Postnatal skeletal deletion of Dickkopf‐1 increases bone formation and bone volume in male and female mice, despite increased sclerostin expression

J Colditz, S Thiele, U Baschant, C Niehrs… - Journal of Bone and …, 2018 - academic.oup.com
J Colditz, S Thiele, U Baschant, C Niehrs, LF Bonewald, LC Hofbauer, M Rauner
Journal of Bone and Mineral Research, 2018academic.oup.com
ABSTRACT The Wnt antagonist Dickkopf‐1 (Dkk1) is a negative regulator of osteoblast
function and bone mass. However, because of the lack of appropriate models, many aspects
of its role in the regulation of postnatal bone turnover and its cellular source have remained
unknown. In this study, we deleted Dkk1 postnatally and in different cell types using various
Cre‐drivers (Rosa26‐ERT2‐Cre, Osx‐cre, Dmp1‐Cre) and assessed to which extent cells of
the osteoblastic lineage contribute to the effects of Dkk1 on bone turnover and homeostasis …
Abstract
The Wnt antagonist Dickkopf‐1 (Dkk1) is a negative regulator of osteoblast function and bone mass. However, because of the lack of appropriate models, many aspects of its role in the regulation of postnatal bone turnover and its cellular source have remained unknown. In this study, we deleted Dkk1 postnatally and in different cell types using various Cre‐drivers (Rosa26‐ERT2‐Cre, Osx‐cre, Dmp1‐Cre) and assessed to which extent cells of the osteoblastic lineage contribute to the effects of Dkk1 on bone turnover and homeostasis. Female and male mice were examined at 12 weeks of age. Mice with a global or cell type–specific deletion of Dkk1 showed a two‐ to threefold higher bone volume compared with their Cre‐negative littermates. The mineral apposition rate and the bone formation rate were increased two‐ to fourfold in all three mouse lines, despite a significant increase in systemic and skeletal levels of sclerostin. Dkk1 deletion further reduced the number of osteoclasts about twofold, which was accompanied by a strong decrease in the receptor activator of nuclear factor‐κB ligand/osteoprotegerin mRNA ratio in femoral bone. Despite similar increases in bone mass, the deletion of Dkk1 in osterix‐expressing cells reduced circulating Dkk1 significantly (males, –79%; females, –77%), whereas they were not changed in Dkk1fl/fl;Dmp1‐Cre mice. However, both lines showed significantly reduced Dkk1 mRNA levels in bone. In summary, we show that lack of Dkk1 in cells of the osteoblastic lineage leads to high bone mass with increased bone formation, despite increased levels of sclerostin. Moreover, the majority of systemic Dkk1 appears to originate from osteoprogenitors but not from mature osteoblasts or osteocytes. Nevertheless, the amount of Dkk1 produced locally by more mature osteogenic cells is sufficient to modulate bone mass. Thus, this study highlights the importance of local Wnt signaling on postnatal bone homeostasis. © 2018 American Society for Bone and Mineral Research.
Oxford University Press