STK35L1 associates with nuclear actin and regulates cell cycle and migration of endothelial cells

PLoS One. 2011 Jan 20;6(1):e16249. doi: 10.1371/journal.pone.0016249.

Abstract

Background: Migration and proliferation of vascular endothelial cells are essential for repair of injured endothelium and angiogenesis. Cyclins, cyclin-dependent kinases (CDKs), and cyclin-dependent kinase inhibitors play an important role in vascular tissue injury and wound healing. Previous studies suggest a link between the cell cycle and cell migration: cells present in the G(1) phase have the highest potential to migrate. The molecular mechanism linking these two processes is not understood.

Methodology/principal findings: In this study, we explored the function of STK35L1, a novel Ser/Thr kinase, localized in the nucleus and nucleolus of endothelial cells. Molecular biological analysis identified a bipartite nuclear localization signal, and nucleolar localization sequences in the N-terminal part of STK35L1. Nuclear actin was identified as a novel binding partner of STK35L1. A class III PDZ binding domains motif was identified in STK35L1 that mediated its interaction with actin. Depletion of STK35L1 by siRNA lead to an accelerated G(1) to S phase transition after serum-stimulation of endothelial cells indicating an inhibitory role of the kinase in G(1) to S phase progression. Cell cycle specific genes array analysis revealed that one gene was prominently downregulated (8.8 fold) in STK35L1 silenced cells: CDKN2A alpha transcript, which codes for p16(INK4a) leading to G(1) arrest by inhibition of CDK4/6. Moreover in endothelial cells seeded on Matrigel, STK35L1 expression was rapidly upregulated, and silencing of STK35L1 drastically inhibited endothelial sprouting that is required for angiogenesis. Furthermore, STK35L1 depletion profoundly impaired endothelial cell migration in two wound healing assays.

Conclusion/significance: The results indicate that by regulating CDKN2A and inhibiting G1- to S-phase transition STK35L1 may act as a central kinase linking the cell cycle and migration of endothelial cells. The interaction of STK35L1 with nuclear actin might be critical in the regulation of these fundamental endothelial functions.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Actins / metabolism*
  • Cell Cycle*
  • Cell Line
  • Cell Movement*
  • Cell Nucleus / metabolism*
  • Cyclin-Dependent Kinase Inhibitor p16 / genetics
  • Endothelial Cells / cytology*
  • G1 Phase
  • Gene Expression Profiling
  • Humans
  • Nuclear Proteins / metabolism
  • Nuclear Proteins / physiology*
  • Protein Kinases / metabolism
  • Protein Kinases / physiology*
  • Protein Serine-Threonine Kinases

Substances

  • Actins
  • Cyclin-Dependent Kinase Inhibitor p16
  • Nuclear Proteins
  • Protein Kinases
  • Protein Serine-Threonine Kinases
  • STK35 protein, human