1
|
The kinase activity of integrin-linked kinase regulates cellular senescence in gastric cancer. Cell Death Dis 2022; 13:577. [PMID: 35778385 PMCID: PMC9249761 DOI: 10.1038/s41419-022-05020-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 06/12/2022] [Accepted: 06/13/2022] [Indexed: 01/21/2023]
Abstract
The activity of integrin-linked kinase (ILK) in cancerous cells is often oncogenic and associated with malignant properties, such as uncontrolled cell cycle progression and evasion from senescence. However, the role of ILK in cellular senescence in gastric cancer (GC) has not been previously examined. We generated single-cell clones of ILK knock-out using CRISPR-Cas9 in human GC lines with mesenchymal or epithelial histology. Cells with no residual ILK expression exhibited strong cellular senescence with diminished clathrin-mediated endocytosis, Surprisingly, ILK loss-induced cellular senescence appeared to be independent of its function in integrin signaling. The low dose of CPD22, a small molecule inhibitor of ILK activity-induced senescence in three GC cell lines with different histologies. Furthermore, senescent cells with ILK depletion transfected with N-terminal truncated ILK mutant remaining catalytic domains displayed the reduction of senescent phenotypes. RNA sequencing and cytokine array results revealed the enrichment of multiple pro-inflammatory signaling pathways in GC lines in the absence of ILK. Our study identified the important role and the potential mechanism of ILK in the cellular senescence of cancerous epithelial cells. The inhibition of ILK activity using small molecule compounds could have a pro-senescent effect as a therapeutic option for GC.
Collapse
|
2
|
Lickert S, Kenny M, Selcuk K, Mehl JL, Bender M, Früh SM, Burkhardt MA, Studt JD, Nieswandt B, Schoen I, Vogel V. Platelets drive fibronectin fibrillogenesis using integrin αIIbβ3. SCIENCE ADVANCES 2022; 8:eabj8331. [PMID: 35275711 PMCID: PMC8916723 DOI: 10.1126/sciadv.abj8331] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Platelets interact with multiple adhesion proteins during thrombogenesis, yet little is known about their ability to assemble fibronectin matrix. In vitro three-dimensional superresolution microscopy complemented by biophysical and biochemical methods revealed fundamental insights into how platelet contractility drives fibronectin fibrillogenesis. Platelets adhering to thrombus proteins (fibronectin and fibrin) versus basement membrane components (laminin and collagen IV) pull fibronectin fibrils along their apical membrane versus underneath their basal membrane, respectively. In contrast to other cell types, platelets assemble fibronectin nanofibrils using αIIbβ3 rather than α5β1 integrins. Apical fibrillogenesis correlated with a stronger activation of integrin-linked kinase, higher platelet traction forces, and a larger tension in fibrillar-like adhesions compared to basal fibrillogenesis. Our findings have potential implications for how mechanical thrombus integrity might be maintained during remodeling and vascular repair.
Collapse
Affiliation(s)
- Sebastian Lickert
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Martin Kenny
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
| | - Kateryna Selcuk
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Johanna L. Mehl
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Markus Bender
- Institute of Experimental Biomedicine – Chair I, University Hospital, and Rudolf Virchow Center, Julius Maximilian University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Susanna M. Früh
- Hahn-Schickard, Georges-Koehler-Allee 103, 79110 Freiburg, Germany
- Laboratory for MEMS Applications, IMTEK Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, 79110 Freiburg, Germany
| | - Melanie A. Burkhardt
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Jan-Dirk Studt
- Division of Hematology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine – Chair I, University Hospital, and Rudolf Virchow Center, Julius Maximilian University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Ingmar Schoen
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
- Corresponding author. (V.V.); (I.S.)
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
- Corresponding author. (V.V.); (I.S.)
| |
Collapse
|
3
|
Integrin-Linked Kinase (ILK) Regulates Urinary Stem Cells Differentiation into Smooth Muscle via NF- κB Signal Pathway. Stem Cells Int 2021; 2021:6633111. [PMID: 33854551 PMCID: PMC8019365 DOI: 10.1155/2021/6633111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/28/2021] [Accepted: 03/09/2021] [Indexed: 11/17/2022] Open
Abstract
Objectives Urinary stem cells (USCs) have the capacity for unlimited growth and are promising tools for the investigations of cell differentiation and urinary regeneration. However, the limited life span significantly restricts their usefulness. This study is aimed at exploring the effect of integrin-linked kinase (ILK) on the smooth muscle cells (SMCs) differentiation of the dog USCs and investigating its molecular mechanism. Methods An immortalized USCs cell line with the molecular markers and biological functions was prepared. After successfully inducing the differentiation of USCs into SMCs, the expression level of the unique key factor and its mechanisms in this process was determined through real-time polymerase chain reaction, Western blot, or Immunofluorescence staining. Results We found that high cell density promoted USCs differentiation SMCs, and ILK was necessary for USCs differentiation into SMCs. Knocking down ILK decreased the expression of SMCs specific-marker, while using a selective ILK agonist increased the expression of SMCs specific-marker. Furthermore, ILK regulated SMCs differentiation in part through the activation of NF-κB pathway in USCs. A NF-κB activity assay showed overexpression of ILK could significantly upregulate NF-κB p50 expression, and NF-κB p50 acts as downstream signal molecular of ILK. Conclusion High cell density induces the differentiation of USCs into SMCs, and ILK is a key regulator of myogenesis. Furthermore, NF-κB signaling pathway might play a crucial role in this process.
Collapse
|
4
|
Nikolopoulou PA, Koufaki MA, Kostourou V. The Adhesome Network: Key Components Shaping the Tumour Stroma. Cancers (Basel) 2021; 13:525. [PMID: 33573141 PMCID: PMC7866493 DOI: 10.3390/cancers13030525] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Beyond the conventional perception of solid tumours as mere masses of cancer cells, advanced cancer research focuses on the complex contributions of tumour-associated host cells that are known as "tumour microenvironment" (TME). It has been long appreciated that the tumour stroma, composed mainly of blood vessels, cancer-associated fibroblasts and immune cells, together with the extracellular matrix (ECM), define the tumour architecture and influence cancer cell properties. Besides soluble cues, that mediate the crosstalk between tumour and stroma cells, cell adhesion to ECM arises as a crucial determinant in cancer progression. In this review, we discuss how adhesome, the intracellular protein network formed at cell adhesions, regulate the TME and control malignancy. The role of adhesome extends beyond the physical attachment of cells to ECM and the regulation of cytoskeletal remodelling and acts as a signalling and mechanosensing hub, orchestrating cellular responses that shape the tumour milieu.
Collapse
Affiliation(s)
| | | | - Vassiliki Kostourou
- Biomedical Sciences Research Centre “Alexander Fleming”, Institute of Bioinnovation, 34 Fleming Str., 16672 Vari-Athens, Greece; (P.A.N.); (M.A.K.)
| |
Collapse
|
5
|
Gagné D, Benoit YD, Groulx JF, Vachon PH, Beaulieu JF. ILK supports RhoA/ROCK-mediated contractility of human intestinal epithelial crypt cells by inducing the fibrillogenesis of endogenous soluble fibronectin during the spreading process. BMC Mol Cell Biol 2020; 21:14. [PMID: 32183701 PMCID: PMC7079544 DOI: 10.1186/s12860-020-00259-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/05/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Fibronectin (FN) assembly into an insoluble fibrillar matrix is a crucial step in many cell responses to extracellular matrix (ECM) properties, especially with regards to the integrin-related mechanosensitive signaling pathway. We have previously reported that the silencing of expression of integrin-linked kinase (ILK) in human intestinal epithelial crypt (HIEC) cells causes significant reductions in proliferation and spreading through concomitantly acquired impairment of soluble FN deposition. These defects in ILK-depleted cells are rescued by growth on exogenous FN. In the present study we investigated the contribution of ILK in the fibrillogenesis of FN and its relation to integrin-actin axis signaling and organization. RESULTS We show that de novo fibrillogenesis of endogenous soluble FN is ILK-dependent. This function seemingly induces the assembly of an ECM that supports increased cytoskeletal tension and the development of a fully spread contractile cell phenotype. We observed that HIEC cell adhesion to exogenous FN or collagen-I (Col-I) is sufficient to restore fibrillogenesis of endogenous FN in ILK-depleted cells. We also found that optimal engagement of the Ras homolog gene family member A (RhoA) GTPase/Rho-associated kinase (ROCK-1, ROCK-2)/myosin light chain (MLC) pathway, actin ventral stress fiber formation, and integrin adhesion complex (IAC) maturation rely primarily upon the cell's capacity to execute FN fibrillogenesis, independent of any significant ILK input. Lastly, we confirm the integrin α5β1 as the main integrin responsible for FN assembly, although in ILK-depleted cells αV-class integrins expression is needed to allow the rescue of FN fibrillogenesis on exogenous substrate. CONCLUSION Our study demonstrates that ILK specifically induces the initiation of FN fibrillogenesis during cell spreading, which promotes RhoA/ROCK-dependent cell contractility and maturation of the integrin-actin axis structures. However, the fibrillogenesis process and its downstream effect on RhoA signaling, cell contractility and spreading are ILK-independent in human intestinal epithelial crypt cells.
Collapse
Affiliation(s)
- David Gagné
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, and Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, J1H 5N4 Canada
| | - Yannick D. Benoit
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5 Canada
| | - Jean-François Groulx
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, California, 92093 USA
| | - Pierre H. Vachon
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4 Canada
| | - Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, and Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, J1H 5N4 Canada
| |
Collapse
|
6
|
Are Integrins Still Practicable Targets for Anti-Cancer Therapy? Cancers (Basel) 2019; 11:cancers11070978. [PMID: 31336983 PMCID: PMC6678560 DOI: 10.3390/cancers11070978] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/27/2019] [Accepted: 07/09/2019] [Indexed: 01/01/2023] Open
Abstract
Correlative clinical evidence and experimental observations indicate that integrin adhesion receptors, in particular those of the αV family, are relevant to cancer cell features, including proliferation, survival, migration, invasion, and metastasis. In addition, integrins promote events in the tumor microenvironment that are critical for tumor progression and metastasis, including tumor angiogenesis, matrix remodeling, and the recruitment of immune and inflammatory cells. In spite of compelling preclinical results demonstrating that the inhibition of integrin αVβ3/αVβ5 and α5β1 has therapeutic potential, clinical trials with integrin inhibitors targeting those integrins have repeatedly failed to demonstrate therapeutic benefits in cancer patients. Here, we review emerging integrin functions and their proposed contribution to tumor progression, discuss preclinical evidence of therapeutic significance, revisit clinical trial results, and consider alternative approaches for their therapeutic targeting in oncology, including targeting integrins in the other cells of the tumor microenvironment, e.g., cancer-associated fibroblasts and immune/inflammatory cells. We conclude that integrins remain a valid target for cancer therapy; however, agents with better pharmacological properties, alternative models for their preclinical evaluation, and innovative combination strategies for clinical testing (e.g., together with immuno-oncology agents) are needed.
Collapse
|
7
|
Soubies E, Radwanska A, Grall D, Blanc-Féraud L, Van Obberghen-Schilling E, Schaub S. Nanometric axial resolution of fibronectin assembly units achieved with an efficient reconstruction approach for multi-angle-TIRF microscopy. Sci Rep 2019; 9:1926. [PMID: 30760745 PMCID: PMC6374485 DOI: 10.1038/s41598-018-36119-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/08/2018] [Indexed: 02/01/2023] Open
Abstract
High resolution imaging of molecules at the cell-substrate interface is required for understanding key biological processes. Here we propose a complete pipeline for multi-angle total internal reflection fluorescence microscopy (MA-TIRF) going from instrument design and calibration procedures to numerical reconstruction. Our custom setup is endowed with a homogeneous field illumination and precise excitation beam angle. Given a set of MA-TIRF acquisitions, we deploy an efficient joint deconvolution/reconstruction algorithm based on a variational formulation of the inverse problem. This algorithm offers the possibility of using various regularizations and can run on graphics processing unit (GPU) for rapid reconstruction. Moreover, it can be easily used with other MA-TIRF devices and we provide it as an open-source software. This ensemble has enabled us to visualize and measure with unprecedented nanometric resolution, the depth of molecular components of the fibronectin assembly machinery at the basal surface of endothelial cells.
Collapse
Affiliation(s)
- Emmanuel Soubies
- Université Côte d'Azur, CNRS, Inria, I3S, France. .,Biomedical Imaging Group, EPFL, Lausanne, Switzerland.
| | | | | | | | | | - Sébastien Schaub
- Université Côte d'Azur, CNRS, Inria, I3S, France. .,Université Côte d'Azur, CNRS, Inserm, iBV, France.
| |
Collapse
|
8
|
Syed SB, Khan FI, Khan SH, Srivastava S, Hasan GM, Lobb KA, Islam A, Ahmad F, Hassan MI. Mechanistic insights into the urea-induced denaturation of kinase domain of human integrin linked kinase. Int J Biol Macromol 2018; 111:208-218. [DOI: 10.1016/j.ijbiomac.2017.12.164] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/29/2017] [Accepted: 12/30/2017] [Indexed: 01/01/2023]
|
9
|
Samuelsson AR, Belvindrah R, Wu C, Müller U, Halfter W. β1-Integrin Signaling is Essential for Lens Fiber Survival. GENE REGULATION AND SYSTEMS BIOLOGY 2017. [DOI: 10.1177/117762500700100016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Integrins have been proposed to play a major role in lens morphogenesis. To determine the role of β1-integrin and its down-stream signaling partner, integrin linked kinase (ILK), in lens morphogenesis, eyes of WT mice and mice with a nestin-linked conditional knockout of β1-integrin or ILK were analyzed for defects in lens development. Mice, lacking the genes encoding the p1-integrin subunit ( Itgb1) or ILK ( Ilk), showed a perinatal degeneration of the lens. Early signs of lens degeneration included vacuolization, random distribution of lens cell nuclei, disrupted fiber morphology and attenuation and separation of the lens capsule. The phenotype became progressively more severe during the first postnatal week eventually leading to the complete loss of the lens. A more severe phenotype was observed in ILK mutants at similar stages. Eyes from embryonic day 13 β1-integrin-mutant embryos showed no obvious signs of lens degeneration, indicating that mutant lens develops normally until peri-recombination. Our findings suggest that β1-integrins and ILK cooperate to control lens cell survival and link lens fibers to the surrounding extracellular matrix. The assembly and integrity of the lens capsule also appears to be reliant on integrin signaling within lens fibers. Extrapolation of these results indicates a novel role of integrins in lens cell-cell adhesions as well as a potential role in the pathogenesis of congenital cataracts.
Collapse
Affiliation(s)
- Andrew R. Samuelsson
- Department of Neurobiology, University of Pittsburgh, 1402 E Biological Science Tower, Pittsburgh PA 15261
| | - Richard Belvindrah
- Department of Cell Biology and Institute for Childhood and Neglected Disease, Scripps Research Institute, La Jolla, CA 92037
| | - Chuanyue Wu
- Department of Pathology, 707 Scaife Hall, University of Pittsburgh, Pittsburgh PA 15261
| | - Uli Müller
- Department of Cell Biology and Institute for Childhood and Neglected Disease, Scripps Research Institute, La Jolla, CA 92037
| | - Willi Halfter
- Department of Neurobiology, University of Pittsburgh, 1402 E Biological Science Tower, Pittsburgh PA 15261
| |
Collapse
|
10
|
Demircioglu F, Hodivala-Dilke K. αvβ3 Integrin and tumour blood vessels-learning from the past to shape the future. Curr Opin Cell Biol 2016; 42:121-127. [PMID: 27474973 DOI: 10.1016/j.ceb.2016.07.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/05/2016] [Accepted: 07/09/2016] [Indexed: 12/14/2022]
Abstract
Angiogenesis, the formation of new blood vessels from pre-existing ones, is thought to enhance tumour growth and these blood vessels can act as conduits of tumour cell metastasis. Integrins, the family of cell surface extracellular matrix receptors, can promote endothelial cell migration and survival, both essential features of angiogenesis, and were thus considered good targets for anti-angiogenic therapy. This sparked the development of agents to block integrin function as new cancer therapies. Here, we review the current status of αvβ3-integrin in tumour angiogenesis. Learning from what we now know about integrin conformational changes and endocytosis, we discuss the possible future of targeting blood vessel αvβ3-integrin in the control of cancer.
Collapse
Affiliation(s)
- Fevzi Demircioglu
- Centre for Tumour Biology, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1 M 6BQ, United Kingdom
| | - Kairbaan Hodivala-Dilke
- Centre for Tumour Biology, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1 M 6BQ, United Kingdom.
| |
Collapse
|
11
|
Blandin AF, Noulet F, Renner G, Mercier MC, Choulier L, Vauchelles R, Ronde P, Carreiras F, Etienne-Selloum N, Vereb G, Lelong-Rebel I, Martin S, Dontenwill M, Lehmann M. Glioma cell dispersion is driven by α5 integrin-mediated cell-matrix and cell-cell interactions. Cancer Lett 2016; 376:328-38. [PMID: 27063097 DOI: 10.1016/j.canlet.2016.04.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 12/11/2022]
Abstract
Glioblastoma multiform (GBM) is the most common and most aggressive primary brain tumor. The fibronectin receptor, α5 integrin is a pertinent novel therapeutic target. Despite numerous data showing that α5 integrin support tumor cell migration and invasion, it has been reported that α5 integrin can also limit cell dispersion by increasing cell-cell interaction. In this study, we showed that α5 integrin was involved in cell-cell interaction and gliomasphere formation. α5-mediated cell-cell cohesion limited cell dispersion from spheroids in fibronectin-poor microenvironment. However, in fibronectin-rich microenvironment, α5 integrin promoted cell dispersion. Ligand-occupied α5 integrin and fibronectin were distributed in fibril-like pattern at cell-cell junction of evading cells, forming cell-cell fibrillar adhesions. Activated focal adhesion kinase was not present in these adhesions but was progressively relocalized with α5 integrin as cell migrates away from the spheroids. α5 integrin function in GBM appears to be more complex than previously suspected. As GBM overexpressed fibronectin, it is most likely that in vivo, α5-mediated dissemination from the tumor mass overrides α5-mediated tumor cell cohesion. In this respect, α5-integrin antagonists may be useful to limit GBM invasion in brain parenchyma.
Collapse
Affiliation(s)
- Anne-Florence Blandin
- Integrins and Cancer, Faculté de Pharmacie, UMR7213 CNRS, LBP, Tumoral Signaling and Therapeutic Targets Department, Université de Strasbourg, Illkirch, France
| | - Fanny Noulet
- Integrins and Cancer, Faculté de Pharmacie, UMR7213 CNRS, LBP, Tumoral Signaling and Therapeutic Targets Department, Université de Strasbourg, Illkirch, France
| | - Guillaume Renner
- Integrins and Cancer, Faculté de Pharmacie, UMR7213 CNRS, LBP, Tumoral Signaling and Therapeutic Targets Department, Université de Strasbourg, Illkirch, France
| | - Marie-Cécile Mercier
- Integrins and Cancer, Faculté de Pharmacie, UMR7213 CNRS, LBP, Tumoral Signaling and Therapeutic Targets Department, Université de Strasbourg, Illkirch, France
| | - Laurence Choulier
- Integrins and Cancer, Faculté de Pharmacie, UMR7213 CNRS, LBP, Tumoral Signaling and Therapeutic Targets Department, Université de Strasbourg, Illkirch, France
| | - Romain Vauchelles
- Integrins and Cancer, Faculté de Pharmacie, UMR7213 CNRS, LBP, Tumoral Signaling and Therapeutic Targets Department, Université de Strasbourg, Illkirch, France
| | - Philippe Ronde
- Integrins and Cancer, Faculté de Pharmacie, UMR7213 CNRS, LBP, Tumoral Signaling and Therapeutic Targets Department, Université de Strasbourg, Illkirch, France
| | - Franck Carreiras
- Equipe de Recherche sur les Relations Matrice Extracellulaire Cellules, ERRMECe (EA 1391), Institut des Matériaux, Université de Cergy-Pontoise, France
| | - Nelly Etienne-Selloum
- Integrins and Cancer, Faculté de Pharmacie, UMR7213 CNRS, LBP, Tumoral Signaling and Therapeutic Targets Department, Université de Strasbourg, Illkirch, France; Department of Pharmacy, Centre Paul Strauss, Strasbourg, France
| | - Gyorgy Vereb
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Hungary
| | - Isabelle Lelong-Rebel
- Integrins and Cancer, Faculté de Pharmacie, UMR7213 CNRS, LBP, Tumoral Signaling and Therapeutic Targets Department, Université de Strasbourg, Illkirch, France
| | - Sophie Martin
- Integrins and Cancer, Faculté de Pharmacie, UMR7213 CNRS, LBP, Tumoral Signaling and Therapeutic Targets Department, Université de Strasbourg, Illkirch, France
| | - Monique Dontenwill
- Integrins and Cancer, Faculté de Pharmacie, UMR7213 CNRS, LBP, Tumoral Signaling and Therapeutic Targets Department, Université de Strasbourg, Illkirch, France
| | - Maxime Lehmann
- Integrins and Cancer, Faculté de Pharmacie, UMR7213 CNRS, LBP, Tumoral Signaling and Therapeutic Targets Department, Université de Strasbourg, Illkirch, France.
| |
Collapse
|
12
|
NOTCH1 intracellular domain negatively regulates PAK1 signaling pathway through direct interaction. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:179-88. [DOI: 10.1016/j.bbamcr.2015.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 11/01/2015] [Accepted: 11/03/2015] [Indexed: 12/28/2022]
|
13
|
Yen CF, Wang HS, Lee CL, Liao SK. Roles of integrin-linked kinase in cell signaling and its perspectives as a therapeutic target. Gynecol Minim Invasive Ther 2014. [DOI: 10.1016/j.gmit.2014.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
14
|
Eckes B, Krieg T, Wickström SA. Role of integrin signalling through integrin-linked kinase in skin physiology and pathology. Exp Dermatol 2014; 23:453-6. [DOI: 10.1111/exd.12429] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2014] [Indexed: 12/31/2022]
Affiliation(s)
- Beate Eckes
- Dermatology; University of Cologne; Cologne Germany
| | - Thomas Krieg
- Dermatology; University of Cologne; Cologne Germany
- CECAD Cologne Excellent in Aging Research; Cologne Germany
- Center for Molecular Medicine Cologne (CMMC); Cologne Germany
| | - Sara A. Wickström
- CECAD Cologne Excellent in Aging Research; Cologne Germany
- Paul Gerson Unna Group ‘Skin Homeostasis and Ageing’; Max Planck Institute for Biology of Ageing; Cologne Germany
| |
Collapse
|
15
|
Honda S, Shirotani-Ikejima H, Tadokoro S, Tomiyama Y, Miyata T. The integrin-linked kinase-PINCH-parvin complex supports integrin αIIbβ3 activation. PLoS One 2013; 8:e85498. [PMID: 24376884 PMCID: PMC3871693 DOI: 10.1371/journal.pone.0085498] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 12/05/2013] [Indexed: 12/22/2022] Open
Abstract
Integrin-linked kinase (ILK) is an important signaling regulator that assembles into the heteroternary complex with adaptor proteins PINCH and parvin (termed the IPP complex). We recently reported that ILK is important for integrin activation in a Chinese hamster ovary (CHO) cell system. We previously established parental CHO cells expressing a constitutively active chimeric integrin (αIIbα6Bβ3) and mutant CHO cells expressing inactive αIIbα6Bβ3 due to ILK deficiency. In this study, we further investigated the underlying mechanisms for ILK-dependent integrin activation. ILK-deficient mutant cells had trace levels of PINCH and α-parvin, and transfection of ILK cDNA into the mutant cells increased not only ILK but also PINCH and α-parvin, resulting in the restoration of αIIbα6Bβ3 activation. In the parental cells expressing active αIIbα6Bβ3, ILK, PINCH, and α-parvin were co-immunoprecipitated, indicating the formation of the IPP complex. Moreover, short interfering RNA (siRNA) experiments targeting PINCH-1 or both α- and β-parvin mRNA in the parent cells impaired the αIIbα6Bβ3 activation as well as the expression of the other components of the IPP complex. In addition, ILK mutants possessing defects in either PINCH or parvin binding failed to restore αIIbα6Bβ3 activation in the mutant cells. Kindlin-2 siRNA in the parental cells impaired αIIbα6Bβ3 activation without disturbing the expression of ILK. For CHO cells stably expressing wild-type αIIbβ3 that is an inactive form, overexpression of a talin head domain (THD) induced αIIbβ3 activation and the THD-induced αIIbβ3 activation was impaired by ILK siRNA through a significant reduction in the expression of the IPP complex. In contrast, overexpression of all IPP components in the αIIbβ3-expressing CHO cells further augmented THD-induced αIIbβ3 activation, whereas they did not induce αIIbβ3 activation without THD. These data suggest that the IPP complex rather than ILK plays an important role and supports integrin activation probably through stabilization of the active conformation.
Collapse
Affiliation(s)
- Shigenori Honda
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Suita, Japan
- * E-mail:
| | | | - Seiji Tadokoro
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshiaki Tomiyama
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Blood Transfusion, Osaka University Hospital, Suita, Osaka, Japan
| | - Toshiyuki Miyata
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Suita, Japan
| |
Collapse
|
16
|
ILK: a pseudokinase with a unique function in the integrin-actin linkage. Biochem Soc Trans 2013; 41:995-1001. [PMID: 23863169 DOI: 10.1042/bst20130062] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ILK (integrin-linked kinase) is a central component of cell-matrix adhesions and an important regulator of integrin function. It forms a ternary complex with two other adaptor proteins, PINCH (particularly interesting cysteine- and histidine-rich protein) and parvin, forming the IPP (ILK-PINCH-parvin) complex that regulates the integrin-actin linkage as well as microtubule dynamics. These functions are essential for processes such as cell migration and matrix remodelling. The present review discusses the recent advances on the structural and functional characterization of ILK and the long-standing debate regarding its reported kinase activity.
Collapse
|
17
|
Serres E, Debarbieux F, Stanchi F, Maggiorella L, Grall D, Turchi L, Burel-Vandenbos F, Figarella-Branger D, Virolle T, Rougon G, Van Obberghen-Schilling E. Fibronectin expression in glioblastomas promotes cell cohesion, collective invasion of basement membrane in vitro and orthotopic tumor growth in mice. Oncogene 2013; 33:3451-62. [DOI: 10.1038/onc.2013.305] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 03/24/2013] [Accepted: 06/04/2013] [Indexed: 01/03/2023]
|
18
|
Elad N, Volberg T, Patla I, Hirschfeld-Warneken V, Grashoff C, Spatz JP, Fässler R, Geiger B, Medalia O. The role of integrin-linked kinase in the molecular architecture of focal adhesions. J Cell Sci 2013; 126:4099-107. [PMID: 23843624 DOI: 10.1242/jcs.120295] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Integrin-mediated focal adhesions (FAs) are large, multi-protein complexes that link the actin cytoskeleton to the extracellular matrix and take part in adhesion-mediated signaling. These adhesions are highly complex and diverse at the molecular level; thus, assigning particular structural or signaling functions to specific components is highly challenging. Here, we combined functional, structural and biophysical approaches to assess the role of a major FA component, namely, integrin-linked kinase (ILK), in adhesion formation. We show here that ILK plays a key role in the formation of focal complexes, early forms of integrin adhesions, and confirm its involvement in the assembly of fibronectin-bound fibrillar adhesions. Examination of ILK-null fibroblasts by cryo-electron tomography pointed to major structural changes in their FAs, manifested as disarray of the associated actin filaments and an increase in the packing density of FA-related particles. Interestingly, adhesion of the mutant cells to the substrate required a higher ligand density than in control cells. These data indicate that ILK has a key role in integrin adhesion assembly and sub-structure, and in the regulation of the FA-associated cytoskeleton.
Collapse
Affiliation(s)
- Nadav Elad
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben Gurion University of the Negev, Beer-Sheva 84120, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Malan D, Elischer A, Hesse M, Wickström SA, Fleischmann BK, Bloch W. Deletion of integrin linked kinase in endothelial cells results in defective RTK signaling caused by caveolin 1 mislocalization. Development 2013; 140:987-95. [PMID: 23404105 DOI: 10.1242/dev.091298] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Integrin linked kinase (ILK) connects the ILK-Pinch-Parvin complex with integrin adhesion sites. Because of the functional relevance of integrin-linked signaling for endothelial cell (EC) biology, we have explored this pathway in Ilk(-/-) embryonic stem (ES) cells differentiated into ECs and vessel-like structures. We have focused in particular on the mechanistic relevance of ILK-Pinch-Parvin complex-related signaling for EC development and tube formation. Our analysis revealed that the formation of vessel-like structures was strongly reduced in Ilk(-/-) ES cells and that this phenotype could be rescued by re-expression of ILK in ES cells. ECs were MACS sorted from wild-type (WT) and Ilk(-/-) ES cells and functional analysis using intracellular calcium imaging as the read-out yielded a complete lack of vascular endothelial growth factor- and epidermal growth factor-dependent responses. The possibility of a caveolin 1-related defect was investigated by transfecting WT and Ilk(-/-) ECs with a caveolin 1-EGFP fusion protein. Time-lapse microscopy showed that the prominent phenotype is due to altered dynamics of caveolin 1 and to a lack of positioning of caveolin 1 in the vicinity of the plasma membrane and that it is rescued by re-expressing ILK in the Ilk(-/-) ES cells. We also found that the defect is caused by the perturbed organization of microtubules and cortical actin filaments. Thus, ILK is required as a scaffold to allow actin-microtubule interactions and correct positioning of caveolin 1 close to the plasma membrane. This is crucial for signaling compartmentalization in ECs and explains the key role of ILK for EC development and function.
Collapse
Affiliation(s)
- Daniela Malan
- Institute of Physiology I, Life and Brain Center, University of Bonn, Bonn, NRW, 53105, Germany
| | | | | | | | | | | |
Collapse
|
20
|
Gupton SL, Riquelme D, Hughes-Alford SK, Tadros J, Rudina SS, Hynes RO, Lauffenburger D, Gertler FB. Mena binds α5 integrin directly and modulates α5β1 function. ACTA ACUST UNITED AC 2012; 198:657-76. [PMID: 22908313 PMCID: PMC3514034 DOI: 10.1083/jcb.201202079] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Mena binds to the cytoplasmic tail of α5 integrin and modulates key
α5β1 integrin functions in adhesion, motility, and
fibrillogenesis. Mena is an Ena/VASP family actin regulator with roles in cell migration,
chemotaxis, cell–cell adhesion, tumor cell invasion, and metastasis.
Although enriched in focal adhesions, Mena has no established function within
these structures. We find that Mena forms an adhesion-regulated complex with
α5β1 integrin, a fibronectin receptor involved in cell adhesion,
motility, fibronectin fibrillogenesis, signaling, and growth factor receptor
trafficking. Mena bound directly to the carboxy-terminal portion of the
α5 cytoplasmic tail via a 91-residue region containing 13 five-residue
“LERER” repeats. In fibroblasts, the Mena–α5 complex
was required for “outside-in” α5β1 functions,
including normal phosphorylation of FAK and paxillin and formation of fibrillar
adhesions. It also supported fibrillogenesis and cell spreading and controlled
cell migration speed. Thus, fibroblasts require Mena for multiple
α5β1-dependent processes involving bidirectional interactions
between the extracellular matrix and cytoplasmic focal adhesion proteins.
Collapse
Affiliation(s)
- Stephanie L Gupton
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
RGD-Dependent Epithelial Cell-Matrix Interactions in the Human Intestinal Crypt. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:248759. [PMID: 22988499 PMCID: PMC3440950 DOI: 10.1155/2012/248759] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 07/05/2012] [Accepted: 07/22/2012] [Indexed: 01/09/2023]
Abstract
Interactions between the extracellular matrix (ECM) and integrin receptors trigger structural and functional bonds between the cell microenvironment and the cytoskeleton. Such connections are essential for adhesion structure integrity and are key players in regulating transduction of specific intracellular signals, which in turn regulate the organization of the cell microenvironment and, consequently, cell function. The RGD peptide-dependent integrins represent a key subgroup of ECM receptors involved in the maintenance of epithelial homeostasis. Here we review recent findings on RGD-dependent ECM-integrin interactions and their roles in human intestinal epithelial crypt cells.
Collapse
|
22
|
Hao YC, Yu LP, Li Q, Zhang XW, Zhao YP, He PY, Xu T, Wang XF. Effects of integrin-linked kinase on human corpus cavernosum smooth muscle cell cytoskeletal organisation. Andrologia 2012; 45:78-85. [PMID: 22616551 DOI: 10.1111/j.1439-0272.2012.01313.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2012] [Indexed: 11/26/2022] Open
Abstract
We investigated the effects of integrin-linked kinase (ILK) on the in vitro attachment, spreading, migration and microfilament dynamics of human corpus cavernosum smooth muscle cells. ILK small interfering RNA (siRNA) was used to transfect human corpus cavernosum smooth muscle cells; and cell attachment, spreading and migration were assessed. Additionally, microfilament dynamics were evaluated using Alexa Fluor 488 and phalloidin staining. We found that ILK gene knock-down significantly inhibited human corpus cavernosum smooth muscle cell attachment, spreading and migration. Moreover, blocking the expression of ILK disturbed actin cytoskeleton reorganisation and morphology in human corpus cavernosum smooth muscle cells. These results show that the targeting of ILK with siRNA significantly inhibited cell attachment, spreading, migration and microfilament dynamics in human corpus cavernosum smooth muscle cells. These findings indicate that ILK might be a potential therapeutic molecular target for the treatment of erectile dysfunction.
Collapse
Affiliation(s)
- Y-C Hao
- Urology Department, Peking University People's Hospital, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Wang F, Wang Y, Zhang L, Zou L. Gene modification with integrin-linked kinase improves function of endothelial progenitor cells in pre-eclampsia in vitro. J Cell Biochem 2012; 112:3103-11. [PMID: 21688303 DOI: 10.1002/jcb.23236] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Integrin-linked kinase (ILK), a multifunctional serine-threonine protein kinase, has been shown to have implications for the treatment of ischemia vascular diseases by promoting angiogenesis in various tissues. However, whether this kinase has therapeutic potential in pre-eclampsia is not well studied. In this report, we determined the changes in the production and action of ILK on endothelial progenitor cells (EPCs) isolated from patients with pre-eclampsia. The effects of ILK transfection on proliferation, migration, and angiogenesis of EPCs were investigated. We showed that EPCs transfected with the ILK gene expressed high levels of ILK protein and mRNA. Transfection with ILK also enhanced the proliferative, migratory, and angiogenic capabilities of EPCs, and promoted the production of VEGF. These results suggest that ILK gene transfection is an effective approach to augment angiogenic properties of EPCs in vitro and providing basis for clinical cell-based gene therapy in patients with pre-eclampsia.
Collapse
Affiliation(s)
- Fang Wang
- Department of Gynecology and Obstetrics, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | | | | | | |
Collapse
|
24
|
Herranz B, Marquez S, Guijarro B, Aracil E, Aicart-Ramos C, Rodriguez-Crespo I, Serrano I, Rodríguez-Puyol M, Zaragoza C, Saura M. Integrin-linked kinase regulates vasomotor function by preventing endothelial nitric oxide synthase uncoupling: role in atherosclerosis. Circ Res 2011; 110:439-49. [PMID: 22194624 DOI: 10.1161/circresaha.111.253948] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Atherosclerotic lesions develop in regions of disturbed flow, whereas laminar flow protects from atherogenesis; however, the mechanisms involved are not completely elucidated. Integrins are mechanosensors of shear stress in endothelial cells, and integrin-linked kinase (ILK) is important for blood vessel integrity and cardiovascular development. OBJECTIVES To explore the role of ILK in vascular function by studying conditionally ILK-deficient (cKO) mice and human atherosclerotic arteries. RESULTS ILK expression was detected in the endothelial cell layer of nonatherosclerotic vessels but was absent from the endothelium of atherosclerotic arteries. Live ultrasound imaging revealed that acetylcholine-mediated vasodilatation was impaired in cKO mice. These mice exhibited lowered agonist-induced nitric oxide synthase (NOS) activity and decreased cyclic guanosine monophosphate and nitrite production. ILK deletion caused endothelial NOS (eNOS) uncoupling, reflected in reduced tetrahydrobiopterin (BH4) levels, increased BH2 levels, decreased dihydrofolate reductase expression, and increased eNOS-dependent generation of superoxide accompanied by extensive vascular protein nitration. ILK reexpression prevented eNOS uncoupling in cKO cells, whereas superoxide formation was unaffected by ILK depletion in eNOS-KO cells, indicating eNOS as a primary source of superoxide anion. eNOS and ILK coimmunoprecipitated in aortic lysates from control animals, and eNOS-ILK-shock protein 90 interaction was detected in human normal mammary arteries but was absent from human atherosclerotic carotid arteries. eNOS-ILK interaction in endothelial cells was prevented by geldanamycin, suggesting heat shock protein 90 as a binding partner. CONCLUSIONS Our results identify ILK as a regulatory partner of eNOS in vivo that prevents eNOS uncoupling, and suggest ILK as a therapeutic target for prevention of endothelial dysfunction related to shear stress-induced vascular diseases.
Collapse
|
25
|
Liang X, Sun Y, Chen J. Particularly interesting cysteine- and histidine-rich protein in cardiac development and remodeling. J Investig Med 2011; 57:842-8. [PMID: 19952891 DOI: 10.2310/jim.0b013e3181c5e31d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Integrin-mediated cell-extracellular matrix interaction plays key roles in tissue morphogenesis and integrity. The Lin11-Isl-1-Mec-3 (LIM) domain-only particularly interesting cysteine- and histidine-rich (PINCH) protein functions as an adaptor essential for the assembly and function of the focal adhesion complex that links integrin signaling to the cytoskeleton and other intracellular signaling pathways and regulates diverse cellular processes such as cell adhesion, migration, growth, differentiation, and survival. Recent biochemical and genetic studies have greatly advanced our knowledge surrounding the molecular interactions and functions of each component of the focal adhesion complex and revealed a requirement for PINCH in early embryogenesis, in morphogenesis of the neural crest and cardiac outflow, and in myocardial growth and remodeling. In this review article, we will provide an overview of the current knowledge of the molecular interactions of PINCH with other components of focal adhesions, highlighting recent discoveries of the in vivo role of PINCH and discuss its potential implication for human heart disease.
Collapse
Affiliation(s)
- Xingqun Liang
- Department of Medicine, University of California at San Diego (UCSD), La Jolla, CA 92093-0613C, USA
| | | | | |
Collapse
|
26
|
Advances in tenascin-C biology. Cell Mol Life Sci 2011; 68:3175-99. [PMID: 21818551 PMCID: PMC3173650 DOI: 10.1007/s00018-011-0783-6] [Citation(s) in RCA: 255] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 07/19/2011] [Accepted: 07/19/2011] [Indexed: 12/11/2022]
Abstract
Tenascin-C is an extracellular matrix glycoprotein that is specifically and transiently expressed upon tissue injury. Upon tissue damage, tenascin-C plays a multitude of different roles that mediate both inflammatory and fibrotic processes to enable effective tissue repair. In the last decade, emerging evidence has demonstrated a vital role for tenascin-C in cardiac and arterial injury, tumor angiogenesis and metastasis, as well as in modulating stem cell behavior. Here we highlight the molecular mechanisms by which tenascin-C mediates these effects and discuss the implications of mis-regulated tenascin-C expression in driving disease pathology.
Collapse
|
27
|
Brunner M, Millon-Frémillon A, Chevalier G, Nakchbandi IA, Mosher D, Block MR, Albigès-Rizo C, Bouvard D. Osteoblast mineralization requires beta1 integrin/ICAP-1-dependent fibronectin deposition. ACTA ACUST UNITED AC 2011; 194:307-22. [PMID: 21768292 PMCID: PMC3144405 DOI: 10.1083/jcb.201007108] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
ICAP-1 prevents recruitment of kindlin-2 to β1 integrin to control
dynamics of fibrillar adhesion sites, fibronectin deposition, and osteoblast
mineralization during bone formation. The morphogenetic and differentiation events required for bone formation are
orchestrated by diffusible and insoluble factors that are localized within the
extracellular matrix. In mice, the deletion of ICAP-1, a modulator of β1
integrin activation, leads to severe defects in osteoblast proliferation,
differentiation, and mineralization and to a delay in bone formation. Deposition
of fibronectin and maturation of fibrillar adhesions, adhesive structures that
accompany fibronectin deposition, are impaired upon ICAP-1 loss, as are type I
collagen deposition and mineralization. Expression of β1 integrin with a
mutated binding site for ICAP-1 recapitulates the ICAP-1–null phenotype.
Follow-up experiments demonstrated that ICAP-1 negatively regulates kindlin-2
recruitment onto the β1 integrin cytoplasmic domain, whereas an excess of
kindlin-2 binding has a deleterious effect on fibrillar adhesion formation.
These results suggest that ICAP-1 works in concert with kindlin-2 to control the
dynamics of β1 integrin–containing fibrillar adhesions and,
thereby, regulates fibronectin deposition and osteoblast mineralization.
Collapse
Affiliation(s)
- Molly Brunner
- Equipe 1 Dynamique des Systèmes d'Adhérence et Différenciation Cellulaire, Institut National de la Santé et de la Recherche Médicale U823, Institut Albert Bonniot, 38042 Grenoble, Cedex 09, France
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Cseh B, Fernandez-Sauze S, Grall D, Schaub S, Doma E, Van Obberghen-Schilling E. Autocrine fibronectin directs matrix assembly and crosstalk between cell–matrix and cell–cell adhesion in vascular endothelial cells. J Cell Sci 2010; 123:3989-99. [DOI: 10.1242/jcs.073346] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Cellular fibronectin (cFN) variants harboring extra FN type 3 repeats, namely extra domains B and A, are major constituents of the extracellular matrix around newly forming blood vessels during development and angiogenesis. Their expression is induced by angiogenic stimuli and their assembly into fibrillar arrays is driven by cell-generated tension at α5β1 integrin-based adhesions. Here, we examined the role and functional redundancy of cFN variants in cultured endothelial cells by isoform-selective RNA interference. We show that FN fibrillogenesis is a cell-autonomous process whereby basally directed secretion and assembly of cellular FN are tightly coupled events that play an important role not only in signaling at cell–matrix adhesions but also at cell–cell contacts. Silencing of cFN variants differentially affects integrin usage, cell spreading, motility and capillary morphogenesis in vitro. cFN-deficient cells undergo a switch from α5β1- to αvβ3-based adhesion, accompanied by a Src-regulated disruption of adherens junctions. These studies identify a crucial role for autocrine FN in subendothelial matrix assembly and junctional integrity that provides spatially and temporally restricted control of endothelial plasticity during angiogenic blood vessel remodeling.
Collapse
Affiliation(s)
- Botond Cseh
- University of Nice-Sophia Antipolis, CNRS UMR 6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| | - Samantha Fernandez-Sauze
- University of Nice-Sophia Antipolis, CNRS UMR 6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| | - Dominique Grall
- University of Nice-Sophia Antipolis, CNRS UMR 6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| | - Sébastien Schaub
- University of Nice-Sophia Antipolis, CNRS UMR 6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| | - Eszter Doma
- University of Nice-Sophia Antipolis, CNRS UMR 6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| | - Ellen Van Obberghen-Schilling
- University of Nice-Sophia Antipolis, CNRS UMR 6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| |
Collapse
|
29
|
Lutz R, Sakai T, Chiquet M. Pericellular fibronectin is required for RhoA-dependent responses to cyclic strain in fibroblasts. J Cell Sci 2010; 123:1511-1521. [PMID: 20375066 DOI: 10.1242/jcs.060905] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
To test the hypothesis that the pericellular fibronectin matrix is involved in mechanotransduction, we compared the response of normal and fibronectin-deficient mouse fibroblasts to cyclic substrate strain. Normal fibroblasts seeded on vitronectin in fibronectin-depleted medium deposited their own fibronectin matrix. In cultures exposed to cyclic strain, RhoA was activated, actin-stress fibers became more prominent, MAL/MKL1 shuttled to the nucleus, and mRNA encoding tenascin-C was induced. By contrast, these RhoA-dependent responses to cyclic strain were suppressed in fibronectin knockdown or knockout fibroblasts grown under identical conditions. On vitronectin substrate, fibronectin-deficient cells lacked fibrillar adhesions containing alpha5 integrin. However, when fibronectin-deficient fibroblasts were plated on exogenous fibronectin, their defects in adhesions and mechanotransduction were restored. Studies with fragments indicated that both the RGD-synergy site and the adjacent heparin-binding region of fibronectin were required for full activity in mechanotransduction, but not its ability to self-assemble. In contrast to RhoA-mediated responses, activation of Erk1/2 and PKB/Akt by cyclic strain was not affected in fibronectin-deficient cells. Our results indicate that pericellular fibronectin secreted by normal fibroblasts is a necessary component of the strain-sensing machinery. Supporting this hypothesis, induction of cellular tenascin-C by cyclic strain was suppressed by addition of exogenous tenascin-C, which interferes with fibronectin-mediated cell spreading.
Collapse
Affiliation(s)
- Roman Lutz
- Friedrich Miescher Institute for Biomedical Research, CH-4056 Basel, Switzerland
| | | | | |
Collapse
|
30
|
Boulter E, Garcia-Mata R, Guilluy C, Dubash A, Rossi G, Brennwald PJ, Burridge K. Regulation of Rho GTPase crosstalk, degradation and activity by RhoGDI1. Nat Cell Biol 2010; 12:477-83. [PMID: 20400958 PMCID: PMC2866742 DOI: 10.1038/ncb2049] [Citation(s) in RCA: 272] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 03/30/2010] [Indexed: 01/07/2023]
Abstract
At steady state, most Rho GTPases are bound in the cytosol to Rho guanine nucleotide dissociation inhibitors (RhoGDIs). RhoGDIs have generally been considered to hold Rho proteins passively in an inactive state within the cytoplasm. Here we describe an evolutionarily conserved mechanism by which RhoGDI1 controls the homeostasis of Rho proteins in eukaryotic cells. We found that depletion of RhoGDI1 promotes misfolding and degradation of the cytosolic geranylgeranylated pool of Rho GTPases while activating the remaining membrane-bound fraction. Because RhoGDI1 levels are limiting, and Rho proteins compete for binding to RhoGDI1, overexpression of an exogenous Rho GTPase displaces endogenous Rho proteins bound to RhoGDI1, inducing their degradation and inactivation. These results raise important questions about the conclusions drawn from studies that manipulate Rho protein levels. In many cases the response observed may arise not simply from the overexpression itself but from additional effects on the levels and activity of other Rho GTPases as a result of competition for binding to RhoGDI1; this may require a re-evaluation of previously published studies that rely exclusively on these techniques.
Collapse
Affiliation(s)
- Etienne Boulter
- Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Rafael Garcia-Mata
- Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Christophe Guilluy
- Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Adi Dubash
- Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Guendalina Rossi
- Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Patrick J. Brennwald
- Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Keith Burridge
- Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Lineberger Comprehensive Cancer Center and UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
31
|
Huang S, Sun Z, Li Z, Martinez-Lemus LA, Meininger GA. Modulation of microvascular smooth muscle adhesion and mechanotransduction by integrin-linked kinase. Microcirculation 2010; 17:113-27. [PMID: 20163538 PMCID: PMC2923833 DOI: 10.1111/j.1549-8719.2009.00011.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
OBJECTIVE In this study, we investigated the involvement of integrin-linked kinase (ILK) in the adhesion of arteriolar vascular smooth muscle cells (VSMC) to fibronectin (FN) and in the mechano-responsiveness of VSMC focal adhesions (FA). METHODS ILK was visualized in VSMC by expressing EGFP-ILK and it was knocked down using ILK-shRNA constructs. Atomic force microscopy (AFM) was used to characterize VSMC interactions with FN, VSMC stiffness and to apply and measure forces at a VSMC single FA site. RESULTS ILK was localized to FA and silencing ILK promoted cell spreading, enhanced cell adhesion, reduced cell proliferation and reduced downstream phosphorylation of GSK-3beta and PKB/Akt. AFM studies demonstrated that silencing ILK enhanced alpha5beta1 integrin adhesion to FN and enhanced VSMC contraction in response to a pulling force applied at the level of a single FN-FA site. CONCLUSIONS ILK functions in arteriolar VSMC appear linked to multiple signaling pathways and processes that inhibit cell spreading, cell adhesion, FA formation, adhesion to FN and the mechano-responsiveness of FN-FA sites.
Collapse
Affiliation(s)
- Shaoxing Huang
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO 65211
| | - Zhe Sun
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO 65211
| | - Zhaohui Li
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO 65211
| | - Luis A. Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO 65211
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO 65211
| | - Gerald A. Meininger
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO 65211
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO 65211
| |
Collapse
|
32
|
Gagné D, Groulx JF, Benoit YD, Basora N, Herring E, Vachon PH, Beaulieu JF. Integrin-linked kinase regulates migration and proliferation of human intestinal cells under a fibronectin-dependent mechanism. J Cell Physiol 2009; 222:387-400. [PMID: 19885839 PMCID: PMC2814089 DOI: 10.1002/jcp.21963] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Integrin-linked kinase (ILK) plays a role in integrin signaling-mediated extracellular matrix (ECM)–cell interactions and also acts as a scaffold protein in functional focal adhesion points. In the present study, we investigated the expression and roles of ILK in human intestinal epithelial cells (IECs) in vivo and in vitro. Herein, we report that ILK and its scaffold-function interacting partners, PINCH-1, α-parvin, and β-parvin, are expressed according to a decreasing gradient from the bottom of the crypt (proliferative/undifferentiated) compartment to the tip of the villus (non-proliferative/differentiated) compartment, closely following the expression pattern of the ECM/basement membrane component fibronectin. The siRNA knockdown of ILK in human IECs caused a loss of PINCH-1, α-parvin, and β-parvin expression, along with a significant decrease in cell proliferation via a loss of cyclin D1 and an increase in p27 and hypophosphorylated pRb expression levels. ILK knockdown severely affected cell spreading, migration, and restitution abilities, which were shown to be directly related to a decrease in fibronectin deposition. All ILK knockdown-induced defects were rescued with exogenously deposited fibronectin. Altogether, our results indicate that ILK performs crucial roles in the control of human intestinal cell and crypt–villus axis homeostasis—especially with regard to basement membrane fibronectin deposition—as well as cell proliferation, spreading, and migration. J. Cell. Physiol. 222: 387–400, 2010. © 2009 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- David Gagné
- CIHR Team on the Digestive Epithelium, Département d'Anatomie et de Biologie Cellulaire, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|
33
|
Kogata N, Tribe RM, Fässler R, Way M, Adams RH. Integrin-linked kinase controls vascular wall formation by negatively regulating Rho/ROCK-mediated vascular smooth muscle cell contraction. Genes Dev 2009; 23:2278-83. [PMID: 19797768 DOI: 10.1101/gad.535409] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Vascular smooth muscle cells (VSMCs) form contractile layers around larger blood vessels in a process that is essential for the formation of a fully functional vasculature. Here, we show that integrin-linked kinase (ILK) is required for the formation of a unitary layer of aligned VSMCs around arterioles and the regulation of blood vessel constriction in mice. In the absence of ILK, activated Rho/ROCK signaling induces the elevated phosphorylation of myosin light chain leading to abnormally enhanced VSMC contraction in vitro and in vivo. Our findings identify ILK as a key component regulating vascular wall formation by negatively modulating VSMC contractility.
Collapse
Affiliation(s)
- Naoko Kogata
- Vascular Development Laboratory, UK London Research Institute, London WC2A 3PX, United Kingdom
| | | | | | | | | |
Collapse
|
34
|
Vecchione C, Carnevale D, Di Pardo A, Gentile MT, Damato A, Cocozza G, Antenucci G, Mascio G, Bettarini U, Landolfi A, Iorio L, Maffei A, Lembo G. Pressure-induced vascular oxidative stress is mediated through activation of integrin-linked kinase 1/betaPIX/Rac-1 pathway. Hypertension 2009; 54:1028-34. [PMID: 19770407 DOI: 10.1161/hypertensionaha.109.136572] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
High blood pressure induces a mechanical stress on vascular walls and evokes oxidative stress and vascular dysfunction. The aim of this study was to characterize the intracellular signaling causing vascular oxidative stress in response to pressure. In carotid arteries subjected to high pressure levels, we observed not only an impaired vasorelaxation, increased superoxide production, and NADPH oxidase activity, but also a concomitant activation of Rac-1, a small G protein. Selective inhibition of Rac-1, with an adenovirus carrying a dominant-negative Rac-1 mutant, significantly reduced NADPH oxidase activity and oxidative stress and, more importantly, rescued vascular function in carotid arteries at high pressure. The analysis of molecular events associated with mechanotransduction demonstrated at high pressure levels an overexpression of integrin-linked kinase 1 and its recruitment to plasma membrane interacting with paxillin. The inhibition of integrin-linked kinase 1 by small interfering RNA impaired Rac-1 activation and rescued oxidative stress-induced vascular dysfunction in response to high pressure. Finally, we showed that betaPIX, a guanine-nucleotide exchange factor, is the intermediate molecule recruited by integrin-linked kinase 1, converging the intracellular signaling toward Rac-1-mediated oxidative vascular dysfunction during pressure overload. Our data demonstrate that biomechanical stress evoked by high blood pressure triggers an integrin-linked kinase 1/betaPIX/Rac-1 signaling, thus generating oxidative vascular dysfunction.
Collapse
Affiliation(s)
- Carmine Vecchione
- Department of Angio-Cardio-Neurology, Neuromed Institute Istituto Di Ricovero e Cura a Carattere Scientifico, Pozzilli, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Blair HC, Yaroslavskiy BB, Robinson LJ, Mapara MY, Pangrazio A, Guo L, Chen K, Vezzoni P, Tolar J, Orchard PJ. Osteopetrosis with micro-lacunar resorption because of defective integrin organization. J Transl Med 2009; 89:1007-17. [PMID: 19546854 PMCID: PMC2856930 DOI: 10.1038/labinvest.2009.58] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In vitro differentiated monocytes were used to characterize the cellular defect in a type of osteopetrosis with minimally functional osteoclasts, in which defects associated with common causes of osteopetrosis were excluded by gene sequencing. Monocytes from the blood of a 28-year-old patient were differentiated in media with RANKL and CSF-1. Cell fusion, acid compartments within cells, and tartrate resistant acid phosphatase (TRAP) activity were normal. However, the osteoclasts made abnormally small pits on the dentine. Phalloidin labeling showed that the cell attachments lacked the peripheral ring structure that supports lacunar resorption. Instead, the osteoclasts had clusters of podosomes near the center of cell attachments. Antibody to the alphavbeta3 integrin pair or to the C-terminal of beta3 did not label podosomes, but antibody to alphav labeled them. Western blots using antibody to the N-terminal of beta3 showed a protein of reduced size. Integrins beta1 and beta5 were upregulated, but, in contrast to observations in beta3 defects, alpha2 had not increased. The rho-GTP exchange protein Vav3, a key attachment organizing protein, did not localize normally with peripheral attachment structures. Vav3 forms of 70 kD and 90 kD were identified on western blots. However, the proteins beta3 integrin, Vav3, Plekhm1, and Src, implicated in attachment defects, had normal exon sequences. In this new type of osteopetrosis, the integrin-organizing complex is dysfunctional, and at least two attachment proteins may be partially degraded.
Collapse
Affiliation(s)
- Harry C Blair
- Department of Pathology, University of Pittsburgh, and Veteran's Affairs Medical Center, Pittsburgh, PA 15261, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Stanchi F, Grashoff C, Nguemeni Yonga CF, Grall D, Fässler R, Van Obberghen-Schilling E. Molecular dissection of the ILK-PINCH-parvin triad reveals a fundamental role for the ILK kinase domain in the late stages of focal-adhesion maturation. J Cell Sci 2009; 122:1800-11. [PMID: 19435803 DOI: 10.1242/jcs.044602] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Integrin-linked kinase (ILK) and cytoplasmic adaptors of the PINCH and parvin families form a ternary complex, termed IPP, that localizes to integrin adhesions. We show here that deletion of the genes encoding ILK or PINCH1 similarly blocks maturation of focal adhesions to tensin-rich and phosphotyrosine-poor fibrillar adhesions (FBs) by downregulating expression or recruitment of tensin and destabilizing alpha5beta1-integrin-cytoskeleton linkages. As IPP components are interdependent for integrin targeting and protein stability, functional dissection of the complex was achieved by fusing ILK, PINCH, parvin or their individual motifs to the cytoplasmic tail of beta3 integrin, normally excluded from FBs. Using this novel gain-of-function approach, we demonstrated that expression of the C-terminal kinase domain of ILK can restore tensin recruitment and prompt focal-adhesion maturation in IPP-null cells. Debilitating mutations in the paxillin- or ATP-binding sites of ILK, together with alpha-parvin silencing, revealed a determinant role for ILK-parvin association, but not for direct paxillin binding, in this function. We propose a model in which the C-terminal domain of ILK promotes integrin sorting by reinforcing alpha5beta1-integrin-actin linkage and controls force transmission by targeting tensin to maturing adhesions.
Collapse
Affiliation(s)
- Fabio Stanchi
- Institute of Developmental Biology and Cancer Research, University of Nice-Sophia Antiopolis, CNRS-UMR6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| | | | | | | | | | | |
Collapse
|
37
|
Fan Y, Gong Y, Ghosh PK, Graham LM, Fox PL. Spatial coordination of actin polymerization and ILK-Akt2 activity during endothelial cell migration. Dev Cell 2009; 16:661-74. [PMID: 19460343 PMCID: PMC2692108 DOI: 10.1016/j.devcel.2009.03.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2008] [Revised: 02/04/2009] [Accepted: 03/11/2009] [Indexed: 10/20/2022]
Abstract
Eukaryotic cell migration proceeds by cycles of protrusion, adhesion, and contraction, regulated by actin polymerization, focal adhesion assembly, and matrix degradation. However, mechanisms coordinating these processes remain largely unknown. Here, we show that local regulation of thymosin-beta4 (Tbeta4) binding to actin monomer (G-actin) coordinates actin polymerization with metalloproteinase synthesis to promote endothelial cell motility. In particular and quite unexpectedly, FRET analysis reveals diminished interaction between Tbeta4 and G-actin at the cell leading edge despite their colocalization there. Profilin-dependent dissociation of G-actin-Tbeta4 complexes simultaneously liberates actin for filament assembly and facilitates Tbeta4 binding to integrin-linked kinase (ILK) in the lamellipodia. Tbeta4-ILK complexes then recruit and activate Akt2, resulting in matrix metalloproteinase-2 production. Thus, the actin-Tbeta4 complex constitutes a latent coordinating center for cell migratory behavior, allowing profilin to initiate a cascade of events at the leading edge that couples actin polymerization to matrix degradation.
Collapse
Affiliation(s)
- Yi Fan
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | | | |
Collapse
|
38
|
Ho B, Bendeck MP. Integrin linked kinase (ILK) expression and function in vascular smooth muscle cells. Cell Adh Migr 2009; 3:174-6. [PMID: 19262169 DOI: 10.4161/cam.3.2.7374] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Vascular smooth muscle cell (SMC) migration and proliferation contribute to arterial wound repair and thickening of the intimal layer in atherosclerosis, restenosis and transplant vascular disease. These processes are influenced by cell adhesion to molecules present in the extracellular matrix, and regulated by the integrin family of cell-surface matrix receptors. An important signaling molecule acting downstream of integrin receptors is integrin-linked kinase (ILK), a serine/threonine kinase and scaffolding protein. ILK has been implicated in cancer cell growth and survival through modulation of downstream targets, notably Akt and glycogen synthase kinase-3beta (GSK3beta). Evidence also exists to establish ILK as a molecular adaptor protein linking integrins to the actin cytoskeleton and regulating actin polymerization, and this function may not necessarily depend upon the kinase activity of ILK. ILK has been implicated in anchorage-independent growth, cell cycle progression, epithelial-mesenchymal transition (EMT), invasion and migration. In addition, ILK has been shown to be involved in vascular development, tumor angiogenesis and cardiac hypertrophy. Despite the documented involvement of integrin signaling in vascular pathologies, the function of ILK has not been well characterized in the SMC response to vascular injury. This brief review summarizes and puts into context the current literature on ILK expression and function in the vascular smooth muscle cell.
Collapse
Affiliation(s)
- Bernard Ho
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, CA
| | | |
Collapse
|
39
|
Dubash AD, Menold MM, Samson T, Boulter E, García-Mata R, Doughman R, Burridge K. Chapter 1 Focal Adhesions: New Angles on an Old Structure. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2009; 277:1-65. [DOI: 10.1016/s1937-6448(09)77001-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
40
|
McDonald PC, Fielding AB, Dedhar S. Integrin-linked kinase--essential roles in physiology and cancer biology. J Cell Sci 2008; 121:3121-32. [PMID: 18799788 DOI: 10.1242/jcs.017996] [Citation(s) in RCA: 282] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Integrin-linked kinase (ILK) is a multifunctional intracellular effector of cell-matrix interactions and regulates many cellular processes, including growth, proliferation, survival, differentiation, migration, invasion and angiogenesis. The use of recently developed Cre-lox-driven recombination and RNA-interference technologies has enabled the evaluation of the physiological roles of ILK in several major organ systems. Significant developmental and tissue-homeostasis defects occur when the gene that encodes ILK is deleted, whereas the expression of ILK is often elevated in human malignancies. Although the cause(s) of ILK overexpression remain to be fully elucidated, accumulating evidence suggests that its oncogenic capacity derives from its regulation of several downstream targets that provide cells with signals that promote proliferation, survival and migration, supporting the concept that ILK is a relevant therapeutic target in human cancer. Furthermore, a global analysis of the ILK 'interactome' has yielded several novel interactions, and has revealed exciting and unexpected cellular functions of ILK that might have important implications for the development of effective therapeutic agents.
Collapse
Affiliation(s)
- Paul C McDonald
- British Columbia Cancer Agency, BC Cancer Research Centre, Department of Cancer Genetics, Vancouver, BC, Canada
| | | | | |
Collapse
|
41
|
Fantozzi I, Grall D, Cagnol S, Stanchi F, Sudaka A, Brunstein MC, Bozec A, Fischel JL, Milano G, Van Obberghen-Schilling E. Overexpression of cortactin in head and neck squamous cell carcinomas can be uncoupled from augmented EGF receptor expression. Acta Oncol 2008; 47:1502-12. [PMID: 18607838 DOI: 10.1080/02841860802089801] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND The gene encoding cortactin, CTTN (locus 11q13), an actin-binding substrate of Src kinases, is frequently amplified in breast and head and neck squamous cell carcinomas (HNSCC) and cortactin overexpression is thought to contribute in a significant way to the invasive phenotype of these tumors. Elevated Epidermal Growth Factor receptor (EGFR) expression is also commonly observed in HNSCC and has been associated with poor prognosis and resistance to cytotoxic agents, including ionizing radiation. It has been suggested that cortactin overexpression may increase EGFR levels in these tumors by affecting receptor downregulation, however we recently found by multivariate analysis, that cortactin expression status remained an independent prognostic factor for local recurrence, disease-free survival, and overall survival. MATERIAL AND METHODS To examine the potential link between cortactin overexpression and EGFR status, we compared cortactin and EGFR levels in a series of tumor lines derived from HNSCC. RNAi-mediated silencing was performed in cortactin overexpressing cells and in vivo tumoral potential with respect to cortactin and EGFR status was analyzed. RESULTS AND DISCUSSION Cortactin and EGFR levels were not strictly coupled in these lines and cortactin depletion did not decrease steady state receptor levels, although it did affect the epithelial to mesenchymal phenotypic conversion of cells. These results, together with clinical findings point to the existence of an EGFR-independent role of cortactin in HNSCC that may have important implications regarding the design of targeted therapies to combat tumor spread.
Collapse
|
42
|
Monferran S, Skuli N, Delmas C, Favre G, Bonnet J, Cohen-Jonathan-Moyal E, Toulas C. Alphavbeta3 and alphavbeta5 integrins control glioma cell response to ionising radiation through ILK and RhoB. Int J Cancer 2008; 123:357-364. [PMID: 18464290 DOI: 10.1002/ijc.23498] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Integrins are extracellular matrix receptors involved in tumour invasion and angiogenesis. Although there is evidence that inhibiting integrins might enhance the efficiency of radiotherapy, little is known about the exact mechanisms involved in the integrin-dependent modulation of tumor radiosensitivity. The purpose of this study was to investigate the role of alphavbeta3 and alphavbeta5 integrins in glioblastoma cell radioresistance and overall to decipher the downstream biological pathways. We first demonstrated that silencing alphavbeta3 and alphavbeta5 integrins with specific siRNAs significantly reduced the survival after irradiation of 2 glioblastoma cell lines: U87 and SF763. We then showed that integrin activity and integrin signalling pathways controlled the glioma cell radiosensitivity. This regulation of glioma cell response to ionising radiation was mediated through the integrin-linked kinase, ILK, and the small GTPase, RhoB, by two mechanisms. The first one, independent of ILK, consists in the regulation of the intracellular level of RhoB by alphavbeta3 or alphavbeta5 integrin. The second pathway involved in cell radiosensitivity consists in RhoB activation by ionising radiation through ILK. Furthermore, we demonstrated that the alphavbeta3/alphavbeta5 integrins/ILK/RhoB pathway controlled the glioma cells radiosensitivity by regulating radiation-induced mitotic cell death. This work identifies a new biological pathway controlling glioblastoma cells radioresistance, activated from the membrane through alphavbeta3 and/or alphavbeta5 integrins via ILK and RhoB. Our results are clues that downstream effectors of alphavbeta3 and alphavbeta5 integrins as ILK and RhoB might also be promising candidate targets for improving the efficiency of radiotherapy and thus the clinical outcome of patients with glioblastoma.
Collapse
Affiliation(s)
- Sylvie Monferran
- Institut Claudius Regaud, INSERMU563, Department of Oncogenesis, Signalling and Therapeutic Innovation, France
| | - Nicolas Skuli
- Institut Claudius Regaud, INSERMU563, Department of Oncogenesis, Signalling and Therapeutic Innovation, France
| | - Caroline Delmas
- Institut Claudius Regaud, INSERMU563, Department of Oncogenesis, Signalling and Therapeutic Innovation, France
| | - Gilles Favre
- Institut Claudius Regaud, INSERMU563, Department of Oncogenesis, Signalling and Therapeutic Innovation, France
| | - Jacques Bonnet
- Department of Radiations, 20-24 rue du Pont St Pierre, 31052 Toulouse, France
| | - Elizabeth Cohen-Jonathan-Moyal
- Institut Claudius Regaud, INSERMU563, Department of Oncogenesis, Signalling and Therapeutic Innovation, France.,Department of Radiations, 20-24 rue du Pont St Pierre, 31052 Toulouse, France
| | - Christine Toulas
- Institut Claudius Regaud, INSERMU563, Department of Oncogenesis, Signalling and Therapeutic Innovation, France
| |
Collapse
|
43
|
Ho B, Hou G, Pickering JG, Hannigan G, Langille BL, Bendeck MP. Integrin-linked kinase in the vascular smooth muscle cell response to injury. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:278-88. [PMID: 18535176 DOI: 10.2353/ajpath.2008.071046] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Integrin-mediated interactions between smooth muscle cells (SMCs) and the extracellular matrix regulate cell migration and proliferation during neointimal hyperplasia. Integrin-linked kinase (ILK) is a serine-threonine kinase and scaffolding molecule that acts downstream of integrin receptors to modulate cell adhesion; therefore, we examined ILK function in SMCs during wound repair. Silencing of ILK expression with siRNA in vitro decreased cell adhesion to fibronectin and accelerated both cell proliferation and wound closure in the cell monolayer; it also resulted in the rearrangement of focal adhesions and diminished central actin stress fibers. Akt and GSK3beta are ILK substrates that are important in cell motility; however, ILK siRNA silencing did not attenuate injury-induced increases in Akt and GSK3beta phosphorylation. Following balloon catheter injury of the rat carotid artery in vivo, a dramatic decrease in ILK levels coincided with both the proliferation and migration of SMCs, which leads to the formation of a thickened neointima. Immunostaining revealed decreased ILK levels in the media and deep layers of the neointima, but increased ILK levels in the subluminal layers of the intima. Taken together, these results suggest that ILK functions to maintain SMC quiescence in the normal artery. A decrease in ILK levels after injury may permit SMC migration, proliferation, and neointimal thickening, and its re-expression at the luminal surface may attenuate this process during later stages of the injury response.
Collapse
Affiliation(s)
- Bernard Ho
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
44
|
Dwivedi A, Sala-Newby GB, George SJ. Regulation of cell-matrix contacts and beta-catenin signaling in VSMC by integrin-linked kinase: implications for intimal thickening. Basic Res Cardiol 2008; 103:244-56. [PMID: 18080083 PMCID: PMC2853711 DOI: 10.1007/s00395-007-0693-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Accepted: 11/09/2007] [Indexed: 10/22/2022]
Abstract
Vascular smooth muscle cell (VSMC) proliferation and migration is responsible for intimal thickening that occurs in restenosis and atherosclerosis. Integrin-linked kinase (ILK) is a serine/threonine protein kinase implicated in signaling pathways involved in cell proliferation and migration. We studied the involvement of ILK in intimal thickening. ILK expression was significantly increased in two models of intimal thickening: balloon-injured rat carotid arteries and human saphenous vein organ cultures. Over-expression of a dominant negative ILK (DN-ILK) significantly reduced intimal thickening by approximately 50% in human saphenous vein organ cultures, demonstrating an important role in intimal thickening. ILK protein and activity was reduced on laminin and up-regulated on fibronectin, indicating ILK protein expression is modulated by extracellular matrix composition. Inhibition of ILK by siRNA knockdown and DN-ILK significantly decreased VSMC proliferation and migration while wild type ILK significantly increased proliferation and migration on laminin, confirming an essential role of ILK in both processes. Localization of paxillin and vinculin and protein levels of FAK and phospho-FAK indicated that inhibition of ILK reduced focal adhesion formation. Additionally, inhibition of ILK significantly attenuated the presence of the cell-cell complex proteins N-cadherin and beta-catenin, and beta-catenin signaling. We therefore suggest ILK modulates VSMC proliferation and migration at least in part by acting as a molecular scaffold in focal adhesions as well as modulating the stability of cell-cell contact proteins and beta-catenin signaling. In summary, ILK plays an important role in intimal thickening by modulating VSMC proliferation and migration via regulation of cell-matrix and cell-cell contacts and beta-catenin signaling.
Collapse
MESH Headings
- Animals
- Carotid Artery Injuries/enzymology
- Carotid Artery Injuries/pathology
- Cell Movement
- Cell Proliferation
- Cell Survival
- Cells, Cultured
- Disease Models, Animal
- Fibronectins/metabolism
- Focal Adhesions/enzymology
- Focal Adhesions/pathology
- Humans
- Laminin/metabolism
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Organ Culture Techniques
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- RNA Interference
- RNA, Small Interfering/metabolism
- Rats
- Saphenous Vein/enzymology
- Saphenous Vein/pathology
- Signal Transduction
- Transduction, Genetic
- Tunica Intima/enzymology
- Tunica Intima/pathology
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Amrita Dwivedi
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Bristol, BS2 8HW, UK
| | | | | |
Collapse
|
45
|
Maier S, Lutz R, Gelman L, Sarasa-Renedo A, Schenk S, Grashoff C, Chiquet M. Tenascin-C induction by cyclic strain requires integrin-linked kinase. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:1150-62. [PMID: 18269918 DOI: 10.1016/j.bbamcr.2008.01.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Revised: 01/11/2008] [Accepted: 01/14/2008] [Indexed: 12/15/2022]
Abstract
Induction of tenascin-C mRNA by cyclic strain in fibroblasts depends on RhoA and Rho dependent kinase (ROCK). Here we show that integrin-linked kinase (ILK) is required upstream of this pathway. In ILK-deficient fibroblasts, RhoA was not activated and tenascin-C mRNA remained low after cyclic strain; tenascin-C expression was unaffected by ROCK inhibition. In ILK wild-type but not ILK-/- fibroblasts, cyclic strain-induced reorganization of actin stress fibers and focal adhesions, as well as nuclear translocation of MAL, a transcriptional co-activator that links actin assembly to gene expression. These findings support a role for RhoA in ILK-mediated mechanotransduction. Rescue of ILK -/- fibroblasts by expression of wild-type ILK restored these responses to cyclic strain. Mechanosensation is not entirely abolished in ILK -/- fibroblasts, since cyclic strain activated Erk-1/2 and PKB/Akt, and induced c-fos mRNA in these cells. Conversely, lysophosphatidic acid stimulated RhoA and induced both c-fos and tenascin-C mRNA in ILK -/- cells. Thus, the signaling pathways controlling tenascin-C expression are functional in the absence of ILK, but are not triggered by cyclic strain. Our results indicate that ILK is selectively required for the induction of specific genes by mechanical stimulation via RhoA-mediated pathways.
Collapse
Affiliation(s)
- Silke Maier
- Friedrich Miescher Institute for Biomedical Research, Novartis Research Foundation, CH-4058 Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
46
|
Rosso L, Pierson PM, Golfier C, Peteri-Brunbäck B, Deroanne C, Van Obberghen-Schilling E, Mienville JM. Pituicyte stellation is prevented by RhoA-or Cdc42-dependent actin polymerization. Cell Mol Neurobiol 2007; 27:791-804. [PMID: 17712627 PMCID: PMC11517133 DOI: 10.1007/s10571-007-9176-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Accepted: 07/28/2007] [Indexed: 11/30/2022]
Abstract
Our aim was to shed light on different steps leading from metabotropic receptor activation to changes in cell shape, such as those that characterize the morphological plasticity of neurohypophysial astrocytes (pituicytes). Using explant cultures of adult rat pituicytes, we have previously established that adenosine A1 receptor activation induces stellation via inhibition of RhoA monomeric GTPase and subsequent disruption of actin stress fibers. Here, we rule out RhoA phosphorylation as a mechanism for that inhibition. Rather, our results are more consistent with involvement of a GTPase-activating protein (GAP). siRNA and pull-down experiments suggest that a step downstream of RhoA might involve Cdc42, another GTPase of the Rho family. However, RhoA activation, e.g., in the presence of serum, induces stress fibers, whereas direct Cdc42 activation appears to confine actin within a submembrane - i.e., cortical - network, which also prevents stellation. Therefore, we propose that RhoA may activate Cdc42 in parallel with an effector, such as p160Rho-kinase, that induces and maintains actin stress fibers in a dominant fashion. Rac1 is not involved in the stellation process per se but appears to induce a dendritogenic effect. Ultimately, it may be stated that pituicyte stellation is inducible upon mere actin depolymerization, and preventable upon actin organization, be it in the form of stress fibers or in a cortical configuration.
Collapse
Affiliation(s)
- Lia Rosso
- CNRS UMR 6548, Laboratoire de Physiologie Cellulaire et Moléculaire Faculté des Sciences, Université de Nice-Sophia Antipolis, 28 Avenue Valrose, Nice Cedex 2, 06108 France
- Present Address: Centre Intégratif de Génomique, Université de Lausanne, Lausanne, 1015 Switzerland
| | - Patricia M. Pierson
- CNRS UMR 6548, Laboratoire de Physiologie Cellulaire et Moléculaire Faculté des Sciences, Université de Nice-Sophia Antipolis, 28 Avenue Valrose, Nice Cedex 2, 06108 France
| | - Claire Golfier
- CNRS UMR 6548, Laboratoire de Physiologie Cellulaire et Moléculaire Faculté des Sciences, Université de Nice-Sophia Antipolis, 28 Avenue Valrose, Nice Cedex 2, 06108 France
| | - Brigitta Peteri-Brunbäck
- CNRS UMR 6548, Laboratoire de Physiologie Cellulaire et Moléculaire Faculté des Sciences, Université de Nice-Sophia Antipolis, 28 Avenue Valrose, Nice Cedex 2, 06108 France
| | - Christophe Deroanne
- CNRS UMR 6543, Centre Antoine Lacassagne, Université de Nice-Sophia Antipolis, Nice, France
- Present Address: Laboratoire de Biologie des Tissus Conjonctifs, Tour de Pathologie B23/3, Sart-Tilman, Liège 1, 4000 Belgium
| | | | - Jean-Marc Mienville
- CNRS UMR 6548, Laboratoire de Physiologie Cellulaire et Moléculaire Faculté des Sciences, Université de Nice-Sophia Antipolis, 28 Avenue Valrose, Nice Cedex 2, 06108 France
| |
Collapse
|
47
|
Mo JS, Kim MY, Han SO, Kim IS, Ann EJ, Lee KS, Seo MS, Kim JY, Lee SC, Park JW, Choi EJ, Seong JY, Joe CO, Faessler R, Park HS. Integrin-linked kinase controls Notch1 signaling by down-regulation of protein stability through Fbw7 ubiquitin ligase. Mol Cell Biol 2007; 27:5565-74. [PMID: 17526737 PMCID: PMC1952089 DOI: 10.1128/mcb.02372-06] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Integrin-linked kinase (ILK) is a scaffold and protein kinase that acts as a pivotal effector in integrin signaling for various cellular functions. In this study, we found that ILK remarkably reduced the protein stability of Notch1 through Fbw7. The kinase activity of ILK was essential for the inhibition of Notch1 signaling. Notably, the protein level and transcriptional activity of the endogenous Notch1 intracellular domain (Notch1-IC) were higher in ILK-null cells than in ILK wild-type cells, and the level of endogenous Notch1-IC was increased by the blocking of the proteasome, suggesting that ILK enhances the proteasomal degradation of Notch1-IC. ILK directly bound and phosphorylated Notch1-IC, thereby facilitating proteasomal protein degradation through Fbw7. Furthermore, we found down-regulation of Notch1-IC and up-regulation of ILK in basal cell carcinoma and melanoma patients but not in squamous cell carcinoma patients. These results suggest that ILK down-regulated the protein stability of Notch1-IC through the ubiquitin-proteasome pathway by means of Fbw7.
Collapse
Affiliation(s)
- Jung-Soon Mo
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Yongbong-dong, Buk-ku, Gwangju 500-757, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Joshi MB, Ivanov D, Philippova M, Erne P, Resink TJ. Integrin-linked kinase is an essential mediator for T-cadherin-dependent signaling via Akt and GSK3beta in endothelial cells. FASEB J 2007; 21:3083-95. [PMID: 17485554 DOI: 10.1096/fj.06-7723com] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Glycosylphosphatidylinositol-anchored T-cadherin (T-cad) influences several parameters of angiogenesis including endothelial cell (EC) differentiation, migration, proliferation, and survival. This presupposes signal transduction networking via mediatory regulators and molecular adaptors since T-cad lacks transmembrane and cytosolic domains. Here, using pharmacological inhibition of PI3K, adenoviral-mediated T-cad-overexpression, siRNA-mediated T-cad-depletion, and agonistic antibody-mediated ligation, we demonstrate signaling by T-cad through PI3K-Akt-GSK3beta pathways in EC. T-cad-overexpressing EC exhibited increased levels and nuclear accumulation of active beta-catenin, which was transcriptionally active as shown by increased Lef/Tcf reporter activity and cyclin D1 levels. Cotransduction of EC with constitutively active GSK3beta (S9A-GSK3beta) abrogated the stimulatory effects of T-cad on active beta-catenin accumulation, proliferation, and survival. Integrin-linked kinase (ILK), a membrane proximal upstream regulator of Akt and GSK3beta, was considered a candidate signaling mediator for T-cad. T-cad was present in anti-ILK immunoprecipitates, and confocal microscopy revealed colocalization of T-cad and ILK within lamellipodia of migrating cells. ILK-siRNA abolished T-cad-dependent effects on (Ser-473)Akt/(Ser-9)GSK3beta phosphorylation, active beta-catenin accumulation, and survival. We conclude ILK is an essential mediator for T-cad signaling via Akt and GSK3beta in EC. This is the first demonstration that ILK can regulate inward signaling by GPI-anchored proteins. Furthermore, ILK-GSK3beta-dependent modulation of active beta-catenin levels by GPI-anchored T-cad represents a novel mechanism for controlling cellular beta-catenin activity.
Collapse
Affiliation(s)
- Manjunath B Joshi
- Department of Research, Cardiovascular Laboratories, Basel University Hospital, Hebelstrasse 20, CH 4031 Basel, Switzerland
| | | | | | | | | |
Collapse
|
49
|
Li JJ, Han M, Wen JK, Li AY. Osteopontin stimulates vascular smooth muscle cell migration by inducing FAK phosphorylation and ILK dephosphorylation. Biochem Biophys Res Commun 2007; 356:13-9. [PMID: 17336930 DOI: 10.1016/j.bbrc.2007.02.092] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2007] [Accepted: 02/12/2007] [Indexed: 10/23/2022]
Abstract
Focal adhesion kinase (FAK) and integrin-linked kinase (ILK) are both involved in integrin-mediated cell migration. However, the molecular mechanism, and the relationship between FAK and ILK activity in signaling transduction for the osteopontin (OPN)-induced migration of vascular smooth muscle cells (VSMCs) remain unclear. Here, we show that treating VSMCs with OPN could result in the dissociation of FAK with ILK by inducing phosphorylation of the former and dephosphorylation of the latter. Furthermore, we demonstrate that FAK phosphorylation induced by OPN is coupled with ILK dephosphorylation. We also provide evidence that ILK acts downstream of FAK in the signaling pathways that mediate OPN-induced VSMC migration. These findings suggest that FAK phosphorylation and ILK dephosphorylation play important roles in VSMC migration induced by OPN.
Collapse
Affiliation(s)
- Jing-Jing Li
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang 050017, China
| | | | | | | |
Collapse
|
50
|
Yoneda A, Ushakov D, Multhaupt HA, Couchman JR. Fibronectin matrix assembly requires distinct contributions from Rho kinases I and -II. Mol Biol Cell 2006; 18:66-75. [PMID: 17065553 PMCID: PMC1751322 DOI: 10.1091/mbc.e06-08-0684] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Extracellular matrix is integral to tissue architecture and regulates many aspects of cell behavior. Fibronectin matrix assembly involves the actin cytoskeleton and the small GTPase RhoA, but downstream signaling is not understood. Here, down-regulation of either rho kinase isoform (ROCK I or -II) by small interfering RNA treatment blocked fibronectin matrix assembly, although the phenotypes were distinct and despite persistence of the alternate kinase. Remnant fibronectin on ROCK-deficient fibroblasts was mostly punctate and more deoxycholate soluble compared with controls. Fibronectin matrix assembly defects in ROCK-deficient cells did not result from decreased synthesis/secretion, altered fibronectin mRNA splicing, metalloproteinase activity, or alpha5beta1 integrin dysfunction. Rescue could be effected by ROCK protein restoration or phosphomimetic myosin light chain expression. However, the effect of ROCK I deficiency on fibronectin matrix assembly was secondary to altered cell surface morphology, rich in filopodia, resulting from high GTP-Cdc42 levels. Total internal reflection microscopy revealed that a submembranous pool of myosin light chain in control cells was missing in ROCK II-deficient cells and replaced by stress fibers. Together, two rho kinases contribute to fibronectin matrix assembly in a different manner and cortical myosin II-driven contractility, but not stress fibers, may be critical in this activity.
Collapse
Affiliation(s)
- Atsuko Yoneda
- Division of Biomedical Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Dmitriy Ushakov
- Division of Biomedical Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Hinke A.B. Multhaupt
- Division of Biomedical Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - John R. Couchman
- Division of Biomedical Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|