1932

Abstract

The goal of tissue engineering is to mitigate the critical shortage of donor organs via in vitro fabrication of functional biological structures. Tissue engineering is one of the most prominent examples of interdisciplinary fields, where scientists with different backgrounds work together to boost the quality of life by addressing critical health issues. Many different fields, such as developmental and molecular biology, as well as technologies, such as micro- and nanotechnologies and additive manufacturing, have been integral for advancing the field of tissue engineering. Over the past 20 years, spectacular advancements have been achieved to harness nature's ability to cure diseased tissues and organs. Patients have received laboratory-grown tissues and organs made out of their own cells, thus eliminating the risk of rejection. However, challenges remain when addressing more complex solid organs such as the heart, liver, and kidney. Herein, we review recent accomplishments as well as challenges that must be addressed in the field of tissue engineering and provide a perspective regarding strategies in further development.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-102715-092331
2017-01-14
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/med/68/1/annurev-med-102715-092331.html?itemId=/content/journals/10.1146/annurev-med-102715-092331&mimeType=html&fmt=ahah

Literature Cited

  1. Langer R, Vacanti JP. 1.  1993. Tissue engineering. Science 260:5110920–26 [Google Scholar]
  2. Lanza R, Langer R, Vacanti JP. 2.  2013. Principles of Tissue Engineering Atlanta, GA: Elsevier Acad, 4th ed..
  3. Persidis A. 3.  1999. Tissue engineering. Nat. Biotechnol. 17:5508–10 [Google Scholar]
  4. Merrill JP, Murray JE, Harrison JH. 4.  et al. 1956. Successful homotransplantation of the kidney in an identical twin. JAMA 160:4277–82 [Google Scholar]
  5. Murray JE, Merrill JP, Harrison JH. 5.  et al. 2001. Renal homotransplantation in identical twins. J. Am. Soc. Nephrol. 12:1201–4 [Google Scholar]
  6. Starzl TE. 6.  1984. The landmark identical twin case. JAMA 251:192572–73 [Google Scholar]
  7. Murray JE, Wilson RE, Tilney NL. 7.  et al. 1968. Five years' experience in renal transplantation with immunosuppressive drugs: survival, function, complications, and the role of lymphocyte depletion by thoracic duct fistula. Ann. Surg. 168:3416–33 [Google Scholar]
  8. Atala A. 8.  2012. Regenerative medicine strategies. J. Pediatr. Surg. 47:117–28 [Google Scholar]
  9. Griffith LG, Naughton G. 9.  2002. Tissue engineering—current challenges and expanding opportunities. Science 295:55571009–14 [Google Scholar]
  10. Lu HH, El-Amin SF, Scott KD. 10.  et al. 2003. Three-dimensional, bioactive, biodegradable, polymer-bioactive glass composite scaffolds with improved mechanical properties support collagen synthesis and mineralization of human osteoblast-like cells in vitro. J. Biomed. Mater. Res. Part A 64:3465–74 [Google Scholar]
  11. Langer R. 11.  2009. Perspectives and challenges in tissue engineering and regenerative medicine. Adv. Mater. 21:32–333235–36 [Google Scholar]
  12. Khademhosseini A, Langer R, Borenstein J. 12.  et al. 2006. Microscale technologies for tissue engineering and biology. PNAS 103:82480–87 [Google Scholar]
  13. Khademhosseini A, Vacanti JP, Langer R. 13.  2009. Progress in tissue engineering. Sci. Am. 300:564–71 [Google Scholar]
  14. Gauvin R, Chen YC, Lee JW. 14.  et al. 2012. Microfabrication of complex porous tissue engineering scaffolds using 3D projection stereolithography. Biomaterials 33:153824–34 [Google Scholar]
  15. Borenstein J, Terai H, King K. 15.  et al. 2002. Microfabrication technology for vascularized tissue engineering. Biomed. Microdevices 4:3167–75 [Google Scholar]
  16. L'Heureux N, Dusserre N, Konig G. 16.  et al. 2006. Human tissue-engineered blood vessels for adult arterial revascularization. Nat. Med. 12:3361–65 [Google Scholar]
  17. Jakab K, Norotte C, Marga F. 17.  et al. 2010. Tissue engineering by self-assembly and bio-printing of living cells. Biofabrication 2:2022001 [Google Scholar]
  18. Vacanti J. 18.  2010. Tissue engineering and regenerative medicine: from first principles to state of the art. J. Pediatr. Surg. 45:2291–94 [Google Scholar]
  19. Mason C, Dunnill P. 19.  2008. A brief definition of regenerative medicine. Regen. Med. 3:11–5 [Google Scholar]
  20. Greenwood HL, Thorsteinsdóttir H, Perry G. 20.  et al. 2006. Regenerative medicine: new opportunities for developing countries. Int. J. Biotechnol. 8:160–77 [Google Scholar]
  21. Sala CC, Ribes MA, Muiños TF. 21.  et al. 2013. Current applications of tissue engineering in biomedicine. J. Biochip Tissue Chip S2:004 [Google Scholar]
  22. Jain RK, Au P, Tam J. 22.  et al. 2005. Engineering vascularized tissue. Nat. Biotechnol. 23:7821–23 [Google Scholar]
  23. Atala A. 23.  2009. Engineering organs. Curr. Opin. Biotechnol. 20:5575–92 [Google Scholar]
  24. Langer R. 24.  2007. Tissue engineering: perspectives, challenges, and future directions. Tissue Eng 13:11–2 [Google Scholar]
  25. Ingber DE, Mow VC, Butler D. 25.  et al. 2006. Tissue engineering and developmental biology: going biomimetic. Tissue Eng 12:123265–83 [Google Scholar]
  26. Lenas P, Moos M Jr., Luyten FP. 26.  2009. Developmental engineering: a new paradigm for the design and manufacturing of cell-based products. Part I: From three-dimensional cell growth to biomimetics of in vivo development. Tissue Eng. Part B Rev. 15:4381–94 [Google Scholar]
  27. Basu J, Ludlow JW. 27.  2012. Developmental engineering the kidney: leveraging principles of morphogenesis for renal regeneration. Birth Defects Res. C Embryo Today 96:130–38 [Google Scholar]
  28. Forgacs G, Newman SA. 28.  2005. Biological Physics of the Developing Embryo. Cambridge, UK: Cambridge Univ. Press
  29. Marga F, Neagu A, Kosztin I. 29.  et al. 2008. Developmental biology and tissue engineering. Birth Defects Res. C Embryo Today 81:4320–28 [Google Scholar]
  30. Steinberg MS. 30.  1963. Reconstruction of tissues by dissociated cells. Science 141:3579401–8 [Google Scholar]
  31. Steinberg MS. 31.  1978. Specific cell ligands and the differential adhesion hypothesis: How do they fit together. ? In Specificity of Embryological Interactions D Garrod 99–129 London: Chapman Hall [Google Scholar]
  32. Steinberg MS. 32.  1998. Goal-directedness in embryonic development. Integr. Biol. 1:249–59 [Google Scholar]
  33. Armstrong PB. 33.  1989. Cell sorting out: the self-assembly of tissues in vitro. Crit. Rev. Biochem. Mol. Biol. 24:2119–49 [Google Scholar]
  34. Mombach JC, Glazier JA, Raphael RC. 34.  et al. 1995. Quantitative comparison between differential adhesion models and cell sorting in the presence and absence of fluctuations. Phys. Rev. Lett. 75:112244–47 [Google Scholar]
  35. Foty RA, Forgacs G, Pfleger CM. 35.  et al. 1994. Liquid properties of embryonic tissues: measurement of interfacial tensions. Phys. Rev. Lett. 72:142298–301 [Google Scholar]
  36. Foty RA, Pfleger CM, Forgacs G. 36.  et al. 1996. Surface tensions of embryonic tissues predict their mutual envelopment behavior. Development 122:51611–20 [Google Scholar]
  37. Duguay D, Foty RA, Steinberg MS. 37.  2003. Cadherin-mediated cell adhesion and tissue segregation: qualitative and quantitative determinants. Dev. Biol. 253:2309–23 [Google Scholar]
  38. Norotte C, Marga F, Neagu A. 38.  et al. 2008. Experimental evaluation of apparent tissue surface tension based on the exact solution of the Laplace equation. Europhys. Lett. 81:446003 [Google Scholar]
  39. Mgharbel A, Delanoë-Ayari H, Rieu J-P. 39.  2009. Measuring accurately liquid and tissue surface tension with a compression plate tensiometer. HFSP J. 3:3213–21 [Google Scholar]
  40. Marmottant P, Mgharbel A, Käfer J. 40.  et al. 2009. The role of fluctuations and stress on the effective viscosity of cell aggregates. PNAS 106:4117271–75 [Google Scholar]
  41. Pérez-Pomares JM, Foty RA. 41.  2006. Tissue fusion and cell sorting in embryonic development and disease: biomedical implications. Bioessays 28:8809–21 [Google Scholar]
  42. McCune M, Shafiee A, Forgacs G. 42.  et al. 2014. Predictive modeling of post bioprinting structure formation. Soft Matter 10:111790–800 [Google Scholar]
  43. Norotte C, Marga FS, Niklason LE. 43.  et al. 2009. Scaffold-free vascular tissue engineering using bioprinting. Biomaterials 30:305910–17 [Google Scholar]
  44. Owens C, Marga F, Forgacs G. 44.  2015. Bioprinting of nerve. Essentials of 3D Biofabrication and Translation A Atala, JJ Yoo 379–94 Atlanta, GA: Elsevier [Google Scholar]
  45. Shafiee A, McCune M, Forgacs G. 45.  et al. 2015. Post-deposition bioink self-assembly: a quantitative study. Biofabrication 7:4045005 [Google Scholar]
  46. Shafiee A, Salleh MM, Yahaya M. 46.  2008. Fabrication of organic solar cells based on a blend of donor-acceptor molecules by inkjet printing technique. IEEE Int. Conf. Semicond. Elect. 2008:319–22 [Google Scholar]
  47. Shafiee A, Salleh MM, Yahaya M. 47.  2009. Fabrication of organic solar cells based on a blend of poly(3-octylthiophene-2,5-diyl) and fullerene derivative using inkjet printing technique. Proc. SPIE 7493:74932D doi: 10.1117/12.843467 [Google Scholar]
  48. Samad WZ, Salleh MM, Shafiee A. 48.  et al. 2010. Preparation nanostructure thin films of fluorine doped tin oxide by inkjet printing technique. AIP Conf. Proc. 1284:83–86 [Google Scholar]
  49. Samad WZ, Salleh MM, Shafiee A. 49.  et al. 2011. Structural, optical and electrical properties of fluorine doped tin oxide thin films deposited using inkjet printing technique. Sains Malaysiana 40:3251–57 [Google Scholar]
  50. Kang H-W, Lee SJ, Ko IK. 50.  et al. 2016. A 3D bioprinting system to produce human-scale tissue constructs with structural integrity. Nat. Biotechnol. 34:3312–19 [Google Scholar]
  51. Shafiee A, Atala A. 51.  2016. Printing technologies for medical applications. Trends Mol. Med. 22:3254–65 [Google Scholar]
  52. Gaebel R, Ma N, Liu J. 52.  et al. 2011. Patterning human stem cells and endothelial cells with laser printing for cardiac regeneration. Biomaterials 32:359218–30 [Google Scholar]
  53. Duan B, Kapetanovic E, Hockaday LA. 53.  et al. 2014. Three-dimensional printed trileaflet valve conduits using biological hydrogels and human valve interstitial cells. Acta Biomater. 10:51836–46 [Google Scholar]
  54. Wu W, DeConinck A, Lewis JA. 54.  2011. Omnidirectional printing of 3D microvascular networks. Adv. Mater. 23:24H178–83 [Google Scholar]
  55. Miller JS, Stevens KR, Yang MT. 55.  et al. 2012. Rapid casting of patterned vascular networks for perfusable engineered three-dimensional tissues. Nat. Mater. 11:9768–74 [Google Scholar]
  56. Samad WZ, Salleh MM, Shafiee A. 56.  2010. Transparent conducting thin films of fluoro doped tin oxide (FTO) deposited using inkjet printing technique. IEEE Int. Conf. Semicond. Electronics 2008:52–55 [Google Scholar]
  57. Samad WZ, Salleh MM, Shafiee A. 57.  et al. 2010. Transparent conductive electrode of fluorine doped tin oxide prepared by inkjet printing technique. Mater. Sci. Forum 663:665694–97 [Google Scholar]
  58. Li J, Wu S, Wu C. 58.  et al. 2016. Versatile surface engineering of porous nanomaterials with bioinspired polyphenol coatings for targeted and controlled drug delivery. Nanoscale 8:168600–6 [Google Scholar]
  59. Kim ES, Ahn EH, Dvir T. 59.  et al. 2014. Emerging nanotechnology approaches in tissue engineering and regenerative medicine. Int. J. Nanomed. 9:1–5 [Google Scholar]
  60. Yang HS, Ieronimakis N, Tsui JH. 60.  et al. 2014. Nanopatterned muscle cell patches for enhanced myogenesis and dystrophin expression in a mouse model of muscular dystrophy. Biomaterials 35:51478–86 [Google Scholar]
  61. Chung BG, Kang L, Khademhosseini A. 61.  2007. Micro- and nanoscale technologies for tissue engineering and drug discovery applications. Expert Opin. Drug Discov. 2:121653–68 [Google Scholar]
  62. Joo S, Kim JY, Lee E. 62.  et al. 2015. Effects of ECM protein micropatterns on the migration and differentiation of adult neural stem cells. Sci. Rep. 5:13043 [Google Scholar]
  63. Erdman N, Schmidt L, Qin W. 63.  et al. 2014. Microfluidics-based laser cell-micropatterning system. Biofabrication 6:3035025 [Google Scholar]
  64. O'Connell CD, Higgins MJ, Moulton SE. 64.  et al. 2015. Nano-bioelectronics via dip-pen nanolithography. J. Mater. Chem. C 3:256431–44 [Google Scholar]
  65. Kim DH, Han K, Gupta K. 65.  et al. 2009. Mechanosensitivity of fibroblast cell shape and movement to anisotropic substratum topography gradients. Biomaterials 30:295433–44 [Google Scholar]
  66. Allen-Hoffmann BL, Schlosser SJ, Ivarie CA. 66.  et al. 2000. Normal growth and differentiation in a spontaneously immortalized near-diploid human keratinocyte cell line, NIKS. J. Investig. Dermatol. 114:3444–55 [Google Scholar]
  67. Centanni JM, Straseski JA, Wicks A. 67.  et al. 2011. StrataGraft skin substitute is well-tolerated and is not acutely immunogenic in patients with traumatic wounds. Ann. Surg. 253:4672–83 [Google Scholar]
  68. Macmull S, Parratt MT, Bentley G. 68.  et al. 2011. Autologous chondrocyte implantation in the adolescent knee. Am. J. Sports Med. 39:81723–30 [Google Scholar]
  69. Jiang Y, Cai Y, Zhang W. 69.  et al. 2016. Human cartilage-derived progenitor cells from committed chondrocytes for efficient cartilage repair and regeneration. Stem Cells Transl. Med. 5:6733–44 [Google Scholar]
  70. Hamilton NJ, Kanani M, Roebuck DJ. 70.  et al. 2015. Tissue-engineered tracheal replacement in a child: a 4-year follow-up study. Am. J. Transplant. 15:102750–57 [Google Scholar]
  71. Zopf DA, Hollister SJ, Nelson ME. 71.  2013. Bioresorbable airway splint created with a three-dimensional printer. N. Engl. J. Med. 368:212043–45 [Google Scholar]
  72. L'Heureux N, Dusserre N, Konig G. 72.  et al. 2006. Human tissue-engineered blood vessels for adult arterial revascularization. Nat. Med. 12:3361–65 [Google Scholar]
  73. McAllister TN, Maruszewski M, Garrido SA. 73.  et al. 2009. Effectiveness of haemodialysis access with an autologous tissue-engineered vascular graft: a multicentre cohort study. Lancet 373:96731440–46 [Google Scholar]
  74. L'Heureux N, McAllister TN. Fuente LM. 74. , de la 2007. Tissue-engineered blood vessel for adult arterial revascularization. N. Engl. J. Med. 357:141451–53 [Google Scholar]
  75. Wystrychowski W, McAllister TN, Zagalski K. 75.  et al. 2014. First human use of an allogeneic tissue-engineered vascular graft for hemodialysis access. J. Vasc. Surg. 60:51353–57 [Google Scholar]
  76. Raya-Rivera A, Esquiliano DR, Yoo JJ. 76.  et al. 2011. Tissue-engineered autologous urethras for patients who need reconstruction: an observational study. Lancet 377:97721175–82 [Google Scholar]
  77. Atala A, Bauer SB, Soker S. 77.  et al. 2006. Tissue-engineered autologous bladders for patients needing cystoplasty. Lancet 367:95181241–46 [Google Scholar]
  78. Yoo JJ, Meng J, Oberpenning F. 78.  et al. 1998. Bladder augmentation using allogenic bladder submucosa seeded with cells. Urology 51:2221–25 [Google Scholar]
  79. Oberpenning F, Meng J, Yoo JJ. 79.  et al. 1999. De novo reconstitution of a functional mammalian urinary bladder by tissue engineering. Nat. Biotechnol. 17:2149–55 [Google Scholar]
  80. Raya-Rivera AM, Esquiliano D, Fierro-Pastrana R. 80.  et al. 2014. Tissue-engineered autologous vaginal organs in patients: a pilot cohort study. Lancet 384:9940329–36 [Google Scholar]
  81. Chen QZ, Harding SE, Ali NN. 81.  et al. 2008. Biomaterials in cardiac tissue engineering: ten years of research survey. Mater. Sci. Eng. Res. 59:1–61–37 [Google Scholar]
  82. Kamata S, Miyagawa S, Fukushima S. 82.  et al. 2015. Targeted delivery of adipocytokines into the heart by induced adipocyte cell-sheet transplantation yields immune tolerance and functional recovery in autoimmune-associated myocarditis in rats. Circ. J. 79:1169–79 [Google Scholar]
  83. Wendel JS, Ye L, Zhang P. 83.  et al. 2014. Functional consequences of a tissue-engineered myocardial patch for cardiac repair in a rat infarct model. Tissue Eng. Part A 20:7–81325–35 [Google Scholar]
  84. Mayfield AE, Tilokee EL, Latham N. 84.  et al. 2014. The effect of encapsulation of cardiac stem cells within matrix-enriched hydrogel capsules on cell survival, post-ischemic cell retention and cardiac function. Biomaterials 35:1133–42 [Google Scholar]
  85. Pati F, Jang J, Ha DH. 85.  et al. 2014. Printing three-dimensional tissue analogues with decellularized extracellular matrix bioink. Nat. Commun. 5:3935 [Google Scholar]
  86. Song JJ, Guyette JP, Gilpin SE. 86.  et al. 2013. Regeneration and experimental orthotopic transplantation of a bioengineered kidney. Nat. Med. 19:5646–51 [Google Scholar]
  87. Hoganson DM, Pryor HI, Vacanti JP. 87.  2008. Tissue engineering and organ structure: a vascularized approach to liver and lung. Pediatr. Res. 63:5520–26 [Google Scholar]
  88. Baptista PM, Siddiqui MM, Lozier G. 88.  et al. 2011. The use of whole organ decellularization for the generation of a vascularized liver organoid. Hepatology 53:2604–17 [Google Scholar]
  89. Ghaemmaghami AM, Hancock MJ, Harrington H. 89.  et al. 2012. Biomimetic tissues on a chip for drug discovery. Drug Discov. Today 17:3–4173–81 [Google Scholar]
  90. Matsusaki M, Sakaue K, Kadowaki K. 90.  et al. 2013. Three-dimensional human tissue chips fabricated by rapid and automatic inkjet cell printing. Adv. Healthc. Mater. 2:4534–39 [Google Scholar]
  91. Chang R, Emami K, Wu H. 91.  et al. 2010. Biofabrication of a three-dimensional liver micro-organ as an in vitro drug metabolism model. Biofabrication 2:4045004 [Google Scholar]
  92. Snyder JE, Hamid Q, Wang C. 92.  et al. 2011. Bioprinting cell-laden matrigel for radioprotection study of liver by pro-drug conversion in a dual-tissue microfluidic chip. Biofabrication 3:3034112 [Google Scholar]
/content/journals/10.1146/annurev-med-102715-092331
Loading
/content/journals/10.1146/annurev-med-102715-092331
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error