PUBLICATION

Fermented Oyster Extract Promotes Osteoblast Differentiation by Activating the Wnt/β-Catenin Signaling Pathway, Leading to Bone Formation

Authors
Molagoda, I.M.N., Karunarathne, W.A.H.M., Choi, Y.H., Park, E.K., Jeon, Y.J., Lee, B.J., Kang, C.H., Kim, G.Y.
ID
ZDB-PUB-191110-2
Date
2019
Source
Biomolecules   9(11): (Journal)
Registered Authors
Keywords
Crassostrea gigas, Wnt/β-catenin, bone formation, mineralization, oyster
MeSH Terms
  • Animals
  • Cell Differentiation/drug effects
  • Cell Proliferation/drug effects
  • Cell Survival/drug effects
  • Core Binding Factor Alpha 1 Subunit/genetics
  • Female
  • Fermentation
  • Gene Expression Regulation, Developmental/drug effects
  • Humans
  • Larva/drug effects
  • Mice
  • Osteoblasts/drug effects
  • Osteocalcin/chemistry
  • Osteocalcin/pharmacology
  • Osteogenesis/drug effects*
  • Osteosarcoma/genetics
  • Osteosarcoma/pathology
  • Ostreidae/chemistry*
  • Sp7 Transcription Factor/chemistry
  • Sp7 Transcription Factor/pharmacology
  • Tissue Extracts/chemistry
  • Tissue Extracts/pharmacology*
  • Wnt Signaling Pathway/drug effects
  • Zebrafish/genetics
  • Zebrafish Proteins/drug effects
  • Zebrafish Proteins/genetics*
PubMed
31698882 Full text @ Biomolecules
Abstract
The Pacific oyster, Crassostrea gigas, is well-known as a nutritious food. Recently, we revealed that fermented extract of C. gigas (FO) inhibited ovariectomy-induced osteoporosis, resulting from suppression of osteoclastogenesis. However, since the beneficial effect of FO on osteogenesis is poorly understood, it was examined in mouse preosteoblast MC3T3-E1 cells, human osteosarcoma MG-63 osteoblast-like cells, and zebrafish larvae in this study. We found that FO increased mitochondrial activity from days 1 to 7; however, total cell number of MC3T3-E1 cells gradually decreased without any change in cell viability, which suggests that FO stimulates the differentiation of MC3T3-E1 cells. FO also promoted the expression of osteoblast marker genes, including runt-related transcription factor 2 (mRUNX2), alkaline phosphatase (mALP), collagen type I α1 (mCol1α1), osteocalcin (mOCN), osterix (mOSX), bone morphogenetic protein 2 (mBMP2), and mBMP4 in MC3T3-E1 cells accompanied by a significant increase in ALP activity. FO also increased nuclear translocation of RUNX2 and OSX transcription factors, ALP activity, and calcification in vitro along with the upregulated expression of osteoblast-specific marker proteins such as RUNX2, ALP, Col1α1, OCN, OSX, and BMP4. Additionally, FO enhanced bone mineralization (calcein intensity) in zebrafish larvae at 9 days post-fertilization comparable to that in the β-glycerophosphate (GP)-treated group. All the tested osteoblast marker genes, including zRUNX2a, zRUNX2b, zALP, zCol1a1, zOCN, zBMP2, and zBMP4, were also remarkably upregulated in the zebrafish larvae in response to FO. It also promoted tail fin regeneration in adult zebrafish as same as the GP-treated groups. Furthermore, not only FO positively regulate β-catenin expression and Wnt/β-catenin luciferase activity, but pretreatment with a Wnt/β-catenin inhibitor (FH535) also significantly decreased FO-mediated bone mineralization in zebrafish larvae, which indicates that FO-induced osteogenesis depends on the Wnt/β-catenin pathway. Altogether, the current study suggests that the supplemental intake of FO has a beneficial effect on osteogenesis.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping