Abstract

Endoplasmic reticulum (ER) stress transducers, such as old astrocyte specifically induced substance (OASIS) and activating transcription factor 6 (ATF6), which are induced by bone morphogenetic protein 2 (BMP2), regulate bone formation and osteoblast differentiation. Here, we examined the role of cAMP response element-binding protein H (CREBH), a member of the same family of ER membrane-bound basic leucine zipper (bZIP) transcription factors as OASIS and ATF6, in osteoblast differentiation and bone formation. Proinflammatory cytokine TNFα increased CREBH expression by up-regulating the nuclear factor-κB (NF-κB) signaling pathway in osteoblasts, increased the level of N-terminal fragment of CREBH in the nucleus, and inhibited BMP2 induction of osteoblast specific gene expression. Overexpression of CREBH suppressed BMP2-induced up-regulation of the osteogenic markers runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP), and osteocalcin (OC) in MC3T3-E1 cells and primary osteoblasts, as well as BMP2-induced ALP activity and OC protein production. In contrast, knockdown of CREBH attenuated the inhibitory effect of TNFα on BMP2-induced osteoblast differentiation. Mechanistic studies revealed that CREBH increased the expression of Smad ubiquitination regulatory factor 1 (Smurf1), leading to ubiquitin-dependent degradation of Smad1, whereas knockdown of CREBH inhibited TNFα-mediated degradation of Smad1 by Smurf1. Consistent with these in vitro findings, administration of Ad-CREBH inhibited BMP2-induced ectopic and orthotopic bone formation in vivo. Taken together, these results suggest that CREBH is a novel negative regulator of osteoblast differentiation and bone formation.

Highlights

  • Severe inflammatory reactions delay wound healing of bone

  • We examined the effects of cAMP response element-binding protein H (CREBH) on the expression levels of the Smad1 downstream factors Runx2 and activating transcription factor 6 (ATF6), which are required for osteoblast differentiation

  • Inflammation is triggered by inflammatory cytokines, such as TNF␣ and LPS, which suppress bone morphogenetic protein 2 (BMP2)-induced osteoblast differentiation in vitro and contribute to bone loss in inflammatory bone diseases, such as rheumatoid arthritis [17, 35]

Read more

Summary

Background

Severe inflammatory reactions delay wound healing of bone. Results: Tumor necrosis factor ␣ (TNF␣) inhibition of osteoblast differentiation is associated with increased cAMP response element-binding protein H (CREBH) and Smurf expression. Endoplasmic reticulum (ER) stress transducers, such as old astrocyte induced substance (OASIS) and activating transcription factor 6 (ATF6), which are induced by bone morphogenetic protein 2 (BMP2), regulate bone formation and osteoblast differentiation. BMP2 stimulates osteoblast differentiation and bone formation by activating several unfolded protein response transducers, including OASIS and ATF6, which induce osteoblast-specific gene expression [9, 10]. Activation of ERK by TNF␣ results in inhibition of the transcription factor osterix, and TNF␣-mediated induction of Smad ubiquitination regulatory factor 1 (Smurf1) and Smurf accelerates the degradation of Runx protein through the proteasomal degradation pathway [25] Despite these findings, the molecular mechanisms underlying inflammatory actions in osteoblast differentiation are not fully understood. We describe a novel signaling pathway that encourages further analyses of the relationship between ER stress and bone formation

Experimental Procedures
Results
Discussion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call