Open access peer-reviewed chapter

Recent Advances in Stem Cell and Tissue Engineering

Written By

Farideh Mohammadian

Submitted: 15 December 2017 Reviewed: 26 February 2018 Published: 06 June 2018

DOI: 10.5772/intechopen.75967

From the Edited Volume

Tissue Regeneration

Edited by Hussein Abdel hay El-Sayed Kaoud

Chapter metrics overview

1,865 Chapter Downloads

View Full Metrics

Abstract

The clinical application of stem cells in tissue engineering and regeneration is becoming more significant. However, its application has been limited by issues like reproducibility of the stem cells, ethical concerns of harvesting some of these stem cells, and controlling the fate of stem cells in vitro and in vivo. The advent of tissue engineering and regeneration has led to the fabrication of advanced biomaterials and scaffolds with enhanced ability to mimic and control the cellular microenvironment similar to that of innate stem cell niche. Combining the use of stem cells with biomaterials and scaffolds especially synthetic hydrogels that have exhibited physicochemical abilities and properties similar to native niche can be the future of tissue engineering in terms of formation of new tissues like bones. Recently, there has an increase in the use of either endothelial progenitor cells (EPCs), induced pluripotent stem cells (iPSCs), or adult mesenchymal stem cells in preclinical studies: however this is yet to be transferred to clinical setups as there are limitations in terms of regulations and ethical considerations. The purpose of this review is to give comprehensive details about the application of stem cells in tissue engineering.

Keywords

  • endothelial progenitor cells
  • induced pluripotent stem cells
  • adult mesenchymal stem
  • tissue engineering
  • scaffolds

1. Introduction

Tissue engineering is a multidisciplinary science that applies the principles of bioengineering for the fabrication of new and improved biomaterials capable of maintaining and restoring the functionality of organs and tissues impaired by disease and trauma. This translational approach has been applied to develop and design patient-specific tissue grafts that mimic the functional properties of native tissues. Three important factors have been accredited to the success of tissue engineering: cocultured stem cells, signaling factor, and the bio-fabricated scaffold. The stem cells are capable of differentiating into several types of tissues and organs, while the bio-fabricated scaffold provides structural support to the seeded stem cells. Signaling factors are responsible for influencing cell phenotype, metabolism, migration, and organization.

Stem cells are undifferentiated cells of embryonic, fetal origin, and they possess the ability to give rise to differentiated cells and then finally develop into organs. Stem cell characteristics include the ability to self-replicate and renew, clonage forming, and high potency ability [1]. In terms of the potency ability of stem cells, stem cells can be totipotent, could differentiate into any cell types (pluripotent) [2], and could differentiate into cells that arise from the three germ layers—ectoderm, endoderm, and mesoderm—from which organs develop [3].

Stem cells can be categorized broadly into embryonic and adult stem cells and are efficient cell sources for tissue regenerative applications. They have also been reported to have the abilities to promote tissue homeostasis, growth, and repair, thereby contributing importantly to tissue and organ regeneration [4]. Bio-fabricated scaffolds consist of decellularized biomaterials to provide structural and anatomical functions to the seeded stem cells, thereby resulting into successful formation of specific tissue. In support of the above report, Kang and colleagues demonstrated that decellularized scaffolds loaded with autologous adipose-derived stem cells (ADSCs) were efficient to repair cartilage defect in an animal model [5]. They concluded that decellularized scaffolds loaded with ADSC induced significant and improved cartilage tissue repair compared to native cartilage.

Advertisement

2. Mesenchymal stem cells seeded for bone tissue engineering

MSCs are stromal stem cells that are heterogeneous and are derived from several tissue sources that include adipose tissue [6], periodontal ligaments [7], bone marrow (Figure 1) [8], umbilical cord (UC) [9], placenta [10], and lungs [11]. MSCs express surface markers like CD73, CD44, CD90, and CD105. The most widely known and used MSCs are bone marrow MSCs and adipose tissue-derived MSCs isolated and purified from the bone marrow and adipose tissue, respectively. Briefly, the anatomy of the bone marrow is made up of the parenchyma and the stroma part. The parenchyma houses the hematopoietic stem cells, and the stoma part consists of the bone marrow stromal cells (MSCs) that have the capability to differentiate into several cell lines like osteoblasts, chondrocytes, adipocytes, etc. The clinical use of both bone-derived mesenchymal stem cells and adipose stem cells in bone tissue engineering has been reported using various models of bone regeneration such as osteogenesis [12, 13], long bone defects [14, 15], and calvarial defects [16, 17]. Furthermore, co-administration of stem cells with cytokines has been reported to be efficient in bone repair as cytokines and growth factors like stromal derived factor-1 (SDF-1) can lead to increased migration and homing of stem cells to the defected site [18]. In a similar report by Ho et al., they demonstrated that co-administration of stromal-derived factor-1 with BM-MSCs would indirectly enhance bone repair by improving migration of innate cells to the site of bone fracture. They concluded that BM-MSCs overexpressing SDF-1 were efficient in improving the new bone formation during the early stage of fracture healing compared to BM-MSC treatment alone [19]. Genes implicated in fracture healing such as osterix [20], hypoxia-inducible factor-1 [21], and BMP-7 [22] have all been reported to be efficient in bone formation when transfected with MSCs.

Figure 1.

Showing in vivo and in vitro stem cell application in engineered tissue (a) In vitro prevascularization methods induce cell-seeded scaffolds to form vasculature (b) In vivo ectopic prevascularization involves implantation of a cell-seeded scaffold into a highly vascularized bed. Adapted from [26] with copyright permission.

Advertisement

3. Advances in MSCs and tissue engineering technology

Recently, bone tissue engineering in combination with novel stem cell-based technologies is yielding promising results as reported by Syed-Picard and colleagues in their experimental study that BM-MSC-derived cell sheets could be used to fabricate functional periosteal tissue [23]. Briefly, culturing BM-MSCs to hyperconfluence to produce abundant extracellular matrix to form robust cell sheets generated the BM-MSC-derived cell sheets. The authors reported that the generated cell sheets supported with calcium phosphate pellets were transplanted subcutaneously into mice for 8 weeks. They concluded that there was significant bone-like tissue formation by the BM-MSC-calcium phosphate pellet structure compared to the non-seeded calcium phosphate scaffold.

In another similar study by [24], BM-MSC cell sheet technology was compared to control cell complex. The authors reported that BM-MSC cell sheet resulted into significant expressed levels of growth factors crucial to bone development like vascular endothelial growth factor and PDGF. In another innovative study of stem cell application in tissue engineering, Ren et al. fabricated and demonstrated a three-dimensional vascularized stem cell sheet construct, composed of both BM-MSCs and human umbilical vein endothelial cells. The authors concluded that there was significant formation of blood vessel formation compared to the control [25].

Advertisement

4. Clinical reports on stem application of bone tissue engineering

A clinical study by Centeno and colleagues reported the application of culture-expanded, autologous BM-MSC for osteoarthritis in more than 300 patients. The authors reported the safety and efficacy of its use. Briefly, the autologous BM-MSCs were cultured in monolayer culture flasks and transplanted into the affected peripheral joints. They concluded that a 50% improvement in clinical symptoms was recorded among the osteoarthritis patients [27]. The application of infrapatellar fat pad-derived ASCs was demonstrated by Koh and colleagues when they reported its efficiency in improving and managing knee osteoarthritis through clinical and radiological results [28].

A mixture of stem cells and progenitor cells with CD90- and CD14-expressing cells resident in the bone marrow called tissue repair cells (TRC) has been demonstrated to be efficient for reconstructing craniofacial bone defects in a controlled feasibility trial [29]. Briefly, the clinical trial was carried out using 24 patients in need of localized osseous reconstruction. The patients were randomized to either guided bone regeneration (GBR) or TRC transplantation and were subsequently assessed. They concluded that TRC therapy resulted in an accelerated and improved alveolar bone regeneration compared to GBR therapy.

Advertisement

5. Induced pluripotent stem cells seeded for bone tissue engineering

Yamanaka’s group is one of the pioneers of studies related to induced pluripotent stem cells. Yamanaka et al. studies like [30] where they reported the possibility of reprogramming of somatic cells into a primordial embryonic stem cell-like state, capable of differentiating into all three germ layers. There are several studies demonstrating the application of iPSCs in tissue engineering like [31], where they reported the ability of polyethersulfone scaffolds seeded with iPSCs to regenerate cranial bone. The authors concluded that iPSCs seeded with polyethersulfone scaffolds promoted and stimulated cranial bone formation compared to scaffold alone. In similar scaffold study design, Liu used an Arg-Gly-Asp-grafted calcium phosphate cement scaffold seeded with iPSC-MSCs overexpressing NELL1 that were efficient to improve osteogenic differentiation process [32]. However, this report was challenged by [33] reporting that osteogenic abilities of iPSCs can only be realized by scaffolds fabricated with calcium phosphate alone in an ex vivo model. The use of iPSCs in tissue engineering has been reported using animal model by Lian and colleagues, in their mouse model of limb ischemia study. They reported that iPSC-MSCs were more efficient compared to adult BM-MSCs [34] based on their more efficient survival and engraftment abilities after transplantation to induce tissue regeneration.

Advertisement

6. Endothelial progenitor cells (EPCs) seeded for bone tissue engineering

EPCs are bone marrow-derived precursor cells and express CD34 molecules. They have the ability to differentiate into endothelial cells and ultimately contribute to the process of angiogenesis [35]. They have been reported to be resident cells in the peripheral blood and potentially contribute to the initiation of neovascularization [36]. There have been several studies demonstrating the use of EPCs in tissue engineering. Zigdon-Giladi and co-workers in their nude mouse model study with calvarial defect demonstrated that human EPCs could enhance the processes of vasculogenesis and osteogenesis [37]. They concluded that there was a significant increase in blood vessel density as well as increased extra-cortical bone height and length in the human EPC-transplanted group compared to the control. Furthermore, EPCs seeded on Gelfoam scaffold were reported to be efficient in stimulating cranial bone formation at the site of injury compared to the unseeded scaffold [38].

In a clinical case carried out by Kuroda and colleagues of tibial surgery, the efficacy of EPCs was demonstrated when autologous, granulocyte colony-stimulating factor (GCSF)-mobilized CD34(+) cells were used in successful tibial autologous bone grafting [39].

Advertisement

7. Stem cells and decellularized scaffolds

Recently, scaffolds have been designed in the form of decellularized tissues and organs and are commonly used in tissue engineering and regenerative medicine (Table 1). Recent and novel advancement in tissue engineering has been the bedrock for the functional replacement of whole organs. Several organs have been bioengineered and implanted into laboratory animal recipients and potentially showing regenerative abilities and functions. Both acellular and decellularized scaffolds have been seeded with stem cells and potentially have exhibited promising clinical results.

Organ/tissue engineered Decellularized scaffolds Cells Type of experiment References
Skin tissue engineering MatrACELL-processed human acellular dermal matrix NA In vivo and clinical [40]
Urethral tissue engineering 3D porous urinary bladder acellular matrix NA Clinical [41]
Bone tissue engineering Decellularized bone cylinders Human-induced pluripotent stem cells In vitro and in vivo [42]
Decellularized bone scaffolds Human adipose-derived stem cells In vitro [43]
Cardiac tissue engineering Decellularized porcine pulmonary valves Autologous bone marrow mesenchymal stem cells In vivo [44]
Pulmonary tissue engineering Acellular porcine and human trachea-lung scaffolds Murine embryonic stem cells; bone marrow-derived mesenchymal stem cells In vitro [45]
Renal tissue engineering
Tracheal tissue engineering
Decellularized rat kidneys Mouse embryonic stem cells In vitro [46]
Genipin cross-linked decellularized rat tracheal scaffold Pluripotent murine embryonic stem cells In vitro [47, 48]
Decellularized leporine tracheal scaffold Bone marrow-derived mesenchymal stromal cells Clinical [49]
Vascular tissue engineering Decellularized vascular scaffold from rat abdominal arteries NA In vivo [50]
Decellularized inferior vena cava of rabbits Adipose-derived mesenchymal stem cells In vitro [51]
Neural tissue engineering Genipin cross-linked gelatin electrospun scaffolds Rat allogeneic mesenchymal stromal cells In vitro [52]
Cartilage tissue engineering Decellularized stem cell matrix Human adult synovium-derived stem cells In vitro [53]
Hepatic tissue engineering Acellular whole liver scaffol Mesenchymal stem cells In vitro and in vivo [54]

Table 1.

Applications of tissue-engineered scaffolds recellularized with stem cells.

Adapted from Rana et al. [55] with copyright permission.

Advertisement

8. Decellularized cardiac tissues and stem cells

There are reports of fabricated decellularized cardiac tissue used as potential scaffolds. For example, human cardiac extracellular cell matrix sheets have been reported to be seeded with mesenchymal stem cells and cardiomyocytes by [56]. The authors concluded that the MSC-cardiomyocyte-derived scaffold efficiently improved and stimulates cardiac tissue regeneration. In another report by [57], decellularized engineered heart valve was successfully implanted for reconstruction of the right ventricular outflow tract. The authors concluded that via echocardiography, the implanted heart valve demonstrated normal physiological pressure gradient after 10 years, with no record of calcification and, in addition, it exhibited an excellent hemodynamic performance.

Advertisement

9. Decellularized respiratory tissues and stem cells

There are reports of fabricated decellularized tissue used as potential scaffolds for respiratory tissue because of their simplified anatomical structure. One of the earliest reports on the fabrication of scaffold for tracheal tissue regeneration is [49] where the authors compared the efficiency of decellularized leporine tracheal scaffold seeded with amniotic-derived mesenchymal stem cells and non-seeded decellularized scaffolds. The authors concluded that MSC-seeded scaffold exhibited a high level of survival of the cells and epithelialization as well as a high level of elastin.

In another study on decellularized tissue, Nichols and colleagues fabricated acellular pig scaffolds using decellularized scaffold seeded with murine embryonic stem cells, pig bone marrow-derived mesenchymal stem cells, and primary human alveolar epithelial type II cells [45]. They concluded that there were recorded changes in type I collagen levels and evidences of cell attachment and viability.

Advertisement

10. Clinical application of tissue-engineered trachea and stem cells

There have been some reports on the successful implantation of bioengineered tissues like tracheal seeded with stem cells clinically. Macchiarini and colleagues first reported the fabrication of human tissue-engineered trachea seeded with autologous epithelial cells and mesenchymal stem cell-derived chondrocytes. They reported that the engineered scaffold was later transplanted into a bronchomalacia patient to replace her left main bronchus. They concluded that there were evidences of functional airway activities and improved mechanical properties of the scaffold within 4 months [58].

In another clinical report by Otti and co-workers following a 5-year study, transplanted tracheal graft exhibited excellent vascularization and recellularization with respiratory epithelium and normal ciliary functions [59]. However, the authors also reported that because of longer production period of the tracheal graft, it might not be suitable for patients in need of urgent transplantation. In a quest to produce a tracheal graft with reduced production time, Baiguera and colleagues designed a human tracheal graft with production period of 3 weeks. The authors reported that the fabricated graft still possess structural and mechanical properties similar to native trachea [47].

In another innovative clinical study carried out by [60], the authors replaced an adult airway with a stem cell-seeded decellularized tracheal scaffold in a patient suffering from congenital tracheal stenosis. They concluded that the graft scaffold showed accelerated revascularization followed by epithelialization after 12 months. Recently, human-derived decellularized trachea seeded with stem cells was demonstrated to be efficient in terms of stability, epithelialization, neovascularization, and chondrocytes formation in a patient suffering from tracheal stenosis [61].

11. ASCs and breast tissue regeneration

Adipose tissue is an important constituent of soft tissues in the body that offers protection to underlying structures. Tissue flap procedures are said to be more efficient in producing a more natural reconstruction; however it is very invasive, while breast implants have been associated with complications like extrusion and lack of contraction of the breast capsule. Adipose-derived stem cells (ASCs) have been identified to be the leading candidate for breast reconstruction, although ASC supplementation has been studied in clinical trials for wound healing therapies [62].

12. Fat graft and ADSC application for breast tissue regeneration

Masuda and colleagues [63] in their report demonstrated that transplanted omental tissue both in the presence and absence of pre-adipocytes isolated from epididymal adipose tissues under the dorsal skin of Wistar rats after 12 weeks efficiently produced high levels of triacylglycerol content, capillary density, and VEGF. They concluded that co-transplantation with pre-adipocytes significantly accelerated adipose tissue formation. In another study by Matsumoto and co-workers [64], they reported that cell-assisted lipotransfer (CAL) fat had an increased survival rate than non-CAL fat, and there were early signs of microvasculature in CAL fat. Moseley and colleagues [65] using a nude mice showed that fat supplemented with ASCs sustained its adipocyte-rich appearance and weighed 2.5× greater compared to non-ASC supplemented grafted fat. Furthermore, Zhu and co-workers reported comparable findings that fat grafts treated with ASCs increase capillary density and neovascularization [66]. Several studies have also reported that cultured human ASCs produce and release several angiogenic growth factors under hypoxia condition [67, 68] and have been associated with increased fat graft microvasculature.

Coleman and colleagues in a retrospective study of 17 breast procedures done from 1995 to 2000 reported that all patients had a significant enhancement in their breast size and shape postoperatively [69]. Coleman and colleagues stated that most patients in their study underwent mammography a year after breast surgery without any known screening complications. Yoshimura and colleagues [70] in their clinical study did CAL on six patients with facial lipoatrophy. The authors concluded that the CAL group had a better clinical improvement score compared to the non-CAL patients. In another related study by Yoshimura and colleagues [71], they conducted two clinical trials, using CAL for breast reconstruction. Yoshimura et al. concluded that after treatment of 55 patients, there was advancement in the clinical results with evidences of graft retention. In addition, reconstruction and retention outcomes were demonstrated by Kitamura and co-workers [72] after CAL treatment in five patients.

In a more recent clinical study by Tissiani and Alonso [73], they investigated the effectiveness of autologous fat grafts supplemented with stromal vascular fraction (SVF) in secondary breast reconstruction surgery. The authors concluded that after 3 years of follow-up of the patients they proved volumetric persistence of this type of fat tissue grafts without any significant clinical complications recorded. In another clinical study by Claro and colleagues, it was reported that the complication rate after autologous fat tissue grafting was low compared to the complication rate after breast reconstruction surgery procedures done with breast implants and/or myocutaneous flaps [74].

13. ASCs and skin tissue engineering

Böttcher-Haberzeth and colleagues have extensively reported the development of a skin substitute known as tissue-engineered dermo-epidermal skin substitutes (DESS). It is made of the two basic native skin layers, epidermal and dermal layers, and it can potentially serve as a near replacement for the natural skin for clinical application [75]. Adipose-derived stem cells are attractive and valuable tools for regenerative skin engineering as they can differentiate into different skin cell lineages as well as secrete paracrine factors responsible for initiating skin tissue repair and regeneration.

Trottier and colleagues demonstrated the endogenous production of the extracellular cell matrix components by various skin cells known as IFATS collection. The authors reported that through this method there was formation of strong multiple layers of cell sheet that lead to increase in the skin graft thickness. The authors recorded satisfactory epidermal thickness and stratification [76]. In another study by [77], the authors seeded ASCs onto different scaffolds to determine the differentiation fate of the respective cells. The ASCs seeded on collagen type 1-based matrix and PEGylated fibrin-based scaffold differentiated into fibroblast-like dermal cells and blood capillary network, respectively. Recently, tropoelastin-based scaffold for skin substitutes was developed by [78]. Briefly, biomimetic scaffold was seeded in vitro with ASCs and transplanted onto the SCID mice. The authors concluded that ASCs grew rapidly and colonized the scaffold that resulted in increased epidermal thickness in vivo.

14. Conclusion

Scaffold-based tissue engineering using stem cells has improved the field of tissue regeneration in medicine; however, it is still at the infancy level. An extensive in-depth scientific knowledge and study of different stem cells will go a long way to translate them to clinical application. In addition, more extensive studies are needed to be done on different scaffold designs because the success of tissue engineering depends on these scaffolds and provides a niche to transplanted cells. Furthermore, most of the use of stem cells in tissue regeneration has been directed toward small tissue defect as such efforts to develop bioengineered grafts to repair larger tissue defects (bone defects) should be made. Several stem cells like induced pluripotent stem cell, mesenchymal stem cells, and ASCs are promising source of patient-specific stem cells with great regenerative potential. However, few or no clinical translation is available as they are potential teratoma and carcinogenic causative agents, and isolation of some of these cells is deemed unethical. Stem cells seeded on decellularized scaffolds have been reported to demonstrate promising and excellent results over the years. However, more clinical evaluations are needed to be properly sure they are safe clinically.

References

  1. 1. Kolios G, Moodley Y. Introduction to stem cells and regenerative medicine. Respiration. 2013;85(1):3-10
  2. 2. Rossant J. Stem cells from the mammalian blastocyst. Stem Cells. 2001;19(6):477-482
  3. 3. De Miguel MP, Fuentes-Julián S, Alcaina Y. Pluripotent stem cells: Origin, maintenance and induction. Stem Cell Reviews. 2010;6(4):633-649
  4. 4. Paschos NK, Brown WE, Eswaramoorthy R, Hu JC, Athanasiou KA. Advances in tissue engineering through stem cell-based co-culture. Journal of Tissue Engineering and Regenerative Medicine. 2015;9(5):488-503
  5. 5. Kang H et al. In vivo cartilage repair using adipose-derived stem cell-loaded decellularized cartilage ECM scaffolds. Journal of Tissue Engineering and Regenerative Medicine. 2014;8(6):442-453
  6. 6. Zuk PA et al. Human adipose tissue is a source of multipotent stem cells. Molecular Biology of the Cell. 2002;13:4279-4295
  7. 7. Seo B-M et al. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet. 2004;364(9429):149-155
  8. 8. Pittenger MF. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143-147
  9. 9. Aktas M et al. Good manufacturing practice-grade production of unrestricted somatic stem cell from fresh cord blood. Cytotherapy. 2010;12(3):338-348
  10. 10. Nazarov I et al. Multipotent stromal stem cells from human placenta demonstrate high therapeutic potential. Stem Cells Translational Medicine. 2012;1(5):359-372
  11. 11. Gong X et al. Isolation and characterization of lung resident mesenchymal stem cells capable of differentiating into alveolar epithelial type II cells. Cell Biology International. 2014;38(4):405-411
  12. 12. Nomura I, Watanabe K, Matsubara H, Hayashi K, Sugimoto N, Tsuchiya H. Uncultured autogenous adipose-derived regenerative cells promote bone formation during distraction osteogenesis in rats. Clinical Orthopaedics and Related Research. 2014;472(12):3798-3806
  13. 13. Sunay O et al. Autologous rabbit adipose tissue-derived mesenchymal stromal cells for the treatment of bone injuries with distraction osteogenesis. Cytotherapy. 2013;15(6):690-702
  14. 14. Ng MH et al. Repair of segmental load-bearing bone defect by autologous mesenchymal stem cells and plasma-derived fibrin impregnated ceramic block results in early recovery of limb function. BioMed Research International. 2014;2014:345910
  15. 15. Hatzistergos KE et al. Bone marrow mesenchymal stem cells stimulate cardiac stem cell proliferation and differentiation. Circulation Research. 2010;107(7):913-922
  16. 16. Liu Y, Yang R, Shi S. Systemic infusion of mesenchymal stem cells improves cell-based bone regeneration via upregulation of regulatory T cells. Tissue Engineering. Part A. 2015;21(3-4):498-509
  17. 17. Im J-Y, Min W-K, You C, Kim H-O, Jin H-K, Bae J. Bone regeneration of mouse critical-sized calvarial defects with human mesenchymal stem cells in scaffold. Laboratory Animal Research. 2013;29(4):196
  18. 18. Yellowley C. CXCL12/CXCR4 signaling and other recruitment and homing pathways in fracture repair. Bonekey Reports. 2013;2(3):300
  19. 19. Ho C-Y, Sanghani A, Hua J, Coathup M, Kalia P, Blunn G. Mesenchymal stem cells with increased stromal cell-derived factor 1 expression enhanced fracture healing. Tissue Engineering. Part A. 2014;21(3-4):594-602. DOI: 10.1089/ten
  20. 20. Tu Q, Valverde P, Li S, Zhang J, Yang P, Chen J. Osterix overexpression in mesenchymal stem cells stimulates healing of critical-sized defects in murine Calvarial bone. Tissue Engineering. 2007;13(10):2431-2440
  21. 21. Zou D et al. The bone-forming effects of HIF-1α-transduced BMSCs promote osseointegration with dental implant in canine mandible. PLoS One. 2012;7(3):e32355
  22. 22. Li J, Li Y, Ma S, Gao Y, Zuo Y, Hu J. Enhancement of bone formation by BMP-7 transduced MSCs on biomimetic nano-hydroxyapatite/polyamide composite scaffolds in repair of mandibular defects. Journal of Biomedial Materials Research Part A. 2010;95(4):973-981
  23. 23. Syed-Picard FN, a Shah G, Costello BJ, Sfeir C. Regeneration of periosteum by human bone marrow stromal cell sheets. Journal of Oral and Maxillofacial Surgery. 2014;72(6):1078-1083
  24. 24. Chen T, Wang Y, Bu L, Li N. Construction of functional tissue-engineered bone using cell sheet technology in a canine model. Experimental and Therapeutic Medicine. 2014;7(4):958-962
  25. 25. Ren L et al. Preparation of three-dimensional vascularized MSC cell sheet constructs for tissue regeneration. BioMed Research International. 2014;2014:301279
  26. 26. Hutton DL, Grayson WL. Stem cell-based approaches to engineering vascularized bone. Current Opinion in Chemical Engineering. 2014;3:75-82
  27. 27. Centeno C, Schultz J, Cheever M, Robinson B, Freeman M, Marasco W. Safety and complications reporting on the re-implantation of culture-expanded mesenchymal stem cells using autologous platelet lysate technique. Current Stem Cell Research & Therapy. 2010;5(1):81-93
  28. 28. Koh YG et al. Mesenchymal stem cell injections improve symptoms of knee osteoarthritis. Arthroscopy: The Journal of Arthroscopic and Related Surgery. 2013;29(4):748-755
  29. 29. Kaigler D et al. Stem cell therapy for craniofacial bone regeneration: A randomized, controlled feasibility trial. Cell Transplantation. 2013;22(5):767-777
  30. 30. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663-676
  31. 31. Ardeshirylajimi A, Dinarvand P, Seyedjafari E, Langroudi L, Jamshidi Adegani F, Soleimani M. Enhanced reconstruction of rat calvarial defects achieved by plasma-treated electrospun scaffolds and induced pluripotent stem cells. Cell and Tissue Research. 2013;354(3):849-860
  32. 32. Liu J, Chen W, Zhao Z, Xu HHK. Effect of NELL1 gene overexpression in iPSC-MSCs seeded on calcium phosphate cement. Acta Biomaterialia. 2014;10(12):5128-5138
  33. 33. Kang H et al. Mineralized gelatin methacrylate-based matrices induce osteogenic differentiation of human induced pluripotent stem cells. Acta Biomaterialia. 2014;10(12):4961-4970
  34. 34. Lian Q et al. Functional mesenchymal stem cells derived from human induced pluripotent stem cells attenuate limb ischemia in mice. Circulation. 2010;121(9):1113-1123
  35. 35. Atesok K et al. An emerging cell-based strategy in orthopaedics: Endothelial progenitor cells. Knee Surgery, Sports Traumatology, Arthroscopy. 2012;20(7):1366-1377
  36. 36. Asahara T. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275(5302):964-966
  37. 37. Zigdon-Giladi H, Michaeli-Geller G, Bick T, Lewinson D, Machtei EE. Human blood-derived endothelial progenitor cells augment vasculogenesis and osteogenesis. Journal of Clinical Periodontology. 2015;42(1):89-95
  38. 38. Li R et al. Endothelial progenitor cells for fracture healing: A microcomputed tomography and biomechanical analysis. Journal of Orthopaedic Trauma. 2011;25(8):467-471
  39. 39. Kuroda R et al. Local transplantation of G-CSF-mobilized CD34 + cells in a patient with tibial nonunion: A case report. Cell Transplantation. 2011;20(9):1491-1496
  40. 40. Moore MA et al. Decellularization of human dermis using non-denaturing anionic detergent and endonuclease: A review. Cell and Tissue Banking. 2015;16(2):249-259
  41. 41. Hung SH, Su CH, Lee FP, Tseng H. Larynx decellularization: Combining freeze-drying and sonication as an effective method. Journal of Voice. 2013;27(3):289-294
  42. 42. de Peppo GM et al. Engineering bone tissue substitutes from human induced pluripotent stem cells. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(21):8680-8685
  43. 43. Fröhlich M, Grayson WL, Marolt D, Gimble JM, Kregar-Velikonja N, Vunjak-Novakovic G. Bone grafts engineered from human adipose-derived stem cells in perfusion bioreactor culture. Tissue Engineering. Part A. 2010;16(1):179-189
  44. 44. Vincentelli A et al. In vivo autologous recellularization of a tissue-engineered heart valve: Are bone marrow mesenchymal stem cells the best candidates? The Journal of Thoracic and Cardiovascular Surgery. 2007;134(2):424-432
  45. 45. Nichols JE et al. Production and assessment of Decellularized pig and human lung scaffolds. Tissue Engineering. Part A. 2013;19(17-18):2045-2062
  46. 46. Ross EA et al. Embryonic stem cells proliferate and differentiate when seeded into kidney scaffolds. Journal of the American Society of Nephrology. 2009;20(11):2338-2347
  47. 47. Baiguera S, Del Gaudio C, Kuevda E, Gonfiotti A, Bianco A, Macchiarini P. Dynamic decellularization and cross-linking of rat tracheal matrix. Biomaterials. 2014;35(24):6344-6350
  48. 48. Baiguera S et al. Electrospun gelatin scaffolds incorporating rat decellularized brain extracellular matrix for neural tissue engineering. Biomaterials. 2014;35(4):1205-1214
  49. 49. Gray FL, Turner CG, Ahmed A, Calvert CE, Zurakowski D, Fauza DO. Prenatal tracheal reconstruction with a hybrid amniotic mesenchymal stem cells-engineered construct derived from decellularized airway. Journal of Pediatric Surgery. 2012;47(6):1072-1078
  50. 50. Nagaoka Y, Yamada H, Kimura T, Kishida A, Fujisato T, Takakuda K. Reconstruction of small diameter arteries using decellularized vascular scaffolds. Journal of Medical and Dental Sciences. 2014;61(1):33-40
  51. 51. Bertanha M et al. Morphofunctional characterization of decellularized vena cava as tissue engineering scaffolds. Experimental Cell Research. 2014;326(1):103-111
  52. 52. Baiguera S et al. Tissue engineered human tracheas for in vivo implantation. Biomaterials. 2010;31(34):8931-8938
  53. 53. Pei M, Zhang Y, Li J, Chen D. Antioxidation of decellularized stem cell matrix promotes human synovium-derived stem cell-based chondrogenesis. Stem Cells and Development. 2013;22(6):889-900
  54. 54. Jiang WC, Cheng YH, Yen MH, Chang Y, Yang VW, Lee OK. Cryo-chemical decellularization of the whole liver for mesenchymal stem cells-based functional hepatic tissue engineering. Biomaterials. 2014;35(11):3607-3617
  55. 55. Rana D, Zreiqat H, Benkirane-Jessel N, Ramakrishna S, Ramalingam M. Development of decellularized scaffolds for stem cell-driven tissue engineering. Journal of Tissue Engineering and Regenerative Medicine. 2017;11(4):942-965
  56. 56. Oberwallner B et al. Preparation of cardiac extracellular matrix scaffolds by decellularization of human myocardium. Journal of Biomedial Materials Research Part A. 2014;102(9):3263-3272
  57. 57. Dohmen PM et al. Mid-term clinical results using a tissue-engineered pulmonary valve to reconstruct the right ventricular outflow tract during the Ross procedure. The Annals of Thoracic Surgery. 2007;84(3):729-736
  58. 58. Macchiarini P et al. Clinical transplantation of a tissue-engineered airway. Lancet. 2008;372(9655):2023-2030
  59. 59. Gonfiotti A et al. The first tissue-engineered airway transplantation: 5-year follow-up results. Lancet. 2014;383(9913):238-244
  60. 60. Elliott MJ et al. Stem-cell-based, tissue engineered tracheal replacement in a child: A 2-year follow-up study. Lancet. 2012;380(9846):994-1000
  61. 61. Berg M et al. Replacement of a tracheal stenosis with a tissue-engineered human trachea using autologous stem cells: A case report. Tissue Engineering. Part A. 2014;20(1-2):389-397
  62. 62. Sterodimas A, de Faria J, Nicaretta B, and Pitanguy I. Tissue engineering with adipose-derived stem cells (ADSCs): Current and future applications. Journal of Plastic, Reconstructive & Aesthetic Surgery. 2010;63(11):1886-1892
  63. 63. Masuda T, Furue M, Matsuda T. Novel strategy for soft tissue augmentation based on transplantation of fragmented omentum and preadipocytes. Tissue Engineering. 2004;10(11-12):1672-1683
  64. 64. Matsumoto D et al. Cell-assisted lipotransfer: Supportive use of human adipose-derived cells for soft tissue augmentation with lipoinjection. Tissue Engineering. 2006;12(12):3375-3382
  65. 65. a Moseley T, Zhu M, Hedrick MH. Adipose-derived stem and progenitor cells as fillers in plastic and reconstructive surgery. Plastic and Reconstructive Surgery. 2006;118:121S-128S
  66. 66. Zhu M et al. Supplementation of fat grafts with adipose-derived regenerative cells improves long-term graft retention. Annals of Plastic Surgery. 2010;64(2):222-228
  67. 67. Rasmussen JG et al. Prolonged hypoxic culture and trypsinization increase the pro-angiogenic potential of human adipose tissue-derived stem cells. Cytotherapy. 2011;13(3):318-328
  68. 68. Rubina K et al. Adipose stromal cells stimulate angiogenesis via promoting progenitor cell differentiation, secretion of Angiogenic factors, and enhancing vessel maturation. Tissue Engineering. Part A. 2009;15(8):2039-2050
  69. 69. Coleman SR, Saboeiro AP. Fat grafting to the breast revisited: Safety and efficacy. Plastic and Reconstructive Surgery. 2007;119(3):775-785-787
  70. 70. Yoshimura K, Sato K, Aoi N, Kurita M, Hirohi T, Harii K. Cell-assisted lipotransfer for cosmetic breast augmentation: Supportive use of adipose-derived stem/stromal cells. Aesthetic Plastic Surgery. 2008;32(1):48-55
  71. 71. Yoshimura K et al. Progenitor-enriched adipose tissue transplantation as rescue for breast implant complications. The Breast Journal. 2010;16(2):169-175
  72. 72. Kitamura SKK, Kajitani K, Hedrick M. Stem cell augmented reconstruction: A new hope for reconstruction after breast conservation therapy. Breast Cancer Research and Treatment. 2011;106
  73. 73. Tissiani LAL, Alonso N. A prospective and controlled clinical trial on stromal vascular fraction enriched fat grafts in secondary breast reconstruction. Stem Cells International. 2016;2016
  74. 74. Claro F, Figueiredo JCA, Zampar AG, Pinto-Neto AM. Applicability and safety of autologous fat for reconstruction of the breast. British Journal of Surgery. 2012;99(6):768-780
  75. 75. Böttcher-Haberzeth S et al. Characterization of pigmented dermo-epidermal skin substitutes in a long-term in vivo assay. Experimental Dermatology. 2015;24(1):16-21
  76. 76. Trottier V, Marceau-Fortier G, Germain L, Vincent C, Fradette J. IFATS collection: Using human adipose-derived stem/stromal cells for the production of new skin substitutes. Stem Cells. 2008;26(10):2713-2723
  77. 77. Chan RK et al. Development of a vascularized skin construct using adipose-derived stem cells from debrided burned skin. Stem Cells International. 2012;2012:1-11
  78. 78. Kellar BER, Diller RB, Machula H, Muller J. Biomimetic skin substitutes created from tropoelastin help to promote wound healing. Frontiers in Bioengineering and Biotechnology. DOI: 10.3389/conf.FBIOE.2016.01.00174

Written By

Farideh Mohammadian

Submitted: 15 December 2017 Reviewed: 26 February 2018 Published: 06 June 2018