Skip to main content
The Open Orthopaedics Journal logoLink to The Open Orthopaedics Journal
editorial
. 2011 Jul 28;5:239–241. doi: 10.2174/1874325001105010239

Tissue Engineering in Orthopaedics and Musculoskeletal Sciences

Anant Mahapatra §,1,*, Wasim S Khan §,2,*
PMCID: PMC3149835  PMID: 21886688

Orthopaedic surgery as a speciality lends itself well to tissue engineering. Many musculoskeletal tissues are frequently damaged or lost in injury and disease, and show a limited capacity for repair. Although orthopaedic surgery has been successful in replacing cartilage, the coming years will establish whether a shift from replacement towards regeneration of tissue is feasible. There has been an increasing interest in tissue engineering approaches to deal with damaged or lost tissue [1-3]. Tissue engineering is an exciting strategy that employs stem cells, scaffolds and growth factors or mediators either in isolation or in combination. It is the expanding science of generating tissue using molecular and cellular techniques, combined with material engineering principles, to replace tissue surgically. This could be in the form of cells with or without matrices. Many tissue engineering advances are being made in orthopaedics and this is partly due to the familiarity with bone marrow derived mesenchymal stem cells. In this review we will cover mesenchymal stem cells, scaffolds, bioreactors and growth factors applicable to orthopaedics and musculoskeletal sciences. This is followed by specific discussions on Dupuytren’s disease where there is early evidence of stem cell etiology, and bone and cartilage tissue engineering.

STEM CELLS

Stem cells are a self-renewing cell population with a high proliferation potential and the ability to undergo chondrogenic, osteogenic and adipogenic differentiation. Protocols for the culture and differentiation of bone marrow derived mesenchymal stem cells have been described [4-6]. Over the last decade, there has been considerable interest in the use of mesenchymal stem cells and tissue engineering principles in orthopaedics and musculoskeletal sciences. The use of autologous postnatal mesenchymal stem cells is generally well accepted by society and does not involve many of the ethical, political and religious considerations that surround embryonic stem cells [7]. Cells with mesenchymal stem cell characteristics have been isolated from many different adult tissues including bone marrow, periosteum, skin, adipose tissue, skeletal muscle, synovial tissue, the infrapatellar fat pad, and more recently, cartilage [8-15], and we provide a systematic review of the literature outlining the sources of mesenchymal stem cell used in musculoskeletal applications.

Human bone marrow-derived mesenchymal stem cells exhibit age-related changes; with increasing age, they are fewer in number [16, 17], have reduced proliferation [18] and reduced differentiation potential [19]. There is uncertainty whether these age-related changes are also exhibited by mesenchymal stem cells from other sources, and what the extent of these changes is in these other sources. There is a need to determine not only the effects of patient age on mesenchymal stem cells [20] but also the effect of senescence. There are many known growth factors involved in controlling and influencing stem cell growth that are also related to cell senescence, and we review these in vitro considerations.

Work by others and by us has shown that mesenchymal stem cells from the synovial fat pad are easier to obtain and are associated with a higher yield of mesenchymal stem cells compared to bone marrow [11, 21, 22]. Although mesenchymal stem cells from different tissues vary in their differentiation potential [23,24], it is possible that there are more similarities between stem cells from various sources than differences; increasing data supports pericytes as candidate mesenchymal stem cells in a number of sources [22,25]. These cells are identified using cell surface markers but there are no papers in the literature that bring together and summarize these cell surface markers. We present a systematic review that summarizes all the available information about the cell surface characterization of adult human MSCs by identifying and evaluating all the published literature in this field. We have found that the most commonly reported positive markers are CD105, CD90, CD44, CD73, CD29, CD13, CD34, CD146, CD106, CD54 and CD166, and the most frequently reported negative markers are CD34, CD14, CD45, CD11b, CD49d, CD106, CD10 and CD31.

SCAFFOLDS, BIOREACTORS AND GROWTH FACTORS

The use of scaffolds remains an integral part of the tissue engineering applications in orthopaedics and musculoskeletal sciences. We provide a summary of the principals of using material scaffolds in musculoskeletal tissue engineering applications and how these materials may eventually come to be incorporated in clinical practice. This is followed by a review of bioreactors that engineered tissue can be generated in. We discuss the role of bioreactors in tissue engineering and evaluate the principles of bioreactor design. We evaluate the methods of cell stimulation and review the bioreactors in common use today. Research has shown the importance of growth factors in guiding and modulating the differentiation of mesenchymal stem cells in order to obtain the required cell type. Gene-based delivery systems have aided the delivery of sustained quantities of these growth factors. The evidence from growth factor enhanced tissue engineering studies for tissue healing looks very positive. This is a multi-disciplinary approach that integrates molecular, biochemical and clinical techniques with developmental and engineering processes. Initial studies indicate an immense potential for cell based strategies to enhance current orthopaedic approaches in skeletal tissue reconstruction and this is also covered in a review.

E-GOVERNANCE

The rules of governance are changing. They are necessarily becoming more stringent as interventions offered to treat conditions carry unpredictable side effects, often associated with novel therapeutic vectors. The clinical relevance of this relates to the obligations of those involved in research, to ensure the best protection for subjects whilst encouraging the development of the field. Existing evidence supports the concept of e-Governance both in operational health research and more broadly in the strategic domain of policy formation. Building on the impact of the UK Comprehensive Research Network and recent EU Directives, it is now possible to focus on the issues of regulation for cell therapies in musculoskeletal science through the development of the Advanced Therapeutic Medicinal Products (ATMP) category of research products. We provide a review that covers the framework that has borne this and the need for more detailed Virtual Research Integration and Collaboration (VRIC) systems to ensure regulatory compliance. Technology research and development plans must develop in close association between tissue engineering and treating clinicians. The scope of this strategy relates to the handling of human tissues the transport and storage of specimens in accordance with current EU directives and the Human Tissue Authority (HTA) regulations.

DUPUYTREN’S DISEASE

Dupuytren’s disease is the most common condition seen by hand surgeons. It is not only prevalent but can also be a most debilitating condition resulting in significant loss of function of the fingers involved. The cause of this disease, however still remains largely unknown although some recent evidence suggests a stem cell etiology. We provide a review article that summarizes the current known knowledge of Dupuytren’s as well as the clinical findings, investigations, treatments available and looks at preliminary evidence of stem cell etiology.

BONE

Tissue engineering of bone has the potential to overcome the limitations of using autologous, allogeneic or synthetic bone grafts to treat extensive bone defects and fracture non-unions [26]. Over a million patients are treated annually to manage and regenerate bone tissue in sites of congenital defects, tumor resection or fractured bones [27]. This regeneration process is very complex and requires a morphogenetic signal, responsive host cells and a suitable scaffold [28]. An increasing variety of synthetic scaffolds for bone regeneration has become commercially available over the last century [27] that aim to provide a three dimensional substrate for cells to populate on and function appropriately. They should have appropriate mechanical properties, biocompatibility and biodegradability at a rate commensurate with remodelling [29].

Stem cells have shown to be excellent carriers for gene transfer having the capability to be transduced. Gene transfer could enable growth factors and bone morphogentic proteins to enhance bone repair. Stem cells are implanted onto scaffolds, which are structures capable of supporting tissue formation by allowing cell migration, proliferation and differentiation. Research aims to produce scaffolds that deliver and retain cells, allow for cell attachment has adequate biodegradability, biocompatibility and non-immunogenicity. However, having tried and testing numerous materials including synthetic and natural products research into the perfect scaffold product continues. We provide a review that aims to explain how stem cells were discovered, the techniques used to isolate stem cells, identify and manipulate them down different cell lineages and discuss the research into using stem cells to reconstruct bone using genetic modification and scaffolds.

CARTILAGE

Osteoarthritis is the most prevalent disorder of the musculoskeletal system [30] and the frequent outcome for arthritis in large joints such as the knee is surgical intervention for joint replacement. A joint replacement tends to be successful in older sedentary patients but the limited lifetime of prostheses makes it much less desirable for younger and more active patients [31]. The numbers of primary and revision total knee replacements are projected to increase six-fold by 2030 [32] and this is the driving force behind numerous ongoing efforts to develop new cell-based strategies for the treatment of focal cartilage defects to prevent secondary osteoarthritis [2]. Current options for the repair of focal cartilage lesions include abrasive chondroplasty, subchondral drilling and microfracture. These result in the formation of fibrocartilage rather than the desired hyaline cartilage, with inferior mechanical and hydroelastic characteristics [33]. Autologous Chondrocyte Implantation is a cell-based strategy currently in clinical practice [34, 35]. Although short-term clinical results have been good, evidence suggests formation of fibrocartilage and progression of degenerative changes in the joint [36].

Tissue engineering applications using mesenchymal stem cells present an interesting new approach for the repair of articular cartilage defects [2,37]. To date there have been only limited reports of human autologous bone marrow derived cell implantation for cartilage repair [38-40]. We include a review that looks at the current treatment strategies for articular cartilage, describes use of mesenchymal stem cells for articular cartilage repair along with the results of clinical studies, and describes the future direction that these strategies are likely to take.

REFERENCES

  • 1.Khan WS, Malik AA, Hardingham TE. Stem cell applications and tissue engineering approaches in surgical Practice. J Perioper Pract. 2009;19(4):130–5. doi: 10.1177/175045890901900401. [DOI] [PubMed] [Google Scholar]
  • 2.Khan WS, Johnson DS, Hardingham DS. The potential use of stem cells for knee articular cartilage repair. Knee. 2010;17(6):369–74. doi: 10.1016/j.knee.2009.12.003. [DOI] [PubMed] [Google Scholar]
  • 3.Khan WS, Marsh D. Tissue engineering approaches for bone repair: cells, scaffolds, growth factors and gene therapy. Stem Cell Res J. 2010;2(1-2):21–32. [Google Scholar]
  • 4.Freidenstein 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;4:393–403. doi: 10.1111/j.1365-2184.1970.tb00347.x. [DOI] [PubMed] [Google Scholar]
  • 5.Johnstone B, Hering TM, Caplan AI, Goldberg VM, Yoo JU. In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp Cell Res. 1998;238:265–72. doi: 10.1006/excr.1997.3858. [DOI] [PubMed] [Google Scholar]
  • 6.Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–7. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
  • 7.Raghunath J, Salacinski HJ, Sales KM, Butler PE, Seifalian M. Advancing cartilage tissue engineering: the application of stem cell technology. Curr Opin Biotech. 2005;16:503–9. doi: 10.1016/j.copbio.2005.08.004. [DOI] [PubMed] [Google Scholar]
  • 8.Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol. 2000;109:235–42. doi: 10.1046/j.1365-2141.2000.01986.x. [DOI] [PubMed] [Google Scholar]
  • 9.De Bari C, Dell’Accio F, Tylzanowski P, Luyten FP. Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis Rheum. 2001;44:1928–42. doi: 10.1002/1529-0131(200108)44:8<1928::AID-ART331>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
  • 10.Zuk PA, Zhu M, Mizino H, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7:211–28. doi: 10.1089/107632701300062859. [DOI] [PubMed] [Google Scholar]
  • 11.Dragoo JL, Samimi B, Zhu M, et al. Tissue-engineered cartilage and bone using stem cells from human infrapatellar fat pads. J Bone Joint Surg Br. 2003;85:740–7. [PubMed] [Google Scholar]
  • 12.Peng H, Huard J. Stem cells in the treatment of muscle and connective tissue diseases. Curr Opin Pharmacol. 2003;3:329–33. doi: 10.1016/s1471-4892(03)00051-1. [DOI] [PubMed] [Google Scholar]
  • 13.Wickham MQ, Erickson GR, Gimble JM, Vail TP, Guilak F. Multipotent stromal cells derived from the infrapatellar fat pad of the knee. Clin Orthop. 2003;412:196–212. doi: 10.1097/01.blo.0000072467.53786.ca. [DOI] [PubMed] [Google Scholar]
  • 14.Dowthwaite GP, Bishop JC, Redman SN, et al. The surface of articular cartilage contains a progenitor cell population. J Cell Sci. 2004;117:889–97. doi: 10.1242/jcs.00912. [DOI] [PubMed] [Google Scholar]
  • 15.Jones EA, Kinsey SE, English A, et al. Isolation and characterisation of bone marrow multipotential mesenchymal progenitor cells. Arthritis Rheum. 2002;46:3349–60. doi: 10.1002/art.10696. [DOI] [PubMed] [Google Scholar]
  • 16.Nishida S, Endo N, Yamagiwa H, Tanizawa T, Takahashi HE. Number of osteoprogenitor cells in human bone marrow markedly decreases after skeletal maturation. J Bone Miner Metab. 1999;17:171–7. doi: 10.1007/s007740050081. [DOI] [PubMed] [Google Scholar]
  • 17.Muschler GF, Nitto H, Boehm CA, Easley KA. Age and gender related changes in the cellularity of human bone marrow and the prevelance of osteoblastic progenitors. J Orthop Res. 2001;19:117–25. doi: 10.1016/S0736-0266(00)00010-3. [DOI] [PubMed] [Google Scholar]
  • 18.Mendes SC, Tibbe JM, Veenhof M, et al. Bone tissue-engineered implants using human bone marrow stromal cells: Effect of culture conditions and donor age. Tissue Eng. 2002;8:911–20. doi: 10.1089/107632702320934010. [DOI] [PubMed] [Google Scholar]
  • 19.Stenderup K, Justesen J, Clausen C, Kassem M. Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells. Bone. 2003;33:919–26. doi: 10.1016/j.bone.2003.07.005. [DOI] [PubMed] [Google Scholar]
  • 20.Khan WS, Marsh DR. A Literature Review on the Effects of Ageing on Mesenchymal Stem Cells. Curr Res J Biol Sci. 2009;1(1):1–6. [Google Scholar]
  • 21.Khan WS, Adesida AB, Hardingham TE. hypoxic conditions increase hif2a and enhance chondrogenesis in stem cells from the infrapatellar fat pad of osteoarthritic patients. Arthritis Res Ther. 2007;9(3):R55. doi: 10.1186/ar2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Khan WS, Tew SR, Adesida AB, Hardingham TE. human infrapatellar fat pad derived stem cells express the pericyte marker 3g5 and show enhanced chondrogenesis after expansion in fibroblast growth factor-2. Arthritis Res Ther. 2008;10(4):R74. doi: 10.1186/ar2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279–95. doi: 10.1091/mbc.E02-02-0105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sakaguchi Y, Sekiya I, Yagishita K, Muneta T. Comparison of human stem cells derived from various mesenchymal tissues-Superiority of synovium as a cell source. Arthritis Rheum. 2005;52:2521–9. doi: 10.1002/art.21212. [DOI] [PubMed] [Google Scholar]
  • 25.Khan WS, Adesida AB, Tew SR, Lowe ET, Hardingham TE. Bone marrow derived mesenchymal stem cells express the pericyte marker 3g5 in culture and show enhanced chondrogenesis in hypoxic conditions. J Orthop Res. 2010;28(6):834–40. doi: 10.1002/jor.21043. [DOI] [PubMed] [Google Scholar]
  • 26.Khan WS, Rayanmarakkar F, Marsh DR. Principles of tissue engineering approaches for bone repair in the hand. In: Beckingsworth RHm., editor. Hand surgery: preoperative expectations, techniques and results. New York: Nova Science Publishers; [Google Scholar]
  • 27.Acarturk TO, Hollinger JO. Commercially available demineralized bone matrix compositions to regenerate calvarial critical-sized bone defects. Plast Reconstr Surg. 2006;118:862–73. doi: 10.1097/01.prs.0000232385.81219.87. [DOI] [PubMed] [Google Scholar]
  • 28.Bostrom R, Mikos A. Tissue engineering of bone. In: Atala A, Mooney DJ, editors. Synthetic biodegradable polymer scaffolds. Berlin, Germany: Birkhaeuser; 1997. pp. 215–34. [Google Scholar]
  • 29.Lanza RP, Langer RS, Vacanti J. Principles of tissue engineering. 3rd. London: Academic; 2007. [Google Scholar]
  • 30.Hardingham TE. Pathophysiology of musculoskeletal disease. In: Maddison PJ, Isenberg DA, Woo P, Glass DN, editors. Oxford textbook of rheumatolog. 2nd. Oxford Medical Publications; 1998. pp. 1–10. [Google Scholar]
  • 31.Parsons IM, Sonnabend DH. What is the role of joint replacement surgery? Best Pract Res Clin Rheumatol. 2004;18:557–72. doi: 10.1016/j.berh.2004.04.003. [DOI] [PubMed] [Google Scholar]
  • 32.Kurtz S, Ong K, Lau E, Mowat F, Halpern M. Projections of primary and revision hip and knee arthroplasty in the United States from 2005 to 2030. J Bone Joint Surg. 2007;89A:780–5. doi: 10.2106/JBJS.F.00222. [DOI] [PubMed] [Google Scholar]
  • 33.Hunziker EB. Articular cartilage repair: basic science and clinical progress; a review of the current status and prospects. Osteoarthritis Cartilage. 2001;10:432–63. doi: 10.1053/joca.2002.0801. [DOI] [PubMed] [Google Scholar]
  • 34.Brittberg M, Peterson L, Sjogren-Jansson E, Tallheden T, Lindahl A. Articular cartilage engineering with autologous chondrocyte transplantation. J Bone Joint Surg. 2003;85A:109–15. doi: 10.2106/00004623-200300003-00017. [DOI] [PubMed] [Google Scholar]
  • 35.Cancedda R, Dozin B, Giannoni P, Quarto R. Tissue engineering and cell therapy of cartilage and bone. Matrix Biol. 2003;22:81–91. doi: 10.1016/s0945-053x(03)00012-x. [DOI] [PubMed] [Google Scholar]
  • 36.Clar C, Cummins E, McIntyre L, et al. Clinical and cost-effectiveness of autologous chondrocyte implantation for cartilage defects in knee joints: systematic review and economic evaluation. Health Technol Assess. 2005;9:1–82. doi: 10.3310/hta9470. [DOI] [PubMed] [Google Scholar]
  • 37.Hardingham T, Tew S, Murdoch A. Tissue engineering: chondrocytes and cartilage. Arthritis Res. 2002;4:S63–8. doi: 10.1186/ar561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wakitani S, Imoto K, Yamamoto T, Saito M, Murata N, Yoneda M. Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthritis Cartilage. 2002;10:199–206. doi: 10.1053/joca.2001.0504. [DOI] [PubMed] [Google Scholar]
  • 39.Wakitani S. Cell transplantation for therapy of rheumatoid arthritis. Nippon Rinsho. 2005;63:672–5. [PubMed] [Google Scholar]
  • 40.Kuroda R, Isada K, Matsumoto T, et al. Treatment of a full-thickness articular cartilage defect in the femoral condyle of an athlete with autologous bone-marrow stromal cells. Osteoarthritis Cartilage. 2007;15:226–31. doi: 10.1016/j.joca.2006.08.008. [DOI] [PubMed] [Google Scholar]

Articles from The Open Orthopaedics Journal are provided here courtesy of Bentham Science Publishers

RESOURCES