Impact of Age on Human Adipose Stem Cells for Bone Tissue Engineering

Denis Dufrane, Denis Dufrane

Abstract

Bone nonunion is a pathological condition in which all bone healing processes have stopped, resulting in abnormal mobility between 2 bone segments. The incidence of bone-related injuries will increase in an aging population, leading to such injuries reaching epidemic proportions. Tissue engineering and cell therapy using mesenchymal stem cells (MSCs) have raised the possibility of implanting living tissue for bone reconstruction. Bone marrow was first proposed as the source of stem cells for bone regeneration. However, as the quantity of MSCs in the bone marrow decreases, the capacity of osteogenic differentiation of bone marrow stem cells is also impaired by the donor's age in terms of reduced MSC replicative capacity; an increased number of apoptotic cells; formation of colonies positive for alkaline phosphatase; and decreases in the availability, growth potential, and temporal mobilization of MSCs for bone formation in case of fracture. Adipose-derived stem cells (ASCs) demonstrate several advantages over those from bone marrow, including a less invasive harvesting procedure, a higher number of stem cell progenitors from an equivalent amount of tissue harvested, increased proliferation and differentiation capacities, and better angiogenic and osteogenic properties in vivo. Subcutaneous native adipose tissue was not affected by the donor's age in terms of cellular senescence and yield of ASC isolation. In addition, a constant mRNA level of osteocalcin and alkaline phosphatase with a similar level of matrix mineralization of ASCs remained unaffected by donor age after osteogenic differentiation. The secretome of ASCs was also unaffected by age when aiming to promote angiogenesis by vascular endothelial growth factor (VEGF) release in hypoxic conditions. Therefore, the use of adipose cells for bone tissue engineering is not limited by the donor's age from the isolation of stem cells up to the manufacturing of a complex osteogenic graft.

Keywords: adipose stem cells; aging; bone; functionality; mesenchymal stem cells.

Conflict of interest statement

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Figures

Fig. 1.
Fig. 1.
(A, B) Impact of age on adipose stem cells differentiation to obtain a 3-dimensional (3D) scaffold-free and oteogenic graft. (A) No impact of the donor’s age was found on the time required to isolate and expand adipose stem cells (from native adipose tissue) and to obtain finally differentiated to the scaffold-free 3D osteogenic graft. The integrity of the 3D graft was histomorphologically assessed at the end of manufacturing by the potency score (before implantation). A score of the graft integrity (for the optimal 3D graft) between −1 and +1 in terms of cell viability counting, interconnective tissue integrity, and demineralized bone matrix content. Note that a final 3D graft was always obtained (individual donors without any impact of donor age, which ranged from 5 and 70 y) for adipose-derived stem cells incubated with demineralized bone matrix, demonstrating the reproducibility of the manufacturing procedures.
Fig. 2.
Fig. 2.
Impact of donor’s age on the integrity of the scaffold-free 3-dimensional (3D) graft. Macroscopically, the 3D osteogenic graft was obtained for donors aged below 18 y old and older 60 y old (upper level). Microscopically, the collagen matrix, the cellularity with 4′,6-diamidino-2-phenylindole (DAPI) staining (middle level, original magnification: ×10), and the osteocalcin expression (lower level, original magnification: ×25) were maintained in the 3D graft for young and old donors of adipose tissue for adipose-derived stem cell isolation, proliferation, and differentiation.

References

    1. Rubin E. Essential pathology, 3rd ed Philadelphia (PA): Lippincott Williams and Wilkins; 2001.
    1. Einhorn TA. The science of fracture healing. J Orthop Trauma. 2005;19(suppl 10):S4–S6.
    1. Gerstenfeld LC, Cullinane DM, Barnes GL, Graves DT, Einhorn TA. Fracture healing as a post-natal developmental process: molecular, spatial, and temporal aspects of its regulation. J Cell Biochem. 2003;88(5):873–884.
    1. Kalfas IH. Principles of bone healing. Neurosurg Focus. 2001;10(4):E1.
    1. Meyer U, Weismann HP. Bone and cartilage In: Schroeder G, editor. Bone and cartilage engineering. New York (NY): Springer; 2006. p. 7–46.
    1. Miclau T, Schneider RA, Eames BF, Helms JA. Common molecular mechanisms regulating fetal bone formation and adult fracture repair In: Lieberman JR, Friedlaender GE, editors. Bone regeneration and repair: biology and clinical application. Totowa (NJ): Humana Press; 2005. p. 45–55.
    1. Sfeir C, Ho L, Doll BA, Azari K, Hollinger JO. Fracture repair In: Lieberman JR, Friedlaender GE, editors. Bone regeneration and repair: biology and clinical applications. Totowa (NJ): Humana Press; 2005. p. 21–44.
    1. Schindele A, McDonald MM, Bokko P, Little DG. Bone remodeling during fracture repair: the cellular picture. Semin Cell Dev Biol. 2008;19(5):459–466.
    1. Mackie EJ, Ahmed YA, Tatarczuch L, Chen KS, Mirams M. Endochondral ossification: how cartilage is converted into bone in the developing skeleton. Int J Biochem Cell Biol. 2008;40(1):46–62.
    1. Giannoudis PV, Einhorn TA, Marsh D. Fracture healing: the diamond concept. Injury. 2007;38(suppl 4):S3–S6.
    1. Megas P, Panagiotis M. Classification of non-union. Injury. 2005;36(suppl 4):S30–S37.
    1. Tzioupis C, Giannoudis PV. Prevalence of long-bone non-unions. Injury. 2007;38(suppl 2):S3–S9.
    1. Adams CI, Keating JF, Court-Brown CM. Cigarette smoking and open tibial fractures. Injury. 2001;32(1):61–65.
    1. Konttinen Y, Imai S, Suda A. Neuropeptides and the puzzle of bone remodeling: state of the art. Acta Orthop Scand. 1996;67(6):632–639.
    1. Rodriguez-Merchan EC, Forriol F. Nonunion: general principles and experimental data. Clin Orthop Relat Res. 2004;(419):4–12.
    1. Rosenwasser MP, Cuellar D. Medical management of osteoporosis and the surgeons’ role. Injury. 2016;47(suppl 1):S62–S64.
    1. Gibon E, Lu L, Goodman SB. Aging, inflammation, stem cells, and bone healing. Stem Cell Res Ther. 2016;7:44.
    1. Mills LA, Simpson AHRW. The relative incidence of fracture non-union in the Scottish population (5.17 million): a 5-year epidemiological study. BMJ Open. 2013;3(2):pii:e002276.
    1. Dahabreh Z, Calori GM, Kanakaris NK, Nikolaou VS, Giannoudis PV. A cost analysis of treatment of tibial fracture nonunion by bone grafting or bone morphogenetic protein-7. Int Orthop. 2009;33(5):1407–1414.
    1. Black CR, Goriainov V, Gibbs D, Kanczler J, Tare RS, Oreffo RO. Bone tissue engineering. Curr Mol Biol Rep. 2015;1(3):132–140.
    1. Friedenstein AY, Chailakhyan RK, Gerasimov UV. Bone marrow osteogenic stem cells: in vitro cultivation and transplantation in diffusion chambers. Cell Tissue Kinetics. 1987;20(3):263−272.
    1. Friedenstein AY, Chailakhyan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinetics. 1970;3(4):393−403.
    1. Dimitriou R, Eleftherios T, Giannoudis P. Current concepts of molecular aspects of bone healing. Injury. 2005;36(12):1392–1404.
    1. Quarto R, Thomas D, Liang CT. Bone progenitor cell deficits and the age-associated decline in bone repair capacity. Calcif Tissue Intl. 1995;56(2):123–129.
    1. Chen TL. Inhibition of growth and differentiation of osteoprogenitors in mouse bone marrow stromal cell cultures by increased donor age and glucocorticoid treatment. Bone. 2004;35(1):83–95.
    1. Shigeno Y, Ashton BA. Human bone-cell proliferation in vitro decreases with human donor age. J Bone Joint Surg (Br). 1995;77(1):139–142.
    1. Muschler GF, Nitto H, Boehm CA, Easley KA. Age- and gender-related changes in the cellularity of human bone marrow and the prevalence of osteoblastic progenitors. J Orthop Res. 2001;19(1):117–125.
    1. 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(5):675–682.
    1. 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(3):335–343.
    1. 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(3):163–173.
    1. Schecroun N, Delloye C. In vitro growth and osteoblastic differentiation of human bone marrow stromal cells supported by autologous plasma. Bone. 2004;35(2):517–524.
    1. Liebergall M, Schroeder J, Mosheiff R, Gazit Z, Yoram Z, Rasooly L, Daskal A, Khoury A, Weil Y, Beyth S. Stem cell-based therapy for prevention of delayed fracture union: a randomized and prospective preliminary study. Mol Ther. 2013;21(8):1631–1638.
    1. Weel H, Mallee WH, van Dijk CN, Blankevoort L, Goedegebuure S, Goslings JC, Kennedy JG, Kerkhoffs GM. The effect of concentrated bone marrow aspirate in operative treatment of fifth metatarsal stress fractures; a double-blind randomized controlled trial. BMC Musculoskelet Disord. 2015;16:211.
    1. Zuk PA, Zhu M, Mizuno H, Huan J, Futrell JW, Katz AJ, Benhaim P, Lorenz HP, Hedrick MH. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7(2):211–228.
    1. 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(12):4279–4295.
    1. Zuk PA. The adipose-derived stem cell: looking back and looking ahead. Mol Biol Cell. 2010;21(11):1783–1787.
    1. Gimble JM, Katz AJ, Bunnell BA. Adipose-derived stem cells for regenerative medicine. Circ Res. 2007;100(9):1249–1260.
    1. Kuhbier JW, Weyand B, Radtke C, Vogt PM, Kasper C, Reimers K. Isolation, characterization, differentiation, and application of adipose-derived stem cells. Adv Biochem Eng Biotechnol. 2010;123:55–105.
    1. Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE, Pell CL, Johnstone BH, Considine RV, March KL. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation. 2004;109(10):1292–1298.
    1. Fraser JK, Wulur I, Alfonso Z, Hedrick MH. Fat tissue: an underappreciated source of stem cells for biotechnology. Trends Biotechnol. 2006;24(4):150–154.
    1. Liao HT, Chen CT. Osteogenic potential: comparison between bone marrow and adipose-derived mesenchymal stem cells. World J Stem Cells. 2014;6(3):288–295.
    1. Barba M, Cicione C, Bernardini C, Michetti F, Lattanzi W. Adipose-derived mesenchymal cells for bone regeneration: state of the art. Biomed Res Intl. 2013;2013:416391.
    1. Schubert T, Xhema D, Vériter S, Schubert M, Behets C, Delloye C, Gianello P, Dufrane D. The enhanced performance of bone allografts using osteogenic-differentiated adipose-derived mesenchymal stem cells. Biomaterials. 2011;32(34):8880–8891.
    1. Schubert T, Lafont S, Beaurin G, Grisay G, Behets C, Gianello P, Dufrane D. Critical size bone defect reconstruction by an autologous 3D osteogenic-like tissue derived from differentiated adipose MSCs. Biomaterials. 2013;34(18):4428–4438.
    1. Schubert T, Poilvache H, Galli C, Gianello P, Dufrane D. Galactosyl-knock-out engineered pig as a xenogenic donor source of adipose MSCs for bone regeneration. Biomaterials. 2013;34(13):3279–3289.
    1. Yoshimura K, Suga H, Eto H. Adipose-derived stem/progenitor cells: roles in adipose tissue remodeling and potential use for soft tissue augmentation. Regenerative Med. 2009;4(2):265–273.
    1. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW., Jr Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112(12):1796–1808.
    1. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112(12):1821–1830.
    1. Prunet-Marcassus B, Cousin B, Caton D, André M, Pénicaud L, Casteilla L. From heterogeneity to plasticity in adipose tissues: site-specific differences. Exp Cell Res. 2006;312(6):727–736.
    1. Yang XF, He X, He J, Zhang LH, Su XJ, Dong ZY, Xu YJ, Li Y, Li YL. High efficient isolation and systematic identification of human adipose-derived mesenchymal stem cells. J Biomed Sci. 2011;18:59.
    1. Minamino T, Orimo M, Shimizu I, Kunieda T, Yokoyama M, Ito T, Nojima A, Nabetani A, Oike Y, Matsubara H, et al. A crucial role for adipose tissue p53 in the regulation of insulin resistance. Nat Med. 2009;15(9):1082–1087.
    1. Shimizu I, Yoshida Y, Suda M, Minamino T. DNA damage response and metabolic disease. Cell Metab. 2014;20(6):967–977.
    1. Wu W, Niklason L, Steinbacher DM. The effect of age on human adipose-derived stem cells. Plast Reconstr Surg. 2013;131(1):27–37.
    1. Kornicka K, Marycz K, Tomaszewski KA, Marędziak M, Śmieszek A. The effect of age on osteogenic and adipogenic differentiation potential of human adipose derived stromal stem cells (hASCs) and the impact of stress factors in the course of the differentiation process. Oxid Med Cell Longev. 2015;2015:309169.
    1. Choudhery MS, Badowski M, Muise A, Pierce J, Harris DT. Donor age negatively impacts adipose tissue-derived mesenchymal stem cell expansion and differentiation. J Transl Med. 2014;12:8.
    1. Dufrane D, Docquier PL, Delloye C, Poirel HA, André W, Aouassar N. Scaffold-free three-dimensional graft from autologous adipose-derived stem cells for large bone defect reconstruction: clinical proof of concept. Medicine (Baltimore). 2015;94(50):e2220.
    1. Vériter S, André W, Aouassar N, Poirel HA, Lafosse A, Docquier PL, Dufrane D. Human adipose-derived mesenchymal stem cells in cell therapy: safety and feasibility in different “hospital exemption” clinical applications. PLoS One. 2015;10(10):e0139566.
    1. Prockop DJ, Brenner M, Fibbe WE, Horwitz E, Le Blanc K, Phinney DG, Simmons PJ, Sensebe L, Keating A. Defining the risks of mesenchymal stromal cell therapy. Cytotherapy. 2010;12(5):576–578.
    1. Bernardo ME, Zaffaroni N, Novara F, Cometa AM, Avanzini MA, Moretta A, Montagna D, Maccario R, Villa R, Daidone MG, et al. 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(19):9142–9149.
    1. Meza-Zepeda LA, Noer A, Dahl JA, Micci F, Myklebost O, Collas P. High-resolution analysis of genetic stability of human adipose tissue stem cells cultured to senescence. J Cell Mol Med. 2008;12(2):553–563.
    1. Chen HT, Lee MJ, Chen CH, Chuang SC, Chang LF, Ho ML, Hung SH, Fu YC, Wang YH, Wang HI, et al. Proliferation and differentiation potential of human adipose-derived mesenchymal stem cells isolated from elderly patients with osteoporotic fractures. J Cell Mol Med. 2012;16(3):582–593.
    1. Lafosse A, Desmet C, Aouassar N, André W, Hanet MS, Beauloye C, Vanwijck R, Poirel HA, Gallez B, Dufrane D. Autologous adipose stromal cells seeded onto a human collagen matrix for dermal regeneration in chronic wounds: clinical proof of concept. Plast Reconstr Surg. 2015;136(2):279–295.
    1. Tarte K, Gaillard J, Lataillade JJ, Fouillard L, Becker M, Mossafa H, Tchirkov A, Rouard H, Henry C, Splingard M, et al. . Clinical-grade production of human mesenchymal stromal cells: occurrence of aneuploidy without transformation. Blood. 2010;115(8):1549–1553.
    1. Trobaugh-Lotrario AD, Kletzel M, Quinones RR, McGavran L, Proytcheva MA, Hunger SP, Malcolm J, Schissel D, Hild E, Giller RH. Monosomy 7 associated with pediatric acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS): successful management by allogeneic hematopoietic stem cell transplant (HSCT). Bone Marrow Transplant. 2005;35(2):143–149.
    1. Nikitina VA, Osipova EY, Katosova LD, Rumyantsev SA, Skorobogatova EV, Shamanskaya TV, Bochkov NP. Study of genetic stability of human bone marrow multipotent mesenchymal stromal cells. Bull Exp Biol Med. 2011;150(5):627–631.
    1. Bochkov NP, Voronina ES, Kosyakova NV, Liehr T, Rzhaninova AA, Katosova LD, Platonova VI, Gol’dshtein DV. Chromosome variability of human multipotent mesenchymal stromal cells. Bull Exp Biol Med. 2007;143(1):122–126.
    1. Haniffa MA, Collin MP, Buckley CD, Dazzi F. Mesenchymal stem cells: the fibroblasts’ new clothes? Haematologica. 2009;94(2):258–263.
    1. Ho AD, Wagner Franke W. Heterogeneity of mesenchymal stromal cell preparations. Cytotherapy. 2008;10(4):320–330.
    1. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143–147.
    1. Bourin P, Bunnell BA, Casteilla L, Dominici M, Katz AJ, March KL, Redl H, Rubin JP, Yoshimura K, Gimble JM. Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: a joint statement of the International Federation for Adipose Therapeutics and Science (IFATS) and the International Society for Cellular Therapy (ISCT). Cytotherapy. 2013;15(6):641–648.
    1. Hematti P. Mesenchymal stromal cells and fibroblasts: a case of mistaken identity? Cytotherapy. 2012;14(5):516–521.
    1. Wagner W, Ho AD, Zenke M. Different facets of aging in human mesenchymal stem cells. Tissue Eng Part B Rev. 2010;16(4):445–453.
    1. Dufrane D, Lafosse A. A simple method to determine the purity of adipose-derived stem cell-based cell therapies. Stem Cells Transl Med. 2016;5(11):1575–1579.
    1. Backly RM, Cancedda R. Bone marrow stem cells in clinical application: harnessing paracrine roles and niche mechanisms. Adv Biochem Eng Biotechnol. 2010;123:265–292.
    1. Salgado AJ, Reis RL, Sousa NJ, Gimble JM. Adipose tissue derived stem cells secretome: soluble factors and their roles in regenerative medicine. Curr Stem Cell Res Ther. 2010;5(2):103–110.
    1. Nakanishi C, Nagaya N, Ohnishi S, Yamahara K, Takabatake S, Konno T, Hayashi K, Kawashiri MA, Tsubokawa T, Yamagishi M. Gene and protein expression analysis of mesenchymal stem cells derived from rat adipose tissue and bone marrow. Circ J. 2011;75(9):2260–2268.
    1. Lee K, Kim H, Kim JM, Kim JR, Kim KJ, Kim YJ, Park SI, Jeong JH, Moon YM, Lim HS, et al. Systemic transplantation of human adipose-derived stem cells stimulates bone repair by promoting osteoblast and osteoclast function. J Cell Mol Med. 2011;15(10):2082–2094.
    1. Langenbach F, Naujoks C, Smeets R, Berr K, Depprich R, Kübler N, Handschel J. Scaffold-free microtissues: differences from monolayer cultures and their potential in bone tissue engineering. Clin Oral Investig. 2013;17(1):9–17.

Source: PubMed

3
Tilaa