Transcriptomic analysis of licensed mesenchymal stem cells reveals a molecular signature associated with an increase of IDO and SOCS3 expression

Cytotherapy ◽  
2021 ◽  
Vol 23 (5) ◽  
pp. S106
Author(s):  
M. Kurte ◽  
J. Cuenca ◽  
E. Martinez ◽  
F. Carrion ◽  
M. Khoury ◽  
...  
2017 ◽  
Vol 106 (1) ◽  
pp. 126-137 ◽  
Author(s):  
Agnese Gugliandolo ◽  
Francesca Diomede ◽  
Paolo Cardelli ◽  
Alessia Bramanti ◽  
Domenico Scionti ◽  
...  

Oncogene ◽  
2007 ◽  
Vol 27 (18) ◽  
pp. 2542-2551 ◽  
Author(s):  
M Galiè ◽  
G Konstantinidou ◽  
D Peroni ◽  
I Scambi ◽  
C Marchini ◽  
...  

2006 ◽  
Vol 17 (8) ◽  
pp. 3543-3556 ◽  
Author(s):  
Agate Noer ◽  
Anita L. Sørensen ◽  
Andrew C. Boquest ◽  
Philippe Collas

Mesenchymal stem cells from adipose tissue can differentiate into mesodermal lineages. Differentiation potential, however, varies between clones of adipose stem cells (ASCs), raising the hypothesis that epigenetic differences account for this variability. We report here a bisulfite sequencing analysis of CpG methylation of adipogenic (leptin [LEP], peroxisome proliferator-activated receptor gamma 2 [PPARG2], fatty acid-binding protein 4 [FABP4], and lipoprotein lipase [LPL]) promoters and of nonadipogenic (myogenin [MYOG], CD31, and GAPDH) loci in freshly isolated human ASCs and in cultured ASCs, in relation to gene expression and differentiation potential. Uncultured ASCs display hypomethylated adipogenic promoters, in contrast to myogenic and endothelial loci, which are methylated. Adipogenic promoters exhibit mosaic CpG methylation, on the basis of heterogeneous methylation between cells and of variation in the extent of methylation of a given CpG between donors, and both between and within clonal cell lines. DNA methylation reflects neither transcriptional status nor potential for gene expression upon differentiation. ASC culture preserves hypomethylation of adipogenic promoters; however, between- and within-clone mosaic methylation is detected. Adipogenic differentiation also maintains the overall CpG hypomethylation of LEP, PPARG2, FABP4, and LPL despite demethylation of specific CpGs and transcriptional induction. Furthermore, enhanced methylation at adipogenic loci in primary differentiated cells unrelated to adipogenesis argues for ASC specificity of the hypomethylated state of these loci. Therefore, mosaic hypomethylation of adipogenic promoters may constitute a molecular signature of ASCs, and DNA methylation does not seem to be a determinant of differentiation potential of these cells.


2019 ◽  
Vol 10 (5) ◽  
Author(s):  
Yin Huang ◽  
Qing Li ◽  
Kunshan Zhang ◽  
Mingyuan Hu ◽  
Yu Wang ◽  
...  

Molecules ◽  
2021 ◽  
Vol 26 (3) ◽  
pp. 606
Author(s):  
Nihal AlMuraikhi ◽  
Hanouf Alaskar ◽  
Sarah Binhamdan ◽  
Amal Alotaibi ◽  
Moustapha Kassem ◽  
...  

Several signalling pathways, including the JAK/STAT signalling pathway, have been identified to regulate the differentiation of human bone marrow skeletal (mesenchymal) stem cells (hBMSCs) into bone-forming osteoblasts. Members of the JAK family mediate the intracellular signalling of various of cytokines and growth factors, leading to the regulation of cell proliferation and differentiation into bone-forming osteoblastic cells. Inhibition of JAK2 leads to decoupling of its downstream mediator, STAT3, and the subsequent inhibition of JAK/STAT signalling. However, the crucial role of JAK2 in hBMSCs biology has not been studied in detail. A JAK2 inhibitor, Fedratinib, was identified during a chemical biology screen of a small molecule library for effects on the osteoblastic differentiation of hMSC-TERT cells. Alkaline phosphatase activity and staining assays were conducted as indicators of osteoblastic differentiation, while Alizarin red staining was used as an indicator of in vitro mineralised matrix formation. Changes in gene expression were assessed using quantitative real-time polymerase chain reaction. Fedratinib exerted significant inhibitory effects on the osteoblastic differentiation of hMSC-TERT cells, as demonstrated by reduced ALP activity, in vitro mineralised matrix formation and downregulation of osteoblast-related gene expression, including ALP, ON, OC, RUNX2, OPN, and COL1A1. To identify the underlying molecular mechanisms, we examined the effects of Fedratinib on a molecular signature of several target genes known to affect hMSC-TERT differentiation into osteoblasts. Fedratinib inhibited the expression of LIF, SOCS3, RRAD, NOTCH3, TNF, COMP, THBS2, and IL6, which are associated with various signalling pathways, including TGFβ signalling, insulin signalling, focal adhesion, Notch Signalling, IL-6 signalling, endochondral ossification, TNF-α, and cytokines and inflammatory response. We identified a JAK2 inhibitor (Fedratinib) as a powerful inhibitor of the osteoblastic differentiation of hMSC-TERT cells, which may be useful as a therapeutic option for treating conditions associated with ectopic bone formation or osteosclerotic metastases.


Sign in / Sign up

Export Citation Format

Share Document