4.4 Article

The SPARC-related factor SMOC-2 promotes growth factor-induced cyclin D1 expression and DNA synthesis via integrin-linked kinase

Journal

MOLECULAR BIOLOGY OF THE CELL
Volume 19, Issue 1, Pages 248-261

Publisher

AMER SOC CELL BIOLOGY
DOI: 10.1091/mbc.E07-05-0510

Keywords

-

Categories

Funding

  1. National Institute of Environmental Health Sciences [ES09558]
  2. Philip Morris USA
  3. Philip Morris International
  4. National Institutes of Health/National Heart, Lung, and Blood Institute [(P01) HL47049]
  5. NATIONAL HEART, LUNG, AND BLOOD INSTITUTE [P01HL047049] Funding Source: NIH RePORTER
  6. NATIONAL INSTITUTE OF ENVIRONMENTAL HEALTH SCIENCES [R29ES009558, R01ES009558] Funding Source: NIH RePORTER

Ask authors/readers for more resources

Secreted modular calcium-binding protein-2 (SMOC-2) is a recently-identified SPARC-related protein of unknown function. In mRNA profiling experiments we, found that SMOC-2 expression was elevated in quiescent (G0) mouse fibroblasts and repressed after mitogenic stimulation with serum. The G0-specific expression of SMOC-2 was similar to that of platelet-derived growth factor-beta receptor (PDGF beta R), a major mitogenic receptor. Therefore, we tested a possible role for SMOC-2 in growth factor-induced cell cycle progression. SMOC-2 overexpression augmented DNA synthesis induced by serum and fibroblast mitogens (including PDGF-BB and basic fibroblast growth factor). Conversely, SMOC-2 ablation by using small interfering RNA attenuated DNA synthesis in response to PDGF-BB and other growth factors. Mitogen-induced expression of cyclin D1 was attenuated in SMOC-2-ablated cells, and cyclin D1-overexpressing cells were resistant to inhibition of mitogenesis after SMOC-2 ablation. Therefore, cyclin D1 is limiting for G1 progression in SMOC-2-deficient cells. SMOC-2 ablation did not inhibit PDGF-induced PDGF beta R autophosphorylation or PDGF-BB dependent activation of mitogen-activated protein kinase and Akt kinases, suggesting that SMOC-2 is dispensable for growth factor receptor activation. However, integrin-linked kinase (ILK) activity was reduced in SMOC-2-ablated cells. Ectopic expression of hyperactive ILK corrected the defective mitogenic response of SMOC-2-deficient cells. Therefore, SMOC-2 contributes to cell cycle progression by maintaining ILK activity during G1. These results identify a novel role for SMOC-2 in cell cycle control.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.4
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available