Signal Transduction
Protein Kinase G and Focal Adhesion Kinase Converge on Src/Akt/β-Catenin Signaling Module in Osteoblast Mechanotransduction*

https://doi.org/10.1074/jbc.M112.347245Get rights and content
Under a Creative Commons license
open access

Mechanical loading of bone induces interstitial fluid flow, leading to fluid shear stress (FSS) of osteoblasts. FSS rapidly increases the intracellular calcium concentration ([Ca2+]) and nitric oxide (NO) synthesis in osteoblasts and activates the protein kinase Akt. Activated Akt stimulates osteoblast proliferation and survival, but the mechanism(s) leading to Akt activation is not well defined. Using pharmacological and genetic approaches in primary human and mouse osteoblasts and mouse MC3T3 osteoblast-like cells, we found that Akt activation by FSS occurred through two parallel pathways; one required calcium stimulation of NO synthase and NO/cGMP/protein kinase G II-dependent activation of Src, and the other required calcium activation of FAK and Src, independent of NO. Both pathways cooperated to increase PI3K-dependent Akt phosphorylation and were necessary for FSS to induce nuclear translocation of β-catenin, c-fos, and cox-2 gene expression and osteoblast proliferation. These data explain how mechanical stimulation of osteoblasts leads to increased signaling through a growth regulatory pathway essential for maintaining skeletal integrity.

beta-Catenin
Calcium
Focal Adhesion Kinase
Mechanotransduction
Osteoblasts
Protein Kinase G (PKG)
Fluid Shear Stress
cGMP-dependent Protein Kinase

Cited by (0)

*

This work was supported, in whole or in part, by National Institutes of Health Grant R01-AR051300 (to R. B. P., H. R., and S. Z.).

This article contains supplemental Figs. 1–6.

1

Both authors contributed equally to this work.

2

Supported by a research fellowship grant from the Deutsche Forschungsgemeinschaft.