3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Inhibiting FAK–Paxillin Interaction Reduces Migration and Invadopodia-Mediated Matrix Degradation in Metastatic Melanoma Cells

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Simple Summary

          The focal adhesion kinase (FAK) is over-expressed in a variety of human tumors and is involved in many aspects of the metastatic process. This has led to the development of small inhibitors of FAK kinase function which are currently evaluated in clinical trials. We demonstrate here that this class of inhibitors, while decreasing melanoma cell migration, increases invadopodia activity in metastatic melanoma cells. Searching for an alternative strategy to inhibit the oncogenic activity of FAK, we show that inhibiting FAK scaffolding function using a small peptide altering FAK–paxillin interactions reduces both migration and invadopodia-mediated matrix degradation in metastatic melanoma cells.

          Abstract

          The nonreceptor tyrosine kinase FAK is a promising target for solid tumor treatment because it promotes invasion, tumor progression, and drug resistance when overexpressed. Investigating the role of FAK in human melanoma cells, we found that both in situ and metastatic melanoma cells strongly express FAK, where it controls tumor cells’ invasiveness by regulating focal adhesion-mediated cell motility. Inhibiting FAK in human metastatic melanoma cells with either siRNA or a small inhibitor targeting the kinase domain impaired migration but led to increased invadopodia formation and extracellular matrix degradation. Using FAK mutated at Y397, we found that this unexpected increase in invadopodia activity is due to the lack of phosphorylation at this residue. To preserve FAK–Src interaction while inhibiting pro-migratory functions of FAK, we found that altering FAK–paxillin interaction, with either FAK mutation in the focal adhesion targeting (FAT) domain or a competitive inhibitor peptide mimicking paxillin LD domains drastically reduces cell migration and matrix degradation by preserving FAK activity in the cytoplasm. In conclusion, our data show that targeting FAK–paxillin interactions could be a potential therapeutic strategy to prevent metastasis formation, and molecules targeting this interface could be alternative to inhibitors of FAK kinase activity which display unexpected effects.

          Related collections

          Most cited references60

          • Record: found
          • Abstract: found
          • Article: not found

          The 'ins' and 'outs' of podosomes and invadopodia: characteristics, formation and function.

          Podosomes and invadopodia are actin-based dynamic protrusions of the plasma membrane of metazoan cells that represent sites of attachment to - and degradation of - the extracellular matrix. The key proteins in these structures include the actin regulators cortactin and neural Wiskott-Aldrich syndrome protein (N-WASP), the adaptor proteins Tyr kinase substrate with four SH3 domains (TKS4) and Tyr kinase substrate with five SH3 domains (TKS5), and the metalloprotease membrane type 1 matrix metalloprotease (MT1MMP; also known as MMP14). Many cell types can produce these structures, including invasive cancer cells, vascular smooth muscle and endothelial cells, and immune cells such as macrophages and dendritic cells. Recently, progress has been made in our understanding of the regulatory and functional aspects of podosome and invadopodium biology and their role in human disease.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            FAK-Src signalling through paxillin, ERK and MLCK regulates adhesion disassembly.

            Cell migration is a complex, highly regulated process that involves the continuous formation and disassembly of adhesions (adhesion turnover). Adhesion formation takes place at the leading edge of protrusions, whereas disassembly occurs both at the cell rear and at the base of protrusions. Despite the importance of these processes in migration, the mechanisms that regulate adhesion formation and disassembly remain largely unknown. Here we develop quantitative assays to measure the rate of incorporation of molecules into adhesions and the departure of these proteins from adhesions. Using these assays, we show that kinases and adaptor molecules, including focal adhesion kinase (FAK), Src, p130CAS, paxillin, extracellular signal-regulated kinase (ERK) and myosin light-chain kinase (MLCK) are critical for adhesion turnover at the cell front, a process central to migration.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              FAK in cancer: mechanistic findings and clinical applications.

              Focal adhesion kinase (FAK) is a cytoplasmic protein tyrosine kinase that is overexpressed and activated in several advanced-stage solid cancers. FAK promotes tumour progression and metastasis through effects on cancer cells, as well as stromal cells of the tumour microenvironment. The kinase-dependent and kinase-independent functions of FAK control cell movement, invasion, survival, gene expression and cancer stem cell self-renewal. Small molecule FAK inhibitors decrease tumour growth and metastasis in several preclinical models and have initial clinical activity in patients with limited adverse events. In this Review, we discuss FAK signalling effects on both tumour and stromal cell biology that provide rationale and support for future therapeutic opportunities.
                Bookmark

                Author and article information

                Contributors
                Role: Academic Editor
                Role: Academic Editor
                Journal
                Cancers (Basel)
                Cancers (Basel)
                cancers
                Cancers
                MDPI
                2072-6694
                14 April 2021
                April 2021
                : 13
                : 8
                : 1871
                Affiliations
                [1 ]Université de Strasbourg, CNRS UMR7021, Laboratoire de Bioimagerie et Pathologies, Migration, Invasion et Microenvironnement, Faculté de Pharmacie, 67401 Illkirch, France; antoine.mousson@ 123456unistra.fr (A.M.); marlene.legrand@ 123456unistra.fr (M.L.); tania.steffan@ 123456unistra.fr (T.S.); philippe.carl@ 123456unistra.fr (P.C.); jean-pierre.gies@ 123456unistra.fr (J.-P.G.); maxime.lehmann@ 123456unistra.fr (M.L.); jan.de-mey@ 123456unistra.fr (J.D.M.); denis.dujardin@ 123456unistra.fr (D.D.); emilie.sick@ 123456unistra.fr (E.S.)
                [2 ]Université de Strasbourg, CNRS UMR7021, Laboratoire de Bioimagerie et Pathologies, Plateforme PIQ, Faculté de Pharmacie, 67401 Illkirch, France; romain.vauchelles@ 123456unistra.fr
                [3 ]Université de Strasbourg, CNRS UMR7242, Intervention Chémobiologique, ESBS, 67412 Illkirch, France; guy.zuber@ 123456unistra.fr
                Author notes
                [* ]Correspondence: philippe.ronde@ 123456unistra.fr ; Tel.: +33-3-6885-4184
                [†]

                These authors contributed equally to this work.

                Author information
                https://orcid.org/0000-0003-2447-7738
                https://orcid.org/0000-0001-6372-5293
                Article
                cancers-13-01871
                10.3390/cancers13081871
                8070677
                33919725
                ba007108-1548-4ab3-bae9-604ffc44ab7b
                © 2021 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( https://creativecommons.org/licenses/by/4.0/).

                History
                : 18 February 2021
                : 08 April 2021
                Categories
                Article

                fak,tyrosine kinase inhibitor,paxillin,invadopodia,migration,melanoma

                Comments

                Comment on this article