35
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Cellular characterization of a novel focal adhesion kinase inhibitor.

      The Journal of Biological Chemistry
      Animals, Apoptosis, drug effects, Breast Neoplasms, drug therapy, enzymology, Cell Line, Tumor, Cell Proliferation, Chemotaxis, Dose-Response Relationship, Drug, Female, Focal Adhesion Protein-Tyrosine Kinases, antagonists & inhibitors, Heterocyclic Compounds with 4 or More Rings, chemistry, pharmacology, therapeutic use, Humans, Male, Prostatic Neoplasms, Protein Kinase Inhibitors, Quinolones, Signal Transduction, Sulfones

      Read this article at

      ScienceOpenPublisherPubMed
      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

          Focal adhesion kinase (FAK) is a member of a family of non-receptor protein-tyrosine kinases that regulates integrin and growth factor signaling pathways involved in cell migration, proliferation, and survival. FAK expression is increased in many cancers, including breast and prostate cancer. Here we describe perturbation of adhesion-mediated signaling with a FAK inhibitor, PF-573,228. In vitro, this compound inhibited purified recombinant catalytic fragment of FAK with an IC(50) of 4 nM. In cultured cells, PF-573,228 inhibited FAK phosphorylation on Tyr(397) with an IC(50) of 30-100 nM. Treatment of cells with concentrations of PF-573,228 that significantly decreased FAK Tyr(397) phosphorylation failed to inhibit cell growth or induce apoptosis. In contrast, treatment with PF-573,228 inhibited both chemotactic and haptotactic migration concomitant with the inhibition of focal adhesion turnover. These studies show that PF-573,228 serves as a useful tool to dissect the functions of FAK in integrin-dependent signaling pathways in normal and cancer cells and forms the basis for the generation of compounds amenable for preclinical and patient trials.

          Related collections

          Author and article information

          Comments

          Comment on this article