We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Stem cells for osteodegenerative diseases: current studies and future outlook

    Darcie L McClelland Descalzo

    Department of Cell Biology & Neuroscience, 1113 Biological Sciences Building, University of California Riverside, Riverside, CA 92521, USA

    ,
    Devon D Ehnes

    Department of Cell Biology & Neuroscience, 1113 Biological Sciences Building, University of California Riverside, Riverside, CA 92521, USA

    &
    Nicole I zur Nieden

    Department of Cell Biology & Neuroscience, 1113 Biological Sciences Building, University of California Riverside, Riverside, CA 92521, USA

    Published Online:https://doi.org/10.2217/rme.13.100

    As the worldwide population grows and life expectancies continue to increase, degenerative diseases of the bones, muscles, and connective tissue are a growing problem for society. Current therapies for osteodegenerative disorders such as hormone replacement therapies, calcium/vitamin D supplements and oral bisphosphonates are often inadequate to stop degeneration and/or have serious negative side effects. Thus, there is an urgent need in the medical community for more effective and safer treatments. Stem cell therapies for osteodegenerative disorders have been rigorously explored over the last decade and are yielding some promising results in animal models and clinical trials. Although much work still needs to be done to ensure the safety and efficacy of these therapies, stem cells represent a new frontier of exciting possibilities for bone and cartilage regeneration.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1 zur Nieden NI. Embryonic stem cell therapy for osteo-degenerative disorders. Biotechnol. Interact. 17, 8–14 (2005). •• Excellent reference for using stem cells to study osteodegenerative diseases.
    • 2 Office of the Surgeon General (US). Bone Health and Osteoporosis: a Report of the Surgeon General. Reports of the Surgeon General, MD, USA (2004 ).
    • 3 Kanis JA. WHO Technical Report: Assessment of Osteoporosis at the Primary Health-care Level. WHO Collaborating Centre for Metabolic Bone Diseases, University of Sheffield, UK, 66 (2007).
    • 4 van der Jagt-Willems HC, van Hengel M, Vis M et al. Why do geriatric outpatients have so many moderate and severe vertebral fractures? Exploring prevalence and risk factors. Age Ageing 41(2), 200–206 (2012).
    • 5 Konan S, Soler A, Haddad FS. Revision hip replacement in patients 55 years of age and younger. Hip Int. 23(2), 162–165 (2013).
    • 6 Isaksson E, Wang H, Sahlin L et al. Expression of estrogen receptors (α, β) and insulin-like growth factor-I in breast tissue from surgically postmenopausal cynomolgus macaques after long-term treatment with HRT and tamoxifen. Breast 11(4), 295–300 (2002).
    • 7 McClung MR, Harris ST, Miller PD et al. Bisphosphonate therapy for osteoporosis: benefits, risks, and drug holiday. Am. J. Med. 126(1), 13–20 (2013).
    • 8 Janovská Z. Bisphosphonate-related osteonecrosis of the jaws. A severe side effect of bisphosphonate therapy. Acta Medica (Hradec Kralove) 55, 111–115 (2012).
    • 9 Anasetti C, Logan BR, Lee SJ et al. Peripheral-blood stem cells versus bone marrow from unrelated donors. N. Engl. J. Med. 367(16), 1487–1496 (2012).
    • 10 Pittenger MF, Mackay AM, Beck SC et al. Multilineage potential of adult human mesenchymal stem cells. Science 284, 143–147 (1999).
    • 11 Seale S, Asakura A, Rudnicki MA. The potential of muscle stem cells. Dev. Cell 1, 333–342 (2001).
    • 12 Gaustad KG, Boquest AC, Anderson BE, Gerdes AM, Collas P. Differentiation of human adispose tissue using extracts of rat cardiomyocytes. Biophys. Biochem. Res. Commun. 314, 420–427 (2003).
    • 13 Fadel L, Viana BR, Tajra Feitosa ML et al. Protocols for obtainment and isolation of two mesenchymal stem cell sources in sheep. Acta Cir. Bras. 26(4), 267–273 (2011).
    • 14 Utsunomiya H, Uchida S, Sekiya I, Sakai A, Moridera K, Nakamura T. Isolation and characterization of human mesenchymal stem cells derived from shoulder tissues. Am. J. Sport Med. 41, 657–668 (2013).
    • 15 Chang H, Knothe Tate ML. Concise review: the periosteum: tapping into a reservoir of clinically useful progenitor cells. Stem Cells Transl. Med. 1, 480–491 (2012).
    • 16 Lopez-Garcia C, Klein AM, Simons BD, Winton DJ. Intestinal stem cell replacement follows a pattern of neutral drift. Science 330, 822–825 (2010).
    • 17 De Rosa L, De Luca M. Dormant and restless skin stem cells. Nature 489, 215–217 (2012).
    • 18 Volponi AA, Kawasaki M, Sharpe PT. Adult human gingival epithelial cells as a source for whole tooth bioengineering. J. Dent. Res. 92, 329–334 (2013).
    • 19 Hess DC, Borlongan CV. Stem cells and neurological diseases. Cell Prolif. 41, 94–114 (2008).
    • 20 Lodi D, Iannitti T, Palmieri B. Stem cells in clinical practice: applications and warnings. J. Exp. Clin. Cancer Res. 30, 9–29 (2011). • Useful insight into both the potential and risk of using embryonic stem cells (ESCs) for therapeutics.
    • 21 Smart N, Riley PR. The stem cell movement. Circ. Res. 102, 1155–1168 (2008).
    • 22 Grande DA, Southerland SS, Manji R, Pate DW, Schwartz RE, Lucas PA. Repair of articular cartilage defects using mesenchymal stem cells. Tissue Eng. 1(1), 345–353 (1995).
    • 23 Brittberg M, Lindahl A, Nilsson A, Ohlsson C, Isaksson O, Peterson L. Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N. Engl. J. Med. 331, 889–895 (1994).
    • 24 Knutsen G, Drogset JO, Engebretsen L et al. A randomized trial comparing autologous chondrocyte implantation with microfracture. Findings at five years. J. Bone Joint Surg. Am. 89, 2105 (2007).
    • 25 Wakitani S, Okabe T, Horibe S et al. Safety of autologous bone marrow-derived mesenchymal stem cell transplantation for cartilage repair in 41 patients with 45 joints followed for up to 11 years and 5 months. J. Tissue Eng. Regen. Med. 5(2), 146–150 (2011).
    • 26 Bhatia M, Wang JC, Kapp U, Bonnet D, Dick JE. Purification of primitive human hematopoietic cells capable of repopulating immune-deficient mice. Proc. Natl Acad. Sci. USA 94, 5320–5325 (1997).
    • 27 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 8, 315–317 (2006).
    • 28 Murphy JM, Dixon K, Beck S, Fabian D, Feldman A, Barry F. Reduced chondrogenic and adipogenic activity of mesenchymal stem cells from patients with advanced osteoarthritis. Arthritis Rheum. 46(3), 704–713 (2002).
    • 29 García-Álvarez F, Alegre-Aguarón E, Desportes P et al. Chondrogenic differentiation in femoral bone marrow-derived mesenchymal cells (MSC) from elderly patients suffering osteoarthritis or femoral fracture. Arch. Gerontol. Geriat. 52, 239–242 (2011).
    • 30 English A, Jones EA, Corscadden D et al. A comparative assessment of cartilage and joint fat pad as a potential source of cells for autologous therapy development in knee osteoarthritis. Rheumatology 46, 1676–1683 (2007).
    • 31 Dudics V, Kunstár A, Kovács J et al. Chondrogenic potential of mesenchymal stem cells from patients with rheumatoid arthritis and osteoarthritis: measurements in a microculture system. Cells Tissues Organs 189, 307–316 (2009).
    • 32 Toghraie FS, Chenari N, Gholipour MA et al. Treatment of osteoarthritis with infrapatellar fat pad derived mesenchymal stem cells in rabbit. Knee 18, 71–75 (2011).
    • 33 Diekman BO, Wu CL, Louer CR et al. Intra-articular delivery of purified mesenchymal stem cells from C57BL/6 or MRL/MpJ superhealer mice prevents post-traumatic arthritis. Cell Transplant. 22(8), 1395–1408 (2012).
    • 34 Koh GY, Choi YJ. Infrapatellar fat pad-derived mesenchymal stem cell therapy for knee osteoarthritis. Knee 19, 902–907 (2012).
    • 35 Koh GY, Jo SB, Kwon OR et al. Mesenchymal stem cell injections improve knee osteoarthritis. Arthroscopy 29(4), 748–755 (2013).
    • 36 NIH Clinical Trials Database. www.clinicaltrials.gov
    • 37 Osteoarthritis and you. www.cdc.gov/features/osteoarthritisplan
    • 38 Holt HL, Katz JN. Reichmann WM. Forecasting the burden of advanced knee osteoarthritis over a 20 year period in a cohort of older US adults: impact of obesity Presented at: American College of Rheumatology Annual Scientific Meeting, San Francisco, CA, USA, 25–29 October 2008 (Astract 204).
    • 39 Roldan M, Macias-Gonzalez M, Garcia R, Tinahones FJ, Martin M. Obesity short circuits stemness gene network in human adipose multipotent stem cells. FASEB J. 25, 4111–4126 (2011).
    • 40 Wu CL, Diekman BO, Jain D, Guilak F. Diet-induced obesity alters the differentiation potential of stem cells isolated from bone marrow, adipose tissue and infrapatellar fad pad: the effects of free fatty acids. Int. J. Obes. (Lond.) 37(8), 1079–1087 (2012).
    • 41 Heldens GT, Bilaney Davidson EN, Vitters EL, Schreurs BW, Piek E, van den Berg WB. Catabolic factors and osteoarthritis conditioned medium inhibit chondrogenesis of human mesenchymal stem cells. Tissue Eng. 18(1–2), 45–54 (2012).
    • 42 Boeuf S, Graf F, Fischer J, Moradi B, Little CB, Richter B. Regulation of aggrecanases from the ADAMTS family and aggrecan neoepitope formation during in vitro chondrogenesis of human mesenchymal stem cells. Eur. Cells Mater. 23, 320–332 (2012).
    • 43 Aggarwal R, Lu J, Kanji S et al. Human umbilical cord blood-derived CD34+ cells reverse osteoporosis in NOD/SCID mice by altering osteoblastic and osteoclastic activities. PLoS ONE 7(6), e39365 (2012).
    • 44 Singh L, Brennan TA, Kim JH et al. Long-term functional engraftment of mesenchymal progenitor cells in a mouse model of accelerated aging. Stem Cells 31(3), 607–611 (2013).
    • 45 Guan M, Yao W, Liu R et al. Directing mesenchymal stem cells to bone to augment bone formation and increase bone mass. Nat. Med. 18(3), 456–462 (2012).
    • 46 Li J, Zhang L, Zhou L. Beneficial effects of non-matched allogeneic cord blood mononuclear cells upon patients with idiopathic osteoporosis. J. Transl. Med. 10, 102–107 (2012).
    • 47 Minguell JJ, Allers C, Lasala GP. Mesenchymal stem cells and the treatment of conditions and diseases: the less glittering side of a conspicuous stem cell for basic research. Stem Cells Dev. 22, 193–203 (2013).
    • 48 Wagner W, Horn P, Castoldi M et al. Replicative senescence of mesenchymal stem cells: a continuous and organized process. PLoS ONE 3, e2213 (2008).
    • 49 Kretlow JD, Jin YQ, Liu W et al. Donor age and cell passage affects differentiation potential of murine bone marrow-derived stem cells. BMC Cell Biol. 9, 60 (2008).
    • 50 Trounson A. The production and directed differentiation of human embryonic stem cells. Endocr. Rev. 27, 208–219 (2006).
    • 51 Robertson EJ. Derivation and maintenance of embryonic stem cell cultures. Methods Mol. Biol. 75, 173–184 (1997).
    • 52 Thomson JA, Itskovitz-Eldor J, Shapiro SS et al. Embryonic stem cell lines derived from human blastocysts. Science 5391(282), 1145–1147 (1998).
    • 53 Rodda DJ, Chew J, Lim L. Transcriptional regulation of nanog by OCT4 and SOX2. J. Biol. Chem. 280(26), 24731–24737 (2005).
    • 54 Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126(4), 663–676 (2006).
    • 55 Takahashi K, Tanabe K, Ohnuki M et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131(5), 861–872 (2007).
    • 56 Huangfu D, Maehr R, Guo W et al. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat. Biotechnol. 26, 795–797 (2008).
    • 57 Hou P, Li Y, Liu C et al. Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science 341(6146), 651–654 (2013).
    • 58 Zhou W, Freed C. Adenoviral gene delivery can reprogram human fibroblasts to induced pluripotent stem cells. Stem Cells 27(11), 2667–2774 (2009).
    • 59 Tsukiyama T, Asano R, Kawaguchi T et al. Simple and efficient method for generation of induced pluripotent stem cells using piggyBac transposition of doxycycline-inducible factors and an EOS reporter system. Genes Cells 16(7), 815–825 (2011).
    • 60 Davis RP, Nemes C, Varga E et al. Generation of induced pluripotent stem cells from human foetal fibroblasts using the Sleeping Beauty transposon gene delivery system. Differentiation 86(1–2), 30–37 (2013).
    • 61 Lin T, Ambasudhan R, Yuan X et al. A chemical platform for improved induction of human iPSCs. Nat. Methods 6(11), 805–808 (2009).
    • 62 Judson RL, Greve TS, Parchem RJ et al. MicroRNA-based discovery of barriers to dedifferentiation of fibroblasts to pluripotent stem cells. Nat. Struct. Mol. Biol. 20(10), 1227–1235 (2013).
    • 63 Egashira T, Yuasa S, Fukuda K. Novel insights into disease modeling using induced pluripotent stem cells. Biol. Pharm. Bull. 36(2), 182–188 (2013). • Thoroughly compiles the various potentials for induced pluripotent stem cells in therapeutics, highlighting the great potential effective drug treatment on a patient-specific basis.
    • 64 Polo JM, Liu S, Figueroa ME et al. Cell type of origin influences the molecular and functional properties of mouse induced pluripotent stem cells. Nat. Biotechnol. 28, 848–855 (2010). • Highlights one of the main problems in using induced pluripotent stem cells.
    • 65 Kim K, Doi A, Wen B et al. Epigenetic memory in induced pluripotent stem cells. Nature 467, 285–290 (2010).
    • 66 Kim K, Zhao R, Doi A et al. Donor cell type can influence the epigenome and differentiation potential of human induced pluripotent stem cells. Nat. Biotechnol. 29, 1117–1119 (2011).
    • 67 Hu BY, Weick JP, Yu J et al. Neural differentiation of human induced pluripotent stem cells follows developmental principles but with variable potency. Proc. Natl Acad. Sci. USA 107, 4335–4340 (2010).
    • 68 Kattman SJ, Witty AD, Gagliardi M et al. Stage-specific optimization of activin/nodal and BMP signaling promotes cardiac differentiation of mouse and human pluripotent stem cell lines. Cell Stem Cell 8(2), 228–240 (2011).
    • 69 Ohnishi H, Oda Y, Aoki T et al. A comparative study of induced pluripotent stem cells generated from frozen, stocked bone marrow- and adipose tissue-derived mesenchymal stem cells. J. Tissue Eng. Regen. Med. 6, 261–271 (2012).
    • 70 Feng L, Bronson S, Niyibizi C. Derivation of murine induced pluripotent stem cells (iPS) and assessment of their differentiation toward osteogenic lineage. J. Cell. Biochem. 109, 643–652 (2010).
    • 71 Nasu A, Ikeya M, Yamamoto T et al. Genetically matched human iPS cells reveal that propensity for cartilage and bone differentiation differs with clones, not cell type of origin. PLoS ONE 8(1), e53771 (2013).
    • 72 Liu J, Chen W, Zhao Z, Xu H. Reprogramming of mesenchymal stem cells derived from iPSCs seeded on biofunctionalized calcium phosphate scaffold for bone engineering. Biomaterials 4(32), 7862–7872 (2013).
    • 73 Xu Y, Liu L, Zhang L et al. Efficient commitment to functional CD34+ progenitor cells from human bone marrow mesenchymal stem-cell-derived induced pluripotent stem cells. PLoS ONE 7(4), e34321 (2012).
    • 74 Jung Y, Bauer G, Nolta JA. Concise review: induced pluripotent stem cell-derived mesenchymal stem cells: progress toward safe clinical products. Stem Cells 30(1), 42–47 (2013).
    • 75 de Peppo GM, Marolt D. Make no bones about it: cells could soon be reprogrammed to grow replacement bones? Expert Opin. Biol. Ther. 14(1), 1–5 (2013).
    • 76 Kelley JM, Daley GQ. Hematopoietic defects and iPSC disease modeling: lessons learned. Immunol. Lett. 155(1–2), 18–20 (2013).
    • 77 Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature 292, 154–156 (1981).
    • 78 Martin GR. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc. Natl Acad. Sci. USA 78(12), 7634–7638 (1981).
    • 79 Alper J. Geron gets green light for human trial of ES cell-derived product. Nat. Biotechnol. 27, 213–214 (2009).
    • 80 Stein R. Stem cells were God’s will, says first recipient of treatment. Washington Post, 15th April (2011).
    • 81 Chapman AR, Scala CC. Evaluating the first-in-human clinical trial of a human embryonic stem cell-based therapy. Kennedy Inst. Ethics J. 22(3), 243–261 (2012).
    • 82 Schwartz S, Hubschman JP, Heilwell G et al. Embryonic stem cell trials for macular degeneration: preliminary report. Lancet 379(9817), 713–720 (2012).
    • 83 Advanced Cell Technology. www.advancedcell.com
    • 84 zur Nieden NI, Kempka G, Ahr HJ. In vitro differentiation of embryonic stem cells into mineralized osteoblasts. Differentiation 17, 18–27 (2003).•• Highlights the promise of using ESCs to regenerate bone.
    • 85 zur Nieden NI. Embryonic stem cell therapy for osteo-degenerative disorders. Biotechnol. Interactions 17, 8–14 (2005).
    • 86 Yamane T, Kunisada T, Yamazaki H, Era T, Nakano T, Hayashi SI. Development of osteoclasts from embryonic stem cells through a pathway that is c-fms but not c-kit dependent. Blood 90(9), 3516–3523 (1997).
    • 87 Raisz LG. Pathogenesis of osteoporosis: concepts, conflicts, and prospects. J. Clin. Invest. 115(12), 3318–3325 (2005).
    • 88 Sovani S, Grogan SP. Osteoarthritis: detection, pathophysiology, and current/future treatment strategies. Orthop. Nurs. 32(1), 25–36 (2013).
    • 89 zur Nieden NI. Embryonic stem cells for osteo-degenerative diseases. Methods Mol. Biol. 690, 1–30 (2011). •• Provides an excellent source of current protocols and research on ESC use for osteodegenerative disorders.
    • 90 Dienelt A, zur Nieden NI. Hyperglycemia impairs skeletogenesis from embryonic stem cells by affecting osteoblast and osteoclast differentiation. Stem Cells Dev. 20, 465–474 (2011).
    • 91 Valdimarsdotter G, Mummery C. Functions of the TGF-β superfamily in human embryonic stem cells. APMIS 113, 773–789 (2005).
    • 92 Morali OG, Jouneau A, McLaughlin KJ, Thiery JP, Larue L. IGF-II promotes mesoderm formation. Dev. Biol. 227, 133–145 (2000).
    • 93 Hwang YS, Chung BG, Ortmann D, Hattori N, Moeller HC, Khademhosseini A. Microwell mediated control of embryoid body size regulates embryonic stem cell fate via differential expression of WNT5a and WNT11. Proc. Natl Acad. Sci. USA 106, 16978–16983 (2009).
    • 94 Lu CC, Brennan J, Robertson EJ. From fertilization to gastrulation: axis formation in the mouse embryo. Curr. Opin. Genet. Dev. 11, 384–392 (2001).
    • 95 Keller KC, zur Nieden NI. Osteogenesis from pluripotent stem cells: neural crest or mesodermal origin? In: Embryonic Stem Cells – Differentiation and Pluripotent Alternatives, Kallos MS (Ed). InTech, Rijeka, Croatia, 323–348 (2011).
    • 96 Kurosawa H. Methods for inducing embryoid body formation: in vitro differentiation system of embryonic stem cells. J. Biosci. Bioeng. 103(5), 389–398 (2007).
    • 97 Itskovitz-Eldor J, Schuldiner M, Karsenti D et al. Differentiation of human embryonic stem cells into embryoid bodies comprising the three embryonic germ layers. Mol. Med. 6, 88–95 (2000).
    • 98 Buttery LD, Bourne S, Xynos JD et al. Differentiation of osteoblasts and in vitro bone formation from murine embryonic stem cells. Tissue Eng. 7, 89–99 (2001).
    • 99 Kawaguchi J, Mee PJ, Smith AG. Osteogenic and chondrogenic differentiation of embryonic stem cells in response to specific growth factors. Bone 36, 758–769 (2005).
    • 100 Woll NL, Heaney JD, Bronson SK. Osteogenic nodule formation from single embryonic stem cell-derived progenitors. Stem Cell Dev. 15, 865–879 (2006).
    • 101 Bielby RC, Pryce RS, Hench LL, Polak JM. Enhanced derivation of osteogenic cells from murine embryonic stem cells after treatment with ionic dissolution products of 58 S bioactive sol-gel glass. Tissue Eng. 11, 479–488 (2005).
    • 102 zur Nieden NI, Kempka G, Ahr HJ. In vitro differentiation of embryonic stem cells into mineralized osteoblasts. Differentiation 17, 18–27 (2003).
    • 103 Warotayanont R, Frenkel B, Snead ML, Zhou Y. Leucine-rich amelogenic peptide induces osteogenesis by activation of the Wnt pathway. Biochem. Biophys. Res. Commun. 387, 558–563 (2009).
    • 104 Arpornmaekilong P, Brown SE, Wang Z, Krebsbach PH. Phenotypic characterization, osteogenic differentiation, and bone regeneration capacity of human embryonic stem cell-derived mesenchymal stem cells. Stem Cell Dev. 18, 1–14 (2009).
    • 105 Rathjen J, Lake JA, Bettess MD, Washington JM, Chapman G, Rathjen PD. Formation of a primitive ectoderm like cell population, EPL cells, from ES cells in response to biologically derived factors. J. Cell Sci. 112, 601–612 (1999).
    • 106 Mummery C, Ward D, van den Brink CE et al. Cardiomyocyte differentiation of mouse and human embryonic stem cells. J. Anat. 200, 233–242 (2002).
    • 107 Hwang YS, Randle WL, Bielby R, Polak JM, Mantalaris A. Enhanced derivation of osteogenic cells from murine embryonic stem cells after treatment with HepG2-conditioned medium and modulation of the embryoid body formation period: application to skeletal tissue engineering. Tissue Eng. 12, 1381–1392 (2006).
    • 108 Lee G, Kim H, Elkabetz Y et al. Isolation and directed differentiation of neural crest stem cells derived from human embryonic stem cells. Nat. Biotechnol. 25(12), 1468–1475 (2007).
    • 109 Tian XF, Heng BC, Ge Z et al. Comparison of osteogenesis of human embryonic stem cells within 2D and 3D culture systems. Scand. J. Clin. Lab. Invest. 68, 58–67 (2008).
    • 110 Garreta E, Genove E, Borros S, Semino CE. Osteogenic differentiation of mouse embryonic stem cells and mouse embryonic fibroblasts in a three-dimensional self-assembling peptide scaffold. Tissue Eng. 12, 2215–2227 (2006).
    • 111 Kim S, Kim SS, Lee SH et al. In vivo bone formation from human embryonic stem cell-derived osteogenic cells in poly (D,L-lactic-co-glycolic acid)/hydroxyapatite composite scaffold. Biomaterials 29, 1043–1053 (2008).
    • 112 Druml C. Stem cell research: toward greater unity in Europe? Cell 139, 649–651 (2009).
    • 113 Dhar D, Hsi-En Ho J. Stem cell research policies around the world. Yale J. Biol. Med. 82(3), 113–115 (2009).
    • 114 Prescott C. The business of exploiting induced pluripotent stem cells. Philos. Trans. R. Soc. Lond. B Biol. Sci. 366, 2323–2328 (2011).
    • 115 HeinOnline: World’s Largest Image-Based Legal Research Database. http://heinonline.org
    • 116 Amariglio N, Hirshberg A, Scheithauer BW et al. Donor-derived brain tumor following neural stem cell transplantation in an ataxia telangiectasia patient. Trends Mol. Med. 6(2), e1000029 (2009).
    • 117 Sverdlov ED, Mineev K. Mutation rate in stem cells: an underestimated barrier on the way to therapy. Trends Mol. Med. 19(5), 273–280 (2013).
    • 118 Gore A, Li Z, Fung HL et al. Somatic coding mutations in human induced pluripotent stem cells. Nature 471, 63–67 (2011).
    • 119 Thomas ED, Lochte HL, Cannon JM, Sahler OD, Ferrebee JW. Supralethal whole body irradiation and isologous bone marrow transplantation in man. J. Clin. Invest. 38, 1709–1716 (1959).
    • 120 Escobedo-Lucea C, Ayuso-Sacedo A, Xiong C et al. Development of a human extracellular matrix for applications related with stem cells and tissue engineering. Stem Cell Rev. 8(1), 170–183 (2012).