Bone regeneration and stem cells

K Arvidson, B M Abdallah, L A Applegate, N Baldini, E Cenni, E Gomez-Barrena, D Granchi, M Kassem, Y T Konttinen, K Mustafa, D P Pioletti, T Sillat, A Finne-Wistrand, K Arvidson, B M Abdallah, L A Applegate, N Baldini, E Cenni, E Gomez-Barrena, D Granchi, M Kassem, Y T Konttinen, K Mustafa, D P Pioletti, T Sillat, A Finne-Wistrand

Abstract

This invited review covers research areas of central importance for orthopaedic and maxillofacial bone tissue repair, including normal fracture healing and healing problems, biomaterial scaffolds for tissue engineering, mesenchymal and foetal stem cells, effects of sex steroids on mesenchymal stem cells, use of platelet-rich plasma for tissue repair, osteogenesis and its molecular markers. A variety of cells in addition to stem cells, as well as advances in materials science to meet specific requirements for bone and soft tissue regeneration by addition of bioactive molecules, are discussed.

© 2011 The Authors Journal of Cellular and Molecular Medicine © 2011 Foundation for Cellular and Molecular Medicine/Blackwell Publishing Ltd.

Figures

Fig 1
Fig 1
Molecular structure of (A) PLA and (B) PLC.
Fig 2
Fig 2
Molecular structure of poly(propylene fumarate).
Fig 3
Fig 3
Molecular structure of poly(1,5-dioxepan-2-one) (DXO).
Fig 4
Fig 4
Development stage, cell type and source of cells currently used in cellular therapy. Cellular source can come from human and animals at different stages of development including embryonic, embryonic-foetal, foetal and adult involving different beginning tissue sources ranging from zygotes to specific tissues (bone marrow, adipose, amniotic fluid, skin, liver, bone, cartilage etc.). Embryonic stem cells are derived from embryos at early stage where less than 100 cells are present, followed by foetal stem cells that are taken from the genital ridge section from 5 to 8 weeks of gestation (drawing modified from http://commons.wikimedia.org/wiki/Image:10_weeks_pregnant.jpg donated by wouter.vengeer@tribal.nl with a free license). Tissue-specific foetal cells are taken following 9 weeks of gestation usually up to 14–16 weeks from normal tissue. Adult stem cells can be isolated from most tissue sources but are rare with only 1 in every 104 to 105 of total cell volume.
Fig 5
Fig 5
Organization of a cell therapy platform. Director of technical and logistics coordinates the program with essential legal and ethical advisors and a medical director for interpretation of medical quality assurance (serology and pathology reports). The separation of hospitals for the organ donation and all other aspects of the platform including serology, pathology and the GLP cell culture laboratory can assure complete anonymous and coded organ donations. Importantly, the Director of Technical and Logistics is not involved in any manner in the organ donation process as required by law. Final approval for use of validated cell banks for human therapy is coordinated and approved with both Hospital Ethics Committees and National regulatory agencies (i.e. European Medicines Association, Food and Drug Administration, SwissMedic [32]).
Fig 6
Fig 6
Schematic representation of the principal signalling networks and transcription factors regulating osteoblast differentiation and osteogenesis (see text for details). Lines with arrowheads indicate a positive action, lines with bars indicate an inhibition and dashed lines indicate that function has to be proven. The lineage commitment and the differentiation of cultured MSC are signed by the acquisition of specific osteoblast functions. In boxes are shown genes that are significantly up-regulated during osteogenic differentiation of MSC. The ‘Gene Ontology Annotation’ of genes listed here is associated to GF-signalling pathways, cell communication and skeletal development. The up-regulated genes have been grouped according to the differentiation status of cultured MSC: ‘Pre-osteoblasts’ are MSC which are able to produce ALP (cytochemical activity, ×10) and generate ‘Colony Forming Units’; ‘Differentiated osteoblasts’ are cells which have the competence to deposit mineral nodules in vitro (von Kossa staining, ×10). Genes are reported with the official symbol, as shown in ‘Gene’ database (http://www.ncbi.nlm.nih.gov/sites/entrez?db=gene).

References

    1. Urist MR, Johnson RW., Jr Calcification and ossification. IV The healing of fractures in man under clinical conditions. J Bone Joint Surg Am. 1943;25:375–426.
    1. Phillips AM. Overview of the fracture healing cascade. Injury. 2005;36:S5–7.
    1. Einhorn TA, Majeska RJ, Rush EB, et al. The expression of cytokine activity by fracture callus. J Bone Miner Res. 1995;10:1272–81.
    1. Gerstenfeld LC, Cho TJ, Kon T, et al. Impaired fracture healing in the absence of TNF-alpha signaling: the role of TNF-alpha in endochondral cartilage resorption. J Bone Miner Res. 2003a;18:1584–92.
    1. Tsiridis E, Upadhyay N, Giannoudis P. Molecular aspects of fracture healing: which are the important molecules. Injury. 2007;38:S11–25.
    1. Gerstenfeld LC, Cullinane DM, Barnes GL, et al. Fracture healing as a post-natal developmental process: molecular, spatial, and temporal aspects of its regulation. J Cell Biochem. 2003b;88:873–84.
    1. Deckers MM, van Bezooijen RL, van der Horst G, et al. Bone morphogenetic proteins stimulate angiogenesis through osteoblast-derived vascular endothelial growth factor A. Endocrinology. 2002;143:1545–53.
    1. Yoshimura Y, Nomura S, Kawasaki S, et al. Colocalization of noggin and bone morphogenetic protein-4 during fracture healing. J Bone Miner Res. 2001;16:876–84.
    1. Onichtchouk D, Chen YG, Dosch R, et al. Silencing of TGF-beta signalling by the pseudoreceptor BAMBI. Nature. 1999;401:480–5.
    1. Nakao A, Afrakhte M, Morén A, et al. Identification of Smad7, a TGFbeta-inducible antagonist of TGF-beta signalling. Nature. 1997;389:631–5.
    1. Bai S, Shi X, Yang X, Cao X. Smad6 as a transcriptional corepressor. J Biol Chem. 2000;275:8267–70.
    1. Ferguson C, Alpern E, Miclau T, Helms JA. Does adult fracture repair recapitulate embryonic skeletal formation. Mech Dev. 1999;87:57–66.
    1. Carter DR, Beaupre GS, Giori NJ, Helms JA. Mechanobiology of skeletal regeneration. Clin Orthop Relat Res. 1998;355:S41–55.
    1. Giannoudis PV, Einhorn TA, Marsh D. Fracture healing: the diamond concept. Injury. 2007;38:S3–6.
    1. Zhuang H, Wang W, Tahernia AD, et al. Mechanical strain-induced proliferation of osteoblastic cells parallels increased TGF-beta 1 mRNA. Biochem Biophys Res Commun. 1996;229:449–53.
    1. Sasaki K, Takagi M, Konttinen Y, et al. Upregulation of matrix metalloproteinase (MMP)-1 and its activator MMP-3 of human osteoblasts by uniaxial cyclic stimulation. J Biomed Mater Res B Appl Biomater. 2007;80B:491–8.
    1. Chiquet M. Regulation of extracellular matrix gene expression by mechanical stress. Matrix Biol. 1999;18:417–26.
    1. Kershaw CJ, Cunningham JL, Kenwright J. Tibial external fixation, weight bearing, and fracture movement. Clin Orthop Relat Res. 1993;293:28–36.
    1. Kwong FN, Harris MB. Recent developments in the biology of fracture repair. J Am Acad Orthop Surg. 2008;16:619–25.
    1. Claes L, Eckert-Hübner K, Augat P. The effect of mechanical stability on local vascularization and tissue differentiation in callus healing. J Orthop Res. 2002;20:1099–105.
    1. Loboa EG, Beaupre GS, Carter DR. Mechanobiology of initial pseudarthrosis formation with oblique fractures. J Orthop Res. 2001;19:1067–72.
    1. Gartner LP, Hiatt JL. Color textbook of histology. 3rd ed. Philadelphia, PA: Saunders Elsevier; 2007. pp. 19103–2899.
    1. Langman J. Medical embryology. human development-normal and abnormal. 3rd ed. Baltimore: The Williams & Wilkins Company; 1975.
    1. ten Cate AR. Oral histology. Development, structure and function. St. Louis, MO: The C.V. Mosby Company; 1980.
    1. Glenny AM, Hooper L, Shaw WC, et al. Feeding interventions for growth and development in infants with cleft lip, cleft palate or cleft lip and palate. Review. Cochrane Database Syst Rev. 2004 CD003315.
    1. Mossey PA, Little J, Munger RG, et al. Cleft lip and palate. Review. Lancet. 2009;9703:1773–85.
    1. Pena WA, Vargervik K, Sharma A, Oberoi S. The role of endosseous implants in the management of alveolar clefts. Review. Pediatr Dent. 2009;31:329–33.
    1. Esposito M, Grusovin MG, Coulthard P, et al. The efficacy of various bone augmentation procedures for dental implants: a cochrane systematic review of randomized controlled clinical trials. Int J Oral Maxillofac Implants. 2006;21:696–710.
    1. Kelly CM, Wilkins RM, Gitelis S, et al. The use of a surgical grade calcium sulfate as a bone graftsubstitute: results of a multicenter trial. Clin Orthop Relat Res. 2001;382:42–50.
    1. Schindler OS, Cannon SR, Briggs TWR, et al. Composite ceramic bone graft substitute in the treatment of locally aggressive benign bone tumours. J Orthop Surg. 2008;16:66–74.
    1. Schieker M, Seitz H, Drosse I, et al. Biomaterials as scaffold for bone tissue engineering. Eur J Trauma. 2006;2:114–24.
    1. Petite H, Viateau V, Bensaid W, et al. Tissue-engineered bone regeneration. Nat Biotechnol. 2000;18:959–63.
    1. Matsubara T, Suardita K, Ishii M, et al. Alveolar bone marrow as a cell source for regenerative medicine: differences between alveolar and iliac bone marrow stromal cells. J Bone Miner Res. 2005;20:399–409.
    1. Shi S, Gronthos S. Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp. J Bone Miner Res. 2003;8:696–704.
    1. Shi S, Bartold PM, Miura M, et al. The efficacy of mesenchymal stem cells to regenerate and repair dental structures. Orthod Craniofac Res. 2005;8:191–9.
    1. Ikeda E, Hirose M, Kotobuki N, et al. Osteogenic differentiation of human dental papilla mesenchymal cells. Biochem Biophys Res Commun. 2006;342:1257–62.
    1. Gronthos S, Brahim J, Li W, et al. Stem cell properties of human dental pulp stem cells. J Dent Res. 2002;81:531–5.
    1. Derubeis AR, Cancedda R. Bone marrow stromal cells (BMSCs) in bone engineering: limitations and recent advances. Ann Biomed Eng. 2004;32:160–5.
    1. Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9:641–50.
    1. Khan Y, Yaszemski MJ, Mikos AG, et al. Tissue engineering of bone: material and matrix consideration. J Bone Joint Surg Am. 2008;90:36–42.
    1. Suzuki O, Imaizumi H, Kamakura S, et al. Bone regeneration by synthetic octacalcium phosphate and its role in biological mineralization. Curr Med Chem. 2008;15:305–13.
    1. Guarino V, Causa F, Ambrosio L. Bioactive scaffolds for connective tissue regeneration. Exp Rev Med Dev. 2007;4:406–18.
    1. Coombes AGA, Rizzi SC, Williamson M, et al. Precipitation casting of polycaprolactone for applica- tions in tissue engineering and drug delivery. Biomaterials. 2004;25:315–25.
    1. Shikinami Y, Okazaki K, Saito M, et al. Bioactive and bioresorbable cellular cubic-composite scaffolds for use in bone reconstruction. J R Soc Interface. 2006;3:805–21.
    1. Mondrinos MJ, Dembzinsky R, Lu L, et al. Porogen-based solid freeform fabrication of polycaprolactone–calcium phosphate scaffold for tissue engineering. Biomaterials. 2006;27:4399–408.
    1. Zhou Y, Hutmacher DW, Varawan S-L, et al. In vitro bone engineering based on polycaprolactone and polycaprolactone-tricalcium phosphate composites. Polym Int. 2007;56:333–42.
    1. Guarino V, Ambrosio L. The synergic effect of polylactide fiber and calcium phosphate particle reinforcement in poly ɛ-caprolactone-based composite scaffolds. Acta Biomaterialia. 2008;4:1778–87.
    1. Cai Z-Y, Yang D-A, Zhang N, et al. Poly(propylene fumarate)/(calcium sulphate/β-tricalcium phosphate) composites: preparation, characterization and in vitro degradation. Acta Biomaterialia. 2009;5:628–35.
    1. Irina M, Pelin, Maier StelianS, et al. Preparation and characterization of a hydroxyapatite–collagen composite as component for injectable bone substitute. Mater Sci Engin. 2009;29:2188–94.
    1. Azevedo MC, Reis RL, Claase MB, et al. Development and properties of polycaprolactone/hydroxyapatite composite biomaterials. J Mater Sci Mat Med. 2003;14:103–7.
    1. Guarino V, Causa F, Netti PA, et al. The role of hydroxyapatite as solid signal on performance of PCL porous scaffolds for bone tissue regeneration. J Biomed Mater Res B. 2008;86:548–57.
    1. Kroeze RJ, Helder MN, Govaert LE, et al. Biodegradable polymers in bone tissue engineering. Materials. 2009;2:833–56.
    1. Li F, Li S, Ghzaoui AE, et al. Synthesis and gelation properties of PEG-PLA-PEG triblock copolymers obtained by coupling monohydroxylated PEG-PLA with adipoyl chloride. Langmuir. 2007a;27:2778–83.
    1. Ryner M, Albertsson A-C. Resorbable and highly elastic block copolymers from 1,5-dioxepan-2-one and L-lactide with controlled tensile properties and hydrophilicity. Biomacromolecules. 2002;3:601–8.
    1. Ryner M, Valdre A, Albertsson A-C. Star-shaped and photo-crosslinked poly(1,5-dioxepan-2-one) – synthesis and characterization. J Polym Sci A: Polym Sci. 2002;40:2049–54.
    1. Idris S, Arvidson K, Plikk P, et al. Polyester copolymer scaffolds enhance expression of bone markers in osteoblast-like cells. J Biomed Mater Res A. 2010;94:631–9.
    1. Dånmark S, Finne-Wistrand A, Albertsson A-C, et al. Osteogenic differentiation in rat bone rarrow derived stromal cells on refined biodegradable polymer scaffolds. J Bioact Compat Pol. 2010;25:207–23.
    1. Schander K, Arvidson K, Mustafa M, et al. Response of Bone and Periodontal Ligament Cells to Biodegradable Polymer Scaffolds in vitro. J Bioact Compat Pol. 2010;25:584–602.
    1. Idris S, Danmark S, Finne-Wistrand A, et al. Biocompatibility of Polyester Scaffolds with Fibroblasts and Osteoblast-like Cells for Bone Tissue Engineering. J Bioact Compat Pol. 2010;25:567–83.
    1. Wang S, Cui W, Bei J. Bulk and surface modifications of polylactide. Anal Bioanal Chem. 2005;381:547–56.
    1. Frost HM. Why should many skeletal scientists and clinicians learn the Utah paradigm of skeletal physiology. J Musculoskel Neuron Interact. 2001;2:121–30.
    1. Parfitt AM. Bone forming cells in clinical conditions. In: Hall BK, editor. Bone, the osteoblast and osteocytes. London, UK: The Telford Press; 1991. pp. 351–426.
    1. Harada S, Rodan GA. Control of osteoblast function and regulation of bone mass. Nature. 2003;423:349–55.
    1. Blair HC, Robinson LJ, Zaidi M. Osteoclast signalling pathways. Biochem Biophys Res Commun. 2005;328:728–38.
    1. Friedenstein AJ, Ivanov-Smolenski AA, Chajlakjan RK, et al. Origin of bone marrow stromal mechanocytes in radiochimeras and heterotopic transplants. Exp Hematol. 1978;6:440–4.
    1. Friedenstein AJ. Osteogenic stem cells in the bone marrow. Bone Miner. 1991;7:243–72.
    1. Benayahu D, Akavia UD, Shur I. Differentiation of bone marrow stroma-derived mesenchymal cells. Curr Med Chem. 2007;14:173–9.
    1. Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea- pig bone marrow and spleen cells. Cell Tissue Kinet. 1970;3:393–403.
    1. Luria EA, Panasyuk AF, Friedenstein AY. Fibroblast colony formation from monolayer cultures of blood cells. Transfusion. 1971;11:345–9.
    1. Kassem M, Mosekilde L, Eriksen EF. 1,25-dihydroxyvitamin D3 potentiates fluoride-stimulated collagen type I production in cultures of human bone marrow stromal osteoblast-like cells. J Bone Miner Res. 1993;8:1453–8.
    1. Rickard DJ, Kassem M, Hefferan TE, et al. Isolation and characterization of osteoblast precursor cells from human bone marrow. J Bone Miner Res. 1996;11:312–24.
    1. Kuznetsov SA, Krebsbach PH, Satomura K, et al. Single-colony derived strains of human marrow stromal fibroblasts form bone after transplantation in vivo. J Bone Miner Res. 1997;12:1335–47.
    1. Post S, Abdallah BM, Bentzon JF, Kassem M. Demonstration of the presence of independent pre-osteoblastic and pre-adipocytic cell populations in bone marrow-derived mesenchymal stem cells. Bone. 2008;43:32–9.
    1. Abdallah BM, Haack-Sorensen M, Burns JS, et al. Maintenance of differentiation potential of human bone marrow mesenchymal stem cells immortalized by human telomerase reverse transcriptase gene despite [corrected] extensive proliferation. Biochem Biophys Res Commun. 2005;326:527–38.
    1. Foster LJ, Zeemann PA, Li C, et al. Differential expression profiling of membrane proteins by quantitative proteomics in a human mesenchymal stem cell line undergoing osteoblast differentiation. Stem Cells. 2005;23:1367–77.
    1. Dominici M, Le BK, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–7.
    1. Gronthos S, Zannettino AC, Hay SJ, et al. Molecular and cellular characterisation of highly purified stromal stem cells derived from human bone marrow. J Cell Sci. 2003a;116:1827–35.
    1. Quirici N, Soligo D, Bossolasco P, et al. Isolation of bone marrow mesenchymal stem cells by anti-nerve growth factor receptor antibodies. Exp Hematol. 2002;30:783–91.
    1. Miura Y, Miura M, Gronthos S, et al. Defective osteogenesis of the stromal stem cells predisposes CD18-null mice to osteoporosis. Proc Natl Acad Sci USA. 2005;102:14022–7.
    1. Gang EJ, Bosnakovski D, Figueiredo CA, et al. SSEA-4 identifies mesenchymal stem cells from bone marrow. Blood. 2007;109:1743–51.
    1. Kuznetsov SA, Mankani MH, Gronthos S, et al. Circulating skeletal stem cells. J Cell Biol. 2001;153:1133–40.
    1. Rosada C, Justesen J, Melsvik D, et al. The human umbilical cord blood: a potential source for osteoblast progenitor cells. Calcif Tissue Int. 2003;72:135–42.
    1. De Bari C, Dell’Accio F, Tylzanowski P, et al. Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis Rheum. 2001;44:1928–42.
    1. Miura M, Gronthos S, Zhao MR, et al. SHED: stem cells from human exfoliated deciduous teeth. Proc Natl Acad Sci USA. 2003;100:5807–12.
    1. De Coppi P, Bartsch G, Jr, Siddiqui MM, et al. Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol. 2007;25:100–6.
    1. Wagner W, Wein F, Seckinger A, et al. Comparative characteristics of mesenchymal stem cells from human bone marrow, adipose tissue, and umbilical cord blood. Exp Hematol. 2005;33:1402–16.
    1. Bianco P, Riminucci M, Gronthos S, et al. Bone marrow stromal stem cells: nature, biology, and potential applications. Stem Cells. 2001;19:180–92.
    1. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–7.
    1. Justesen J, Stenderup K, Eriksen EF, et al. Maintenance of osteoblastic and adipocytic differentiation potential with age and osteoporosis in human marrow stromal cell cultures. Calcif Tissue Int. 2002;71:36–44.
    1. Abdallah BM, Kassem M. Human mesenchymal stem cells: from basic biology to clinical applications. Gene Ther. 2008;15:109–16.
    1. Kuznetsov SA, Krebsbach PH, Satomura K, et al. Single-colony derived strains of human marrow stromal fibroblasts form bone after transplantation in vivo. J Bone Miner Res. 1997;12:1335–47.
    1. Piersanti S, Sacchetti B, Funari A, et al. Lentiviral transduction of human postnatal skeletal (stromal, mesenchymal) stem cells: in vivo transplantation and gene silencing. Calcif Tissue Int. 2006;78:372–84.
    1. Larsen KH, Frederiksen CM, Burns JS, et al. Identifying a molecular phenotype for bone marrow stromal cells with in vivo bone-forming capacity. J Bone Miner Res. 2010;25:796–808.
    1. Komori T, Yagi H, Nomura S, et al. Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell. 1997;89:755–64.
    1. Nakashima K, Zhou X, Kunkel G, et al. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell. 2002;108:17–29.
    1. Qiu W, Andersen TE, Bollerslev J, et al. Patients with high bone mass phenotype exhibit enhanced osteoblast differentiation and inhibition of adipogenesis of human mesenchymal stem cells. J Bone Miner Res. 2007;22:1720–31.
    1. Lecka-Czernik B, Moerman EJ, Grant DF, et al. Divergent effects of selective peroxisome proliferator-activated receptor-gamma 2 ligands on adipocyte versus osteoblast differentiation. Endocrinology. 2002;143:2376–84.
    1. Ng LJ, Wheatley S, Muscat GE, et al. SOX9 binds DNA, activates transcription, and coexpresses with type II collagen during chondrogenesis in the mouse. Dev Biol. 1997;183:108–21.
    1. Luu HH, Song WG, Luo X, et al. Distinct roles of bone morphogenetic proteins in osteogenic differentiation of mesenchymal stem cells. J Orthop Res. 2007;25:665–77.
    1. Gaur T, Lengner CJ, Hovhannisyan H, et al. Canonical WNT signaling promotes osteogenesis by directly stimulating Runx2 gene expression. J Biol Chem. 2005;280:33132–40.
    1. Abdallah BM, Jensen CH, Gutierrez G, et al. Regulation of human skeletal stem cells differentiation by Dlk1/Pref-1. J Bone Miner Res. 2004;19:841–52.
    1. Rifas L. The role of noggin in human mesenchymal stem cell differentiation. J Cell Biochem. 2007;100:824–34.
    1. Kratchmarova I, Blagoev B, Haack-Sorensen M, et al. Mechanism of divergent growth factor effects in mesenchymal stem cell differentiation. Science. 2005;308:1472–7.
    1. Barrett-Connor E, Kritz-Silverstein D. Does hyperinsulinemia preserve bone. Diabetes Care. 1996;19:1388–92.
    1. Mukherjee A, Murray RD, Shalet SM. Impact of growth hormone status on body composition and the skeleton. Horm Res. 2004;62:35–41.
    1. Takeda S, Elefteriou F, Levasseur R, et al. Leptin regulates bone formation via the sympathetic nervous system. Cell. 2002;111:305–17.
    1. Chan GK, Duque G. Age-related bone loss: old bone, new facts. Gerontology. 2002;48:62–71.
    1. Tchkonia T, Giorgadze N, Pirtskhalava T, et al. Fat depot origin affects adipogenesis in primary cultured and cloned human preadipocytes. Am J Physiol Regul Integr Comp Physiol. 2002;282:R1286–96.
    1. Hong JH, Hwang ES, McManus MT, et al. TAZ, a transcriptional modulator of mesenchymal stem cell differentiation. Science. 2005;309:1074–8.
    1. Akune T, Ohba S, Kamekura S, et al. PPARgamma insufficiency enhances osteogenesis through osteoblast formation from bone marrow progenitors. J Clin Invest. 2004;113:846–55.
    1. Sabatakos G, Sims NA, Chen J, et al. Overexpression of DeltaFosB transcription factor(s) increases bone formation and inhibits adipogenesis. Nat Med. 2000;6:985–90.
    1. Takada I, Kouzmenko AP, Kato S. Wnt and PPARgamma signaling in osteoblastogenesis and adipogenesis. Nat Rev Rheumatol. 2009;5:442–7.
    1. Gimble JM, Zvonic S, Floyd ZE, et al. Playing with bone and fat. J Cell Biochem. 2006;98:251–66.
    1. Rosen CJ, Bouxsein ML. Mechanisms of disease: is osteoporosis the obesity of bone. Nat Clin Pract Rheumatol. 2006;2:35–43.
    1. Lee NK, Sowa H, Hinoi E, et al. Endocrine regulation of energy metabolism by the skeleton. Cell. 2007;130:456–69.
    1. Ferron M, Hinoi E, Karsenty G, et al. Osteocalcin differentially regulates beta cell and adipocyte gene expression and affects the development of metabolic diseases in wild-type mice. Proc Natl Acad Sci USA. 2008;105:5266–70.
    1. Abdallah BM, Ding M, Jensen CH, et al. Dlk1/FA1 is a novel endocrine regulator of bone and fat mass and its serum level is modulated by growth hormone. Endocrinology. 2007a;148:3111–21.
    1. Abdallah BM, Boissy P, Tan Q, et al. dlk1/FA1 regulates the function of human bone marrow mesenchymal stem cells by modulating gene expression of pro-inflammatory cytokines and immune response-related factors. J Biol Chem. 2007b;282:7339–51.
    1. Compston JE. Sex steroids and bone. Physiol Rev. 2001;81:419–47.
    1. Douchi T, Oki T, Nakamura S, et al. The effect of body composition on bone density in pre- and postmenopausal women. Maturitas. 1997;27:55–60.
    1. Ray R, Novotny NM, Crisostomo PR, et al. Sex steroids and stem cell function. Mol Med. 2008;14:493–501.
    1. Heim M, Frank O, Kampmann G, et al. The phytoestrogen genistein enhances osteogenesis and represses adipogenic differentiation of human primary bone marrow stromal cells. Endocrinology. 2004;145:848–59.
    1. Ramalho AC, Jullienne A, Couttet P, et al. Effect of oestradiol on cytokine production in immortalized human marrow stromal cell lines. Cytokine. 2001;16:126–30.
    1. Hong L, Colpan A, Peptan IA. Modulations of 17-beta estradiol on osteogenic and adipogenic differentiations of human mesenchymal stem cells. Tissue Eng. 2006;12:2747–53.
    1. Leskelä HV, Olkku A, Lehtonen S, et al. Estrogen receptor alpha genotype confers interindividual variability of response to estrogen and testosterone in mesenchymal-stem-cell-derived osteoblasts. Bone. 2006;39:1026–34.
    1. Boissy P, Andersen TL, Abdallah BM, et al. Resveratrol inhibits myeloma cell growth, prevents osteoclast formation, and promotes osteoblast differentiation. Cancer Res. 2005;65:9943–52.
    1. Dai Z, Li Y, Quarles LD, et al. Resveratrol enhances proliferation and osteoblastic differentiation in human mesenchymal stem cells via ER-dependent ERK1/2 activation. Phytomedicine. 2007;14:806–14.
    1. Pino AM, Rodriguez JM, Rios S, et al. Aromatase activity of human mesenchymal stem cells is stimulated by early differentiation, vitamin D and leptin. J Endocrinol. 2006;191:715–25.
    1. Sillat T, Pöllänen R, Lopes JR, et al. Intracrine androgenic apparatus in human bone marrow stromal cells. J Cell Mol Med. 2009;13:3296–302.
    1. Compston J. Local biosynthesis of sex steroids in bone. J Clin Endocrinol Metab. 2002;87:5398–400.
    1. Brockstedt H, Kassem M, Eriksen EF, et al. Age- and sex-related changes in iliac cortical bone mass and remodelling. Bone. 1993;14:681–91.
    1. Stenderup K, Justesen J, Eriksen EF, et al. Number and proliferative capacity of osteogenic stem cells are maintained during aging and in patients with osteoporosis. J Bone Miner Res. 2001;16:1120–9.
    1. Stenderup K, Justesen J, Clausen C, et al. Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells. Bone. 2003;33:919–26.
    1. Abdallah BM, Haack-Sorensen M, Fink T, et al. Inhibition of osteoblast differentiation but not adipocyte differentiation of mesenchymal stem cells by sera obtained from aged females. Bone. 2006;39:181–8.
    1. Simonsen JL, Rosada C, Serakinci N, et al. Telomerase expression extends the proliferative life-span and maintains the osteogenic potential of human bone marrow stromal cells. Nat Biotechnol. 2002;20:592–6.
    1. Gronthos S, Chen SQ, Wang CY, et al. Telomerase accelerates osteogenesis of bone marrow stromal stem cells by upregulation of CBFA1, osterix, and osteocalcin. J Bone Miner Res. 2003b;18:716–22.
    1. Bianco P, Robey PG. Stem cells in tissue engineering. Nature. 2001;414:118–21.
    1. Guillot PV, Cui W, Fisk NM, et al. Stem cell differentiation and expansion for clinical applications of tissue engineering. J Cell Mol Med. 2007;11:935–44.
    1. Kaviani A, Perry TE, Dzakovic A, et al. The amniotic fluid as a source of cells for fetal tissue engineering. J Pediatr Surg. 2001;36:1662–5.
    1. Kaviani A, Perry TE, Barnes CM, et al. The placenta as a cell source in fetal tissue engineering. J Pediatr Surg. 2002;37:995–9.
    1. Murry CE, Keller G. Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell. 2008;132:661–80.
    1. Quintin A, Schizas C, Scaletta C, et al. Isolation and in vitro chondrogenic potential of human foetal spine cells. J Cell Mol Med. 2009;13:2559–69.
    1. Tobita M, Uysal AC, Ogawa R, et al. Periodontal tissue regeneration with adipose-derived stem cells. Tissue Eng Part A. 2008;14:945–53.
    1. Hohlfeld J, de Buys Roessingh A, Hirt-Burri N, et al. Tissue engineered fetal skin constructs for paediatric burns. Lancet. 2005;366:840–2.
    1. Kent J, Pfeffer N. Regulating the collection and use of fetal stem cells. BMJ. 2006;332:866–7.
    1. Applegate LA, Scaletta C, Hirt-Burri N, et al. Whole-cell bioprocessing of human fetal cells for tissue engineering of skin. Skin Pharmacol Physiol. 2009;22:63–73.
    1. Hirt-Burri N, de Buys Roessingh AS, Scaletta C, et al. Human muscular fetal cells: a potential cell source for muscular therapies. Pediatr Surg Int. 2008;24:37–47.
    1. Pioletti DP, Montjovent MO, Zambelli PY, et al. Bone tissue engineering using foetal cell therapy. Swiss Med Wkly. 2006;136:557–60.
    1. Bruder SP, Fink DJ, Caplan AI. Mesenchymal stem cells in bone development, bone repair, and skeletal regeneration therapy. J Cell Biochem. 1994;56:283–94.
    1. Connolly JF, Guse R, Tiedeman J, et al. Autologous marrow injection as a substitute for operative grafting of tibial nonunions. Clin Orthop Relat Res. 1991;266:259–70.
    1. Kruyt M, De Bruijn J, Rouwkema J Van Bliterswijk C, et al. Analysis of the dynamics of bone formation, effect of cell seeding density, and potential of allogeneic cells in cell-based bone tissue engineering in goats. Tissue Eng A. 2008;14:1081–8.
    1. Connolly J, Guse R, Lippiello L, et al. Development of an osteogenic bone-marrow preparation. J Bone Joint Surg Am. 1989;71:684–91.
    1. Barry FP, Murphy JM. Mesenchymal stem cells: clinical applications and biological characterization. Int J Biochem Cell Biol. 2004;36:568–84.
    1. Montjovent MO, Bocelli-Tyndall C, Scaletta C, et al. In vitro characterization of immune-related properties of human fetal bone cells for potential tissue engineering applications. Tissue Eng A. 2009;15:1523–32.
    1. Quintin A, Hirt-Burri N, Scaletta C, et al. Consistency and safety of cell banks for research and clinical use: preliminary analysis of fetal skin banks. Cell Transplant. 2007;16:675–84.
    1. Mirmalek-Sani SH, Tare RS, Morgan SM, et al. Characterization and multipotentiality of human fetal femur-derived cells: implications for skeletal tissue regeneration. Stem Cells. 2006;24:1042–53.
    1. Montjovent MO, Burri N, Mark S, et al. Fetal bone cells for tissue engineering. Bone. 2004;35:1323–33.
    1. Montjovent MO, Mathieu L, Hinz B, et al. Biocompatibility of bioresorbable poly(L-lactic acid) composite scaffolds obtained by supercritical gas foaming with human fetal bone cells. Tissue Eng. 2005;11:1640–9.
    1. SwissMedics. Swiss Federal Council Transplantation Law. TxL;SR 810.21, 2007.
    1. EU. Community code relating to medicinal products for hum use Parliament E editor. Directive 2001/83/EC, 2001.
    1. Parliament E, editor. EU, Implementing Directive 2004/23/EC of the European Parliament and of the Council as regards certain technical requirements for the donation, procurement and testing of human tissues and cells. editor. Directive 2006/17/EC, 2006.
    1. Parliament E, editor. EU, Implementing Directive 2004/23/EC of the European Parliament and of the Council as regards traceability requirements, notification of serious adverse reactions and events and certain technical requirements for the coding, processing, preservation, storage and distribution of human tissues and cells. editor. Directive 2006/86/EC, 2006.
    1. EU. Advanced therapy medicinal products amending Directive 2001/83/EC and Regulation (EC) No 726/2004, Parliament, editor. Regulation (EC) No 1394/2007, 2007.
    1. FDA. Human cells, tissues, and cellular and tissue-based products. 21 CFR 1271, 2006.
    1. EU. Setting standards of quality and safety for the collection, testing, processing, storage and distribution of human blood and blood components, Directive 2002/98/EC, 2002.
    1. EU. Setting standards of quality and safety for the donation, procurement, testing, processing, preservation, storage and distribution of human tissues and cells Parliament E editor. Directive 2004/23/EC, 2004.
    1. Heinonen M, Oila O, Nordstrom K. Current issues in the regulation of human tissue-engineering products in the European Union. Tissue Eng. 2005;11:1905–11.
    1. Trommelmans L, Selling J, Dierickx K. A critical assessment of the directive on tissue engineering of the European union. Tissue Eng. 2007;13:667–72.
    1. Marx RE, Carlson ER, Eichstaedt RM, et al. Platelet-rich plasma: growth factor enhancement for bone grafts. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 1998;85:638–46.
    1. Foster TE, Puskas BL, Mandelbaum BR, et al. Platelet-rich plasma: from basic science to clinical applications. Am J Sports Med. 2009;37:2259–72.
    1. Mazzucco L, Balbo V, Cattana E, et al. Not every PRP-gel is born equal. Evaluation of growth factor availability for tissues through four PRP-gel preparations: fibrinet, RegenPRP-Kit, Plateltex and one manual procedure. Vox Sang. 2009;97:110–8.
    1. Dohan DM, Choukrou J, Diss A, et al. Platelet-rich fibrin (PRF): a second-generation platelet concentrate. Part I: technological concepts and evolution. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2006a;101:e37–44.
    1. Anitua E, Sánchez M, Orive G, et al. The potential impact of the preparation rich in growth factors (PRGF) in different medical fields. Biomaterials. 2007;28:4551–60.
    1. Mazzucco L, Balbo V, Cattana E, et al. Platelet-rich plasma and platelet gel preparation using Plateltex. Vox Sang. 2008;94:202–8.
    1. Anitua E, Aguirre JJ, Algorta J, et al. Effectiveness of autologous preparation rich in growth factors for the treatment of chronic cutaneous ulcers. J Biomed Mater Res B Appl Biomater. 2008;84:415–21.
    1. Moojen DJ, Everts PA, Schure RM, et al. Antimicrobial activity of platelet-leukocyte gel against Staphylococcus aureus. J Orthop Res. 2008;26:404–10.
    1. Dohan DM, Choukroun J, Diss A, et al. Platelet-rich fibrin (PRF): a second-generation platelet concentrate. Part III: leucocyte activation: a new feature for platelet concentrates. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2006b;101:e51–55.
    1. Vogel JP, Szalay K, Geiger F, et al. Platelet-rich plasma improves expansion of human mesenchymal stem cells and retains differentiation capacity and in vivo bone formation in calcium phosphate. Platelets. 2006;17:462–9.
    1. Lucarelli E, Beccheroni A, Donati D, et al. Platelet-derived growth factors enhance proliferation of human stromal stem cells. Biomaterials. 2003;24:3095–100.
    1. Gruber R, Varga F, Fischer MB, et al. Platelets stimulate proliferation of bone cells: involvement of platelet-derived growth factor, microparticles and membranes. Clin Oral Implants Res. 2002a;13:529–35.
    1. Ogino Y, Ayukawa Y, Kukita T, et al. The contribution of platelet-derived growth factor, transforming growth factor-beta1, and insulin-like growth factor-I in platelet-rich plasma to the proliferation of osteoblast-like cells. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2006;101:724–9.
    1. Gruber R, Karreth F, Kandler B, et al. Platelet-released supernatants increase migration and proliferation, and decrease osteogenic differentiation of bone marrow-derived mesenchymal progenitor cells under in vitro conditions. Platelets. 2004;15:29–35.
    1. Gruber R, Kandler B, Fischer MB, et al. Osteogenic differentiation induced by bone morphogenetic proteins can be suppressed by platelet-released supernatant in vitro. Clin Oral Implants Res. 2006;17:188–93.
    1. He L, Lin Y, Hu X, et al. A comparative study of platelet-rich fibrin (PRF) and platelet-rich plasma (PRP) on the effect of proliferation and differentiation of rat osteoblasts in vitro. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2009;108:707–13.
    1. Butcher A, Milner R, Ellis K, et al. Interaction of platelet-rich concentrate with bone graft materials: an in vitro study. J Orthop Trauma. 2009;23:195–200.
    1. Cenni E, Perut F, Ciapetti G, et al. In vitro evaluation of freeze-dried bone allografts combined with platelet rich plasma and human bone marrow stromal cells for tissue engineering. J Mater Sci Mater Med. 2009a;20:45–50.
    1. Kasten P, Vogel J, Geiger F, et al. The effect of platelet-rich plasma on healing in critical-size long-bone defects. Biomaterials. 2008;29:3983–92.
    1. Nair MB, Varma HK, John A. Platelet-rich plasma and fibrin glue-coated bioactive ceramics enhance growth and differentiation of goat bone marrow-derived stem cells. Tissue Eng A. 2009a;15:1619–31.
    1. Zhang Z, Chen J, Jin D. Platelet-derived growth factor (PDGF)-BB stimulates osteoclastic bone resorption directly: the role of receptor beta. Biochem Biophys Res Commun. 1998;251:190–4.
    1. Niida S, Kaku M, Amano H, et al. Vascular endothelial growth factor can substitute for macrophage colony-stimulating factor in the support of osteoclastic bone resorption. J Exp Med. 1999;190:293–8.
    1. Weicht B, Maitz P, Kandler B, et al. Activated platelets positively regulate RANKL-mediated osteoclast differentiation. J Cell Biochem. 2007;102:1300–7.
    1. Gruber R, Karreth F, Fischer MB, et al. Platelet-released supernatants stimulate formation of osteoclast-like cells through a prostaglandin/RANKL-dependent mechanism. Bone. 2002b;30:726–32.
    1. Ogino Y, Ayukawa Y, Kukita T, et al. Platelet-rich plasma suppresses osteoclastogenesis by promoting the secretion of osteoprotegerin. J Periodontal Res. 2009;44:217–24.
    1. Cenni E, Avnet S, Fotia C, et al. Platelet-rich plasma impairs osteoclast generation from human precursors of peripheral blood. J Orthop Res. 2010;28:792–7.
    1. Gerard D, Carlson ER, Gotcher JE, et al. Effects of platelet-rich plasma at the cellular level on healing of autologous bone-grafted mandibular defects in dogs. J Oral Maxillofac Surg. 2007;65:721–7.
    1. Pilkington MF, Sims SM, Dixon SJ. Transforming growth factor-β induces osteoclast ruffling and chemotaxis: potential role in osteoclast recruitment. J Bone Miner Res. 2001;16:1237–47.
    1. Takai H, Kanematsu M, Yano K, et al. Transforming growth factor-β stimulates the production of osteoprotegerin/osteoclastogenesis inhibitory factor by bone marrow stromal cells. J Biol Chem. 1998;273:27091–6.
    1. Towler DA. The osteogenic-angiogenic interface: novel insights into the biology of bone formation and fracture repair. Curr Osteoporos Rep. 2008;6:67–71.
    1. Brandi ML, Collin-Osdoby P. Vascular biology and the skeleton. J Bone Miner Res. 2006;21:183–92.
    1. von Schroeder HP, Veillette CJ, Payandeh J, et al. Endothelin-1 promotes osteoprogenitor proliferation and differentiation in fetal rat calvarial cell cultures. Bone. 2003;33:673–84.
    1. Bouletreau PJ, Warren SM, Spector JA, et al. Hypoxia and VEGF up-regulate BMP-2 mRNA and protein expression in microvascular endothelial cells: implications for fracture healing. Plast Reconstr Surg. 2002;109:2384–97.
    1. Bertrand-Duchesne MP, Grenier D, Gagnon G. Epidermal growth factor released from platelet-rich plasma promotes endothelial cell proliferation in vitro. J Periodontal Res. 2009;45:87–93.
    1. Kilian O, Flesch I, Wenisch S, et al. Effects of platelet growth factors on human mesenchymal stem cells and human endothelial cells in vitro. Eur J Med Res. 2004;9:337–44.
    1. Kandler B, Fischer MB, Watzek G, et al. Platelet-released supernatant increases matrix metalloproteinase-2 production, migration, proliferation and tube formation of human umbilical vascular endothelial cells. J Periodontol. 2004;75:1255–61.
    1. Bir SC, Esaki J, Marui A, et al. Angiogenic properties of sustained release platelet-rich plasma: characterization in-vitro and in the ischemic hind limb of the mouse. J Vasc Surg. 2009;50:870–9.
    1. Fiedler J, Etzel N, Brenner RE. To go or not to go: migration of human mesenchymal progenitor cells stimulated by isoforms of PDGF. J Cell Biochem. 2004;93:990–8.
    1. Cenni E, Ciapetti G, Granchi D, et al. Endothelial cells incubated with platelet-rich plasma express PDGF-B and ICAM-1 and induce bone marrow stromal cell migration. J Orthop Res. 2009b;27:1493–8.
    1. Aghaloo TL, Moy PK, Freymiller EG. Evaluation of platelet-rich plasma in combination with anorganic bovine bone in the rabbit cranium: a pilot study. Int J Oral Maxillofac Implants. 2004;19:59–65.
    1. Jensen TB, Rahbek O, Overgaard S, et al. Platelet rich plasma and fresh frozen bone allograft as enhancement of implant fixation. An experimental study in dogs. J Orthop Res. 2004;22:653–8.
    1. Roldán JC, Jepsen S, Miller J, et al. Bone formation in the presence of platelet-rich plasma versus bone morphogenetic protein-7. Bone. 2004;34:80–90.
    1. Yamada Y, Ueda M, Hibi H, et al. Translational research for injectable tissue-engineered bone regeneration using mesenchymal stem cells and platelet-rich plasma: from basic research to clinical case study. Cell Transplant. 2004a;4:343–55.
    1. Dallari D, Fini M, Stagni C, et al. In vivo study on the healing of bone defects treated with bone marrow stromal cells, platelet-rich plasma, and freeze-dried bone allografts, alone and in combination. J Orthop Res. 2006;24:877–88.
    1. Nagata M, Messora M, Okamoto R, et al. Influence of the proportion of particulate autogenous bone graft/platelet-rich plasma on bone healing in critical-size defects: an immunohistochemical analysis in rat calvaria. Bone. 2009a;45:339–45.
    1. Suba Z, Takacs D, Gyulai-Gaal S, et al. Facilitation of β-tricalcium phosphate-induced alveolar bone regeneration by platelet-rich plasma in beagle dogs: a histologic and histomorphometric study. Int J Oral Maxillofac Implants. 2004;19:832–8.
    1. Nair MB, Varma HK, Menon KV, et al. Reconstruction of goat femur segmental defects using triphasic ceramic-coated hydroxyapatite in combination with autologous cells and platelet-rich plasma. Acta Biomater. 2009b;5:1742–55.
    1. Geuze RE, Everts PA, Kruyt MC. Orthotopic location has limited benefit from allogeneic or autologous multipotent stromal cells seeded on ceramic scaffolds. Tissue Engineering A. 2009;15:3231–9.
    1. Lieberman JR, Daluiski A, Einhorn TA. The role of growth factors in the repair of bone. Biology and clinical applications. J Bone Joint Surg Am. 2002;84-A:1032–44.
    1. Yamada Y, Ueda M, Naiki T, et al. Autogenous injectable bone for regeneration with mesenchymal stem cells and platelet-rich plasma: tissue-engineered bone regeneration. Tissue Eng. 2004b;10:955–64.
    1. Ito K, Yamada Y, Naiki T, et al. Simultaneous implant placement and bone regeneration around dental implants using tissue-engineered bone with fibrin glue, mesenchymal stem cells and platelet-rich plasma. Clin Oral Implants Res. 2006;5:579–86.
    1. Fuerst G, Gruber R, Tangl S, et al. Enhanced bone-to-implant contact by platelet-released growth factors in mandibular cortical bone: a histomorphometric study in minipigs. Int J Oral Maxillofac Implants. 2003;18:685–90.
    1. Nagata MJ, Melo LG, Messora MR, et al. Effect of platelet-rich plasma on bone healing of autogenous bone grafts in critical-size defects. J Clin Periodontol. 2009b;36:775–83.
    1. Wiltfang J, Kloss FR, Kessler P, et al. Effects of platelet-rich plasma on bone healing in combination with autogenous bone and bone substitutes in critical-size defects. Clin Oral Implants Res. 2004;15:187–93.
    1. Fennis JP, Stoelinga PJ, Jansen JA. Mandibular reconstruction: a histological and histomorphometric study on the use of autogenous scaffolds, particulate cortico-cancellous bone grafts and platelet rich plasma in goats. Int J Oral Maxillofac Surg. 2004;33:48–55.
    1. Choi BH, Im CJ, Huh JY, et al. Effect of platelet-rich plasma on bone regeneration in autogenous bone graft. Int J Oral Maxillofac Surg. 2004;33:56–9.
    1. Thor A, Franke-Stenport V, Johansson CB, et al. Early bone formation in human bone grafts treated with platelet-rich plasma: preliminary histomorphometric results. Int J Oral Maxillofac Surg. 2007;36:1164–71.
    1. Intini G. The use of platelet-rich plasma in bone reconstruction therapy. Biomaterials. 2009;30:4956–66.
    1. Alsousou J, Thompson M, Hulley P, et al. The biology of platelet-rich plasma and its application in trauma and orthopaedic surgery: a review of the literature. J Bone Joint Surg Br. 2009;91:987–96.
    1. Galasso O, Mariconda M, Romano G, et al. Expandable intramedullary nailing and platelet rich plasma to treat long bone non-unions. J Orthop Traumatol. 2008;9:129–34.
    1. Kitoh H, Kawasumi M, Kaneko H, et al. Differential effects of culture-expanded bone marrow cells on the regeneration of bone between the femoral and the tibial lengthenings. J Pediatr Orthop. 2009;29:643–9.
    1. Gandhi A, Bibbo C, Pinzur M, et al. The role of platelet-rich plasma in foot and ankle surgery. Foot Ankle Clin. 2005;10:621–37.
    1. Bielecki T, Gazdzik TS, Szczepanski T. Benefit of percutaneous injection of autologous platelet-leukocyte-rich gel in patients with delayed union and nounion. Eur Surg Res. 2008;40:289–96.
    1. Mariconda M, Cozzolino F, Cozzolino A, et al. Platelet gel supplementation in long bone nonunions treated by external fixation. J Orthop Trauma. 2008;22:342–5.
    1. Calori GM, Tagliabue L, Gala L, et al. Application of rhBMP-7 and platelet-rich plasma in the treatment of long bone non-unions: a prospective randomised clinical study on 120 patients. Injury. 2008;39:1391–402.
    1. Kitoh H, Kitakoji T, Tsuchiya H, et al. Transplantation of culture expanded bone marrow cells and platelet rich plasma in distraction osteogenesis of the long bones. Bone. 2007;40:522–8.
    1. Jenis LG, Banco RJ, Kwon B. A prospective study of autologous growth factors (AGF) in lumbar interbody. Spine J. 2006;6:14–20.
    1. Carreon LY, Glassman SD, Anekstein Y, et al. Platelet gel (AGF) fails to increase fusion rates in instrumented posterolateral fusions. Spine. 2005;30:E243–6.
    1. Coetzee JC, Pomeroy GC, Watts JD, et al. The use of autologous concentrated growth factors to promote syndesmosis fusion in the Agility total ankle replacement. A preliminary study. Foot Ankle Int. 2005;26:840–6.
    1. Barrow CR, Pomeroy GC. Enhancement of syndesmotic fusion rates in total ankle arthroplasty with the use of autologous platelet concentrate. Foot Ankle Int. 2005;26:458–61.
    1. Bibbo C, Bono CM, Lin SS. Union rates using autologous platelet concentrate alone and with bone graft in high-risk foot and ankle surgery patients. J Surg Orthop Adv. 2005;14:17–22.
    1. Berghoff W, Pietrzak W, Rhodes R. Platelet-rich plasma application during closure following total knee arthroplasty. Orthopedics. 2006;29:590–8.
    1. Yamada Y, Ueda M, Naiki T, et al. Tissue-engineered injectable bone regeneration for osseointegrated dental implants. Clin Oral Implants Res. 2004c;5:589–597.
    1. Ueda M, Yamada Y, Ozawa R, et al. Clinical case reports of injectable tissue-engineered bone for alveolar augmentation with simultaneous implant placement. Int J Periodont Restorative Dent. 2005;2:129–37.
    1. Marx RE. Reconstruction of defects caused by bisphosphonate-induced osteonecrosis of the jaws. J Oral Maxillofac Surg. 2009;67:107–19.
    1. Plachokova AS, Nikolidakis D, Mulder J, et al. Effect of platelet-rich plasma on bone regeneration in dentistry: a systematic review. Clin Oral Implants Res. 2008;19:539–45.
    1. Griffin XL, Smith CM, Costa ML. The clinical use of platelet-rich plasma in the promotion of bone healing: a systematic review. Injury. 2009;40:158–62.
    1. Dallari D, Savarino L, Stagni C, et al. Enhanced tibial osteotomy healing with use of bone grafts supplemented with platelet gel or platelet gel and bone marrow stromal cells. J Bone Joint Surg Am. 2007;89:2413–20.
    1. Smith SE, Roukis TS. Bone and wound healing augmentation with platelet-rich plasma. Clin Podiatr Med Surg. 2009;26:559–88.
    1. Li X, Quigg RJ, Zhou J, et al. Early signals for fracture healing. J Cell Biochem. 2005;95:189–205.
    1. Li X, Wang H, Touma E, et al. Genetic network and pathway analysis of differentially expressed proteins during critical cellular events in fracture repair. J Cell Biochem. 2007;100:527–43.
    1. Deschaseaux F, Sensébé L, Heymann D. Mechanisms of bone repair and regeneration. Trends Mol Med. 2009;15:417–29.
    1. Kulterer B, Friedl G, Jandrositz A, et al. Gene expression profiling of human mesenchymal stem cells derived from bone marrow during expansion and osteoblast differentiation. BMC Genomics. 2007;8:70–85.
    1. Granchi D, Ochoa G, Leonardi E, et al. Gene expression patterns related to osteogenic differentiation of bone marrow-derived mesenchymal stem cells during ex vivo expansion. Tissue Eng Part C Methods. 2010;16:511–24.
    1. Gronthos S, Simmons PJ, Graves SE, et al. Integrin-mediated interactions between human bone marrow stromal precursor cells and the extracellular matrix. Bone. 2001;28:174–81.
    1. Hadjiargyrou M, Lombardo F, Zhao S, et al. Transcriptional profiling of bone regeneration. Insight into the molecular complexity of wound repair. J Biol Chem. 2002;277:30177–82.
    1. Huang W, Yang S, Shao J, et al. Signaling and transcriptional regulation in osteoblast commitment and differentiation. Front Biosci. 2007;12:3068–92.
    1. Civitelli R. Cell-cell communication in the osteoblast/osteocyte lineage. Arch Biochem Biophys. 2008;473:188–92.
    1. Deng ZL, Sharff KA, Tang N, et al. Regulation of osteogenic differentiation during skeletal development. Front Biosci. 2008;13:2001–21.
    1. Papachroni KK, Karatzas DN, Papavassiliou KA, et al. Mechanotransduction in osteoblast regulation and bone disease. Trends Mol Med. 2009;15:208–16.
    1. Dimitriou R, Tsiridis E, Giannoudis PV. Current concepts of molecular aspects of bone healing. Injury. 2005;36:1392–404.
    1. Chen D, Zhao M, Mundy GR. Bone morphogenetic proteins. Growth Factors. 2004;22:233–41.
    1. Wan M, Cao X. BMP signaling in skeletal development. Biochem Biophys Res Commun. 2005;328:651–7.
    1. Lai CF, Cheng SL. Signal transductions induced by bone morphogenetic protein-2 and transforming growth factor-beta in normal human osteoblastic cells. J Biol Chem. 2002;277:15514–22.
    1. Su N, Du X, Chen L. FGF signaling: its role in bone development and human skeleton diseases. Front Biosci. 2008;13:2842–65.
    1. Jacob AL, Smith C, Partanen J, et al. Fibroblast growth factor receptor 1 signaling in the osteochondrogenic cell lineage regulates sequential steps of osteoblast maturation. Dev Biol. 2006;296:315–28.
    1. Montero A, Okada Y, Tomita M, et al. Disruption of the fibroblast growth factor-2 gene results in decreased bone mass and bone formation. J Clin Invest. 2000;105:1085–93.
    1. Ornitz DM. FGF signaling in the developing endochondral skeleton. Cytokine Growth Factor Rev. 2005;16:205–13.
    1. Giustina A, Mazziotti G, Canalis E. Growth hormone, insulin-like growth factors, and the skeleton. Endocr Rev. 2008;29:535–59.
    1. Nakae J, Yoshiaki Kido Y, Accili D. Distinct and overlapping functions of Insulin and IGF-I receptors. Endocrine Reviews. 2001;22:818–35.
    1. Tokunaga A, Oya T, Ishii Y, et al. PDGF receptor beta is a potent regulator of mesenchymal stromal cell function. J Bone Miner Res. 2008;23:1519–28.
    1. Kumar A, Salimath BP, Stark GB, et al. Platelet-derived growth factor receptor signaling is not involved in osteogenic differentiation of human mesenchymal stem cells. Tissue Eng A. 2010;16:983–93.
    1. Chang L, Karin M. Mammalian MAP kinase signaling cascades. Nature. 2001;410:37–40.
    1. Westendorf JJ, Kahler RA, Schroeder TM. Wnt signaling in osteoblasts and bone diseases. Gene. 2004;341:19–39.
    1. Jackson A, Vayssiere B, Garcia T, et al. Gene array analysis of Wnt-regulated genes in C3H10T1/2 cells. Bone. 2005;36:585–98.
    1. Fujita K, Janz S. Attenuation of WNT signaling by DKK-1 and -2 regulates BMP2-induced osteoblast differentiation and expression of OPG, RANKL and M-CSF. Mol Cancer. 2007;6:71–83.
    1. Day TF, Guo X, Garrett-Beal L, Yang Y. Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev Cell. 2005;8:739–50.
    1. Chung UI, Schipani E, McMahon AP, et al. Indian hedgehog couples chondrogenesis to osteogenesis in endochondral bone development. J Clin Invest. 2001;107:295–304.
    1. Long F, Chung UI, Ohba S, et al. Ihh signaling is directly required for the osteoblast lineage in the endochondral skeleton. Development. 2004;131:1309–18.
    1. Day TF, Yang Y. Wnt and hedgehog signaling pathways in bone development. J Bone Joint Surg Am. 2008;90:19–24.
    1. Rodda SJ, McMahon AP. Distinct roles for Hedgehog and canonical Wnt signaling in specification, differentiation and maintenance of osteoblast progenitors. Development. 2006;133:3231–44.
    1. Canalis E. Notch signaling in osteoblasts. Sci Signal. 2008;1:1–4.
    1. Chau JF, Leong WF, Li B. Signaling pathways governing osteoblast proliferation, differentiation and function. Histol Histopathol. 2009;24:1593–606.
    1. Engin F, Yao Z, Yang T, et al. Dimorphic effects of Notch signaling in bone homeostasis. Nat Med. 2008;14:299–305.
    1. Mundy GR, Elefteriou F. Boning up on ephrin signaling. Cell. 2006;126:441–3.
    1. Zhao C, Irie N, Takada Y, et al. Bidirectional ephrinB2-EphB4 signaling controls bone homeostasis. Cell Metab. 2006;4:111–21.
    1. Irie N, Takada Y, Watanabe Y, et al. Bidirectional signaling through ephrinA2-EphA2 enhances osteoclastogenesis and suppresses osteoblastogenesis. J Biol Chem. 2009;284:14637–44.
    1. Marie PJ. Transcription factors controlling osteoblastogenesis. Arch Biochem Biophys. 2008;473:98–105.
    1. Lian JB, Javed A, Zaidi SK, et al. Regulatory controls for osteoblast growth and differentiation: role of Runx/Cbfa/AML factors. Crit Rev Eukaryot Gene Expr. 2004;14:1–41.
    1. Koga T, Matsui Y, Asagiri M, et al. NFAT and Osterix cooperatively regulate bone formation. Nat Med. 2005;11:880–5.
    1. Hartmann C. Transcriptional networks controlling skeletal development. Curr Opin Genet Dev. 2009;19:437–43.

Source: PubMed

3
Abonner