Abstract

ObjectivesAdipose-derived stem cells are frequently used for bone regeneration both in vitro and in vivo. N6-methyladenosine (m6A) is the most abundant post-transcriptional modification on eukaryotic RNAs and plays multifaceted roles in development and diseases. However, the regulatory mechanisms of m6A in osteogenic differentiation of human adipose-derived stem cells (hASCs) remain elusive. The present study aimed to build the transcriptome-wide m6A methylome during the osteogenic differentiation of hASCs.Materials and methodshASCs were harvested after being cultured in a basic or osteogenic medium for 7 days, and the osteogenic differentiation was validated by alkaline phosphatase (ALP) and Alizarin Red S staining, ALP activity assay, and qRT-PCR analysis of ALP, RUNX2, BGLAP, SPP1, SP7, and COL1A1 genes. The m6A level was colorimetrically measured, and the expression of m6A regulators was confirmed by qRT-PCR and western blot. Moreover, m6A MeRIP-seq and RNA-seq were performed to build the transcriptome and m6A methylome. Furthermore, bioinformatic analyses including volcano plots, Venn plots, clustering analysis, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, gene sets enrichment analysis, and protein-protein interaction analysis were conducted.ResultsIn total, 1145 differentially methylated peaks, 2261 differentially expressed genes, and 671 differentially methylated and expressed genes (DMEGs) were identified. GO and KEGG pathway analyses conducted for these DMEGs revealed extensive and osteogenic biological functions. The “PI3K-Akt signaling pathway”; “MAPK signaling pathway”; “parathyroid hormone synthesis, secretion, and action”; and “p53 signaling pathway” were significantly enriched, and the DMEGs in these pathways were identified as m6A-specific key genes. A protein-protein interaction network based on DMEGs was built, and VEGFA, CD44, MMP2, HGF, and SPARC were speculated as the hub DMEGs.ConclusionsThe total m6A level was reduced with osteogenic differentiation of hASCs. The transcriptome-wide m6A methylome built in the present study indicated quite a few signaling pathways, and hub genes were influenced by m6A modification. Future studies based on these epigenetic clues could promote understanding of the mechanisms of osteogenic differentiation of hASCs.

Highlights

  • Large bone defects, caused by trauma, bone loss, and tumors, lead to heavy social and economic burdens

  • A protein-protein interaction network based on differentially methylated and expressed genes (DMEGs) was built, and VEGFA, CD44, MMP2, HGF, and SPARC were speculated as the hub DMEGs

  • The transcriptome-wide m6A methylome built in the present study indicated quite a few signaling pathways, and hub genes were influenced by m6A modification

Read more

Summary

Introduction

Large bone defects, caused by trauma, bone loss, and tumors, lead to heavy social and economic burdens. The clinical gold standard for the treatment of skeletal defects is autogenous or allogeneic bone grafts, which suffers disadvantages of a limited amount of harvested bone and donor site morbidity [1, 2]. Built on stem cells or osteoprogenitor cells, osteoconductive biomaterials, and osteoconductive cytokines facilitating cellular proliferation and osteogenic differentiation, offers a promising solution [2, 3]. Human bone marrow-derived mesenchymal stem cells (hBMSCs), naturally resident in the bone marrow, are pluripotent and have been widely applied in bone tissue engineering for years [4, 5]. The access to autogenous hBMSCs is invasive and subject to limited cell incidence. The self-renewal and proliferative ability of hBMSCs weakens with donor aging and diseases such as osteoporosis and arthritis [6,7,8]

Objectives
Methods
Results
Discussion
Conclusion
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