J Vet Sci.  2017 Jun;18(2):141-148. 10.4142/jvs.2017.18.2.141.

Effect of donor age on the proliferation and multipotency of canine adipose-derived mesenchymal stem cells

Affiliations
  • 1Viral Disease Research Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea. virusmania@korea.kr

Abstract

Research into adipose tissue-derived mesenchymal stem cells (AD-MSCs) has demonstrated the feasibility of their use in clinical applications due to their ease of isolation and abundance in adipose tissue. We isolated AD-MSCs from young and old dogs, and the cells were subjected to sequential sub-passaging from passage 1 (P1) to P7. Canine AD-MSCs (cAD-MSCs) were examined for proliferation kinetics, expression of molecules associated with self-renewal, expression of cell surface markers, and differentiation potentials at P3. Cumulative population doubling level was significantly higher in cAD-MSCs of young donors than in those of old donors. In addition, expressions of CD73, CD80, Oct3/4, Nanog, cell survival genes and differentiation potentials were significantly higher in young donors than in old donors. The present study suggests that donor age should be considered when developing cell-based therapies for clinical application of cAD-MSCs.

Keyword

adipose mesenchymal stem cells; age; canine; differentiation; multipotency

MeSH Terms

Adipose Tissue/*cytology/physiology
Age Factors
Animals
Cell Differentiation/physiology
Cell Proliferation/*physiology
Dogs
Gene Expression Profiling/veterinary
Mesenchymal Stromal Cells/*physiology
Real-Time Polymerase Chain Reaction/veterinary
Reverse Transcriptase Polymerase Chain Reaction/veterinary

Figure

  • Fig. 1 Morphology and proliferation of canine adipose tissue-derived mesenchymal stem cells (cAD-MSCs). (A) Morphology of cells passage 1–6 (P1–P6) derived from adipose tissues typically appeared as fibroblast-like. (B) Cumulative population doubling level (CPDL) of cAD-MSCs during continuous passages. CPDL of the cAD-MSCs increased at each passage until P3. Cells were grown in two age groups, YAD-MSCs and OAD-MSCs. GAPDH was used as a housekeeping control gene. The results are shown as the mean ± standard error of the mean (n = 5) obtained by three determinations. YAD, 7-month-old dogs; OAD, 10- to 11-year-old dogs. 100× (A).

  • Fig. 2 Expression pluripotency markers (Oct3/4, Sox2, and Nanog) of canine adipose tissue-derived mesenchymal stem cells (cAD-MSCs). Using reverse transcriptase polymerase chain reaction (RT-PCR; A) and real-time quantitative RT-PCR (qRT-PCR; B), the expressions of stemness genes of cAD-MSCs were examined in two age groups (young and old) at passage 3. GAPDH was used as a housekeeping control gene. All mRNA data were normalized to the GAPDH levels, and the relative fold change in expression level is shown as a mean ± SEM (n = 3). *p < 0.05. YAD, 7-month-old dogs; OAD, 10- to 11-year-old dogs.

  • Fig. 3 Surface markers of canine adipose tissue-derived mesenchymal stem cells (cAD-MSCs). MSC-specific cell surface markers (positive: CD44, CD54, CD61, CD73, CD80, CD90, and CD105; not detectable: CD29, CD34, CD117, and MHC-class II) of cAD-MSCs were observed by real-time quantitative reverse transcriptase polymerase chain reaction in two age groups (young and old) at passage 3. GAPDH was used as a housekeeping control gene. All mRNA data were normalized to the levels of GAPDH, and relative fold changes in expression levels are shown as the mean ± SEM (n = 3). *p < 0.05. YAD, 7-month-old dogs; OAD, 10- to 11-year-old dogs.

  • Fig. 4 Differentiation potentials of canine adipose tissue-derived mesenchymal stem cells (cAD-MSCs). Adipocytes (Oil Red O; A), osteocytes (Alizarin Red; B), and chondrocytes (Alcian Blue; C) were positively stained in two age groups (young and old) at passage 3. (D) The mRNA expressions of adipocyte, osteocyte, and chondrocyte-related genes were detected by real-time quantitative reverse transcriptase polymerase chain reaction and compared between undifferentiated and differentiated cells in the two groups at passage 3. All mRNA data were normalized to the levels of GAPDH, and relative fold changes in expression levels are shown as the mean ± SEM (n = 3). *p < 0.05. 100× (A–C). YAD, 7-month-old dogs; OAD, 10- to 11-year-old dogs; LPL, lipoprotein lipase; OP, osteopontin; OC, osteocalcin; COL2A, type II-collagen.

  • Fig. 5 Cell survival-related genes of canine adipose tissue-derived mesenchymal stem cells (cAD-MSCs). The mRNA expressions of cell survival-related genes such as TERT, DKC1, HDAC1, DNMT1, Bcl-2, LDHA, and SLC2A1 were examined by real-time quantitative reverse transcriptase polymerase chain reaction in two groups (young and old) at passage 3. GAPDH was used as a housekeeping control gene. All mRNA data were normalized to the levels of GAPDH, and relative fold changes in expression levels are shown as the mean ± SEM (n = 3). *p < 0.05. YAD, 7-month-old dogs; OAD, 10- to 11-year-old dogs; TERT, telomerase reverse transcriptase; DKC1, dyskerin pseudouridine synthase 1; HDAC1, histone deacetylase 1; DNMT1, DNA (cytosine-5)-methyltransferase 1; LDHA, lactate dehydrogenase A; SLC2A1, glucose transporter member 1.


Reference

1. Alt EU, Senst C, Murthy SN, Slakey DP, Dupin CL, Chaffin AE, Kadowitz PJ, Izadpanah R. Aging alters tissue resident mesenchymal stem cells properties. Stem Cell Res. 2012; 8:215–225.
2. Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells. 2007; 25:2739–2749.
Article
3. Choudhery MS, Khan M, Mahmood R, Mehmood A, Khan SN, Riazuddin S. Bone marrow derived mesenchymal stem cells from aged mice have reduced wound healing, angiogenesis, proliferation and anti-apoptosis capabilities. Cell Biol Int. 2012; 36:747–753.
Article
4. Cristofalo VJ, Allen RG, Pignolo RJ, Martin BG, Beck JC. Relationship between donor age and the replicative lifespan of human cells in culture: a reevaluation. Proc Natl Acad Sci U S A. 1998; 95:10614–10619.
Article
5. Feng X, Xing J, Feng G, Sang A, Shen B, Xu Y, Jiang J, Liu S, Tan W, Gu Z, Li L. Age dependent impaired neurogenic differentiation capacity of dental stem cell is associated with Wnt/XMLLink_XYZ-catenin signaling. Cell Mol Neurobiol. 2013; 33:1023–1031.
Article
6. Fraser JK, Wulur I, Alfonso Z, Hedrick MH. Fat tissue: an underappreciated source of stem cells for biotechnology. Trends Biotechnol. 2006; 24:150–154.
Article
7. Gimble J, Guilak F. Adipose-derived adult stem cells: isolation, characterization, and differentiation potential. Cytotheraphy. 2003; 5:362–369.
Article
8. Guercio A, Di Bella S, Casella S, Di Marco P, Russo C, Piccione G. Canine mesenchymal stem cells (MSCs): characterization in relation to donor age and adipose tissue-harvesting site. Cell Biol Int. 2013; 37:789–798.
Article
9. Iohara K, Murakami M, Nakata K, Nakashima M. Age-dependent decline in dental pulp regeneration after pulpectomy in dogs. Exp Gerontol. 2014; 52:39–45.
Article
10. Itahana Y, Han R, Barbier S, Lei Z, Rozen S, Itahana K. The uric acid transporter SLC2A9 is a direct target gene of the tumor suppressor p53 contributing to antioxidant defense. Oncogene. 2015; 34:1799–1810.
Article
11. Kanawa M, Igarashi A, Ronald VS, Higashi Y, Kurihara H, Sugiyama M, Saskianti T, Pan H, Kato Y. Age-dependent decrease in the chondrogenic potential of human bone marrow mesenchymal stromal cells expanded with fibroblast growth factor-2. Cytotherapy. 2013; 15:1062–1072.
Article
12. Kretlow JD, Jin YQ, Liu W, Zhang WJ, Hong TH, Zhou G, Baggett LS, Mikos AG, Cao Y. Donor age and cell passage affects differentiation potential of murine bone marrow-derived stem cells. BMC Cell Biol. 2008; 9:60.
Article
13. Lee KS, Cha SH, Kang HW, Song JY, Lee KW, Ko KB, Lee HT. Effects of serial passage on the characteristics and chondrogenic differentiation of canine umbilical cord matrix derived mesenchymal stem cells. Asian-Australas J Anim Sc. 2013; 26:588–595.
Article
14. Lee KS, Kang HW, Lee HT, Kim HJ, Kim CL, Song JY, Lee KW, Cha SH. Sequential sub-passage decreases the differentiation potential of canine adipose-derived mesenchymal stem cells. Res Vet Sci. 2014; 96:267–275.
Article
15. Lee KS, Nah JJ, Lee BC, Lee HT, Lee HS, So BJ, Cha SH. Maintenance and characterization of multipotent mesenchymal stem cells isolated from canine umbilical cord matrix by collagenase digestion. Res Vet Sci. 2013; 94:144–151.
Article
16. Li Q, Qi LJ, Guo ZK, Li H, Zuo HB, Li NN. CD73+ adipose-derived mesenchymal stem cells possess higher potential to differentiate into cardiomyocytes in vitro. J Mol Histol. 2013; 44:411–422.
Article
17. Li Y, Charif N, Mainard D, Bensoussan D, Stoltz JF, de Isla N. Donor's age dependent proliferation decrease of human bone marrow mesenchymal stem cells is linked to diminished clonogenicity. Biomed Mater Eng. 2014; 24:Suppl 1. 47–52.
Article
18. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT Method. Methods. 2001; 25:402–408.
Article
19. Mizuno H. Adipose-derived stem cells for tissue repair and regeneration: ten years of research and a literature review. J Nippon Med Sch. 2009; 76:55–66.
Article
20. Neupane M, Chang CC, Kiupel M, Yuzbasiyan-Gurkan V. Isolation and characterization of canine adipose-derived mesenchymal stem cells. Tissue Eng Part A. 2008; 14:1007–1015.
Article
21. Ode A, Kopf J, Kurtz A, Schmidt-Bleek K, Schrade P, Kolar P, Buttgereit F, Lehmann K, Hutmacher DW, Duda GN, Kasper G. CD73 and CD29 concurrently mediate the mechanically induced decrease of migratory capacity of mesenchymal stromal cells. Eur Cell Mater. 2011; 6:26–42.
Article
22. Oh YS, Jeong SG, Cho GW. Anti-senescence effects of DNA methyltransferase inhibitor RG108 in human bone marrow mesenchymal stromal cells. Biotechnol Appl Biochem. 2015; 62:583–590.
Article
23. Ohsaki H, Sawa T, Sasazaki S, Kano K, Taniguchi M, Mukai F, Mannen H. Stearoyl-CoA desaturase mRNA expression during bovine adipocyte differentiation in primary culture derived from Japanese Black and Holstein cattle. Comp Biochem Physiol A Mol Integr Physiol. 2007; 148:629–634.
Article
24. Paxson JA, Gruntman AM, Davis AM, Parkin CM, Ingenito EP, Hoffman AM. Age dependence of lung mesenchymal stromal cell dynamics following pneumonectomy. Stem Cells Dev. 2013; 22:3214–3225.
Article
25. Requicha JF, Viegas CA, Albuquerque CM, Azevedo JM, Reis RL, Gomes ME. Effect of anatomical origin and cell passage number on the stemness and osteogenic differentiation potential of canine adipose-derived stem cells. Stem Cell Rev. 2012; 8:1211–1222.
Article
26. 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; 129:163–173.
Article
27. Takemitsu H, Zhao D, Yamamoto I, Harada Y, Michishita M, Arai T. Comparison of bone marrow and adipose tissue-derived canine mesenchymal stem cells. BMC Vet Res. 2012; 8:150.
Article
28. Vaags AK, Rosic-Kablar S, Gartley CJ, Zheng YZ, Chesney A, Villagómez DAF, Kruth SA, Hough MR. Derivation and characterization of canine embryonic stem cell lines with in vitro and in vivo differentiation potential. Stem Cells. 2009; 27:329–340.
Article
29. Vieira NM, Brandalise V, Zucconi E, Secco M, Strauss BE, Zatz M. Isolation, characterization, and differentiation potential of canine adipose-derived stem cells. Cell Transplant. 2010; 19:279–289.
Article
30. Wagner W, Bork S, Horn P, Krunic D, Walenda T, Diehlmann A, Benes V, Blake J, Huber FX, Eckstein V, Boukamp P, Ho AD. Aging and replicative senescence have related effects on human stem and progenitor cells. PLoS One. 2009; 4:e5846.
Article
31. Wang YH, Israelsen WJ, Lee D, Yu VWC, Jeanson NT, Clish CB, Cantley LC, Vander Heiden MG, Scadden DT. Cell-state-specific metabolic dependency in hematopoiesis and leukemogenesis. Cell. 2014; 158:1309–1323.
Article
32. Wenceslau CV, Miglino MA, Martins DS, Ambrósio CE, Lizier NF, Pignatari GC, Kerkis I. Mesenchymal progenitor cells from canine fetal tissues: yolk sac, liver, and bone marrow. Tissue Eng Part A. 2011; 17:2165–2176.
Article
33. Yang CC, Ellis SE, Xu F, Burg KJL. In vitro regulation of adipogenesis: tunable engineered tissues. J Tissue Eng Regen Med. 2007; 1:146–153.
Article
34. Zheng B, Cao B, Li G, Huard J. Mouse adipose-derived stem cells undergo multilineage differentiation in vitro but primarily osteogenic and chondrogenic differentiation in vivo. Tissue Eng. 2006; 12:1891–1901.
Article
35. Zhou S, Greenberger JS, Epperly MW, Goff JP, Adler C, Leboff MS, Glowacki J. Age-related intrinsic changes in human bone-marrow-derived mesenchymal stem cells and their differentiation to osteoblasts. Aging Cell. 2008; 7:335–343.
Article
36. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002; 13:4279–4295.
Article
Full Text Links
  • JVS
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr