Browse > Article
http://dx.doi.org/10.5713/ajas.19.0796

Transdifferentiation of α-1,3-galactosyltransferase knockout pig bone marrow derived mesenchymal stem cells into pancreatic β-like cells by microenvironment modulation  

Ullah, Imran (Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration)
Lee, Ran (Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration)
Oh, Keon Bong (Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration)
Hwang, Seongsoo (Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration)
Kim, Youngim (Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration)
Hur, Tai-Young (Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration)
Ock, Sun A (Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration)
Publication Information
Asian-Australasian Journal of Animal Sciences / v.33, no.11, 2020 , pp. 1837-1847 More about this Journal
Abstract
Objective: To evaluate the pancreatic differentiation potential of α-1,3-galactosyltransferase knockout (GalTKO) pig-derived bone marrow-derived mesenchymal stem cells (BM-MSCs) using epigenetic modifiers with different pancreatic induction media. Methods: The BM-MSCs have been differentiated into pancreatic β-like cells by inducing the overexpression of key transcription regulatory factors or by exposure to specific soluble inducers/small molecules. In this study, we evaluated the pancreatic differentiation of GalTKO pig-derived BM-MSCs using epigenetic modifiers, 5-azacytidine (5-Aza) and valproic acid (VPA), and two types of pancreatic induction media - advanced Dulbecco's modified Eagle's medium (ADMEM)-based and N2B27-based media. GalTKO BM-MSCs were treated with pancreatic induction media and the expression of pancreas-islets-specific markers was evaluated by real-time quantitative polymerase chain reaction, Western blotting, and immunofluorescence. Morphological changes and changes in the 5'-C-phosphate-G-3' (CpG) island methylation patterns were also evaluated. Results: The expression of the pluripotent marker (POU class 5 homeobox 1 [OCT4]) was upregulated upon exposure to 5-Aza and/or VPA. GalTKO BM-MSCs showed increased expression of neurogenic differentiation 1 in the ADMEM-based (5-Aza) media, while the expression of NK6 homeobox 1 was elevated in cells induced with the N2B27-based (5-Aza) media. Moreover, the morphological transition and formation of islets-like cellular clusters were also prominent in the cells induced with the N2B27-based media with 5-Aza. The higher insulin expression revealed the augmented trans-differentiation ability of GalTKO BM-MSCs into pancreatic β-like cells in the N2B27-based media than in the ADMEM-based media. Conclusion: 5-Aza treated GalTKO BM-MSCs showed an enhanced demethylation pattern in the second CpG island of the OCT4 promoter region compared to that in the GalTKO BM-MSCs. The exposure of GalTKO pig-derived BM-MSCs to the N2B27-based microenvironment can significantly enhance their trans-differentiation ability into pancreatic β-like cells.
Keywords
N2B27; GalTKO BM-MSCs; Epigenetic Modifications; Cytidine; Pancreatic ${\beta}$-like Cells;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Bouwens L, Houbracken I, Mfopou JK. The use of stem cells for pancreatic regeneration in diabetes mellitus. Nat Rev Endocrinol 2013;9:598-606. https://doi.org/10.1038/nrendo.2013.145   DOI
2 Godfrey KJ, Mathew B, Bulman JC, Shah O, Clement S, Gallicano GI. Stem cell-based treatments for Type 1 diabetes mellitus: Bone marrow, embryonic, hepatic, pancreatic and induced pluripotent stem cells. Diabet Med 2012;29:14-23. https://doi.org/10.1111/j.1464-5491.2011.03433.x   DOI
3 Millman JR, Xie C, Van Dervort A, Gurtler M, Pagliuca FW, Melton DA. Generation of stem cell-derived ${\beta}$-cells from patients with type 1 diabetes. Nat Commun 2016;7:11463. https://doi.org/10.1038/ncomms11463   DOI
4 Bellin MD, Barton FB, Heitman A, et al. Potent induction immunotherapy promotes long-term insulin independence after islet transplantation in type 1 diabetes. Am J Transplant 2012;12:1576-83. https://doi.org/10.1111/j.1600-6143.2011.03977.x   DOI
5 Lumelsky N, Blondel O, Laeng P, Velasco I, Ravin R, McKay R. Differentiation of embryonic stem cells to insulin-secreting structures similar to pancreatic Islets. Science 2001;292:1389-94. https://doi.org/10.1126/science.1058866   DOI
6 Jiang J, Au M, Lu K, et al. Generation of insulin-producing islet-like clusters from human embryonic stem cells. Stem Cells 2007;25:1940-53. https://doi.org/10.1634/stemcells.2006-0761   DOI
7 Gottlicher M, Minucci S, Zhu P, et al. Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. EMBO J 2001;20:6969-78. https://doi.org/10.1093/emboj/20.24.6969   DOI
8 Sprangers B, Waer M, Billiau AD. Xenotransplantation: where are we in 2008? Kidney Int 2008;74:14-21. https://doi.org/10.1038/ki.2008.135   DOI
9 Ahn KS, Kim YJ, Kim M, et al. Resurrection of an alpha-1,3-galactosyltransferase gene-targeted miniature pig by recloning using postmortem ear skin fibroblasts. Theriogenology 2011;75:933-9. https://doi.org/10.1016/j.theriogenology.2010.11. 001   DOI
10 Ock SA, Jeon BJ, Rho GJ. Comparative characterization of porcine mesenchymal stem cells derived from bone marrow extract and skin tissues. Tissue Eng Part C Methods 2010;16:1481-91. http://doi.org/10.1089/ten.tec.2010.0149   DOI
11 Wu SC, Zhang Y. Active DNA demethylation: many roads lead to Rome. Nat Rev Mol Cell Biol 2010;11:607-20. https://doi.org/10.1038/nrm2950   DOI
12 Chiu CP, Blau HM. 5-Azacytidine permits gene activation in a previously noninducible cell type. Cell 1985;40:417-24. https://doi.org/10.1016/0092-8674(85)90155-2   DOI
13 Gao Y, Jammes H, Rasmussen MA, et al. Epigenetic regulation of gene expression in porcine epiblast, hypoblast, trophectoderm and epiblast-derived neural progenitor cells. Epigenetics 2011;6:1149-61. https://doi.org/10.4161/epi.6.9.16954   DOI
14 Peister A, Mellad JA, Larson BL, Hall BM, Gibson LF, Prockop DJ. Adult stem cells from bone marrow (MSCs) isolated from different strains of inbred mice vary in surface epitopes, rates of proliferation, and differentiation potential. Blood 2004;103:1662-8. https://doi.org/10.1182/blood-2003-09-3070   DOI
15 Jones PA. Effects of 5-azacytidine and its 2'-deoxyderivative on cell differentiation and DNA methylation. Pharmacol Ther 1985;28:17-27. https://doi.org/10.1016/0163-7258(85)90080-4   DOI
16 Conrad E, Stein R, Hunter CS. Revealing transcription factors during human pancreatic ${\beta}$ cell development. Trends Endocrinol Metab 2014;25:407-14. https://doi.org/10.1016/j.tem.2014. 03.013   DOI
17 Yao S, Chen S, Clark J, et al. Long-term self-renewal and directed differentiation of human embryonic stem cells in chemically defined conditions. Proc Natl Acad Sci USA 2006;103:6907-12. https://doi.org/10.1073/pnas.0602280103   DOI
18 Hansson M, Tonning A, Frandsen U, et al. Artifactual insulin release from differentiated embryonic stem cells. Diabetes 2004;53:2603-9. https://doi.org/10.2337/diabetes.53.10.2603   DOI
19 Maherali N, Sridharan R, Xie W, et al. Directly reprogrammed fibroblasts show global epigenetic remodeling and widespread tissue contribution. Cell Stem Cell 2007;1:55-70. https://doi.oarg/10.1016/j.stem.2007.05.014   DOI
20 Deb-Rinker P, Ly D, Jezierski A, Sikrska M, Walker PR. Sequential DNA methylation of the Nanog and Oct-4 upstream regions in human NT2 cells during neuronal differentiation. J Biol Chem 2005;280:6257-60. https://doi.org/10.1074/jbc.C400479200   DOI
21 Fan A, Ma K, An X, et al. Effects of TET1 knockdown on gene expression and DNA methylation in porcine induced pluripotent stem cells. Reproduction 2013;146:569-79 https://doi.org/10.1530/REP-13-0212   DOI
22 Ueyama H, Horibe T, Hinotsu S, et al. Chromosomal variability of human mesenchymal stem cells cultured under hypoxic conditions. J Cell Mol Med 2012;16:72-82. https://doi.org/10.1111/j.1582-4934.2011.01303.x   DOI
23 Pennarossa G, Maffei S, Campagnol M, et al. Brief demethylation step allows the conversion of adult human skin fibroblasts into insulin-secreting cells. Proc Natl Acad Sci USA 2013;110:8948-53. https://doi.org/10.1073/pnas.1220637110   DOI
24 Jafarian A, Taghikhani M, Abroun S, Pourpak Z, Allahverdi A, Soleimani M. Generation of high-yield insulin producing cells from human bone marrow mesenchymal stem cells. Mol Biol Rep 2014;4:4783-94. https://doi.org/10.1007/s11033- 014-3349-5
25 Gabr MM, Zakaria MM, Refaie AF, et al. Insulin producing cells from adult human bone marrow mesenchymal stem cells control streptozotocin-induced diabetes in nude mice. Cell Transplant 2013;22:133-45. https://doi.org/10.3727/09636 8912X647162   DOI
26 Belame Shivakumar S, Bharti D, Baregundi Subbarao R, et al. Pancreatic endocrine-like cells differentiated from human umbilical cords Wharton's jelly mesenchymal stem cells using small molecules. J Cell Physiol 2019;234:3933-47. https://doi.org/10.1002/jcp.27184   DOI
27 Takemitsu H, Zhao D, Ishikawa S, Michishita M, Arai T, Yamamoto I. Mechanism of insulin production in canine bone marrow derived mesenchymal stem cells. Gen Comp Endocrinol 2013;189:1-6. https://doi.org/10.1016/j.ygcen.2013.04.009   DOI
28 Xie H, Wang Y, Zhang H, Qi H, Zhou H, Li FR. Role of injured pancreatic extract promotes bone marrow-derived mesenchymal stem cells efficiently differentiate into insulin-producing cells. PloS One 2013;8:e76056. https://doi.org/10.1371/journal.pone.0076056   DOI
29 Li M, Liu G-H, Izpisua Belmonte JC. Navigating the epigenetic landscape of pluripotent stem cells. Nat Rev Mol Cell Biol 2012;13:524-35. https://doi.org/10.1038/nrm3393   DOI