DOI QR코드

DOI QR Code

Genome-scale DNA methylation pattern profiling of human bone marrow mesenchymal stem cells in long-term culture

  • Choi, Mi Ran (Department of Molecular and Life Sciences, Hanyang University) ;
  • In, Yong-Ho (Medicinal Bioconvergence Research Center, Seoul National University) ;
  • Park, Jungsun (Medicinal Bioconvergence Research Center, Seoul National University) ;
  • Park, Taesung (Interdisciplinary Program in Bioinformatics, Seoul National University) ;
  • Jung, Kyoung Hwa (Department of Molecular and Life Sciences, Hanyang University) ;
  • Chai, Jin Choul (Department of Molecular and Life Sciences, Hanyang University) ;
  • Chung, Mi Kyung (Fertility Center, CHA Gangnam Medical Center, CHA University) ;
  • Lee, Young Seek (Department of Molecular and Life Sciences, Hanyang University) ;
  • Chai, Young Gyu (Department of Molecular and Life Sciences, Hanyang University)
  • Accepted : 2012.06.05
  • Published : 2012.08.31

Abstract

Human bone marrow mesenchymal stem cells (MSCs) expanded in vitro exhibit not only a tendency to lose their proliferative potential, homing ability and telomere length but also genetic or epigenetic modifications, resulting in senescence. We compared differential methylation patterns of genes and miRNAs between early-passage [passage 5 (P5)] and late-passage (P15) cells and estimated the relationship between senescence and DNA methylation patterns. When we examined hypermethylated genes (methylation peak ${\geq}$ 2) at P5 or P15, 2,739 genes, including those related to fructose and mannose metabolism and calcium signaling pathways, and 2,587 genes, including those related to DNA replication, cell cycle and the PPAR signaling pathway, were hypermethylated at P5 and P15, respectively. There was common hypermethylation of 1,205 genes at both P5 and P15. In addition, genes that were hypermethylated at P5 (CPEB1, GMPPA, CDKN1A, TBX2, SMAD9 and MCM2) showed lower mRNA expression than did those hypermethylated at P15, whereas genes that were hypermethylated at P15 (MAML2, FEN1 and CDK4) showed lower mRNA expression than did those that were hypermethylated at P5, demonstrating that hypermethylation at DNA promoter regions inhibited gene expression and that hypomethylation increased gene expression. In the case of hypermethylation on miRNA, 27 miRNAs were hypermethylated at P5, whereas 44 miRNAs were hypermethylated at P15. These results show that hypermethylation increases at genes related to DNA replication, cell cycle and adipogenic differentiation due to long-term culture, which may in part affect MSC senescence.

Keywords

Acknowledgement

Supported by : National Research Foundation of Korea (NRF)

References

  1. Antequera F. Structure, function and evolution of CpG island promoters. Cell Mol Life Sci 2003;60:1647-1658 https://doi.org/10.1007/s00018-003-3088-6
  2. Banfi A, Muraglia A., Dozin B, Mastrogiacomo M, Cancedda R, Quarto R. Proliferation kinetics and differentiation potential of ex vivo expanded human bone marrow stromal cells: Implications for their use in cell therapy. Exp Hematol 2000;28:707-715 https://doi.org/10.1016/S0301-472X(00)00160-0
  3. Baxter MA, Wynn RF, Jowitt SN, Wraith JE, Fairbairn LJ, Bellantuono I. Study of telomere length reveals rapid aging of human marrow stromal cells following in vitro expansion. Stem Cells 2004;22:675-682 https://doi.org/10.1634/stemcells.22-5-675
  4. Beausejour CM, Krtolica A, Galimi F, Narita M, Lowe SW, Yaswen P, Campisi J. Reversal of human cellular senescence: roles of the p53 and p16 pathways. EMBO J 2003;22:4212-4222 https://doi.org/10.1093/emboj/cdg417
  5. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Statist Soc B 1995;57:289-300
  6. Bernardo ME, Zaffaroni N, Novara F, Cometa AM, Avanzini MA, Moretta A, Montagna D, Maccario R, Villa R, Daidone MG, Zuffardi O, Locatelli F. Human bone marrow derived mesenchymal stem cells do not undergo transformation after long-term in vitro culture and do not exhibit telomere maintenance mechanisms. Cancer Res 2007;67:9142-9149 https://doi.org/10.1158/0008-5472.CAN-06-4690
  7. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev 2002;16:6-21 https://doi.org/10.1101/gad.947102
  8. Bonab MM, Alimoghaddam K, Talebian F, Ghaffari SH, Ghavamzadeh A, Nikbin B. Aging of mesenchymal stem cell in vitro. BMC Cell Biol 2006;7:14 https://doi.org/10.1186/1471-2121-7-14
  9. Bork S, Pfister S, Witt H, Horn P, Korn B, Ho AD, Wagner W. DNA methylation pattern changes upon long-term culture and aging of human mesenchymal stromal cells. Aging Cell 2010;9:54-63 https://doi.org/10.1111/j.1474-9726.2009.00535.x
  10. Bruder SP, Jaiswal N, Haynesworth SE. Growth kinetics, self-renewal, and the osteogenic potential of purified human mesenchymal stem cells during extensive subcultivation and following cryopreservation. J Cell Biochem 1997;64:278-294 https://doi.org/10.1002/(SICI)1097-4644(199702)64:2<278::AID-JCB11>3.0.CO;2-F
  11. Cawthon RM. Telomere measurement by quantitative PCR. Nucleic Acids Res 2002;30:e47 https://doi.org/10.1093/nar/30.10.e47
  12. Choi MR, Kim HY, Park JY, Lee TY, Baik CS, Chai YG, Jung KH, Park KS, Roh W, Kim KS, Kim SH. Selection of optimal passage of bone marrow-derived mesenchymal stem cells for stem cell therapy in patients with amyotrophic lateral sclerosis. Neurosci Lett 2010;472:94-98 https://doi.org/10.1016/j.neulet.2010.01.054
  13. Crosby ME, Kulshreshtha R, Ivan M, Glazer PM. MicroRNA regulation of DNA repair gene expression in hypoxic stress. Cancer Res 2009;69:1221-1229 https://doi.org/10.1158/0008-5472.CAN-08-2516
  14. Dahl JA, Duggal S, Coulston N, Millar D, Melki J, Shahdadfar A, Brinchmann JE, Collas P. Genetic and epigenetic instability of human bone marrow mesenchymal stem cells expanded in autologous serum or fetal bovine serum. Int J Dev Biol 2008;52:1033-1042 https://doi.org/10.1387/ijdb.082663jd
  15. Digirolamo CM, Stokes D, Colter D, Phinney DG, Class R, Prockop DJ. Propagation and senescence of human marrow stromal cells in culture: a simple colony-forming assay identifies samples with the greatest potential to propagate and differentiate. Br J Haematol 1999;107:275-281 https://doi.org/10.1046/j.1365-2141.1999.01715.x
  16. Elango N, Yi SV. DNA methylation and structural and functional bimodality of vertebrate promoters. Mol Biol Evol 2008;25:1602-1608 https://doi.org/10.1093/molbev/msn110
  17. Godlewski J, Newton HB, Chiocca EA, Lawler SE. MicroRNAs and glioblastoma; the stem cell connection. Cell Death Differ 2010;17:221-228 https://doi.org/10.1038/cdd.2009.71
  18. Guillot PV, Gotherstrom C, Chan J, Kurata H, Fisk NM. Human first-trimester fetal MSC express pluripotency markers and grow faster and have longer telomeres than adult MSC. Stem Cells 2007;25:646-654
  19. Hammond SM, Sharpless NE. HMGA2, microRNAs, and stem cell aging. Cell 2008;135:1013-1016 https://doi.org/10.1016/j.cell.2008.11.026
  20. Ibanez-Ventoso C, Yang M, Guo S, Robins H, Padgett RW, Driscoll M. Modulated microRNA expression during adult lifespan in Caenorhabditis elegans. Aging Cell 2006;5: 235-246 https://doi.org/10.1111/j.1474-9726.2006.00210.x
  21. Lara-Castro C, Fu Y, Chung BH, Garvey WT. Adiponectin and the metabolic syndrome: mechanisms mediating risk for metabolic and cardiovascular disease. Curr Opin Lipidol 2007;18:263-270 https://doi.org/10.1097/MOL.0b013e32814a645f
  22. Liang R, Bates DJ, Wang E. Epigenetic control of microRNA expression and aging. Curr Genomics 2009;10:184-193 https://doi.org/10.2174/138920209788185225
  23. Nelson WJ, Nusse R. Convergence of Wnt, beta-catenin, and cadherin pathways. Science 2004;303:1483-1487 https://doi.org/10.1126/science.1094291
  24. Nilsson O, Mitchum RD Jr, Schrier L, Ferns SP, Barnes KM, Troendle JF, Baron J. Growth plate senescence is associated with loss of DNA methylation. J Endocrinol 2005;186:241-249 https://doi.org/10.1677/joe.1.06016
  25. Noer A, Sorensen AL, Boquest AC, Collas P. Stable CpG hypomethylation of adipogenic promoters in freshly isolated, cultured, and differentiated mesenchymal stem cells from adipose tissue. Mol Biol Cell 2006;17:3543-3556 https://doi.org/10.1091/mbc.E06-04-0322
  26. Noer A, Boquest AC, Collas P. Dynamics of adipogenic promoter DNA methylation during clonal culture of human adipose stem cells to senescence. BMC Cell Biol 2007;8:18 https://doi.org/10.1186/1471-2121-8-18
  27. Pallasch CP, Patz M, Park YJ, Hagist S, Eggle D, Claus R, Debey-Pascher S, Schulz A, Frenzel LP, Claasen J, Kutsch N, Krause G, Mayr C, Rosenwald A, Plass C, Schultze JL, Hallek M, Wendtner CM. miRNA deregulation by epigenetic silencing disrupts suppression of the oncogene PLAG1 in chronic lymphocytic leukemia. Blood 2009;114:3255-3264 https://doi.org/10.1182/blood-2009-06-229898
  28. Rombouts WJ, Ploemacher RE. Primary murine MSC show highly efficient homing to the bone marrow but lose homing ability following culture. Leukemia 2003;17:160-170 https://doi.org/10.1038/sj.leu.2402763
  29. Rubinstein JC, Tran N, Ma S, Halaban R, Krauthammer M. Genome-wide methylation and expression profiling identifies promoter characteristics affecting demethylation-induced gene up-regulation in melanoma. BMC Med Genomics 2010;3:4 https://doi.org/10.1186/1755-8794-3-4
  30. Schaefer A, Jung M, Miller K, Lein M, Kristiansen G, Erbersdobler A, Jung K. Suitable reference genes for relative quantification of miRNA expression in prostate cancer. Exp Mol Med 2010;42:749-758 https://doi.org/10.3858/emm.2010.42.11.076
  31. Shibata KR, Aoyama T, Shima Y, Fukiage K, Otsuka S, Furu M, Kohno Y, Ito K, Fujibayashi S, Neo M, Nakayama T, Nakamura T, Toguchida J. Expression of the p16INK4A gene is associated closely with senescence of human mesenchymal stem cells and is potentially silenced by DNA methylation during in vitro expansion. Stem Cells 2007;25: 2371-2382 https://doi.org/10.1634/stemcells.2007-0225
  32. Sinkkonen L, Hugenschmidt T, Berninger P, Gaidatzis D, Mohn F, Artus-Revel CG, Zavolan M, Svoboda P, Filipowicz W. MicroRNAs control de novo DNA methylation through regulation of transcriptional repressors in mouse embryonic stem cells. Nat Struct Mol Biol 2008;15:259-267 https://doi.org/10.1038/nsmb.1391
  33. Suzuki MM, Bird A. DNA methylation landscapes: provocative insights from epigenomics. Nat Rev Genet 2008;9:465-476
  34. Te Pas MF, Hulsegge I, Coster A, Pool MH, Heuven HH, Janss LL. Biochemical pathways analysis of microarray results: regulation of myogenesis in pigs. BMC Dev Biol 2007;7:66 https://doi.org/10.1186/1471-213X-7-66
  35. Wagner W, Horn P, Castoldi M, Diehlmann A, Bork S, Saffrich R, Benes V, Blake J, Pfister S, Eckstein V, Ho AD. Replicative senescence of mesenchymal stem cells: a continuous and organized process. PLoS One 2008;3:e2213 https://doi.org/10.1371/journal.pone.0002213
  36. Weber M, Hellmann I, Stadler MB, Ramos L, Paabo S, Rebhan M, Schubeler D. Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat Genet 2007;39:457-466 https://doi.org/10.1038/ng1990
  37. Zhang W, Ji W, Yang J, Yang L, Chen W, Zhuang Z. Comparison of global DNA methylation profiles in replicative versus premature senescence. Life Sci 2008;83:475-480 https://doi.org/10.1016/j.lfs.2008.07.015

Cited by

  1. DNA Methylation Changes during In Vitro Propagation of Human Mesenchymal Stem Cells: Implications for Their Genomic Stability? vol.2013, pp.None, 2012, https://doi.org/10.1155/2013/192425
  2. hiPSCs: Reprogramming towards cell-based therapies vol.2, pp.3, 2013, https://doi.org/10.4236/ojrm.2013.23010
  3. Senescent cells harbour features of the cancer epigenome vol.15, pp.12, 2012, https://doi.org/10.1038/ncb2879
  4. Generation and Characterization of an Immortalized Human Mesenchymal Stromal Cell Line vol.23, pp.19, 2012, https://doi.org/10.1089/scd.2013.0599
  5. Germline Mutations in MAP3K6 Are Associated with Familial Gastric Cancer vol.10, pp.10, 2012, https://doi.org/10.1371/journal.pgen.1004669
  6. The role of Alk-1 and Alk-5 in the mechanosensing of chondrocytes vol.19, pp.4, 2012, https://doi.org/10.2478/s11658-014-0220-6
  7. Role of endoscopic ultrasound in the molecular diagnosis of pancreatic cancer vol.20, pp.31, 2012, https://doi.org/10.3748/wjg.v20.i31.10758
  8. Lung Fibroblasts from Patients with Idiopathic Pulmonary Fibrosis Exhibit Genome-Wide Differences in DNA Methylation Compared to Fibroblasts from Nonfibrotic Lung vol.9, pp.9, 2012, https://doi.org/10.1371/journal.pone.0107055
  9. H3K4me1 marks DNA regions hypomethylated during aging in human stem and differentiated cells vol.25, pp.1, 2012, https://doi.org/10.1101/gr.169011.113
  10. Placental mesenchymal stem cells of fetal origin deposit epigenetic alterations during long-term culture under serum-free condition vol.15, pp.2, 2012, https://doi.org/10.1517/14712598.2015.960837
  11. 중간엽줄기세포의 노화에 따른 후생유전학적 변화 vol.25, pp.6, 2012, https://doi.org/10.5352/jls.2015.25.6.724
  12. Human Mesenchymal Stem Cells Retain Multilineage Differentiation Capacity Including Neural Marker Expression after Extended In Vitro Expansion vol.10, pp.9, 2012, https://doi.org/10.1371/journal.pone.0137255
  13. Derivation and growth characteristics of dental pulp stem cells from patients of different ages vol.12, pp.4, 2012, https://doi.org/10.3892/mmr.2015.4106
  14. Anti-senescence effects of DNA methyltransferase inhibitor RG108 in human bone marrow mesenchymal stromal cells : Anti-Senescence Effects of RG108 vol.62, pp.5, 2012, https://doi.org/10.1002/bab.1393
  15. Epigenetic Modifications upon Senescence of Mesenchymal Stem Cells vol.2, pp.3, 2012, https://doi.org/10.1007/s40778-016-0051-7
  16. The DNA methylation profile of activated human natural killer cells vol.11, pp.5, 2012, https://doi.org/10.1080/15592294.2016.1163454
  17. Identity, proliferation capacity, genomic stability and novel senescence markers of mesenchymal stem cells isolated from low volume of human bone marrow vol.7, pp.10, 2016, https://doi.org/10.18632/oncotarget.7456
  18. Antisenescence activity of G9a inhibitor BIX01294 on human bone marrow mesenchymal stromal cells vol.40, pp.2, 2012, https://doi.org/10.3906/biy-1507-11
  19. DNA methylation is critical for tooth agenesis: implications for sporadic non-syndromic anodontia and hypodontia vol.6, pp.None, 2012, https://doi.org/10.1038/srep19162
  20. Inhibiting DNA methylation switches adipogenesis to osteoblastogenesis by activating Wnt10a vol.6, pp.None, 2012, https://doi.org/10.1038/srep25283
  21. A Multi-Step miRNA-mRNA Regulatory Network Construction Approach Identifies Gene Signatures Associated with Endometrioid Endometrial Carcinoma vol.7, pp.6, 2016, https://doi.org/10.3390/genes7060026
  22. Senescence in Human Mesenchymal Stem Cells: Functional Changes and Implications in Stem Cell-Based Therapy vol.17, pp.7, 2012, https://doi.org/10.3390/ijms17071164
  23. Comprehensive DNA Methylation and Gene Expression Profiling in Differentiating Human Adipocytes vol.117, pp.12, 2012, https://doi.org/10.1002/jcb.25568
  24. Age-associated hydroxymethylation in human bone-marrow mesenchymal stem cells vol.14, pp.None, 2012, https://doi.org/10.1186/s12967-016-0966-x
  25. Methylome Analysis of Human Bone Marrow MSCs Reveals Extensive Age- and Culture-Induced Changes at Distal Regulatory Elements vol.9, pp.3, 2012, https://doi.org/10.1016/j.stemcr.2017.07.018
  26. Epigenetic Regulation of Bone Marrow Stem Cell Aging: Revealing Epigenetic Signatures associated with Hematopoietic and Mesenchymal Stem Cell Aging vol.10, pp.1, 2012, https://doi.org/10.14336/ad.2017.1213
  27. Impact of DNA methyltransferase inhibitor 5‐azacytidine on cardiac development of zebrafish in vivo and cardiomyocyte proliferation, apoptosis, and the homeostasis of gene expression in vitro vol.120, pp.10, 2012, https://doi.org/10.1002/jcb.29010
  28. Molecular Mechanisms Contributing to Mesenchymal Stromal Cell Aging vol.10, pp.2, 2020, https://doi.org/10.3390/biom10020340
  29. Senescent mesenchymal stem/stromal cells and restoring their cellular functions vol.12, pp.9, 2020, https://doi.org/10.4252/wjsc.v12.i9.966
  30. Challenges and advances in clinical applications of mesenchymal stromal cells vol.14, pp.1, 2012, https://doi.org/10.1186/s13045-021-01037-x