Clin Orthop Surg.  2018 Sep;10(3):271-278. 10.4055/cios.2018.10.3.271.

Mesenchymal Stem Cell Therapy for Bone Regeneration

Affiliations
  • 1Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Korea. spinelee@snu.ac.kr
  • 2Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul, Korea.
  • 3Institute of Medical and Biological Engineering, Seoul National University Medical Research Center, Seoul, Korea.

Abstract

Mesenchymal stem cells (MSCs) have been used in clinic for approximately 20 years. During this period, various new populations of MSCs have been found or manipulated. However, their characters and relative strength for bone regeneration have not been well known. For a comprehensive understanding of MSCs, we reviewed the literature on the multipotent cells ranging from the definition to the current research progress for bone regeneration. Based on our literature review, bone marrow MSCs have been most widely studied and utilized in clinical settings. Among other populations of MSCs, adipose-derived MSCs and perivascular MSCs might be potential candidates for bone regeneration, whose efficacy and safety still require further investigation.

Keyword

Stem cells; Osteogenesis; Bone diseases; Review

MeSH Terms

Bone Diseases
Bone Marrow
Bone Regeneration*
Mesenchymal Stromal Cells*
Osteogenesis
Stem Cells

Cited by  2 articles

Clinical Use of Mesenchymal Stem Cells in Bone Regeneration
Chan-Woo Park, Seung-Jae Lim, Youn-Soo Park
J Korean Orthop Assoc. 2019;54(6):490-497.    doi: 10.4055/jkoa.2019.54.6.490.

Current perspectives in stem cell therapies for osteoarthritis of the knee
Gi Beom Kim, Oog-Jin Shon
Yeungnam Univ J Med. 2020;37(3):149-158.    doi: 10.12701/yujm.2020.00157.


Reference

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(4):315–317.
Article
2. Hernigou P, Bernaudin F, Reinert P, Kuentz M, Vernant JP. Bone-marrow transplantation in sickle-cell disease: effect on osteonecrosis: a case report with a four-year follow-up. J Bone Joint Surg Am. 1997; 79(11):1726–1730.
3. Lin CY, Lin KJ, Kao CY, et al. The role of adipose-derived stem cells engineered with the persistently expressing hybrid baculovirus in the healing of massive bone defects. Biomaterials. 2011; 32(27):6505–6514.
Article
4. Crisan M, Corselli M, Chen WC, Peault B. Perivascular cells for regenerative medicine. J Cell Mol Med. 2012; 16(12):2851–2860.
Article
5. Klontzas ME, Kenanidis EI, Heliotis M, Tsiridis E, Mantalaris A. Bone and cartilage regeneration with the use of umbilical cord mesenchymal stem cells. Expert Opin Biol Ther. 2015; 15(11):1541–1552.
Article
6. Csobonyeiova M, Polak S, Zamborsky R, Danisovic L. iPS cell technologies and their prospect for bone regeneration and disease modeling: a mini review. J Adv Res. 2017; 8(4):321–327.
Article
7. Park JS, Suryaprakash S, Lao YH, Leong KW. Engineering mesenchymal stem cells for regenerative medicine and drug delivery. Methods. 2015; 84:3–16.
Article
8. Asatrian G, Pham D, Hardy WR, James AW, Peault B. Stem cell technology for bone regeneration: current status and potential applications. Stem Cells Cloning. 2015; 8:39–48.
9. Horwitz EM, Prockop DJ, Fitzpatrick LA, et al. Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat Med. 1999; 5(3):309–313.
Article
10. Horwitz EM, Prockop DJ, Gordon PL, et al. Clinical responses to bone marrow transplantation in children with severe osteogenesis imperfecta. Blood. 2001; 97(5):1227–1231.
Article
11. Horwitz EM, Gordon PL, Koo WK, et al. Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: implications for cell therapy of bone. Proc Natl Acad Sci U S A. 2002; 99(13):8932–8937.
Article
12. Ajiboye RM, Eckardt MA, Hamamoto JT, Plotkin B, Daubs MD, Wang JC. Outcomes of demineralized bone matrix enriched with concentrated bone marrow aspirate in lumbar fusion. Int J Spine Surg. 2016; 10:35.
Article
13. Neen D, Noyes D, Shaw M, Gwilym S, Fairlie N, Birch N. Healos and bone marrow aspirate used for lumbar spine fusion: a case controlled study comparing healos with autograft. Spine (Phila Pa 1976). 2006; 31(18):E636–E640.
14. Connolly JF, Guse R, Tiedeman J, Dehne R. Autologous marrow injection as a substitute for operative grafting of tibial nonunions. Clin Orthop Relat Res. 1991; (266):259–270.
Article
15. Garg NK, Gaur S, Sharma S. Percutaneous autogenous bone marrow grafting in 20 cases of ununited fracture. Acta Orthop Scand. 1993; 64(6):671–672.
Article
16. Goel A, Sangwan SS, Siwach RC, Ali AM. Percutaneous bone marrow grafting for the treatment of tibial non-union. Injury. 2005; 36(1):203–206.
Article
17. Ismail HD, Phedy P, Kholinne E, et al. Mesenchymal stem cell implantation in atrophic nonunion of the long bones: a translational study. Bone Joint Res. 2016; 5(7):287–293.
Article
18. Gangji V, Hauzeur JP, Matos C, De Maertelaer V, Toungouz M, Lambermont M. Treatment of osteonecrosis of the femoral head with implantation of autologous bone-marrow cells: a pilot study. J Bone Joint Surg Am. 2004; 86(6):1153–1160.
Article
19. Gangji V, De Maertelaer V, Hauzeur JP. Autologous bone marrow cell implantation in the treatment of non-traumatic osteonecrosis of the femoral head: five year follow-up of a prospective controlled study. Bone. 2011; 49(5):1005–1009.
Article
20. Mao Q, Jin H, Liao F, Xiao L, Chen D, Tong P. The efficacy of targeted intraarterial delivery of concentrated autologous bone marrow containing mononuclear cells in the treatment of osteonecrosis of the femoral head: a five year follow-up study. Bone. 2013; 57(2):509–516.
Article
21. Hernigou P, Trousselier M, Roubineau F, et al. Stem cell therapy for the treatment of hip osteonecrosis: a 30-year review of progress. Clin Orthop Surg. 2016; 8(1):1–8.
Article
22. Hernigou P, Flouzat-Lachaniette CH, Delambre J, et al. Osteonecrosis repair with bone marrow cell therapies: state of the clinical art. Bone. 2015; 70:102–109.
Article
23. Kawate K, Yajima H, Ohgushi H, et al. Tissue-engineered approach for the treatment of steroid-induced osteonecrosis of the femoral head: transplantation of autologous mesenchymal stem cells cultured with beta-tricalcium phosphate ceramics and free vascularized fibula. Artif Organs. 2006; 30(12):960–962.
Article
24. Sen RK, Tripathy SK, Aggarwal S, Marwaha N, Sharma RR, Khandelwal N. Early results of core decompression and autologous bone marrow mononuclear cells instillation in femoral head osteonecrosis: a randomized control study. J Arthroplasty. 2012; 27(5):679–686.
Article
25. Ma Y, Wang T, Liao J, et al. Efficacy of autologous bone marrow buffy coat grafting combined with core decompression in patients with avascular necrosis of femoral head: a prospective, double-blinded, randomized, controlled study. Stem Cell Res Ther. 2014; 5(5):115.
Article
26. Hernigou P, Poignard A, Beaujean F, Rouard H. Percutaneous autologous bone-marrow grafting for nonunions: influence of the number and concentration of progenitor cells. J Bone Joint Surg Am. 2005; 87(7):1430–1437.
Article
27. Griffin M, Iqbal SA, Bayat A. Exploring the application of mesenchymal stem cells in bone repair and regeneration. J Bone Joint Surg Br. 2011; 93(4):427–434.
Article
28. Gimble JM, Katz AJ, Bunnell BA. Adipose-derived stem cells for regenerative medicine. Circ Res. 2007; 100(9):1249–1260.
Article
29. Baer PC, Geiger H. Adipose-derived mesenchymal stromal/ stem cells: tissue localization, characterization, and heterogeneity. Stem Cells Int. 2012; 2012:812693.
30. Baer PC. Adipose-derived mesenchymal stromal/stem cells: an update on their phenotype in vivo and in vitro. World J Stem Cells. 2014; 6(3):256–265.
Article
31. Cai X, Lin Y, Hauschka PV, Grottkau BE. Adipose stem cells originate from perivascular cells. Biol Cell. 2011; 103(9):435–447.
Article
32. Maumus M, Peyrafitte JA, D'Angelo R, et al. Native human adipose stromal cells: localization, morphology and phenotype. Int J Obes (Lond). 2011; 35(9):1141–1153.
Article
33. Cowan CM, Shi YY, Aalami OO, et al. Adipose-derived adult stromal cells heal critical-size mouse calvarial defects. Nat Biotechnol. 2004; 22(5):560–567.
Article
34. Levi B, James AW, Nelson ER, et al. Human adipose derived stromal cells heal critical size mouse calvarial defects. PLoS One. 2010; 5(6):e11177.
Article
35. Cui L, Liu B, Liu G, et al. Repair of cranial bone defects with adipose derived stem cells and coral scaffold in a canine model. Biomaterials. 2007; 28(36):5477–5486.
Article
36. Abudusaimi A, Aihemaitijiang Y, Wang YH, Cui L, Maimaitiming S, Abulikemu M. Adipose-derived stem cells enhance bone regeneration in vascular necrosis of the femoral head in the rabbit. J Int Med Res. 2011; 39(5):1852–1860.
Article
37. Peterson B, Zhang J, Iglesias R, et al. Healing of critically sized femoral defects, using genetically modified mesenchymal stem cells from human adipose tissue. Tissue Eng. 2005; 11(1-2):120–129.
Article
38. Sunay O, Can G, Cakir Z, et al. Autologous rabbit adipose tissue-derived mesenchymal stromal cells for the treatment of bone injuries with distraction osteogenesis. Cytotherapy. 2013; 15(6):690–702.
Article
39. Hsu WK, Wang JC, Liu NQ, et al. Stem cells from human fat as cellular delivery vehicles in an athymic rat posterolateral spine fusion model. J Bone Joint Surg Am. 2008; 90(5):1043–1052.
Article
40. Liu G, Zhang Y, Liu B, Sun J, Li W, Cui L. Bone regeneration in a canine cranial model using allogeneic adipose derived stem cells and coral scaffold. Biomaterials. 2013; 34(11):2655–2664.
Article
41. Sandor GK, Numminen J, Wolff J, et al. Adipose stem cells used to reconstruct 13 cases with cranio-maxillofacial hardtissue defects. Stem Cells Transl Med. 2014; 3(4):530–540.
Article
42. Prins HJ, Schulten EA, Ten Bruggenkate CM, Klein-Nulend J, Helder MN. Bone regeneration using the freshly isolated autologous stromal vascular fraction of adipose tissue in combination with calcium phosphate ceramics. Stem Cells Transl Med. 2016; 5(10):1362–1374.
Article
43. Bui KH, Duong TD, Nguyen NT, et al. Symptomatic knee osteoarthritis treatment using autologous adipose derived stem cells and platelet-rich plasma: a clinical study. Biomed Res Ther. 2014; 1(1):2–8.
Article
44. Pers YM, Rackwitz L, Ferreira R, et al. Adipose mesenchymal stromal cell-based therapy for severe osteoarthritis of the knee: a phase I dose-escalation trial. Stem Cells Transl Med. 2016; 5(7):847–856.
Article
45. Dufrane D, Docquier PL, Delloye C, Poirel HA, Andre 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.
46. Yousefi AM, James PF, Akbarzadeh R, Subramanian A, Flavin C, Oudadesse H. Prospect of stem cells in bone tissue engineering: a review. Stem Cells Int. 2016; 2016:6180487.
Article
47. 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(4):1211–1222.
Article
48. Bellotti C, Stanco D, Ragazzini S, et al. Analysis of the karyotype of expanded human adipose-derived stem cells for bone reconstruction of the maxillo-facial region. Int J Immunopathol Pharmacol. 2013; 26:1 Suppl. 3–9.
Article
49. 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.
Article
50. 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.
Article
51. Corselli M, Crisan M, Murray IR, et al. Identification of perivascular mesenchymal stromal/stem cells by flow cytometry. Cytometry A. 2013; 83(8):714–720.
Article
52. Chung CG, James AW, Asatrian G, et al. Human perivascular stem cell-based bone graft substitute induces rat spinal fusion. Stem Cells Transl Med. 2014; 3(10):1231–1241.
Article
53. Askarinam A, James AW, Zara JN, et al. Human perivascular stem cells show enhanced osteogenesis and vasculogenesis with Nel-like molecule I protein. Tissue Eng Part A. 2013; 19(11-12):1386–1397.
Article
54. James AW, Zara JN, Corselli M, et al. An abundant perivascular source of stem cells for bone tissue engineering. Stem Cells Transl Med. 2012; 1(9):673–684.
Article
55. Kargozar S, Lotfibakhshaiesh N, Ai J, et al. Strontium- and cobalt-substituted bioactive glasses seeded with human umbilical cord perivascular cells to promote bone regeneration via enhanced osteogenic and angiogenic activities. Acta Biomater. 2017; 58:502–514.
Article
56. Tsang WP, Shu Y, Kwok PL, et al. CD146+ human umbilical cord perivascular cells maintain stemness under hypoxia and as a cell source for skeletal regeneration. PLoS One. 2013; 8(10):e76153.
Article
57. Bosch J, Houben AP, Radke TF, et al. Distinct differentiation potential of “MSC” derived from cord blood and umbilical cord: are cord-derived cells true mesenchymal stromal cells? Stem Cells Dev. 2012; 21(11):1977–1988.
Article
58. Jin HJ, Bae YK, Kim M, et al. Comparative analysis of human mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord blood as sources of cell therapy. Int J Mol Sci. 2013; 14(9):17986–18001.
Article
59. Bosch J, Houben AP, Hennicke T, et al. Comparing the gene expression profile of stromal cells from human cord blood and bone marrow: lack of the typical “bone” signature in cord blood cells. Stem Cells Int. 2013; 2013:631984.
Article
60. Maher S, Kolieb E, Sabik NA, Abd-Elhalim D, El-Serafi AT, El-Wazir Y. Comparison of the osteogenic differentiation potential of mesenchymal cells isolated from human bone marrow, umbilical cord blood and placenta derived stem cells. Beni-Suef Univ J Basic Appl Sci. 2015; 4(1):80–85.
Article
61. Worringer KA, Rand TA, Hayashi Y, et al. The let-7/LIN-41 pathway regulates reprogramming to human induced pluripotent stem cells by controlling expression of prodifferentiation genes. Cell Stem Cell. 2014; 14(1):40–52.
Article
62. Xie J, Peng C, Zhao Q, et al. Osteogenic differentiation and bone regeneration of iPSC-MSCs supported by a biomimetic nanofibrous scaffold. Acta Biomater. 2016; 29:365–379.
Article
63. Ardeshirylajimi A, Soleimani M, Hosseinkhani S, Parivar K, Yaghmaei P. A comparative study of osteogenic differentiation human induced pluripotent stem cells and adipose tissue derived mesenchymal stem cells. Cell J. 2014; 16(3):235–244.
64. Chen IP. The use of patient-specific induced pluripotent stem cells (iPSCs) to identify osteoclast defects in rare genetic bone disorders. J Clin Med. 2014; 3(4):1490–1510.
Article
65. Sheyn D, Ben-David S, Shapiro G, et al. Human induced pluripotent stem cells differentiate into functional mesenchymal stem cells and repair bone defects. Stem Cells Transl Med. 2016; 5(11):1447–1460.
Article
66. Tang M, Chen W, Liu J, Weir MD, Cheng L, Xu HH. Human induced pluripotent stem cell-derived mesenchymal stem cell seeding on calcium phosphate scaffold for bone regeneration. Tissue Eng Part A. 2014; 20(7-8):1295–1305.
Article
67. Barruet E, Hsiao EC. Using human induced pluripotent stem cells to model skeletal diseases. Methods Mol Biol. 2016; 1353:101–118.
Article
68. Saitta B, Passarini J, Sareen D, et al. Patient-derived skeletal dysplasia induced pluripotent stem cells display abnormal chondrogenic marker expression and regulation by BMP2 and TGFβ1. Stem Cells Dev. 2014; 23(13):1464–1478.
Article
69. Matsumoto Y, Hayashi Y, Schlieve CR, et al. Induced pluripotent stem cells from patients with human fibrodysplasia ossificans progressiva show increased mineralization and cartilage formation. Orphanet J Rare Dis. 2013; 8:190.
Article
70. Matsumoto Y, Ikeya M, Hino K, et al. New protocol to optimize iPS cells for genome analysis of fibrodysplasia ossificans progressiva. Stem Cells. 2015; 33(6):1730–1742.
Article
71. Barruet E, Morales BM, Lwin W, et al. The ACVR1 R206H mutation found in fibrodysplasia ossificans progressiva increases human induced pluripotent stem cell-derived endothelial cell formation and collagen production through BMP-mediated SMAD1/5/8 signaling. Stem Cell Res Ther. 2016; 7(1):115.
Article
72. Quarto N, Leonard B, Li S, et al. Skeletogenic phenotype of human Marfan embryonic stem cells faithfully phenocopied by patient-specific induced-pluripotent stem cells. Proc Natl Acad Sci U S A. 2012; 109(1):215–220.
Article
73. Yu YS, Shen ZY, Ye WX, et al. AKT-modified autologous intracoronary mesenchymal stem cells prevent remodeling and repair in swine infarcted myocardium. Chin Med J (Engl). 2010; 123(13):1702–1708.
74. Li Y, Yu X, Lin S, Li X, Zhang S, Song YH. Insulin-like growth factor 1 enhances the migratory capacity of mesenchymal stem cells. Biochem Biophys Res Commun. 2007; 356(3):780–784.
Article
75. Tsubokawa T, Yagi K, Nakanishi C, et al. Impact of antiapoptotic and anti-oxidative effects of bone marrow mesenchymal stem cells with transient overexpression of heme oxygenase-1 on myocardial ischemia. Am J Physiol Heart Circ Physiol. 2010; 298(5):H1320–H1329.
Article
76. Hodgkinson CP, Gomez JA, Mirotsou M, Dzau VJ. Genetic engineering of mesenchymal stem cells and its application in human disease therapy. Hum Gene Ther. 2010; 21(11):1513–1526.
Article
77. Lu CH, Chang YH, Lin SY, Li KC, Hu YC. Recent progresses in gene delivery-based bone tissue engineering. Biotechnol Adv. 2013; 31(8):1695–1706.
Article
78. Kimelman Bleich N, Kallai I, Lieberman JR, Schwarz EM, Pelled G, Gazit D. Gene therapy approaches to regenerating bone. Adv Drug Deliv Rev. 2012; 64(12):1320–1330.
Article
79. Lien CY, Chih-Yuan Ho K, Lee OK, Blunn GW, Su Y. Restoration of bone mass and strength in glucocorticoid-treated mice by systemic transplantation of CXCR4 and cbfa-1 co-expressing mesenchymal stem cells. J Bone Miner Res. 2009; 24(5):837–848.
Article
80. Cho SW, Sun HJ, Yang JY, et al. Transplantation of mesenchymal stem cells overexpressing RANK-Fc or CXCR4 prevents bone loss in ovariectomized mice. Mol Ther. 2009; 17(11):1979–1987.
Article
81. Guan M, Yao W, Liu R, et al. Directing mesenchymal stem cells to bone to augment bone formation and increase bone mass. Nat Med. 2012; 18(3):456–462.
Article
82. Yao W, Lane NE. Targeted delivery of mesenchymal stem cells to the bone. Bone. 2015; 70:62–65.
Article
83. He X, Dziak R, Yuan X, et al. BMP2 genetically engineered MSCs and EPCs promote vascularized bone regeneration in rat critical-sized calvarial bone defects. PLoS One. 2013; 8(4):e60473.
Article
84. Zou D, Zhang Z, He J, et al. Repairing critical-sized calvarial defects with BMSCs modified by a constitutively active form of hypoxia-inducible factor-1α and a phosphate cement scaffold. Biomaterials. 2011; 32(36):9707–9718.
Article
85. Zou D, Zhang Z, Ye D, et al. Repair of critical-sized rat calvarial defects using genetically engineered bone marrow-derived mesenchymal stem cells overexpressing hypoxiainducible factor-1α. Stem Cells. 2011; 29(9):1380–1390.
Article
86. Zou D, Zhang Z, He J, et al. Blood vessel formation in the tissue-engineered bone with the constitutively active form of HIF-1α mediated BMSCs. Biomaterials. 2012; 33(7):2097–2108.
Article
87. Yu L, Lu J, Zhang B, et al. miR-26a inhibits invasion and metastasis of nasopharyngeal cancer by targeting EZH2. Oncol Lett. 2013; 5(4):1223–1228.
Article
88. Tashiro K, Inamura M, Kawabata K, et al. Efficient adipocyte and osteoblast differentiation from mouse induced pluripotent stem cells by adenoviral transduction. Stem Cells. 2009; 27(8):1802–1811.
Article
89. Ye JH, Xu YJ, Gao J, et al. Critical-size calvarial bone defects healing in a mouse model with silk scaffolds and SATB2-modified iPSCs. Biomaterials. 2011; 32(22):5065–5076.
Article
Full Text Links
  • CIOS
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