract
Coronary vessel development is a highly coordinated process during heart formation. Abnormal development and dysfunction of the coronary network are contributory factors in the majority of heart disease. Understanding the molecular mechanisms that regulate coronary vessel formation is crucial for preventing and treating the disease. We report a zebrafish gene-trap vinculin b (vclb) mutant that displays abnormal coronary vessel development among multiple cardiac defects. The mutant shows overproliferation of epicardium-derived cells and disorganization of coronary vessels, and they eventually die off at juvenile stages. Mechanistically, Vclb deficiency results in the release of another cytoskeletal protein, paxillin, from the Vclb complex and the upregulation of ERK and FAK phosphorylation in epicardium and endocardium, causing disorganization of endothelial cells and pericytes during coronary vessel development. By contrast, cardiac muscle development is relatively normal, probably owing to redundancy with Vcla, a vinculin paralog that is expressed in the myocardium but not epicardium. Together, our results reveal a previously unappreciated function of vinculin in epicardium and endocardium and reinforce the notion that well-balanced FAK activity is essential for coronary vessel development.
Collapse
Affiliation(s)
- Feng Cheng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liyun Miao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Wu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Xia Gong
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jingwei Xiong
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Jian Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
39
|
Kraft-Sheleg O, Zaffryar-Eilot S, Genin O, Yaseen W, Soueid-Baumgarten S, Kessler O, Smolkin T, Akiri G, Neufeld G, Cinnamon Y, Hasson P. Localized LoxL3-Dependent Fibronectin Oxidation Regulates Myofiber Stretch and Integrin-Mediated Adhesion. Dev Cell 2016; 36:550-61. [PMID: 26954549 DOI: 10.1016/j.devcel.2016.02.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 01/21/2016] [Accepted: 02/07/2016] [Indexed: 02/07/2023]
Abstract
For muscles to function, myofibers have to stretch and anchor at the myotendinous junction (MTJ), a region rich in extracellular matrix (ECM). Integrin signaling is required for MTJ formation, and mutations affecting the cascade lead to muscular dystrophies in mice and humans. Underlying mechanisms for integrin activation at the MTJ and ECM modifications regulating its signaling are unclear. We show that lysyl oxidase-like 3 (LoxL3) is a key regulator of integrin signaling that ensures localized control of the cascade. In LoxL3 mutants, myofibers anchor prematurely or overshoot to adjacent somites, and are loose and lack tension. We find that LoxL3 complexes with and directly oxidizes Fibronectin (FN), an ECM scaffold protein and integrin ligand enriched at the MTJ. We identify a mechanism whereby localized LoxL3 secretion from myofiber termini oxidizes FN, enabling enhanced integrin activation at the tips of myofibers and ensuring correct positioning and anchoring of myofibers along the MTJ.
Collapse
Affiliation(s)
- Ortal Kraft-Sheleg
- The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Shelly Zaffryar-Eilot
- The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Olga Genin
- Department of Poultry and Aquaculture, Institute of Animal Sciences, Agricultural Research Organization, The Volcani Center, Bet Dagan 50250, Israel
| | - Wesal Yaseen
- The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Sharon Soueid-Baumgarten
- The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Ofra Kessler
- Cancer Research and Vascular Biology Center, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Tatyana Smolkin
- Cancer Research and Vascular Biology Center, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Gal Akiri
- Cancer Research and Vascular Biology Center, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Gera Neufeld
- Cancer Research and Vascular Biology Center, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Yuval Cinnamon
- Department of Poultry and Aquaculture, Institute of Animal Sciences, Agricultural Research Organization, The Volcani Center, Bet Dagan 50250, Israel
| | - Peleg Hasson
- The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
40
|
Mechanosensitive components of integrin adhesions: Role of vinculin. Exp Cell Res 2015; 343:21-27. [PMID: 26607713 PMCID: PMC4856733 DOI: 10.1016/j.yexcr.2015.11.017] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 11/18/2015] [Indexed: 12/14/2022]
Abstract
External forces play a key role in shaping development and normal physiology. Aberrant responses to forces, or changes in the nature of such forces, are implicated in a variety of diseases. Cells contain several types of adhesions, linking them to their external environment. It is through these adhesions that forces are both sensed (from the outside inwards) and applied (from inside to out). Furthermore, several adhesion-based proteins are sensitive to changes in intracellular forces, utilising them for activation and regulation. Here, we outline how vinculin, a key component of integrin-mediated adhesions linking the actin cytoskeleton to the extracellular matrix (ECM), is regulated by force and acts as force transducing protein. We discuss the role of vinculin in vivo and its place in health and disease; summarise the proposed mechanisms by which vinculin is recruited to and activated at integrin-ECM adhesions; and discuss recent findings that place vinculin as the major force sensing and transmitting component of cell–matrix adhesion complexes. Finally, we discuss the role of vinculin in regulating the cellular responses to both the physical properties of the external environment and to externally applied physical stimuli.
Collapse
|
41
|
Actomyosin-dependent formation of the mechanosensitive talin-vinculin complex reinforces actin anchoring. Nat Commun 2015; 5:3095. [PMID: 24452080 PMCID: PMC3916842 DOI: 10.1038/ncomms4095] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Accepted: 12/12/2013] [Indexed: 01/06/2023] Open
Abstract
The force generated by the actomyosin cytoskeleton controls focal adhesion dynamics during cell migration. This process is thought to involve the mechanical unfolding of talin to expose cryptic vinculin-binding sites. However, the ability of the actomyosin cytoskeleton to directly control the formation of a talin–vinculin complex and the resulting activity of the complex are not known. Here we develop a microscopy assay with pure proteins in which the self-assembly of actomyosin cables controls the association of vinculin to a talin-micropatterned surface in a reversible manner. Quantifications indicate that talin refolding is limited by vinculin dissociation and modulated by the actomyosin network stability. Finally, we show that the activation of vinculin by stretched talin induces a positive feedback that reinforces the actin–talin–vinculin association. This in vitro reconstitution reveals the mechanism by which a key molecular switch senses and controls the connection between adhesion complexes and the actomyosin cytoskeleton. The interaction between focal adhesion proteins vinculin and talin is stimulated by mechanical stretching. Here the authors reconstitute actomyosin-dependent stretching of talin in vitro, and show that the resulting activation of vinculin reinforces anchoring of the adhesion complex to actin.
Collapse
|
42
|
Bruntz RC, Lindsley CW, Brown HA. Phospholipase D signaling pathways and phosphatidic acid as therapeutic targets in cancer. Pharmacol Rev 2015; 66:1033-79. [PMID: 25244928 DOI: 10.1124/pr.114.009217] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Phospholipase D is a ubiquitous class of enzymes that generates phosphatidic acid as an intracellular signaling species. The phospholipase D superfamily plays a central role in a variety of functions in prokaryotes, viruses, yeast, fungi, plants, and eukaryotic species. In mammalian cells, the pathways modulating catalytic activity involve a variety of cellular signaling components, including G protein-coupled receptors, receptor tyrosine kinases, polyphosphatidylinositol lipids, Ras/Rho/ADP-ribosylation factor GTPases, and conventional isoforms of protein kinase C, among others. Recent findings have shown that phosphatidic acid generated by phospholipase D plays roles in numerous essential cellular functions, such as vesicular trafficking, exocytosis, autophagy, regulation of cellular metabolism, and tumorigenesis. Many of these cellular events are modulated by the actions of phosphatidic acid, and identification of two targets (mammalian target of rapamycin and Akt kinase) has especially highlighted a role for phospholipase D in the regulation of cellular metabolism. Phospholipase D is a regulator of intercellular signaling and metabolic pathways, particularly in cells that are under stress conditions. This review provides a comprehensive overview of the regulation of phospholipase D activity and its modulation of cellular signaling pathways and functions.
Collapse
Affiliation(s)
- Ronald C Bruntz
- Department of Pharmacology (R.C.B., C.W.L., H.A.B.) and Vanderbilt Center for Neuroscience Drug Discovery (C.W.L.), Vanderbilt University Medical Center; Department of Chemistry, Vanderbilt Institute of Chemical Biology (C.W.L., H.A.B.); Vanderbilt Specialized Chemistry for Accelerated Probe Development (C.W.L.); and Department of Biochemistry, Vanderbilt-Ingram Cancer Center (H.A.B.), Vanderbilt University, Nashville, Tennessee
| | - Craig W Lindsley
- Department of Pharmacology (R.C.B., C.W.L., H.A.B.) and Vanderbilt Center for Neuroscience Drug Discovery (C.W.L.), Vanderbilt University Medical Center; Department of Chemistry, Vanderbilt Institute of Chemical Biology (C.W.L., H.A.B.); Vanderbilt Specialized Chemistry for Accelerated Probe Development (C.W.L.); and Department of Biochemistry, Vanderbilt-Ingram Cancer Center (H.A.B.), Vanderbilt University, Nashville, Tennessee
| | - H Alex Brown
- Department of Pharmacology (R.C.B., C.W.L., H.A.B.) and Vanderbilt Center for Neuroscience Drug Discovery (C.W.L.), Vanderbilt University Medical Center; Department of Chemistry, Vanderbilt Institute of Chemical Biology (C.W.L., H.A.B.); Vanderbilt Specialized Chemistry for Accelerated Probe Development (C.W.L.); and Department of Biochemistry, Vanderbilt-Ingram Cancer Center (H.A.B.), Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
43
|
Maartens AP, Brown NH. Anchors and signals: the diverse roles of integrins in development. Curr Top Dev Biol 2015; 112:233-72. [PMID: 25733142 DOI: 10.1016/bs.ctdb.2014.11.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Integrins mediate cell adhesion by providing a link between the actin cytoskeleton and the extracellular matrix. As well as acting to anchor cells, integrin adhesions provide sensory input via mechanotransduction and synergism with signaling pathways, and provide the cell with the conditions necessary for differentiation in a permissive manner. In this review, we explore how integrins contribute to development, and what this tells us about how they work. From a signaling perspective, the influence of integrins on cell viability and fate is muted in a developmental context as compared to cell culture. Integrin phenotypes tend to arise from a failure of normally specified cells to create tissues properly, due to defective adhesion. The diversity of integrin functions in development shows how cell adhesion is continuously adjusted, both within and between animals, to fit developmental purpose.
Collapse
Affiliation(s)
- Aidan P Maartens
- Department of Physiology, Development and Neuroscience, The Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas H Brown
- Department of Physiology, Development and Neuroscience, The Gurdon Institute, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
44
|
Sterner O, Giazzon M, Zürcher S, Tosatti S, Liley M, Spencer ND. Delineating fibronectin bioadhesive micropatterns by photochemical immobilization of polystyrene and poly(vinylpyrrolidone). ACS APPLIED MATERIALS & INTERFACES 2014; 6:18683-18692. [PMID: 25253530 DOI: 10.1021/am5042093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Bioadhesive micropatterns, capable of laterally confining cells to a 2D lattice, have proven effective in simulating the in vivo tissue environment. They reveal fundamental aspects of the role of adhesion in cell mechanics, proliferation, and differentiation. Here we present an approach based on photochemistry for the fabrication of synthetic polymer micropatterns. Perfluorophenyl azide (PFPA), upon deep-UV exposure, forms a reactive nitrene capable of covalently linking to a molecule that is in close proximity. PFPA has been grafted onto a backbone of poly(allyl amine), which readily forms a self-assembled monolayer on silicon wafers or glass. A film of polystyrene was applied by spin-coating, and by laterally confining the UV exposure through a chromium-on-quartz photomask, monolayers of polymers could be immobilized in circular microdomains. Poly(vinylpyrrolidone) (PVP) was attached to the background to form a barrier to nonspecific protein adsorption and cell adhesion. Micropatterns were characterized with high-lateral-resolution time-of-flight secondary ion mass spectrometry (TOF-SIMS), which confirmed the formation of polystyrene domains within a PVP background. Fluorescence-microscopy adsorption assays with rhodamine-labeled bovine serum albumin demonstrated the nonfouling efficiency of PVP and, combined with TOF-SIMS, allowed for a comprehensive characterization of the pattern geometry. The applicability of the micropatterned platform in single-cell assays was tested by culturing two cell types, WM 239 melanoma cells and SaOs-2 osteoblasts, on micropatterned glass, either with or without backfilling of the patterns with fibronectin. It was demonstrated that the platform was efficient in confining cells to the fibronectin-backfilled micropatterns for at least 48 h. PVP is thus proposed as a viable, highly stable alternative to poly(ethylene glycol) for nonfouling applications. Due to the versatility of the nitrene-insertion reaction, the platform could be extended to other polymer pairs or proteins and the surface chemistry adapted to specific applications.
Collapse
Affiliation(s)
- Olof Sterner
- Laboratory for Surface Science and Technology, Department of Materials, ETH Zurich , Vladimir-Prelog-Weg 5, CH-8093, Zürich, Switzerland
| | | | | | | | | | | |
Collapse
|
45
|
Meddens MBM, van den Dries K, Cambi A. Podosomes revealed by advanced bioimaging: what did we learn? Eur J Cell Biol 2014; 93:380-7. [PMID: 25454791 DOI: 10.1016/j.ejcb.2014.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/04/2014] [Accepted: 09/28/2014] [Indexed: 02/01/2023] Open
Abstract
Podosomes are micrometer-sized, circular adhesions formed by cells such as osteoclasts, macrophages, dendritic cells, and endothelial cells. Because of their small size and the lack of methods to visualize individual proteins and protein complexes, podosomes have long been considered a simple two-module structure with a protrusive actin core and a surrounding adhesive ring composed of integrins and cytoskeletal adaptor proteins such as vinculin and talin. In the past decade, the applications of fluorescence based techniques that circumvent the diffraction limit of conventional light microscopy took a major leap forward. Podosomes have been imaged by a variety of these super-resolution methods, and in this concise review we discuss how these super-resolution data have increased our understanding of the podosome ultra-structure and function.
Collapse
Affiliation(s)
- Marjolein B M Meddens
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, The Netherlands
| | - Koen van den Dries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, The Netherlands
| | - Alessandra Cambi
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, The Netherlands.
| |
Collapse
|
46
|
Márquez MG, Brandán YR, Guaytima EDV, Paván CH, Favale NO, Sterin-Speziale NB. Physiologically induced restructuring of focal adhesions causes mobilization of vinculin by a vesicular endocytic recycling pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2991-3003. [PMID: 25241342 DOI: 10.1016/j.bbamcr.2014.09.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 09/11/2014] [Accepted: 09/13/2014] [Indexed: 11/19/2022]
Abstract
In epithelial cells, vinculin is enriched in cell adhesion structures but is in equilibrium with a large cytosolic pool. It is accepted that when cells adhere to the extracellular matrix, a part of the soluble cytosolic pool of vinculin is recruited to specialized sites on the plasma membrane called focal adhesions (FAs) by binding to plasma membrane phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2). We have previously shown that bradykinin (BK) induces both a reversible dissipation of vinculin from FAs, by the phospholipase C (PLC)-mediated hydrolysis of PtdIns(4,5)P2, and the concomitant internalization of vinculin. Here, by using an immunomagnetic method, we isolated vinculin-containing vesicles induced by BK stimulation. By analyzing the presence of proteins involved in vesicle traffic, we suggest that vinculin can be delivered in the site of FA reassembly by a vesicular endocytic recycling pathway. We also observed the formation of vesicle-like structures containing vinculin in the cytosol of cells treated with lipid membrane-affecting agents, which caused dissipation of FAs due to their deleterious effect on membrane microdomains where FAs are inserted. However, these vesicles did not contain markers of the recycling endosomal compartment. Vinculin localization in vesicles has not been reported before, and this finding challenges the prevailing model of vinculin distribution in the cytosol. We conclude that the endocytic recycling pathway of vinculin could represent a physiological mechanism to reuse the internalized vinculin to reassembly new FAs, which occurs after long time of BK stimulation, but not after treatment with membrane-affecting agents.
Collapse
Affiliation(s)
- María Gabriela Márquez
- Instituto de Investigaciones en Ciencias de la Salud Humana (IICSHUM), Universidad Nacional de La Rioja, Av. Luis Vernet 1000, 5300 La Rioja, Argentina; Instituto de Química y Físico-Química Biológica (IQUIFIB)-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113AAD Buenos Aires, Argentina
| | - Yamila Romina Brandán
- Instituto de Investigaciones en Ciencias de la Salud Humana (IICSHUM), Universidad Nacional de La Rioja, Av. Luis Vernet 1000, 5300 La Rioja, Argentina; Instituto de Química y Físico-Química Biológica (IQUIFIB)-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113AAD Buenos Aires, Argentina
| | - Edith Del Valle Guaytima
- Instituto de Investigaciones en Ciencias de la Salud Humana (IICSHUM), Universidad Nacional de La Rioja, Av. Luis Vernet 1000, 5300 La Rioja, Argentina; Instituto de Química y Físico-Química Biológica (IQUIFIB)-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113AAD Buenos Aires, Argentina
| | - Carlos Humberto Paván
- Instituto de Química y Físico-Química Biológica (IQUIFIB)-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113AAD Buenos Aires, Argentina
| | - Nicolás Octavio Favale
- Instituto de Química y Físico-Química Biológica (IQUIFIB)-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113AAD Buenos Aires, Argentina; Cátedra de Biología Celular, Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113AAD Buenos Aires, Argentina
| | - Norma B Sterin-Speziale
- Instituto de Química y Físico-Química Biológica (IQUIFIB)-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113AAD Buenos Aires, Argentina.
| |
Collapse
|
47
|
Optineurin associates with the podocyte Golgi complex to maintain its structure. Cell Tissue Res 2014; 358:567-83. [PMID: 25096716 DOI: 10.1007/s00441-014-1968-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 07/09/2014] [Indexed: 12/22/2022]
Abstract
Optineurin, a cytosolic protein associated with the actin cytoskeleton, microtubules, and the Golgi complex, appears to have an important function in neurons, as mutations in its gene are causative for neurodegenerative diseases such as primary open-angle glaucoma and amyotrophic lateral sclerosis. Here, we report that optineurin is localized in podocytes of the kidney and induced upon injury following treatment with puromycin aminonucleoside. In cultured human podocytes, optineurin localizes to the Golgi complex. Optineurin depletion by RNA interference causes Golgi fragmentation. Moreover, if the Golgi complex is fragmented following microtubule destabilization induced by nocodazole treatment, optineurin dissociates from Golgi vesicles. Furthermore, optineurin colocalizes with vinculin-labeled focal contacts of cultured podocytes and with lysosome-like structures. Optineurin is essential for the survival of cultured podocytes, as optineurin depletion causes cell death. Thus, optineurin appears to play an important role in the maintenance of the podocyte Golgi complex and in the trafficking of vesicles to focal contacts and lysosomes.
Collapse
|
48
|
Regulation of focal adhesion formation by a vinculin-Arp2/3 hybrid complex. Nat Commun 2014; 5:3758. [DOI: 10.1038/ncomms4758] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/28/2014] [Indexed: 12/26/2022] Open
|
49
|
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
|
50
|
Simon NC, Barbieri JT. Bacillus cereus Certhrax ADP-ribosylates vinculin to disrupt focal adhesion complexes and cell adhesion. J Biol Chem 2014; 289:10650-10659. [PMID: 24573681 DOI: 10.1074/jbc.m113.500710] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Bacillus cereus is often associated with mild to moderate gastroenteritis; however, some recent isolates cause inhalational anthrax-like diseases and death. These potential emerging human pathogens express multiple virulence factors. B. cereus strain G9241 expresses anthrax toxin, several polysaccharide capsules, and the novel ADP-ribosyltransferase, Certhrax. In this study, we show that Certhrax ADP-ribosylates Arg-433 of vinculin, a protein that coordinates actin cytoskeleton and extracellular matrix interactions. ADP-ribosylation of vinculin disrupted focal adhesion complexes and redistributed vinculin to the cytoplasm. Exogenous vinculin rescued these phenotypes. This provides a mechanism for strain G9241 to breach host barrier defenses and promote bacterial growth and spread. Certhrax is the first bacterial toxin to add a post-translational modification to vinculin to disrupt the actin cytoskeleton.
Collapse
Affiliation(s)
- Nathan C Simon
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Joseph T Barbieri
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226.
| |
Collapse
|