Open access

Oral and Maxillofacial Tissue Engineering with Adipose- Derived Stem Cells

Written By

Morikuni Tobita and Hiroshi Mizuno

Submitted: 16 May 2012 Published: 22 May 2013

DOI: 10.5772/55899

From the Edited Volume

Regenerative Medicine and Tissue Engineering

Edited by Jose A. Andrades

Chapter metrics overview

2,649 Chapter Downloads

View Full Metrics

1. Introduction

Oral and maxillofacial tissues are a complex array of bone, cartilage, soft tissue, nerves and vasculature. Damage to these structures, even when minimal, usually leads to noticeable deformities. Therefore, the repair of large segmental bone defects of the jaw or mandible due to trauma, inflammation, or tumor surgery remains a major clinical problem. For many years, simple autogenic, allogenic, or xenogenic bone grafts, or combinations thereof, have been the mainstay for tissue replacement [1]. However, when large bone defects are present, advanced approaches such as free tissue transfer with microvascular reanastomosis of vascularized flaps from distant sites including the fibula, iliac crest, scapula, and radius are needed to repair or regenerate a functionally complex tissue such as maxillofacial tissue [2, 3]. While these procedures have proven to be reliable and effective, they require extended hospitalization, and a secondary donor site with the associated morbidity and complications. As an alternative to current surgical techniques or approaches, developments in tissue engineering using the gene therapy and stem cell biology strive to utilize cells, biomaterial scaffolds and cell signaling factors to regenerate large oral and maxillofacial tissues defect with precise replication of normal body contours. A tissue engineering approach offers several potential benefits, including a decrease in donor site morbidity, a decrease in technical sensitivity of the repair, and the ability to closely mimic the in vivo microenvironment in an attempt to recapitulate normal craniofacial development [1].

Mesenchymal stem cells (MSCs) derived from bone marrow have been used experimentally for tissue engineering applications [4-6]. MSCs can differentiate into several different cell types, such as those that produce bone, cartilage, tendon, and other connective tissues, as well as muscle, adipose, and dermal cells [7-10]. MSCs can be expanded in culture while maintaining their multipotency.

The concept of prefabricated bone engineering with MSCs for large bone defects may play a pivotal role in future therapies. However, bone marrow-derived MSCs have been reported to require selective sera lots and growth factor supplements for culture expansion [11]. Furthermore, traditional bone marrow procurement, particularly in volumes larger than a few milliliters may be painful, frequently requiring general or spinal anesthesia [12-14].

Bone marrow tissue provides the most universal and attractive source of MSCs; however, other tissues such as periosteal [15], muscle [16], synovial membrane [17] and adipose [18-20] tissues also appear to possess MSCs. Particularly, adipose tissue is an important source of stem cells because subcutaneous adipose tissue is an abundant and accessible source of both uncultured stromal vascular fraction (SVF) cells and cultured homogeneous adipose-derived stem cells (ASCs) (21). ASCs obtained from lipoaspirates have multilineage potential and will differentiate into adipogenic, chondrogenic, myogenic, osteogenic, and neurogenic cells [19, 22, 23]. Thus, ASCs have great potential for clinical applications such as the repair of damaged tissues and angiogenic therapy. Injection of human ASCs was recently shown to improve neovascularization in an ischemic hindlimb mouse model and osteoid matrix formation in immunotolerant mice [24-26]. Further, ASCs have been shown to increase the functional capacity of damaged skeletal muscle in vivo [27]. Therefore, these reports suggest that ASCs may also have the potential for use in large bone tissue engineering techniques such as prefabrication. Recently, prefabricated bone engineered with ASCs was reported both with in vivo studies in rat and a clinical human case. Thus, the use of ASCs in maxillofacial tissue reconstruction should be viewed favorably and these novel approaches may have advantages for tissue reconstruction.

In this chapter, the current approaches and the biomaterials used for repair of large bone defects are presented, and the novel approach of prefabricated bone engineering with MSCs and ASCs is introduced.

Advertisement

2. Current therapy for large bone reconstruction

Bone tissue is composed of heterogeneous cell types embedded in a three-dimensional mineralized extracellular matrix. The scaffolds for repair of large bone defects, including autogenous bone grafts or biomaterials, must provide the necessary support for cells to proliferate while maintaining their potential to differentiate, and must possess an architecture suitable for matching the final shape of the newly formed bone [28].

2.1. Autologous bone reconstruction

The current standard of care for repair of critical large bone defects consists of autogenous bone grafting using bone from the rib or iliac crest of the patient. An autologous bone graft is still the ideal material for the repair of craniofacial defects; however, the availability of autologous bone is limited and harvesting can be associated with complications [29]. Vascularized and avascular autogenous bone has a greater osteogenic capacity than any other bone replacement material, as revascularization attracts mesenchymal differentiation into osteogenic, chondrogenic and other cell types. Autogenous bone transplants possess an inherent biocompatibility and are therefore more easily incorporated without immunogenic responses [30]. However, the clinical use of autologous bone transplants is limited by considerable donor site morbidity, which increases with the amount of harvested bone. Bleeding, hematomas, infections, and chronic pain are common complications of autologous bone graft harvests [31, 32].

2.2. Allogenic/Xenogenic bone reconstruction

Demineralized bone matrix (DMB) is the de-cellularized and organic component of bone, and is a commercially available osteoinductive and osteoconductive biomaterial. DMB represents a concentrated source of bone morphogenetic proteins (BMPs) and has been used in numerous animals systems since its initial description in 1965 [33]. The widespread use of DMB in humans still remains restricted since the immunologic properties of donor DMB are unknown [34].

With the disadvantages of host morbidity and the limits in suitable harvesting sites and material for autologous grafts, the use of xenografts might be considered for large bone reconstructions, although the histocompatibility issues between the human recipient and animal donor preclude the use of bone xenografts [34]. However, bovine-derived DMB is currently used in oral and maxillofacial surgery [35].

2.3. Synthetic scaffolds for bone reconstruction

A wide variety of synthetic (alloplastic) scaffolds such as ceramics and polymers are used clinically for bone grafting [30]. Ceramics are crystalline, inorganic, nonmetallic minerals that are held together by ionic bonds and usually densified by sintering [36]. Ceramics such as hydroxyapatite and β-tricalcium phosphate (TCP) are currently in use clinically for bone tissue regeneration of large bone defects.

Various synthetic polymer scaffolds exhibit different structural, mechanical and degradation properties that make then suitable for bone tissue engineering [36]. Blending polymers of different molecular weights can achieve both optimal degradation rates and mechanical properties [37]. Some synthetic polymer scaffolds such as polycaprolactone (PCL) scaffold, polylactic acid (PLLA), polyglycolic acid (PGA) and polylactic-co-glycolic acid (PLGA) materials have been approved by the FDA for craniofacial applications or as absorbable sutures and bone pins/screws [36].

2.4. Gene therapy for bone reconstruction

The use of exogenous cytokines and growth factors, which are essential for bone regeneration, promotes cell adhesion, proliferation, migration and osteogenic differentiation [28]. Growth factors such as BMPs, fibroblast growth factors (FGFs), insulin-like growth factors (IGF), vascular endothelial growth factors (VEGF) and platelet-derived growth factors (PDGF) have been used in bone regeneration [28, 36].

Recently the use of combinations of growth factors, such as BMP-2 and NEL-like molecule-1 (NELL-1), was tested in rapid distraction osteogenesis in a rabbit model. The combined treatment produced significantly greater bone healing compared to single growth factor treatments after four weeks of treatment [38]. However, some reports have cautioned that the clinical use of BMPs and VEGF is in its infancy, and some risks may accompany their use. VEGF is commonly upregulated in various types of tumors to enhance their vascularization, and subcutaneous sarcomas were found in some rats administered recombinant human BMP-7 [39, 40], although no clinical relationship has been established between the use of these growth factors and tumor formation.

2.5. Prefabricated bone engineering for oral and maxillofacial tissue reconstruction

Prefabrication is an interesting area of oral and maxillofacial surgery and plastic and reconstructive surgery, because it represents a bridge between conventional reconstructive surgery and tissue engineering [41, 42]. The purpose of prefabrication is to build a tissue (muscle, bone, skin, or composite) with characteristics as similar as possible to those of the defect that is to be repaired [43]. Conventional osteomyocutaneous flaps do not always meet the requirements for repairing a composite defect. A prefabricated composite flap can be created according to the complex geometry of the defect. Prefabrication of multi-component flaps is a well established procedure in plastic and reconstructive surgery [41]. This concept is based on the revascularization phenomenon directly related to host tissue vascularity [44] and has significantly expanded the frontiers of reconstructive surgery.

Hirase et al. were the first to report the use of prefabricated myocutaneous and osteomyocutaneous tissue in a rat model [45]. Flap prefabrication using conventional bone grafts allows for generation of new types of flaps independent of the vascular anatomy of the bone transplant. However, the donor site morbidity after harvesting of bone for grafting is still a problem. Recently, biomaterials, osteogenic cells and osteoinductive growth factors have been used for generation of vascularized bone tissues in combination with a vascular axis or vascularized flaps. An inflammatory wound healing response as a reaction to the surgical implantation induces vascularization of the scaffolds [31]. Induction of axial vascularization protected the porous biomaterials from bacterial infection and transfer of this vascularized hard tissue as a free flap has been demonstrated [46]. Prefabricated vascularized bone grafts have been used in a clinical setting for mandibular reconstruction following thorough in vivo evaluation in a pig model [47-49]. In these studies, granules of xenogenic bone minerals soaked with recombinant Osteogenic protein-1 were implanted into the latissimus dorsi muscle and the neo-tissue was subsequently transferred to sites of mandibular defects using microsurgical techniques.

Advertisement

3. Mesenchymal stem cells for oral and maxillofacial tissue reconstruction

3.1. Mesenchymal stem cells for bone engineering

The bone marrow is not only the site where hematopoiesis occurs in postnatal life, it is also a reservoir of pluripotent stem cells for mesenchymal tissues [50]. Plated at low densities, single precursor cells derived from bone marrow, and referred to as colony-forming units, give rise to distinct and heterogeneous colonies. These colonies have been shown to undergo osteogenic, chondrogenic and adipogenic differentiation [51].

Chang and colleagues showed that MSCs can produce ectopic bone generation in a mouse model [52]. A suspension of osteogenically induced MSCs was added to 2% alginate, which was then gelled by mixing with calcium sulfate. The gel was injected subcutaneously on the dorsal side of the experimental animals. Histological examination of the implants revealed signs of endochondrosis with woven bone deposition. The equilibrium modulus of the newly formed bone increased with time up to 678 kPa at 30 weeks, as determined by biomechanical analysis. This value is approximately 1.62% of native bovine cancellous bone. In another study [53] of large mandibular bone defect repair, dog MSCs cultured with ß-TCP to generate osteogenic cells were co-implanted with a titanium plate into a 30 mm segmental mandible defect. Biomechanical tests showed a significant difference between the experimental group (with cells) and the control group (without cells), highlighting the importance of the MSCs in bone formation. Pedicled bone flaps based on collagen I scaffolds, bone marrow stromal cells and a PTFE membrane have been successfully generated using the carotid artery and jugular vein or the saphenous bundle as a vascular axis in a mouse model [54]. The osteogenetic stimulus was supplied by the injection of mouse MSCs cultured in osteogenic medium inside the space delimited by the PTFE membrane. After only 4 weeks islands of bone tissue were present inside the membrane.

3.2. Clinical trials for bone engineering with mesenchymal stem cells

There is some clinical experience with bone reconstruction using expanded MSCs combined with scaffolds. Constructs of expanded autologous MSCs in macroporous hydroxyapatite were used in three patients with large segmental bone defects [55, 56].

Warnke and Terheyden have developed a two stage procedure for mandible reconstruction in humans [57]. This study used prefabrication in the latissimus dorsi muscle with the aim of reconstructing a 70 mm defect in the mandible of a man who underwent a tumor resection years previously. The entire construction of the mandible was built using blocks of Bio-Oss® and MSCs that had been cultured in the presence of BMP-7. The Bio-Oss® and MSCs were placed in a titanium cage, and implanted into the latissimus dorsi of the patient and maintained in situ for 7 weeks. Subsequently, this unit, together with the vascular bundle that supplied it, was removed and re-implanted in the mandible defect by fixation with titanium plates and microvascular sutures connecting the vasculature to the external carotid artery and the cephalic vein.

Advertisement

4. Adipose-derived stem cells for oral and maxillofacial tissue engineering

4.1. Characterization of adipose-derived stem cells

There is a general consensus that SVF cells are a heterogeneous population, and no specific ranges for each subpopulation have been agreed upon formally [21]. In contrast, the International Society for Cell Therapy has provided guidelines for the definition of MSCs, as follows. (1) MSCs must be plastic-adherent when maintained in standard culture conditions. (2) MSCs must express CD105, CD73 and CD90, and lack expression of CD45, CD34, CD14 or CD11b, CD79α or CD19 and HLA-DR surface antigens. (3) MSCs must differentiate into osteoblasts, adipocytes and chondroblasts in vitro [58].

SVF cells include preadipocytes, fibroblasts, vascular smooth muscle cells, endothelial cells (ECs), resident monocytes/macrophages, lymphocytes and ASCs [59]. Although the criteria to define SVF cells remain in contention, the heterogeneous SVF cell population includes putative ASCs (CD31-, CD34+/-, CD45-, CD90+, CD105-, CD146-), endothelial (progenitor) cells (CD31+, CD34+, CD45-, CD90+, CD105-, CD146+), vascular smooth muscle cells or pericytes (CD31-, CD34+/-, CD45-, CD90+, CD105-, CD146+), and hematopoietic cells (CD45+) in uncultured conditions [60]. Cultured ASCs show an extensive proliferative ability in an uncommitted state while retaining their multilineage differentiation potential. ASCs express the mesenchymal stem cell markers CD10, CD13, CD29, CD34, CD44, CD54, CD71, CD90, CD105, CD106, CD117, CD166 and STRO-1. They are negative for the hematopoietic lineage markers CD45, CD14, CD16, CD56, CD61, CD62E, CD104, and CD106 and for the EC markers CD31, CD144, and von Willebrand factor [20, 61, 62]. Morphologically, cultured ASCs are fibroblast-like and preserve their shape after expansion in vitro [20, 63]. The ASC specific surface markers CD29, CD90, and CD166 increase during culture [64]. In later passages, ASC cultures are homogeneous and exhibit fibroblastoid morphology. The composition of the subpopulations, therefore, may change during expansion [65, 66]. Therefore ASCs match the standard criteria for MSCs.

4.2. Differentiation potential of osteogenic cells in vitro and in vivo

Numerous studies have presented results that clearly show that ASCs can differentiate into osteoblasts [20, 59, 63, 67, 68]. ASCs exhibit a time-dependent expression of genes and proteins associated with the osteoblast phenotype, including ALP, Type I Collagen, OPN, ON, RUNX2, BMP-2, BMP-4 and BMP receptors I and II [20, 67, 69, 70]. Additionally, between 2 and 4 weeks of culture, mineralization of the extracellular matrix begins and proceeds via the activity of ALP, an enzyme that hydrolyzes phosphate esters making available inorganic phosphate to form hydroxyapatite [19, 20, 71].

Furthermore, recent reports have shown that ASCs co-cultured with ECs exhibit enhanced osteogenesis [72, 73]. ASCs exhibited increased secretion of alkaline phosphatase and osteocalcin, and an overall increase in osteogenesis in the co-cultured situation compare with other experimental groups. These interactions may be important to regenerate bone in large bone defects since angiogenesis plays a key role in regeneration of large amounts of tissue.

4.3. Fabricated bone engineering with adipose-derived stem cells

To make a functional prefabricated bone, three elements are required: scaffolds to provide a three-dimensional support, growth factors to stimulate neovascularization, and MSCs to give an osteoinductive stimulus. Okuda et al. have reported prefabrication of tissue engineered bone grafts using ASCs in a rat model [74]. ASCs and porous β-TCP as scaffold material were implanted into the superficial inferior epigastric artery flap. After prefabrication for eight weeks, the prefabricated flaps were elevated and the pedicles were clamped for 4 h; prefabricated tissue was harvested two weeks later. The osteogenic capacity of the prefabricated graft was not significantly different from non-prefabricated grafts examined after two weeks in a rat model. Furthermore, an analysis of angiogenesis suggested that the prefabricated model possessed significantly greater capillary density than the non-prefabricated model.

Recently, repair of a large bony defect using ASCs was clinically reported [75-77] (Table. 1). Mesima°ki and colleagues published a clinical case report of prefabricated bone tissue engineering [77]. The large bony defect was reconstructed with a microvascular flap using autologous ASCs, β-TCP and BMP-2, 36 months after a hemimaxillectomy due to a large keratocyst. After expansion of ASCs and cultivation with β-TCP and BMP-2 in vitro, a titanium cage filled with ASCs and β-TCP was inserted through a vertical incision into a pouch prepared in the patient’s left rectus abdominis muscle. The rectus abdominis free flap was raised. Before severing the vascular supply to the muscle, the muscle pouch was carefully opened and the titanium cage was opened. After severing the vessels, the flap was placed in the maxillary defect; the inferior epigastric artery was anastomosed end-to-end to the facial artery and the vein end-to-end to the facial vein.

Clinical reports/trials with ASCs Design Results Ref
Widespread calvarial defect Autologous SVFs with fibrin glue Success, follow-up: 3 months after operation [75]
Large calvarial defect Implant autologous cultured ASCs with β-TCP No complications, follow-up: 3 months after operations [76]
Maxillary reconstruction Fabricated bone tissue using autologous cultured ASCs with β-TCP and BMP-2 Success, follow-up: 8 months after operations [77]
Large osseous defect Autologous ASCs with different scaffolds Recruiting NCT01218945*
Avascular Necrosis of the Femoral Head Autologous adipose tissue derived MSCs transplantation Phase 1, 2 NCT01532076*

Table 1.

Clinical reports/trials for large bony defect using adipose-derived stem cells

Abbreviations: SVF; Stromal Vascular Fraction, ASCs; Adipose-derived Stem Cells, β-TCP; Beta-tricalcium phosphate, MSCs; mesenchymal stem cells, BMP; bone morphogenetic protein


(*Identifier on Clinical trials website: *http://clinicaltrials.gov/ct2/results?term=adipose+derived+cells+bone).


Advertisement

5. Future perspective

In the past decade, basic research characterizing ASCs shows that these cells have the potential to regenerate tissue defects such as large bone defects, and clinical studies have examined the potential use of ASCs to reconstruct oral and maxillofacial tissue. Although clinical studies have only just begun, the use of ASCs in the clinical setting is extremely promising because ASCs are a readily available, multipotent, and abundant cell type with the capability to undergo robust osteogenesis. However, further studies, including research to determine the mechanism of osteogenic differentiation and studies to evaluate the safety of ASC usage, will be necessary to realize the potential of ASCs in clinical regenerative medicine of the future.

References

  1. 1. Ward BB, Brown SE, Krebsbach PH. Bioengineering strategies for regeneration of craniofacial bone: a review of emerging technologies. Oral Dis 2010;16(8):709-716.
  2. 2. Disa JJ, Cordeiro PG. Mandible reconstruction with microvascular surgery. Semin Surg Oncol 2000;19(3):226-234.
  3. 3. Emerick KS, Teknos TN. State-of-the-art mandible reconstruction using revascularized free-tissue transfer. Expert Rev Anticancer Ther 2007;7(12):1781-1788.
  4. 4. Caplan AI. Mesenchymal stem cells. J Orthop Res 1991;9(5):641-650.
  5. 5. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284(5411):143-147.
  6. 6. Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science 1997;276(5309):71-74.
  7. 7. Caplan AI. Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J Cell Physiol 2007;213(2):341-347.
  8. 8. Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells 2007;25(11):2739-2749.
  9. 9. Nixon AJ, Watts AE, Schnabel LV. Cell- and gene-based approaches to tendon regeneration. J Shoulder Elbow Surg 2012;21(2):278-294.
  10. 10. Markowicz M, Koellensperger E, Neuss S, Koenigschulte S, Bindler C, Pallua N. Human bone marrow mesenchymal stem cells seeded on modified collagen improved dermal regeneration in vivo. Cell Transplant 2006;15(8-9):723-732.
  11. 11. Donald P. Lennon SEH, Scott P. Bruder, Neelam Jaiswal and Arnold I. Caplan. Human and animal mesenchymal progenitor cells from bone marrow: Identification of serum for optimal selection and proliferation. In Vitro Cell Dev Biol 1996;32(10):602-611.
  12. 12. Auquier P, Macquart-Moulin G, Moatti JP, Blache JL, Novakovitch G, Blaise D, et al. Comparison of anxiety, pain and discomfort in two procedures of hematopoietic stem cell collection: leukacytapheresis and bone marrow harvest. Bone Marrow Transplant 1995;16(4):541-547.
  13. 13. Nishimori M, Yamada Y, Hoshi K, Akiyama Y, Hoshi Y, Morishima Y, et al. Health-related quality of life of unrelated bone marrow donors in Japan. Blood 2002;99(6):1995-2001.
  14. 14. De Ugarte DA, Morizono K, Elbarbary A, Alfonso Z, Zuk PA, Zhu M, et al. Comparison of multi-lineage cells from human adipose tissue and bone marrow. Cells Tissues Organs 2003;174(3):101-109.
  15. 15. Ball MD, Bonzani IC, Bovis MJ, Williams A, Stevens MM. Human periosteum is a source of cells for orthopaedic tissue engineering: a pilot study. Clin Orthop Relat Res 2011;469(11):3085-3093.
  16. 16. Wu X, Wang S, Chen B, An X. Muscle-derived stem cells: isolation, characterization, differentiation, and application in cell and gene therapy. Cell Tissue Res. 2010;340(3):549-567.
  17. 17. Jo CH, Ahn HJ, Kim HJ, Seong SC, Lee MC. Surface characterization and chondrogenic differentiation of mesenchymal stromal cells derived from synovium. Cytotherapy 2007;9(4):316-327.
  18. 18. Tobita M, Orbay H, Mizuno H. Adipose-derived stem cells: current findings and future perspectives. Discov Med 2011;11(57):160-170.
  19. 19. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 2001;7(2):211-228.
  20. 20. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 2002;13(12):4279-4295.
  21. 21. Gimble JM, Bunnell BA, Chiu ES, Guilak F. Concise review: Adipose-derived stromal vascular fraction cells and stem cells: let's not get lost in translation. Stem Cells 2011;29(5):749-754.
  22. 22. Mizuno H, Zuk PA, Zhu M, Lorenz HP, Benhaim P, Hedrick MH. Myogenic differentiation by human processed lipoaspirate cells. Plast Reconstr Surg 2002;109(1):199-209.
  23. 23. Orbay H, Tobita M, Mizuno H. Mesenchymal stem cells isolated from adipose and other tissues: basic biological properties and clinical applications. Stem Cells Int 2012;2012:461718.
  24. 24. Puissant B, Barreau C, Bourin P, Clavel C, Corre J, Bousquet C, et al. Immunomodulatory effect of human adipose tissue-derived adult stem cells: comparison with bone marrow mesenchymal stem cells. Br J Haematol 2005;129(1):118-129.
  25. 25. Hicok KC, Du Laney TV, Zhou YS, Halvorsen YD, Hitt DC, Cooper LF, et al. Human adipose-derived adult stem cells produce osteoid in vivo. Tissue Eng 2004;10(3-4):371-380.
  26. 26. Miranville A, Heeschen C, Sengenes C, Curat CA, Busse R, Bouloumie A. Improvement of postnatal neovascularization by human adipose tissue-derived stem cells. Circulation 2004;110(3):349-355.
  27. 27. Bacou F, el Andalousi RB, Daussin PA, Micallef JP, Levin JM, Chammas M, et al. Transplantation of adipose tissue-derived stromal cells increases mass and functional capacity of damaged skeletal muscle. Cell Transplant 2004;13(2):103-111.
  28. 28. Zhang Z. Bone regeneration by stem cell and tissue engineering in oral and maxillofacial region. Front Med 2011;5(4):401-413.
  29. 29. Kolk A, Handschel J, Drescher W, Rothamel D, Kloss F, Blessmann M, et al. Current trends and future perspectives of bone substitute materials - From space holders to innovative biomaterials. J Craniomaxillofac Surg 2012. DOI; 10.1016/j.jcms.2012.01.002
  30. 30. den Boer FC, Wippermann BW, Blokhuis TJ, Patka P, Bakker FC, Haarman HJ. Healing of segmental bone defects with granular porous hydroxyapatite augmented with recombinant human osteogenic protein-1 or autologous bone marrow. J Orthop Res 2003;21(3):521-528.
  31. 31. Kneser U, Schaefer DJ, Polykandriotis E, Horch RE. Tissue engineering of bone: the reconstructive surgeon's point of view. J Cell Mol Med 2006;10(1):7-19.
  32. 32. Arrington ED, Smith WJ, Chambers HG, Bucknell AL, Davino NA. Complications of iliac crest bone graft harvesting. Clin Orthop Relat Res 1996;(329):300-309.
  33. 33. Urist MR. Bone: formation by autoinduction. Science 1965;150(3698):893-899.
  34. 34. Zuk PA. Tissue engineering craniofacial defects with adult stem cells? Are we ready yet? Pediatr Res 2008;63(5):478-486.
  35. 35. Browaeys H, Bouvry P, De Bruyn H. A literature review on biomaterials in sinus augmentation procedures. Clin Implant Dent Relat Res 2007;9(3):166-177.
  36. 36. Bueno EM, Glowacki J. Cell-free and cell-based approaches for bone regeneration. Nat Rev Rheumatol 2009;5(12):685-697.
  37. 37. Piskin E, Isoglu IA, Bolgen N, Vargel I, Griffiths S, Cavusoglu T, et al. In vivo performance of simvastatin-loaded electrospun spiral-wound polycaprolactone scaffolds in reconstruction of cranial bone defects in the rat model. J Biomed Mater Res A 2009;90(4):1137-1151.
  38. 38. Zhu S, Song D, Jiang X, Zhou H, Hu J. Combined effects of recombinant human BMP-2 and Nell-1 on bone regeneration in rapid distraction osteogenesis of rabbit tibia. Injury 2011;42(12):1467-1473.
  39. 39. Kirker-Head CA. Development and application of bone morphogenetic proteins for the enhancement of bone healing. J Orthopaed Traumatol 2005;6(1):1-9.
  40. 40. Runyan CM, Taylor JA. Clinical applications of stem cells in craniofacial surgery. Facial Plast Surg 2010;26(5):385-395.
  41. 41. Khouri RK, Upton J, Shaw WW. Principles of flap prefabrication. Clin Plast Surg 1992;19(4):763-771.
  42. 42. Morrison WA, Penington AJ, Kumta SK, Callan P. Clinical applications and technical limitations of prefabricated flaps. Plast Reconstr Surg 1997;99(2):378-385.
  43. 43. Di Bella C, Lucarelli E, Donati D. Historical review of bone prefabrication. Chir Organi Mov 2008;92(2):73-78.
  44. 44. Khouri RK, Upton J, Shaw WW. Prefabrication of composite free flaps through staged microvascular transfer: an experimental and clinical study. Plast Reconstr Surg 1991;87(1):108-115.
  45. 45. Hirase Y, Valauri FA, Buncke HJ. Prefabricated sensate myocutaneous and osteomyocutaneous free flaps: an experimental model. Preliminary report. Plast Reconstr Surg 1988;82(3):440-446.
  46. 46. Bernard SL, Picha GJ. The use of coralline hydroxyapatite in a "biocomposite" free flap. Plast Reconstr Surg 1991;87(1):96-105.
  47. 47. Terheyden H, Warnke P, Dunsche A, Jepsen S, Brenner W, Palmie S, et al. Mandibular reconstruction with prefabricated vascularized bone grafts using recombinant human osteogenic protein-1: an experimental study in miniature pigs. Part II: transplantation. Int J Oral Maxillofac Surg 2001;30(6):469-478.
  48. 48. Terheyden H, Menzel C, Wang H, Springer IN, Rueger DR, Acil Y. Prefabrication of vascularized bone grafts using recombinant human osteogenic protein-1--part 3: dosage of rhOP-1, the use of external and internal scaffolds. Int J Oral Maxillofac Surg 2004;33(2):164-172.
  49. 49. Terheyden H, Knak C, Jepsen S, Palmie S, Rueger DR. Mandibular reconstruction with a prefabricated vascularized bone graft using recombinant human osteogenic protein-1: an experimental study in miniature pigs. Part I: Prefabrication. Int J Oral Maxillofac Surg 2001;30(5):373-379.
  50. 50. Cancedda R, Bianchi G, Derubeis A, Quarto R. Cell therapy for bone disease: a review of current status. Stem Cells 2003;21(5):610-619.
  51. 51. Muraglia A, Cancedda R, Quarto R. Clonal mesenchymal progenitors from human bone marrow differentiate in vitro according to a hierarchical model. J Cell Sci 2000;113 (Pt 7):1161-1166.
  52. 52. Chang SC, Tai CL, Chung HY, Lin TM, Jeng LB. Bone marrow mesenchymal stem cells form ectopic woven bone in vivo through endochondral bone formation. Artif Organs 2009;33(4):301-308.
  53. 53. He Y, Zhang ZY, Zhu HG, Qiu W, Jiang X, Guo W. Experimental study on reconstruction of segmental mandible defects using tissue engineered bone combined bone marrow stromal cells with three-dimensional tricalcium phosphate. J Craniofac Surg 2007;18(4):800-805.
  54. 54. Mankani MH, Krebsbach PH, Satomura K, Kuznetsov SA, Hoyt R, Robey PG. Pedicled bone flap formation using transplanted bone marrow stromal cells. Arch Surg 2001;136(3):263-270.
  55. 55. Quarto R, Mastrogiacomo M, Cancedda R, Kutepov SM, Mukhachev V, Lavroukov A, et al. Repair of large bone defects with the use of autologous bone marrow stromal cells. N Engl J Med 2001;344(5):385-386.
  56. 56. Marcacci M, Kon E, Moukhachev V, Lavroukov A, Kutepov S, Quarto R, et al. Stem cells associated with macroporous bioceramics for long bone repair: 6- to 7-year outcome of a pilot clinical study. Tissue Eng 2007;13(5):947-955.
  57. 57. Warnke PH, Springer IN, Wiltfang J, Acil Y, Eufinger H, Wehmoller M, et al. Growth and transplantation of a custom vascularised bone graft in a man. Lancet 2004;364(9436):766-770.
  58. 58. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006;8(4):315-317.
  59. 59. Levi B, Longaker MT. Concise review: adipose-derived stromal cells for skeletal regenerative medicine. Stem Cells 2011;29(4):576-582.
  60. 60. Mizuno H, Tobita M, Uysal AC. Concise review: Adipose-derived stem cells as a novel tool for future regenerative medicine. Stem Cells 2012;30(5):804-810.
  61. 61. Musina RA, Bekchanova ES, Sukhikh GT. Comparison of mesenchymal stem cells obtained from different human tissues. Bull Exp Biol Med 2005;139(4):504-509.
  62. 62. Romanov YA, Darevskaya AN, Merzlikina NV, Buravkova LB. Mesenchymal stem cells from human bone marrow and adipose tissue: isolation, characterization, and differentiation potentialities. Bull Exp Biol Med 2005;140(1):138-143.
  63. 63. Arrigoni E, Lopa S, de Girolamo L, Stanco D, Brini AT. Isolation, characterization and osteogenic differentiation of adipose-derived stem cells: from small to large animal models. Cell Tissue Res 2009;338(3):401-411.
  64. 64. Mitchell JB, McIntosh K, Zvonic S, Garrett S, Floyd ZE, Kloster A, et al. Immunophenotype of human adipose-derived cells: temporal changes in stromal-associated and stem cell-associated markers. Stem Cells 2006;24(2):376-385.
  65. 65. Baer PC, Geiger H. Adipose-derived mesenchymal stromal/stem cells: tissue localization, characterization, and heterogeneity. Stem Cells Int 2012;2012:812693.
  66. 66. Schellenberg A, Stiehl T, Horn P, Joussen S, Pallua N, Ho AD, et al. Population dynamics of mesenchymal stromal cells during culture expansion. Cytotherapy 2012;14(4):401-411.
  67. 67. Rada T, Reis RL, Gomes ME. Adipose tissue-derived stem cells and their application in bone and cartilage tissue engineering. Tissue Eng Part B Rev 2009;15(2):113-125.
  68. 68. Safwani WK, Makpol S, Sathapan S, Chua KH. Alteration of gene expression levels during osteogenic induction of human adipose derived stem cells in long-term culture. Cell Tissue Bank 2012. DOI; 10.1007/s10561-012-9309-1.
  69. 69. Halvorsen YD, Franklin D, Bond AL, Hitt DC, Auchter C, Boskey AL, et al. Extracellular matrix mineralization and osteoblast gene expression by human adipose tissue-derived stromal cells. Tissue Eng 2001;7(6):729-741.
  70. 70. Zhao Y, Lin H, Zhang J, Chen B, Sun W, Wang X, et al. Crosslinked three-dimensional demineralized bone matrix for the adipose-derived stromal cell proliferation and differentiation. Tissue Eng Part A 2009;15(1):13-21.
  71. 71. Ciuffi S, Fabbri S, Zonefrati R, Galli G, Tanini A, Brandi ML. Subcutaneous adipocytes may become osteoblasts. Clin Cases Miner Bone Metab 2012;9(1):28-30.
  72. 72. Zhao X, Liu L, Wang FK, Zhao DP, Dai XM, Han XS. Coculture of vascular endothelial cells and adipose-derived stem cells as a source for bone engineering. Ann Plast Surg 2012;69(1):91-98.
  73. 73. Wang J, Ye Y, Tian H, Yang S, Jin X, Tong W, et al. In vitro osteogenesis of human adipose-derived stem cells by coculture with human umbilical vein endothelial cells. Biochem Biophys Res Commun 2011;412(1):143-149.
  74. 74. Okuda T, Uysal AC, Tobita M, Hyakusoku H, Mizuno H. Prefabrication of tissue engineered bone grafts: an experimental study. Ann Plast Surg 2010;64(1):98-104.
  75. 75. Lendeckel S, Jodicke A, Christophis P, Heidinger K, Wolff J, Fraser JK, et al. Autologous stem cells (adipose) and fibrin glue used to treat widespread traumatic calvarial defects: case report. J Craniomaxillofac Surg 2004;32(6):370-373.
  76. 76. Thesleff T, Lehtimaki K, Niskakangas T, Mannerstrom B, Miettinen S, Suuronen R, et al. Cranioplasty with adipose-derived stem cells and biomaterial: a novel method for cranial reconstruction. Neurosurgery 2011;68(6):1535-1540.
  77. 77. Mesimaki K, Lindroos B, Tornwall J, Mauno J, Lindqvist C, Kontio R, et al. Novel maxillary reconstruction with ectopic bone formation by GMP adipose stem cells. Int J Oral Maxillofac Surg 2009;38(3):201-209.

Written By

Morikuni Tobita and Hiroshi Mizuno

Submitted: 16 May 2012 Published: 22 May 2013