scholarly journals Stable CpG Hypomethylation of Adipogenic Promoters in Freshly Isolated, Cultured, and Differentiated Mesenchymal Stem Cells from Adipose Tissue

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 ◽  
Author(s):  
Yalan Yang ◽  
Zhiguo Liu ◽  
Weimin Zhao ◽  
Lei Huang ◽  
Tianwen Wu ◽  
...  

Abstract Background Bone marrow (BM) and umbilical cord (UC) are the main sources of mesenchymal stem cells (MSCs). These two MSCs display significant differences in many biological characteristics, yet the underlying molecular mechanisms need to be explored. Results In this study, to better understanding the biological features of MSCs, we isolated BMMSCs and UCMSCs from inbred Wuzhishan miniature pigs and generated the first global DNA methylation and gene expression profiles of porcine MSCs. The results showed that the osteogenic and adipogenic differentiation ability of porcine BMMSCs is stronger than that of UCMSCs. Stem cell surface marker CD90 were positively detected in both BMMSCs and UCMSCs. 587 genes were differentially methylated (280 hypermethylated and 307 hypomethylated) at the promoter regions between BMMSCs and UCMSCs. Meanwhile, 1,979 differentially expressed genes (1,407 up-regulated and 572 down-regulated) were identified between BMMSCs and UCMSCs. Integrative analysis reveals that 120 genes displayed differences in both gene expression and promoter methylation. Gene Ontology enrichment analysis revealed that these differential genes were associated with cell differentiation, cell migration, and immunogenicity properties. Remarkably, skeletal system development related genes were significantly hypomethylated and up-regulated in UCMSCs, while cell cycle genes were significantly higher down-regulated and hypermethylated, implying UCMSCs have higher cell proliferative activity and lower osteogenic differentiation potential than BMMSCs. Conclusions Our results indicate that DNA methylation plays an important role in regulating the biological characteristics differences between BMMSCs and UCMSCs. The study might provide a molecular theory basis for the application of porcine MSCs in human.


2021 ◽  
Vol 22 (1) ◽  
Author(s):  
Zheng Feng ◽  
Yalan Yang ◽  
Zhiguo Liu ◽  
Weimin Zhao ◽  
Lei Huang ◽  
...  

Abstract Background Bone marrow (BM) and umbilical cord (UC) are the main sources of mesenchymal stem cells (MSCs). These two MSCs display significant differences in many biological characteristics, yet the underlying regulation mechanisms of these cells remain largely unknown. Results BMMSCs and UCMSCs were isolated from inbred Wuzhishan miniature pigs and the first global DNA methylation and gene expression profiles of porcine MSCs were generated. The osteogenic and adipogenic differentiation ability of porcine BMMSCs is greater than that of UCMSCs. A total of 1979 genes were differentially expressed and 587 genes were differentially methylated at promoter regions in these cells. Integrative analysis revealed that 102 genes displayed differences in both gene expression and promoter methylation. Gene ontology enrichment analysis showed that these genes were associated with cell differentiation, migration, and immunogenicity. Remarkably, skeletal system development-related genes were significantly hypomethylated and upregulated, whereas cell cycle genes were opposite in UCMSCs, implying that these cells have higher cell proliferative activity and lower differentiation potential than BMMSCs. Conclusions Our results indicate that DNA methylation plays an important role in regulating the differences in biological characteristics of BMMSCs and UCMSCs. Results of this study provide a molecular theoretical basis for the application of porcine MSCs in human medicine.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Kulisara Marupanthorn ◽  
Chairat Tantrawatpan ◽  
Pakpoom Kheolamai ◽  
Duangrat Tantikanlayaporn ◽  
Sirikul Manochantr

AbstractMesenchymal stem cells (MSCs) are important in regenerative medicine because of their potential for multi-differentiation. Bone marrow, chorion and placenta have all been suggested as potential sources for clinical application. However, the osteogenic differentiation potential of MSCs derived from chorion or placenta is not very efficient. Bone morphogenetic protein-2 (BMP-2) plays an important role in bone development. Its effect on osteogenic augmentation has been addressed in several studies. Recent studies have also shown a relationship between miRNAs and osteogenesis. We hypothesized that miRNAs targeted to Runt-related transcription factor 2 (Runx-2), a major transcription factor of osteogenesis, are responsible for regulating the differentiation of MSCs into osteoblasts. This study examines the effect of BMP-2 on the osteogenic differentiation of MSCs isolated from chorion and placenta in comparison to bone marrow-derived MSCs and investigates the role of miRNAs in the osteogenic differentiation of MSCs from these sources. MSCs were isolated from human bone marrow, chorion and placenta. The osteogenic differentiation potential after BMP-2 treatment was examined using ALP staining, ALP activity assay, and osteogenic gene expression. Candidate miRNAs were selected and their expression levels during osteoblastic differentiation were examined using real-time RT-PCR. The role of these miRNAs in osteogenesis was investigated by transfection with specific miRNA inhibitors. The level of osteogenic differentiation was monitored after anti-miRNA treatment. MSCs isolated from chorion and placenta exhibited self-renewal capacity and multi-lineage differentiation potential similar to MSCs isolated from bone marrow. BMP-2 treated MSCs showed higher ALP levels and osteogenic gene expression compared to untreated MSCs. All investigated miRNAs (miR-31, miR-106a and miR148) were consistently downregulated during the process of osteogenic differentiation. After treatment with miRNA inhibitors, ALP activity and osteogenic gene expression increased over the time of osteogenic differentiation. BMP-2 has a positive effect on osteogenic differentiation of chorion- and placenta-derived MSCs. The inhibition of specific miRNAs enhanced the osteogenic differentiation capacity of various MSCs in culture and this strategy might be used to promote bone regeneration. However, further in vivo experiments are required to assess the validity of this approach.


2011 ◽  
Vol 6 (8) ◽  
pp. 772-788 ◽  
Author(s):  
P. Hepsibha ◽  
T.V. Meenambiga ◽  
A. Mangalagow ◽  
A. Palanisamy ◽  
A. Stalin ◽  
...  

2018 ◽  
Vol 2018 ◽  
pp. 1-11 ◽  
Author(s):  
Yangyang Cao ◽  
Haoqing Yang ◽  
Luyuan Jin ◽  
Juan Du ◽  
Zhipeng Fan

Bone marrow mesenchymal stem cells (BMSCs) nowadays are regarded as promising candidates in cell-based therapy for the regeneration of damaged bone tissues that are either incurable or intractable due to the insufficiency of current therapies. Recent studies suggest that BMSCs differentiate into osteoblasts, and that this differentiation is regulated by some specific patterns of epigenetic modifications, such as DNA methylation. However, the potential role of DNA methylation modification in BMSC osteogenic differentiation is unclear. In this study, we performed a genome-wide study of DNA methylation between the noninduced and induced osteogenic differentiation of BMSCs at day 7. We found that the majority of cytosines in a CpG context were methylated in induced BMSCs. Our results also revealed that, along with the induced osteogenic differentiation in BMSCs, the average genomic methylation levels and CpG methylation in transcriptional factor regions (TFs) were increased, the CpG methylation level of various genomic elements was mainly in the medium-high methylation section, and CpG methylation levels in the repeat element had highly methylated levels. The GO analysis of differentially methylated region- (DMR-) associated genes (DMGs) showed that GO terms, including cytoskeletal protein binding (included in Molecular Function GO terms), skeletal development (included in Biological Process GO terms), mesenchymal cell differentiation (included in Biological Process GO terms), and stem cell differentiation (included in Biological Process), were enriched in the hypermethylated DMGs. Then, the KEGG analysis results showed that the WNT pathway, inositol phosphate metabolism pathway, and cocaine addiction pathway were more correlative with the DMRs during the induced osteogenic differentiation in BMSCs. In conclusion, this study revealed the difference of methylated levels during the noninduced and induced osteogenic differentiation of BMSCs and provided useful information for future works to characterize the important function of epigenetic mechanisms on BMSCs’ differentiation.


2016 ◽  
Vol 33 (10) ◽  
pp. 1395-1403 ◽  
Author(s):  
Moysés S. Miranda ◽  
Hamilton S. Nascimento ◽  
Mayra P. R. Costa ◽  
Nathália N. Costa ◽  
Karynne N. L. Brito ◽  
...  

2007 ◽  
Vol 19 (1) ◽  
pp. 227
Author(s):  
A. C. Boquest ◽  
A. Noer ◽  
A. L. Sørensen ◽  
K. Vekterud ◽  
P. Collas

Mesenchymal stem cells (MSCs) have received intense research interest due to their perceived potential application in regenerative medicine; nevertheless, MSCs are primarily restricted to form mesodermal cell types. Adipose stem cells (ASCs) with a CD34+ CD105+ CD45– CD31– immunophenotype can be obtained in an uncultured state with high purity from the stromal vascular fraction of human liposuction material (Boquest et al. 2005 Mol. Biol. Cell 16, 1131–1141). While ASCs differentiate readily into adipocytes, their endothelial lineage commitment has been scarcely reported, and controversy remains regarding ASC contribution to vascularization. To address the epigenetic commitment of ASCs to adipogenic and endothelial lineages, we carried out a bisulfite sequencing analysis of CpG methylation in the promoters of adipogenic (LEP, PPARG2, FABP4, LPL), endothelial (CD31, CD144), and myogenic (MYOG) genes in freshly isolated and in clonal ASC cultures in relation to gene expression and differentiation potential. Uncultured ASCs display mosaic hypomethylation of adipogenic promoters, in contrast to MYOG, CD31, or CD144 which are methylated (Noer et al. 2006 Mol. Biol. Cell 17, in press). Nevertheless, CpG methylation does not reflect transcriptional status of these genes in undifferentiated cells. Culture and adipogenic differentiation of ASCs maintains the hypomethylated profile of adipogenic promoters and the hypermethylation of non-adipogenic promoters. Endothelial stimulation of ASCs in methylcellulose elicits tubule-like networks, up-regulation of CD31 and CD144, and restrictive induction of a CD31+ CD144+ immunophenotype. Discrete and lineage-specific changes in CpG methylation in the CD31 and CD144 promoters take place but no global demethylation that marks endothelial cells occurs. Promoters not involved in endothelial differentiation retain a methylation profile characteristic of undifferentiated cells. Hypermethylation of CD31 and CD144 suggests a restricted commitment of ASCs to the endothelial lineage. This contrasts with hypomethylation of adipogenic promoters which reflects a propensity toward adipogenic differentiation. Despite the up-regulation of lineage-specific transcripts, overall maintenance of promoter methylation after adipogenic, osteogenic, and endothelial differentiation suggests the maintenance of an epigenetic signature characteristic of undifferentiated cells. Analysis of CpG methylation at lineage-specific promoters should provide a robust assessment of epigenetic commitment of stem cells to a specific lineage.


2019 ◽  
Vol 120 (7) ◽  
pp. 11842-11852 ◽  
Author(s):  
Simone Ortiz Moura Fideles ◽  
Adriana Cassia Ortiz ◽  
Amanda Freire Assis ◽  
Max Jordan Duarte ◽  
Fabiola Singaretti Oliveira ◽  
...  

2020 ◽  
Vol 2020 ◽  
pp. 1-14
Author(s):  
Seung-Cheol Lee ◽  
Yoo-Jung Lee ◽  
Min Kyoung Shin ◽  
Jung-Suk Sung

Human mesenchymal stem cells derived from adipose tissue (hADMSCs) are a desirable candidate in regenerative medicine. hADMSCs secrete growth factors, cytokines, and chemokines and also express various receptors that are important in cell activation, differentiation, and migration to injured tissue. We showed that the expression level of chemokine receptor CXCR6 was significantly increased by ~2.5-fold in adipogenic-differentiated cells (Ad), but not in osteogenic-differentiated cells (Os) when compared with hADMSCs. However, regulation of CXCR6 expression on hADMSCs by using lentiviral particles did not affect the differentiation potential of hADMSCs. Increased expression of CXCR6 on Ad was mediated by both receptor recycling, which was in turn regulated by secretion of CXCL16, and de novo synthesis. The level of soluble CXCL16 was highly increased in both Ad and Os in particular, which inversely correlates with the expression on a transmembrane-bound form of CXCL16 that is cleaved by disintegrin and metalloproteinase. We concluded that the expression of CXCR6 is regulated by receptor degradation or recycling when it is internalized by interaction with CXCL16 and by de novo synthesis of CXCR6. Overall, our study may provide an insight into the molecular mechanisms of the CXCR6 reciprocally expressed on differentiated cells from hADMSCs.


Sign in / Sign up

Export Citation Format

Share Document