2021
DOI: 10.1155/2021/8888416
|View full text |Cite
|
Sign up to set email alerts
|

Chaetocin Promotes Osteogenic Differentiation via Modulating Wnt/Beta-Catenin Signaling in Mesenchymal Stem Cells

Abstract: Mesenchymal stemXin cells (MSCs) are a great cell source for bone regeneration. Although combining MSCs with growth factors and scaffolds provides a useful clinical strategy for bone tissue engineering, the efficiency of MSC osteogenic differentiation remains to be improved. Epigenetic modification is related to the differentiation ability of MSCs during osteogenic induction. In this study, we evaluate the effect of Chaetocin, an inhibitor of lysine-specific histone methyltransferases, on the differentiation o… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

0
15
0

Year Published

2021
2021
2023
2023

Publication Types

Select...
8

Relationship

0
8

Authors

Journals

citations
Cited by 14 publications
(15 citation statements)
references
References 20 publications
0
15
0
Order By: Relevance
“…Suppression of EZH2 prevents BMSCs from differentiating into adipocytes rather than osteoblasts, resulting in increased bone formation during OP [ 42 ]. Knockdown of SUV39H1, a methyltransferase of H3K9me3, or its auxiliary factor HP1α will down-regulate H3K9me3 and lead to BMSC senescence, while overexpression of HP1α would up-regulate H3K9me3 level, thus promoting BMSC osteogenic differentiation [ 41 ]. The regulation modes of histone modification enzymes (summarized in Table 2 ) can provide viewpoints for further exploration.…”
Section: Treating Target For Histone Modificationmentioning
confidence: 99%
See 1 more Smart Citation
“…Suppression of EZH2 prevents BMSCs from differentiating into adipocytes rather than osteoblasts, resulting in increased bone formation during OP [ 42 ]. Knockdown of SUV39H1, a methyltransferase of H3K9me3, or its auxiliary factor HP1α will down-regulate H3K9me3 and lead to BMSC senescence, while overexpression of HP1α would up-regulate H3K9me3 level, thus promoting BMSC osteogenic differentiation [ 41 ]. The regulation modes of histone modification enzymes (summarized in Table 2 ) can provide viewpoints for further exploration.…”
Section: Treating Target For Histone Modificationmentioning
confidence: 99%
“…Both G9a and SUV39h1/2 could catalyze the methylation of H3K9, but the same inhibitory histone modification H3K9me3 produces different results. G9a inhibitors (e.g., A366, BIX01294) promote adipogenesis [ 37 ], but SUV39h1/2 inhibitors (e.g., chaetocin) promote osteogenesis [ 41 ]. In conclusion, different inhibitors exert different effects, which is critical and calls for researchers’ attention in selecting inhibitors.…”
Section: Treating Target For Histone Modificationmentioning
confidence: 99%
“…Recent studies have shown that Wnt/β-catenin signaling plays an important role in the osteogenic differentiation of MSCs ( Li et al, 2019 ; Liang et al, 2021 ). To explore whether Gli1+ progenitors are regulated by Wnt/β-catenin signaling, we harvested tdTomato + cells from Gli1-Cre ERT2 ;tdTomato condyles 1 week after induction.…”
Section: Resultsmentioning
confidence: 99%
“…When DNA methylation was modulated in vitro in osteogenic progenitors, it was demonstrated that DNA methyltransferase 1 (DNMT1) inhibition in periodontal ligament cells restored RUNX2 expression and their osteogenic capacity, and inhibition of methylation in MSC supported osteogenic differentiation at the cost of adipogenic capacity. For KDM7A, a role in a reciprocal mechanism for the switch between osteogenic and adipogenic differentiation was also reported [44,57,58]. On the other hand, osteogenic receptors like PTH1R or the vitamin D receptor VDR may, vice versa, modulate the expression of epigenetically relevant components [59,60].…”
Section: Alu Elements Highmentioning
confidence: 86%