DOI QR코드

DOI QR Code

Subpopulations of miniature pig mesenchymal stromal cells with different differentiation potentials differ in the expression of octamer-binding transcription factor 4 and sex determining region Y-box 2

  • Jeon, Ryounghoon (Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University) ;
  • Park, Sungjo (Department of Cardiovascular Medicine, Mayo Clinic) ;
  • Lee, Sung-Lim (Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University) ;
  • Rho, Gyu-Jin (Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University)
  • Received : 2019.05.23
  • Accepted : 2019.09.03
  • Published : 2020.03.01

Abstract

Objective: Human mesenchymal stromal cells (MSCs) exhibit variable differentiation potential and can be divided accordingly into distinct subpopulations whose ratios vary with donor age. However, it is unknown whether the same is true in pigs. This study investigated MSC subpopulations in miniature pig and compared their characteristics in young (2 to 3 months) and adult (27 to 35 months) pigs. Methods: Osteogenic, chondrogenic, and adipogenic capacity of isolated MSCs was evaluated by von Kossa, Alcian blue, and oil red O staining, respectively. Cell surface antigen expression was determined by flow cytometry. Proliferative capacity was assessed with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Expression of marker genes was detected by quantitative real-time polymerase chain reaction. Results: Porcine MSCs comprised cells with trilineage and bilineage differentiation potential (tMSCs and bMSCs, respectively) and non-differentiating stromal cells (NDSCs). The tMSC and bMSC fractions were smaller in adult than in young pigs (63.0% vs 71.2% and 11.6% vs 24.0%, respectively, p<0.05); NDSCs showed the opposite trend (25.4% vs 4.8%; p<0.05). Subpopulations showed no differences in morphology, cell surface antigen expression, or proliferative capacity, but octamer-binding transcription factor 4 (OCT4) expression was higher in tMSCs than in bMSCs and NDSCs (p<0.05), whereas sex determining region Y-box 2 (SOX2) expression was higher in tMSCs and bMSCs than in NDSCs (p<0.05). Aging had no effect on these trends. Conclusion: Porcine MSCs comprise distinct subpopulations that differ in their differentiation potential and OCT4 and SOX2 expression. Aging does not affect the characteristics of each subpopulation but alters their ratios.

Keywords

References

  1. Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement. Cytotherapy 2006;8:315-7. https://doi.org/10.1080/14653240600855905
  2. Maxson S, Lopez EA, Yoo D, Danilkovitch-Miagkova A, Leroux MA. Concise review: role of mesenchymal stem cells in wound repair. Stem Cells Transl Med 2012;1:142-9. https://doi.org/10.5966/sctm.2011-0018
  3. da Silva Meirelles L, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci 2006;119:2204-13. https://doi.org/10.1242/jcs.02932
  4. Nombela-Arrieta C, Ritz J, Silberstein LE. The elusive nature and function of mesenchymal stem cells. Nat Rev Mol Cell Biol 2011;12:126-31. https://doi.org/10.1038/nrm3049
  5. Russell KC, Phinney DG, Lacey MR, Barrilleaux BL, Meyertholen KE, O'Connor KC. In vitro high-capacity assay to quantify the clonal heterogeneity in trilineage potential of mesenchymal stem cells reveals a complex hierarchy of lineage commitment. Stem Cells 2010;28:788-98. https://doi.org/10.1002/stem.312
  6. Mo M, Wang S, Zhou Y, Li H, Wu Y. Mesenchymal stem cell subpopulations: phenotype, property and therapeutic potential. Cell Mol Life Sci 2016;73:3311-21. https://doi.org/10.1007/s00018-016-2229-7
  7. Muraglia A, Cancedda R, Quarto R. Clonal mesenchymal progenitors from human bone marrow differentiate in vitro according to a hierarchical model. J Cell Sci 2000;113:1161-6. https://doi.org/10.1242/jcs.113.7.1161
  8. Lee WC, Shi H, Poon Z, et al. Multivariate biophysical markers predictive of mesenchymal stromal cell multipotency. Proc Natl Acad Sci USA 2014;111:E4409-18. https://doi.org/10.1073/pnas.1402306111
  9. McLeod CM, Mauck RL. On the origin and impact of mesenchymal stem cell heterogeneity: new insights and emerging tools for single cell analysis. Eur Cell Mater 2017;34:217-31. https://doi.org/10.22203/eCM.v034a14
  10. Zaim M, Karaman S, Cetin G, Isik S. Donor age and long-term culture affect differentiation and proliferation of human bone marrow mesenchymal stem cells. Ann Hematol 2012;91:1175-86. https://doi.org/10.1007/s00277-012-1438-x
  11. Stolzing A, Scutt A. Age-related impairment of mesenchymal progenitor cell function. Aging Cell 2006;5:213-24. https://doi.org/10.1111/j.1474-9726.2006.00213.x
  12. Mueller SM, Glowacki J. Age-related decline in the osteogenic potential of human bone marrow cells cultured in three-dimensional collagen sponges. J Cell Biochem 2001;82:583-90. https://doi.org/10.1002/jcb.1174
  13. Ganguly P, El-Jawhari JJ, Giannoudis PV, Burska AN, Ponchel F, Jones EA. Age-related changes in bone marrow mesenchymal stromal cells: A potential impact on osteoporosis and osteoarthritis development. Cell Transplant 2017;26:1520-9. https://doi.org/10.1177/0963689717721201
  14. Steindler DA. Stem cells, regenerative medicine, and animal models of disease. ILAR J 2007;48:323-38. https://doi.org/10.1093/ilar.48.4.323
  15. Heino TJ, Alm JJ, Moritz N, Aro HT. Comparison of the osteogenic capacity of minipig and human bone marrow-derived mesenchymal stem cells. J Orthop Res 2012;30:1019-25. https://doi.org/10.1002/jor.22049
  16. Lee WJ, Kim JY, Wu TP, Park LS. The establishment of a porcine rheumatoid arthritis model: Collagen induced arthritis minipig model. J Pharmacol Sci 2016;132:41-7. https://doi.org/10.1016/j.jphs.2016.04.012
  17. Zhang L, Chan C. Isolation and enrichment of rat mesenchymal stem cells (MSCs) and separation of single-colony derived MSCs. J Vis Exp 2010;37:e1852. https://doi.org/10.3791/1852
  18. Lee JH, Lee WJ, Jeon RH, et al. Development and gene expression of porcine cloned embryos derived from bone marrow stem cells with overexpressing Oct4 and Sox2. Cell Reprogram 2014;16:428-38. https://doi.org/10.1089/cell.2014.0036
  19. Stolzing A, Jones E, McGonagle D, Scutt A. Age-related changes in human bone marrow-derived mesenchymal stem cells: consequences for cell therapies. Mech Ageing Dev 2008;129:163-73. https://doi.org/10.1016/j.mad.2007.12.002
  20. Park SB, Seo KW, So AY, et al. SOX2 has a crucial role in the lineage determination and proliferation of mesenchymal stem cells through Dickkopf-1 and c-MYC. Cell Death Differ 2012; 19:534-45. https://doi.org/10.1038/cdd.2011.137
  21. Weissman IL. Stem cells: units of development, units of regeneration, and units in evolution. Cell 2000;100:157-68. https://doi.org/10.1016/S0092-8674(00)81692-X
  22. Walker PA, Shah SK, Harting MT, Cox CS Jr. Progenitor cell therapies for traumatic brain injury: barriers and opportunities in translation. Dis Model Mech 2009;2:23-38. https://doi.org/10.1242/dmm.001198
  23. Stadtfeld M, Hochedlinger K. Induced pluripotency: history, mechanisms, and applications. Genes Dev 2010;24:2239-63. https://doi.org/10.1101/gad.1963910
  24. Boyer LA, Lee TI, Cole MF, et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 2005;122: 947-56. https://doi.org/10.1016/j.cell.2005.08.020
  25. Yu P, Nie Q, Tang C, Zhang L. Nanog induced intermediate state in regulating stem cell differentiation and reprogramming. BMC Syst Biol 2018;12:22. https://doi.org/10.1186/s12918-018-0552-3
  26. Pierantozzi E, Gava B, Manini I, et al. Pluripotency regulators in human mesenchymal stem cells: expression of NANOG but not of OCT-4 and SOX-2. Stem Cells Dev 2011;20:915-23. https://doi.org/10.1089/scd.2010.0353
  27. Rui Y, Xu L, Chen R, et al. Epigenetic memory gained by priming with osteogenic induction medium improves osteogenesis and other properties of mesenchymal stem cells. Sci Rep 2015;5:11056. https://doi.org/10.1038/srep11056
  28. Liu TM, Wu YN, Guo XM, Hui JH, Lee EH, Lim B. Effects of ectopic Nanog and Oct4 overexpression on mesenchymal stem cells. Stem Cells Dev 2009;18:1013-22. https://doi.org/10.1089/scd.2008.0335
  29. Liu S, Bou G, Sun R, et al. Sox2 is the faithful marker for pluripotency in pig: evidence from embryonic studies. Dev Dyn 2015;244:619-27. https://doi.org/10.1002/dvdy.24248
  30. Han J, Mistriotis P, Lei P, Wang D, Liu S, Andreadis ST. Nanog reverses the effects of organismal aging on mesenchymal stem cell proliferation and myogenic differentiation potential. Stem Cells 2012;30:2746-59. https://doi.org/10.1002/stem.1223

Cited by

  1. The Peripherin Gene Regulates the Migration of Bone Marrow Mesenchymal Stem Cells in Wuzhishan Mini Pigs vol.2020, 2020, https://doi.org/10.1155/2020/8856388