microRNA-103a functions as a mechanosensitive microRNA to inhibit bone formation through targeting Runx2

J Bone Miner Res. 2015 Feb;30(2):330-45. doi: 10.1002/jbmr.2352.

Abstract

Emerging evidence indicates that microRNAs (miRNAs) play essential roles in regulating osteoblastogenesis and bone formation. However, the role of miRNA in osteoblast mechanotransduction remains to be defined. In this study, we aimed to investigate whether miRNAs regulate mechanical stimulation-triggered osteoblast differentiation and bone formation through modulation of Runx2, the master transcription factor for osteogenesis. We first investigated the role of mechanical loading both in a mouse model and in an osteoblast culture system and the outcomes clearly demonstrated that mechanical stimuli can regulate osteogenesis and bone formation both in vivo and in vitro. Using bioinformatic analyses and subsequent confirmation by quantitative real-time PCR (qRT-PCR), we found that multiple miRNAs that potentially target Runx2 were responding to in vitro mechanical stimulation, among which miR-103a was fully characterized. miR-103a and its host gene PANK3 were both downregulated during cyclic mechanical stretch (CMS)-induced osteoblast differentiation, whereas Runx2 protein expression was upregulated. Overexpression of miR-103a significantly decreased and inhibition of miR-103a increased Runx2 protein level, suggesting that miR-103a acts as an endogenous attenuator of Runx2 in osteoblasts. Mutation of putative miR-103a binding sites in Runx2 mRNA abolishes miR-103a-mediated repression of the Runx2 3'-untranslated region (3'UTR) luciferase reporter activity, suggesting that miR-103a binds to Runx2 3'UTR. Osteoblast marker gene profiling and osteogenic phenotype assays demonstrated that miR-103a negatively correlates with CMS-induced osteogenesis. Further, the perturbation of miR-103a also has a significant effect on osteoblast activity and matrix mineralization. More importantly, we found an inhibitory role of miR-103a in regulating bone formation in hindlimb unloading mice, and pretreatment with antagomir-103a partly rescued the osteoporosis caused by mechanical unloading. Taken together, our data suggest that miR-103a is the first identified mechanosensitive miRNA that regulates osteoblast differentiation by directly targeting Runx2, and therapeutic inhibition of miR-103a may be an efficient anabolic strategy for skeletal disorders caused by pathological mechanical loading.

Keywords: CYCLIC MECHANICAL STRETCH; MECHANOTRANSDUCTION; MICRORNAS; OSTEOBLAST; RUNX2.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adult
  • Animals
  • Cell Differentiation
  • Cell Line
  • Core Binding Factor Alpha 1 Subunit / genetics
  • Core Binding Factor Alpha 1 Subunit / metabolism*
  • Disease Models, Animal
  • Extracellular Matrix / metabolism
  • Gene Expression Regulation
  • Hindlimb Suspension
  • Humans
  • Male
  • Mechanotransduction, Cellular*
  • Mice, Inbred C57BL
  • MicroRNAs / genetics
  • MicroRNAs / metabolism*
  • Osteoblasts / pathology
  • Osteogenesis*
  • Osteoporosis / pathology
  • Osteoporosis / physiopathology
  • Stress, Mechanical
  • Transcription, Genetic
  • Weight-Bearing

Substances

  • Core Binding Factor Alpha 1 Subunit
  • MIRN103 microRNA, human
  • MIRN103 microRNA, mouse
  • MicroRNAs