Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Building gut from scratch — progress and update of intestinal tissue engineering

Abstract

Short bowel syndrome (SBS), a condition defined by insufficient absorptive intestinal epithelium, is a rare disease, with an estimated prevalence up to 0.4 in 10,000 people. However, it has substantial morbidity and mortality for affected patients. The mainstay of treatment in SBS is supportive, in the form of intravenous parenteral nutrition, with the aim of achieving intestinal autonomy. The lack of a definitive curative therapy has led to attempts to harness innate developmental and regenerative mechanisms to engineer neo-intestine as an alternative approach to addressing this unmet clinical need. Exciting advances have been made in the field of intestinal tissue engineering (ITE) over the past decade, making a review in this field timely. In this Review, we discuss the latest advances in the components required to engineer intestinal grafts and summarize the progress of ITE. We also explore some key factors to consider and challenges to overcome when transitioning tissue-engineered intestine towards clinical translation, and provide the future outlook of ITE in therapeutic applications and beyond.

Key points

  • Intestinal tissue engineering has the potential to offer curative therapy in patients with short bowel syndrome.

  • Multiple components, including an absorptive mucosa, smooth muscle, enteric nerves and vasculature are required to generate a functional full-thickness intestinal graft.

  • Advances in intestinal tissue engineering include endothelial cell reprogramming and vascular engineering, generation of mucosal grafts using patient-derived materials and colon mucosal repurposing using small intestinal organoids.

  • Vascularization and lymphatic engineering, generation of multilayered personalized intestinal grafts and scaling-up of graft size present some of the future challenges in intestinal tissue engineering.

  • A collaborative approach, combining expertise in stem cell biology, engineering and biotechnology, is fundamental to advance engineered intestine towards clinical translation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Intestinal structure and components of engineered intestinal grafts.
Fig. 2: Timeline highlighting significant advances in the field of intestinal tissue engineering.
Fig. 3: Cellular and scaffold sources used to generate TESI and summary of engineering strategies to date.

Similar content being viewed by others

References

  1. Massironi, S. et al. Understanding short bowel syndrome: current status and future perspectives. Dig. Liver Dis. 52, 253–261 (2020).

    Article  PubMed  Google Scholar 

  2. Brandt, C. F. et al. Home parenteral nutrition in adult patients with chronic intestinal failure: the evolution over 4 decades in a tertiary referral center. JPEN J. Parenter. Enteral Nutr. 41, 1178–1187 (2017).

    Article  PubMed  Google Scholar 

  3. Wales, P. W. et al. Neonatal short bowel syndrome: population-based estimates of incidence and mortality rates. J. Pediatr. Surg. 39, 690–695 (2004).

    Article  PubMed  Google Scholar 

  4. Spencer, A. U. et al. Pediatric short bowel syndrome: redefining predictors of success. Ann. Surg. 242, 403–409 (2005). discussion 409–412.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Arhip, L., Serrano-Moreno, C., Romero, I., Camblor, M. & Cuerda, C. The economic costs of home parenteral nutrition: systematic review of partial and full economic evaluations. Clin. Nutr. 40, 339–349 (2021).

    Article  CAS  PubMed  Google Scholar 

  6. Grant, D. et al. Intestinal transplant registry report: global activity and trends. Am. J. Transpl. 15, 210–219 (2015).

    Article  CAS  Google Scholar 

  7. Kesseli, S. & Sudan, D. Small bowel transplantation. Surg. Clin. North. Am. 99, 103–116 (2019).

    Article  PubMed  Google Scholar 

  8. Schwartz, S. D. et al. Human embryonic stem cell-derived retinal pigment epithelium in patients with age-related macular degeneration and Stargardt’s macular dystrophy: follow-up of two open-label phase 1/2 studies. Lancet 385, 509–516 (2015).

    Article  PubMed  Google Scholar 

  9. Trounson, A. & McDonald, C. Stem cell therapies in clinical trials: progress and challenges. Cell Stem Cell 17, 11–22 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. Feldman, E. L. et al. Intraspinal neural stem cell transplantation in amyotrophic lateral sclerosis: phase 1 trial outcomes. Ann. Neurol. 75, 363–373 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Hirsch, T. et al. Regeneration of the entire human epidermis using transgenic stem cells. Nature 551, 327–332 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Brittberg, M. et al. Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N. Engl. J. Med. 331, 889–895 (1994).

    Article  CAS  PubMed  Google Scholar 

  13. European Medicines Agency. Holoclar: ex vivo expanded autologous human corneal epithelial cells containing stem cells. https://www.ema.europa.eu/en/medicines/human/EPAR/holoclar (2015).

  14. European Medicines Agency. EU/3/08/579: Orphan designation for the corneal lesions, with associated corneal (limbal) stem cell deficiency, due to ocular burns. Ex-vivo-expanded autologous human corneal epithelium-containing stem cells. https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu308579 (2015).

  15. National Institute for Health and Care Excellence. Holoclar for treating limbal stem cell deficiency after eye burns. Technology appraisal guidance [TA467]. https://www.nice.org.uk/guidance/ta467 (2017).

  16. Rama, P. et al. Limbal stem-cell therapy and long-term corneal regeneration. N. Engl. J. Med. 363, 147–155 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Atala, A., Bauer, S. B., Soker, S., Yoo, J. J. & Retik, A. B. Tissue-engineered autologous bladders for patients needing cystoplasty. Lancet 367, 1241–1246 (2006).

    Article  PubMed  Google Scholar 

  18. Ott, H. C. et al. Regeneration and orthotopic transplantation of a bioartificial lung. Nat. Med. 16, 927–933 (2010).

    Article  CAS  PubMed  Google Scholar 

  19. Elliott, M. J. et al. Stem-cell-based, tissue engineered tracheal replacement in a child: a 2-year follow-up study. Lancet 380, 994–1000 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Urbani, L. et al. Multi-stage bioengineering of a layered oesophagus with in vitro expanded muscle and epithelial adult progenitors. Nat. Commun. 9, 4286 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Ponsky, J. L. & Strong, A. T. in Gray’s Anatomy: The Anatomical Basis of Clinical Practice. 42nd edn (ed. Standring, S.) 1173–1184 (Elsevier, 2020).

  22. Clevers, H. et al. Tissue-engineering the intestine: the trials before the trials. Cell Stem Cell 24, 855–859 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. Meran, L. et al. Engineering transplantable jejunal mucosal grafts using patient-derived organoids from children with intestinal failure. Nat. Med. 26, 1593–1601 (2020).

    Article  CAS  PubMed  Google Scholar 

  24. Palikuqi, B. et al. Adaptable haemodynamic endothelial cells for organogenesis and tumorigenesis. Nature 585, 426–432 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Nikolaev, M. et al. Homeostatic mini-intestines through scaffold-guided organoid morphogenesis. Nature 585, 574–578 (2020).

    Article  PubMed  Google Scholar 

  26. Sugimoto, S. et al. An organoid-based organ-repurposing approach to treat short bowel syndrome. Nature 592, 99–104 (2021).

    CAS  PubMed  Google Scholar 

  27. Goodman, B. E. Insights into digestion and absorption of major nutrients in humans. Adv. Physiol. Educ. 34, 44–53 (2010).

    Article  PubMed  Google Scholar 

  28. Turner, J. R. Intestinal mucosal barrier function in health and disease. Nat. Rev. Immunol. 9, 799–809 (2009).

    Article  CAS  PubMed  Google Scholar 

  29. Furness, J. B. The enteric nervous system and neurogastroenterology. Nat. Rev. Gastroenterol. Hepatol. 9, 286–294 (2012).

    Article  CAS  PubMed  Google Scholar 

  30. Gehart, H. & Clevers, H. Tales from the crypt: new insights into intestinal stem cells. Nat. Rev. Gastroenterol. Hepatol. 16, 19–34 (2019).

    Article  PubMed  Google Scholar 

  31. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Yen, T. H. & Wright, N. A. The gastrointestinal tract stem cell niche. Stem Cell Rev. 2, 203–212 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Powell, D. W., Pinchuk, I. V., Saada, J. I., Chen, X. & Mifflin, R. C. Mesenchymal cells of the intestinal lamina propria. Annu. Rev. Physiol. 73, 213–237 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Meran, L., Baulies, A. & Li, V. S. W. Intestinal stem cell niche: the extracellular matrix and cellular components. Stem Cell Int. 2017, 7970385 (2017).

    Google Scholar 

  35. Clevers, H. & Nusse, R. Wnt/β-catenin signaling and disease. Cell 149, 1192–1205 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. VanDussen, K. L. et al. Notch signaling modulates proliferation and differentiation of intestinal crypt base columnar stem cells. Development 139, 488–497 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. van den Brink, G. R. Hedgehog signaling in development and homeostasis of the gastrointestinal tract. Physiol. Rev. 87, 1343–1375 (2007).

    Article  PubMed  Google Scholar 

  38. Cifarelli, V. & Eichmann, A. The intestinal lymphatic system: functions and metabolic implications. Cell Mol. Gastroenterol. Hepatol. 7, 503–513 (2019).

    Article  PubMed  Google Scholar 

  39. Bjerknes, M. & Cheng, H. Modulation of specific intestinal epithelial progenitors by enteric neurons. Proc. Natl Acad. Sci. USA 98, 12497–12502 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Obata, Y. et al. Neuronal programming by microbiota regulates intestinal physiology. Nature 578, 284–289 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Rooks, M. G. & Garrett, W. S. Gut microbiota, metabolites and host immunity. Nat. Rev. Immunol. 16, 341–352 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Schlieve, C. R. et al. Neural crest cell implantation restores enteric nervous system function and alters the gastrointestinal transcriptome in human tissue-engineered small intestine. Stem Cell Rep. 9, 883–896 (2017).

    Article  CAS  Google Scholar 

  43. Choi, R. S. & Vacanti, J. P. Preliminary studies of tissue-engineered intestine using isolated epithelial organoid units on tubular synthetic biodegradable scaffolds. Transplant. Proc. 29, 848–851 (1997).

    Article  CAS  PubMed  Google Scholar 

  44. Grikscheit, T. C. et al. Tissue-engineered small intestine improves recovery after massive small bowel resection. Ann. Surg. 240, 748–754 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Evans, G. S., Flint, N., Somers, A. S., Eyden, B. & Potten, C. S. The development of a method for the preparation of rat intestinal epithelial cell primary cultures. J. Cell Sci. 101, 219–231 (1992).

    Article  PubMed  Google Scholar 

  46. Hou, X. et al. Short-term and long-term human or mouse organoid units generate tissue-engineered small intestine without added signalling molecules. Exp. Physiol. 103, 1633–1644 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Martin, G. R. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc. Natl Acad. Sci. USA 78, 7634–7638 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Thomson, J. A. et al. Embryonic stem cell lines derived from human blastocysts. Science 282, 1145–1147 (1998).

    Article  CAS  PubMed  Google Scholar 

  49. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  PubMed  Google Scholar 

  51. Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917–1920 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Sato, T. et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology 141, 1762–1772 (2011).

    Article  CAS  PubMed  Google Scholar 

  53. Sato, T. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).

    Article  CAS  PubMed  Google Scholar 

  54. Spence, J. R. et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature 470, 105–109 (2011).

    Article  PubMed  Google Scholar 

  55. Watson, C. L. et al. An in vivo model of human small intestine using pluripotent stem cells. Nat. Med. 20, 1310–1314 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Finkbeiner, S. R. et al. Generation of tissue-engineered small intestine using embryonic stem cell-derived human intestinal organoids. Biol. Open. 4, 1462–1472 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Baulies, A., Angelis, N. & Li, V. S. W. Hallmarks of intestinal stem cells. Development 147, dev182675 (2020).

    Article  CAS  PubMed  Google Scholar 

  58. Hageman, J. H. et al. Intestinal regeneration: regulation by the microenvironment. Dev. Cell 54, 435–446 (2020).

    Article  CAS  PubMed  Google Scholar 

  59. Roulis, M. & Flavell, R. A. Fibroblasts and myofibroblasts of the intestinal lamina propria in physiology and disease. Differentiation 92, 116–131 (2016).

    Article  CAS  PubMed  Google Scholar 

  60. Lei, N. Y. et al. Intestinal subepithelial myofibroblasts support the growth of intestinal epithelial stem cells. PLoS ONE 9, e84651 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  61. McCarthy, N., Kraiczy, J. & Shivdasani, R. A. Cellular and molecular architecture of the intestinal stem cell niche. Nat. Cell Biol. 22, 1033–1041 (2020).

    Article  CAS  PubMed  Google Scholar 

  62. Aoki, R. et al. Foxl1-expressing mesenchymal cells constitute the intestinal stem cell niche. Cell Mol. Gastroenterol. Hepatol. 2, 175–188 (2016).

    Article  PubMed  Google Scholar 

  63. Shoshkes-Carmel, M. et al. Subepithelial telocytes are an important source of Wnts that supports intestinal crypts. Nature 557, 242–246 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Degirmenci, B., Valenta, T., Dimitrieva, S., Hausmann, G. & Basler, K. GLI1-expressing mesenchymal cells form the essential Wnt-secreting niche for colon stem cells. Nature 558, 449–453 (2018).

    Article  CAS  PubMed  Google Scholar 

  65. Stzepourginski, I. et al. CD34+ mesenchymal cells are a major component of the intestinal stem cells niche at homeostasis and after injury. Proc. Natl Acad. Sci. USA 114, E506–E513 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Greicius, G. et al. PDGFRα+ pericryptal stromal cells are the critical source of Wnts and RSPO3 for murine intestinal stem cells in vivo. Proc. Natl Acad. Sci. USA 115, E3173–E3181 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. McCarthy, N. et al. Distinct mesenchymal cell populations generate the essential intestinal BMP signaling gradient. Cell Stem Cell 26, 391–402.e5 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Bondurand, N., Natarajan, D., Thapar, N., Atkins, C. & Pachnis, V. Neuron and glia generating progenitors of the mammalian enteric nervous system isolated from foetal and postnatal gut cultures. Development 130, 6387–6400 (2003).

    Article  CAS  PubMed  Google Scholar 

  69. Almond, S., Lindley, R. M., Kenny, S. E., Connell, M. G. & Edgar, D. H. Characterisation and transplantation of enteric nervous system progenitor cells. Gut 56, 489–496 (2007).

    Article  PubMed  Google Scholar 

  70. Metzger, M. et al. Expansion and differentiation of neural progenitors derived from the human adult enteric nervous system. Gastroenterology 137, 2063–2073.e4 (2009).

    Article  CAS  PubMed  Google Scholar 

  71. Cooper, J. E. et al. In vivo transplantation of fetal human gut-derived enteric neural crest cells. Neurogastroenterol. Motil. 29, e12900 (2017).

    Article  Google Scholar 

  72. Metzger, M., Caldwell, C., Barlow, A. J., Burns, A. J. & Thapar, N. Enteric nervous system stem cells derived from human gut mucosa for the treatment of aganglionic gut disorders. Gastroenterology 136, 2214–2225.e3 (2009).

    Article  CAS  PubMed  Google Scholar 

  73. Workman, M. J. et al. Engineered human pluripotent-stem-cell-derived intestinal tissues with a functional enteric nervous system. Nat. Med. 23, 49–59 (2017).

    Article  CAS  PubMed  Google Scholar 

  74. Findlay, Q., Yap, K. K., Bergner, A. J., Young, H. M. & Stamp, L. A. Enteric neural progenitors are more efficient than brain-derived progenitors at generating neurons in the colon. Am. J. Physiol. Gastrointest. Liver Physiol. 307, G741–G748 (2014).

    Article  CAS  PubMed  Google Scholar 

  75. Yamashita, J. et al. Flk1-positive cells derived from embryonic stem cells serve as vascular progenitors. Nature 408, 92–96 (2000).

    Article  CAS  PubMed  Google Scholar 

  76. Zeng, L. & Childs, S. J. The smooth muscle microRNA miR-145 regulates gut epithelial development via a paracrine mechanism. Dev. Biol. 367, 178–186 (2012).

    Article  CAS  PubMed  Google Scholar 

  77. Tattoli, I. et al. Optimisation of isolation of richly pure and homogeneous primary human colonic smooth muscle cells. Dig. Liver Dis. 36, 735–743 (2004).

    Article  CAS  PubMed  Google Scholar 

  78. Walthers, C. M., Lee, M., Wu, B. M. & Dunn, J. C. Smooth muscle strips for intestinal tissue engineering. PLoS ONE 9, e114850 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Kobayashi, M. et al. Bioengineering functional smooth muscle with spontaneous rhythmic contraction in vitro. Sci. Rep. 8, 13544 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Nair, D. G., Han, T. Y., Lourenssen, S. & Blennerhassett, M. G. Proliferation modulates intestinal smooth muscle phenotype in vitro and in colitis in vivo. Am. J. Physiol. Gastrointest. Liver Physiol. 300, G903–G913 (2011).

    Article  CAS  PubMed  Google Scholar 

  81. Wang, Q. et al. Bioengineered intestinal muscularis complexes with long-term spontaneous and periodic contractions. PLoS ONE 13, e0195315 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  82. Hirschi, K. K. & Majesky, M. W. Smooth muscle stem cells. Anat. Rec. A Discov. Mol. Cell Evol. Biol. 276, 22–33 (2004).

    Article  PubMed  Google Scholar 

  83. Minasi, M. G. et al. The meso-angioblast: a multipotent, self-renewing cell that originates from the dorsal aorta and differentiates into most mesodermal tissues. Development 129, 2773–2783 (2002).

    Article  CAS  PubMed  Google Scholar 

  84. Dellavalle, A. et al. Pericytes of human skeletal muscle are myogenic precursors distinct from satellite cells. Nat. Cell Biol. 9, 255–267 (2007).

    Article  CAS  PubMed  Google Scholar 

  85. Thurner, M. et al. Generation of myogenic progenitor cell-derived smooth muscle cells for sphincter regeneration. Stem Cell Res. Ther. 11, 233 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Ghionzoli, M. et al. Human amniotic fluid stem cell differentiation along smooth muscle lineage. FASEB J. 27, 4853–4865 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Tamama, K., Sen, C. K. & Wells, A. Differentiation of bone marrow mesenchymal stem cells into the smooth muscle lineage by blocking ERK/MAPK signaling pathway. Stem Cell Dev. 17, 897–908 (2008).

    Article  CAS  Google Scholar 

  88. Shaffiey, S. A. et al. Intestinal stem cell growth and differentiation on a tubular scaffold with evaluation in small and large animals. Regen. Med. 11, 45–61 (2016).

    Article  CAS  PubMed  Google Scholar 

  89. Kitano, K. et al. Bioengineering of functional human induced pluripotent stem cell-derived intestinal grafts. Nat. Commun. 8, 765 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  90. Grant, C. N. et al. Human and mouse tissue-engineered small intestine both demonstrate digestive and absorptive function. Am. J. Physiol. Gastrointest. Liver Physiol. 308, G664–G677 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Asahara, T. et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science 275, 964–967 (1997).

    Article  CAS  PubMed  Google Scholar 

  92. Shi, Q. et al. Evidence for circulating bone marrow-derived endothelial cells. Blood 92, 362–367 (1998).

    Article  CAS  PubMed  Google Scholar 

  93. Rafii, S. & Lyden, D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat. Med. 9, 702–712 (2003).

    Article  CAS  PubMed  Google Scholar 

  94. Lin, Y., Weisdorf, D. J., Solovey, A. & Hebbel, R. P. Origins of circulating endothelial cells and endothelial outgrowth from blood. J. Clin. Invest. 105, 71–77 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Kalashnik, L. et al. A cell kinetic analysis of human umbilical vein endothelial cells. Mech. Ageing Dev. 120, 23–32 (2000).

    Article  CAS  PubMed  Google Scholar 

  96. Siow, R. C. Culture of human endothelial cells from umbilical veins. Methods Mol. Biol. 806, 265–274 (2012).

    Article  CAS  PubMed  Google Scholar 

  97. Levenberg, S., Golub, J. S., Amit, M., Itskovitz-Eldor, J. & Langer, R. Endothelial cells derived from human embryonic stem cells. Proc. Natl Acad. Sci. USA 99, 4391–4396 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Holloway, E. M. et al. Differentiation of human intestinal organoids with endogenous vascular endothelial cells. Dev. Cell 54, 516–528.e7 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Lee, S. J. et al. Generation of pure lymphatic endothelial cells from human pluripotent stem cells and their therapeutic effects on wound repair. Sci. Rep. 5, 11019 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Kono, T. et al. Differentiation of lymphatic endothelial cells from embryonic stem cells on OP9 stromal cells. Arterioscler. Thromb. Vasc. Biol. 26, 2070–2076 (2006).

    Article  CAS  PubMed  Google Scholar 

  101. Gibot, L. et al. Cell-based approach for 3D reconstruction of lymphatic capillaries in vitro reveals distinct functions of HGF and VEGF-C in lymphangiogenesis. Biomaterials 78, 129–139 (2016).

    Article  CAS  PubMed  Google Scholar 

  102. Helm, C. L., Zisch, A. & Swartz, M. A. Engineered blood and lymphatic capillaries in 3-D VEGF-fibrin-collagen matrices with interstitial flow. Biotechnol. Bioeng. 96, 167–176 (2007).

    Article  CAS  PubMed  Google Scholar 

  103. Tammela, T. et al. Therapeutic differentiation and maturation of lymphatic vessels after lymph node dissection and transplantation. Nat. Med. 13, 1458–1466 (2007).

    Article  CAS  PubMed  Google Scholar 

  104. Visuri, M. T. et al. VEGF-C and VEGF-C156S in the pro-lymphangiogenic growth factor therapy of lymphedema: a large animal study. Angiogenesis 18, 313–326 (2015).

    Article  CAS  PubMed  Google Scholar 

  105. Olsson, A. K., Dimberg, A., Kreuger, J. & Claesson-Welsh, L. VEGF receptor signalling–in control of vascular function. Nat. Rev. Mol. Cell Biol. 7, 359–371 (2006).

    Article  CAS  PubMed  Google Scholar 

  106. Joukov, V. et al. A novel vascular endothelial growth factor, VEGF-C, is a ligand for the Flt4 (VEGFR-3) and KDR (VEGFR-2) receptor tyrosine kinases. EMBO J. 15, 1751 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Karkkainen, M. J. et al. Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat. Immunol. 5, 74–80 (2004).

    Article  CAS  PubMed  Google Scholar 

  108. Saha, S. et al. Macrophage-derived extracellular vesicle-packaged WNTs rescue intestinal stem cells and enhance survival after radiation injury. Nat. Commun. 7, 13096 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Biton, M. et al. T helper cell cytokines modulate intestinal stem cell renewal and differentiation. Cell 175, 1307–1320.e22 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Lynch, S. V. & Pedersen, O. The human intestinal microbiome in health and disease. N. Engl. J. Med. 375, 2369–2379 (2016).

    Article  CAS  PubMed  Google Scholar 

  111. Noel, G. et al. A primary human macrophage-enteroid co-culture model to investigate mucosal gut physiology and host-pathogen interactions. Sci. Rep. 7, 45270 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Gjorevski, N. et al. Neutrophilic infiltration in organ-on-a-chip model of tissue inflammation. Lab. Chip 20, 3365–3374 (2020).

    Article  CAS  PubMed  Google Scholar 

  113. Nozaki, K. et al. Co-culture with intestinal epithelial organoids allows efficient expansion and motility analysis of intraepithelial lymphocytes. J. Gastroenterol. 51, 206–213 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Shah, P. et al. A microfluidics-based in vitro model of the gastrointestinal human-microbe interface. Nat. Commun. 7, 11535 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Kim, R. et al. An in vitro intestinal platform with a self-sustaining oxygen gradient to study the human gut/microbiome interface. Biofabrication 12, 015006 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  116. Sasaki, N. et al. Development of a scalable coculture system for gut anaerobes and human colon epithelium. Gastroenterology 159, 388–390.e5 (2020).

    Article  CAS  PubMed  Google Scholar 

  117. Wu, H. et al. Lactobacillus reuteri maintains intestinal epithelial regeneration and repairs damaged intestinal mucosa. Gut Microbes 11, 997–1014 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  118. Mileto, S. J. et al. Clostridioides difficile infection damages colonic stem cells via TcdB, impairing epithelial repair and recovery from disease. Proc. Natl Acad. Sci. USA 117, 8064–8073 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Obata, Y. & Pachnis, V. The effect of microbiota and the immune system on the development and organization of the enteric nervous system. Gastroenterology 151, 836–844 (2016).

    Article  CAS  PubMed  Google Scholar 

  120. Mowat, A. M. & Agace, W. W. Regional specialization within the intestinal immune system. Nat. Rev. Immunol. 14, 667–685 (2014).

    Article  CAS  PubMed  Google Scholar 

  121. Cromeens, B. P. et al. Production of tissue-engineered intestine from expanded enteroids. J. Surg. Res. 204, 164–175 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Zakhem, E., Tamburrini, R., Orlando, G., Koch, K. L. & Bitar, K. N. Transplantation of a human tissue-engineered bowel in an athymic rat model. Tissue Eng. C. Methods 23, 652–660 (2017).

    Article  CAS  Google Scholar 

  123. Ju, Y. M. et al. Electrospun vascular scaffold for cellularized small diameter blood vessels: a preclinical large animal study. Acta Biomater. 59, 58–67 (2017).

    Article  CAS  PubMed  Google Scholar 

  124. Murphy, S. V., De Coppi, P. & Atala, A. Opportunities and challenges of translational 3D bioprinting. Nat. Biomed. Eng. 4, 370–380 (2020).

    Article  PubMed  Google Scholar 

  125. Perez-Gonzalez, C. et al. Mechanical compartmentalization of the intestinal organoid enables crypt folding and collective cell migration. Nat. Cell Biol. 23, 745–757 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Gjorevski, N. et al. Designer matrices for intestinal stem cell and organoid culture. Nature 539, 560–564 (2016).

    Article  CAS  PubMed  Google Scholar 

  127. Totonelli, G. et al. A rat decellularized small bowel scaffold that preserves villus-crypt architecture for intestinal regeneration. Biomaterials 33, 3401–3410 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Organ, G. M., Mooney, D. J., Hansen, L. K., Schloo, B. & Vacanti, J. P. Transplantation of enterocytes utilizing polymer-cell constructs to produce a neointestine. Transplant. Proc. 24, 3009–3011 (1992).

    CAS  PubMed  Google Scholar 

  129. Li, M. L. et al. Influence of a reconstituted basement membrane and its components on casein gene expression and secretion in mouse mammary epithelial cells. Proc. Natl Acad. Sci. USA 84, 136–140 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Cromeens, B. P., Wang, Y., Liu, Y., Johnson, J. & Besner, G. E. Critical intestinal cells originate from the host in enteroid-derived tissue-engineered intestine. J. Surg. Res. 223, 155–164 (2018).

    Article  PubMed  Google Scholar 

  131. Giobbe, G. G. et al. Extracellular matrix hydrogel derived from decellularized tissues enables endodermal organoid culture. Nat. Commun. 10, 5658 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  132. Cao, L. et al. Intestinal lineage commitment of embryonic stem cells. Differentiation 81, 1–10 (2011).

    Article  CAS  PubMed  Google Scholar 

  133. Yui, S. et al. Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5(+) stem cell. Nat. Med. 18, 618–623 (2012).

    Article  CAS  PubMed  Google Scholar 

  134. Yuan, H. et al. Premigratory neural crest stem cells generate enteric neurons populating the mouse colon and regulating peristalsis in tissue-engineered intestine. Stem Cells Transl. Med. 10, 922–938 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Perin, S., McCann, C. J., De Coppi, P. & Thapar, N. Isolation and characterisation of mouse intestinal mesoangioblasts. Pediatr. Surg. Int. 35, 29–34 (2019).

    Article  PubMed  Google Scholar 

  136. Liu, Y. et al. Comparison of different in vivo incubation sites to produce tissue-engineered small intestine. Tissue Eng. Part. A 24, 1138–1147 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Brassard, J. A., Nikolaev, M., Hubscher, T., Hofer, M. & Lutolf, M. P. Recapitulating macro-scale tissue self-organization through organoid bioprinting. Nat. Mater. 20, 22–29 (2021).

    Article  CAS  PubMed  Google Scholar 

  138. Urciuolo, A. et al. Intravital three-dimensional bioprinting. Nat. Biomed. Eng. 4, 901–915 (2020).

    Article  CAS  PubMed  Google Scholar 

  139. Schweinlin, M. et al. Development of an advanced primary human in vitro model of the small intestine. Tissue Eng. C. Methods 22, 873–883 (2016).

    Article  CAS  Google Scholar 

  140. Cossu, G. et al. Lancet Commission: Stem cells and regenerative medicine. Lancet 391, 883–910 (2018).

    Article  PubMed  Google Scholar 

  141. Schwank, G. et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell 13, 653–658 (2013).

    Article  CAS  PubMed  Google Scholar 

  142. Fukuda, M. et al. Small intestinal stem cell identity is maintained with functional Paneth cells in heterotopically grafted epithelium onto the colon. Genes Dev. 28, 1752–1757 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Fordham, R. P. et al. Transplantation of expanded fetal intestinal progenitors contributes to colon regeneration after injury. Cell Stem Cell 13, 734–744 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Sugimoto, S. et al. Reconstruction of the human colon epithelium in vivo. Cell Stem Cell 22, 171–176.e5 (2018).

    Article  CAS  PubMed  Google Scholar 

  145. McCann, C. J. et al. Transplantation of enteric nervous system stem cells rescues nitric oxide synthase deficient mouse colon. Nat. Commun. 8, 15937 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Fattahi, F. et al. Deriving human ENS lineages for cell therapy and drug discovery in Hirschsprung disease. Nature 531, 105–109 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. King, N. M. & Perrin, J. Ethical issues in stem cell research and therapy. Stem Cell Res. Ther. 5, 85 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  148. Gore, A. et al. Somatic coding mutations in human induced pluripotent stem cells. Nature 471, 63–67 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Hussein, S. M. et al. Copy number variation and selection during reprogramming to pluripotency. Nature 471, 58–62 (2011).

    Article  CAS  PubMed  Google Scholar 

  150. Churko, J. M. et al. Transcriptomic and epigenomic differences in human induced pluripotent stem cells generated from six reprogramming methods. Nat. Biomed. Eng. 1, 826–837 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Finkbeiner, S. R. et al. Transcriptome-wide analysis reveals hallmarks of human intestine development and maturation in vitro and in vivo. Stem. Cell Rep. 4, 1140–1155 (2015).

    Article  CAS  Google Scholar 

  152. Poling, H. M. et al. Mechanically induced development and maturation of human intestinal organoids in vivo. Nat. Biomed. Eng. 2, 429–442 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Hewes, S. A. et al. In vitro models of the small intestine: engineering challenges and engineering solutions. Tissue Eng. B Rev. 26, 313–326 (2020).

    Article  Google Scholar 

  154. Doss, M. X. & Sachinidis, A. Current challenges of iPSC-based disease modeling and therapeutic implications. Cells 8, 403 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  155. Sala, F. G. et al. A multicellular approach forms a significant amount of tissue-engineered small intestine in the mouse. Tissue Eng. A 17, 1841–1850 (2011).

    Article  CAS  Google Scholar 

  156. Cruz-Acuna, R. et al. Synthetic hydrogels for human intestinal organoid generation and colonic wound repair. Nat. Cell Biol. 19, 1326–1335 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. McCann, C. J. & Thapar, N. Enteric neural stem cell therapies for enteric neuropathies. Neurogastroenterol. Motil. 30, e13369 (2018).

    Article  CAS  PubMed  Google Scholar 

  158. Quiros-Tejeira, R. E. et al. Long-term parenteral nutritional support and intestinal adaptation in children with short bowel syndrome: a 25-year experience. J. Pediatr. 145, 157–163 (2004).

    Article  PubMed  Google Scholar 

  159. Tappenden, K. A. Intestinal adaptation following resection. JPEN. J. Parenter. Enteral Nutr.38, 23S–31S (2014).

    Article  PubMed  Google Scholar 

  160. Amiot, A., Messing, B., Corcos, O., Panis, Y. & Joly, F. Determinants of home parenteral nutrition dependence and survival of 268 patients with non-malignant short bowel syndrome. Clin. Nutr. 32, 368–374 (2013).

    Article  PubMed  Google Scholar 

  161. Pfeifle, V. A., Gros, S. J., Frongia, G., Schafer, K. H. & Holland-Cunz, S. Regenerative capacity of the enteric nervous system after ileoileal anastomoses in a rat model. Eur. J. Pediatr. Surg. 27, 200–205 (2017).

    PubMed  Google Scholar 

  162. Yanagida, H., Yanase, H., Sanders, K. M. & Ward, S. M. Intestinal surgical resection disrupts electrical rhythmicity, neural responses, and interstitial cell networks. Gastroenterology 127, 1748–1759 (2004).

    Article  PubMed  Google Scholar 

  163. Soave, F. Hirschsprung’s disease. Technique and results of Soave’s operation. Br. J. Surg. 53, 1023–1027 (1966).

    Article  CAS  PubMed  Google Scholar 

  164. Chambers, W. M. & Mc, C. M. N. J. Should ileal pouch-anal anastomosis include mucosectomy? Colorectal Dis. 9, 384–392 (2007).

    Article  CAS  PubMed  Google Scholar 

  165. Kandel, P. & Wallace, M. B. Colorectal endoscopic mucosal resection (EMR). Best. Pract. Res. Clin. Gastroenterol. 31, 455–471 (2017).

    Article  PubMed  Google Scholar 

  166. Manceau, G. et al. Elective subtotal colectomy with ileosigmoid anastomosis for colon cancer preserves bowel function and quality of life. Colorectal Dis. 15, 1078–1085 (2013).

    CAS  PubMed  Google Scholar 

  167. Urbani, L. et al. Long-term cryopreservation of decellularised oesophagi for tissue engineering clinical application. PLoS ONE 12, e0179341 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  168. Wright, N. J., Zani, A. & Ade-Ajayi, N. Epidemiology, management and outcome of gastroschisis in Sub-Saharan Africa: results of an international survey. Afr. J. Paediatr. Surg. 12, 1–6 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  169. Fowler, K. L. et al. Marked stem/progenitor cell expansion occurs early after murine ileostomy: a new model. J. Surg. Res. 220, 182–196 (2017).

    Article  PubMed  Google Scholar 

  170. Schweiger, P. J. et al. Lrig1 marks a population of gastric epithelial cells capable of long-term tissue maintenance and growth in vitro. Sci. Rep. 8, 15255 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  171. Tetteh, P. W. et al. Replacement of lost Lgr5-positive stem cells through plasticity of their enterocyte-lineage daughters. Cell stem Cell 18, 203–213 (2016).

    Article  CAS  PubMed  Google Scholar 

  172. Tian, H. et al. A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature 478, 255–259 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. van Es, J. H. et al. Dll1+ secretory progenitor cells revert to stem cells upon crypt damage. Nat. Cell Biol. 14, 1099–1104 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  174. Guiu, J. et al. Tracing the origin of adult intestinal stem cells. Nature 570, 107–111 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Yui, S. et al. YAP/TAZ-dependent reprogramming of colonic epithelium links ECM remodeling to tissue regeneration. Cell Stem Cell 22, 35–49.e7 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Kim, H. B. et al. Serial transverse enteroplasty (STEP): a novel bowel lengthening procedure. J. Pediatr. Surg. 38, 425–429 (2003).

    Article  PubMed  Google Scholar 

  177. Ramos-Gonzalez, G. & Kim, H. B. Autologous intestinal reconstruction surgery. Semin. Pediatr. Surg. 27, 261–266 (2018).

    Article  PubMed  Google Scholar 

  178. Corro, C., Novellasdemunt, L. & Li, V. S. W. A brief history of organoids. Am. J. Physiol. Cell Physiol. 319, C151–C165 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Roerink, S. F. et al. Intra-tumour diversification in colorectal cancer at the single-cell level. Nature 556, 457–462 (2018).

    Article  CAS  PubMed  Google Scholar 

  180. Matano, M. et al. Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids. Nat. Med. 21, 256–262 (2015).

    Article  CAS  PubMed  Google Scholar 

  181. Neal, J. T. et al. Organoid modeling of the tumor immune microenvironment. Cell 175, 1972–1988.e16 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Bigorgne, A. E. et al. TTC7A mutations disrupt intestinal epithelial apicobasal polarity. J. Clin. Invest. 124, 328–337 (2014).

    Article  CAS  PubMed  Google Scholar 

  183. Chen, H. J. et al. A recellularized human colon model identifies cancer driver genes. Nat. Biotechnol. 34, 845–851 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  184. Pironi, L. Definitions of intestinal failure and the short bowel syndrome. Best. Pract. Res. Clin. Gastroenterol. 30, 173–185 (2016).

    Article  PubMed  Google Scholar 

  185. Batra, A., Keys, S. C., Johnson, M. J., Wheeler, R. A. & Beattie, R. M. Epidemiology, management and outcome of ultrashort bowel syndrome in infancy. Arch. Dis. Child. Fetal Neonatal Ed. 102, F551–F556 (2017).

    Article  PubMed  Google Scholar 

  186. Sulkowski, J. P. & Minneci, P. C. Management of short bowel syndrome. Pathophysiology 21, 111–118 (2014).

    Article  PubMed  Google Scholar 

  187. McDuffie, L. A. et al. Intestinal adaptation after small bowel resection in human infants. J. Pediatr. Surg. 46, 1045–1051 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  188. Carroll, R. E., Benedetti, E., Schowalter, J. P. & Buchman, A. L. Management and complications of short bowel syndrome: an updated review. Curr. Gastroenterol. Rep. 18, 40 (2016).

    Article  PubMed  Google Scholar 

  189. Kelly, D. A. Intestinal failure-associated liver disease: what do we know today? Gastroenterology 130, S70–S77 (2006).

    Article  CAS  PubMed  Google Scholar 

  190. Hollwarth, M. E. Surgical strategies in short bowel syndrome. Pediatr. Surg. Int. 33, 413–419 (2017).

    Article  PubMed  Google Scholar 

  191. Drucker, D. J. The discovery of GLP-2 and development of teduglutide for short bowel syndrome. ACS Pharmacol. Transl. Sci. 2, 134–142 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Martinez Rivera, A. & Wales, P. W. Intestinal transplantation in children: current status. Pediatr. Surg. Int. 32, 529–540 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  193. Daly, A. C. et al. 3D bioprinting for cartilage and osteochondral tissue engineering. Adv. Healthc. Mater. 6, 1700298 (2017).

    Article  Google Scholar 

  194. Cubo, N., Garcia, M., Del Canizo, J. F., Velasco, D. & Jorcano, J. L. 3D bioprinting of functional human skin: production and in vivo analysis. Biofabrication 9, 015006 (2016).

    Article  PubMed  Google Scholar 

  195. Markstedt, K. et al. 3D bioprinting human chondrocytes with nanocellulose-alginate bioink for cartilage tissue engineering applications. Biomacromolecules 16, 1489–1496 (2015).

    Article  CAS  PubMed  Google Scholar 

  196. Gao, M. et al. Tissue-engineered trachea from a 3D-printed scaffold enhances whole-segment tracheal repair. Sci. Rep. 7, 5246 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  197. Costello, C. M. et al. Synthetic small intestinal scaffolds for improved studies of intestinal differentiation. Biotechnol. Bioeng. 111, 1222–1232 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Jensen, T. et al. Polyurethane scaffolds seeded with autologous cells can regenerate long esophageal gaps: an esophageal atresia treatment model. J. Pediatr. Surg. 54, 1744–1754 (2019).

    Article  PubMed  Google Scholar 

  199. Catry, J. et al. Circumferential esophageal replacement by a tissue-engineered substitute using mesenchymal stem cells: an experimental study in mini pigs. Cell Transpl. 26, 1831–1839 (2017).

    Article  Google Scholar 

  200. Medicines and Healthcare products Regulatory Agency and Department of Health and Social Care. Guidance: good manufacturing practice and good distribution practice. https://www.gov.uk/guidance/good-manufacturing-practice-and-good-distribution-practice (2014).

  201. European Medicines Agency. Good manufacturing practice. https://www.ema.europa.eu/en/human-regulatory/research-development/compliance/good-manufacturing-practice (2021).

  202. US Food and Drug Administration. Current good manufacturing practice (CGMP) regulations. https://www.fda.gov/drugs/pharmaceutical-quality-resources/current-good-manufacturing-practice-cgmp-regulations (2020).

  203. European Medicines Agency. Advanced therapy medicinal products: overview. https://www.ema.europa.eu/en/human-regulatory/overview/advanced-therapy-medicinal-products-overview (2021).

  204. European Commission. Regulation no 1394/2007 on advanced therapy medicinal products. https://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:L:2007:324:0121:0137:en:PDF (2007).

Download references

Acknowledgements

This Review is a snapshot of the current state of intestinal tissue engineering and we apologize to the many colleagues whose work could not be cited here due to space limitations. The authors (V.S.W.L., P.De C. and L.T.) were funded by Horizon 2020 grant INTENS 668294 on the project ‘Intestinal Tissue Engineering Solution for Children with Short Bowel Syndrome’. The laboratory of V.S.W.L. is supported by the Francis Crick Institute, which receives its core funding from Cancer Research UK (FC001105), the UK Medical Research Council (FC001105) and the Wellcome Trust (FC001105). P.De C. is supported by an NIHR Professorship, NIHR UCL BRC-GOSH, the Great Ormond Street Hospital Children’s Charity and the Oak Foundation. L.T. is funded by NIHR UCL BRC-GOSH Crick Clinical Research Training Fellowship. B.C.J. is supported by the General Sir John Monash Foundation, Australia and University College London.

Author information

Authors and Affiliations

Authors

Contributions

L.T. and B.C.J. researched data for the article. L.T., B.C.J. and V.S.W.L. contributed substantially to discussion of the content. All authors wrote the article and reviewed and/or edited the manuscript before submission.

Corresponding authors

Correspondence to Paolo De Coppi or Vivian S. W. Li.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Gastroenterology & Hepatology thanks Hans Clevers; Hjalte Larsen, who co-reviewed with Kim Jensen; Simon Vales, who co-reviewed with Maxime Mahe; and Toshiro Sato for their contribution to the peer review of this work.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Stem cells

Cells with the ability to divide and produce further stem cells (self-renewal) and cells that can differentiate into specialized cell types (potency).

Organoid units

Aggregates of intestinal epithelial cells with a core of mesenchyme obtained by mechanical and enzymatic digestion of small intestinal mucosa.

Pluripotent stem cells

(PSCs). Cells with the ability to be cultured indefinitely in an undifferentiated state, whilst retaining the ability to differentiate into endoderm, mesoderm and ectoderm

Organoids

Cluster of cells growing in 3D containing stem, progenitor and differentiated cells that self-organize to resemble aspects of native tissue.

Epithelial organoids

Organoids containing stem, progenitor and differentiated cells from epithelium only (single germ layer).

Multi-tissue organoids

Organoids containing cells of multiple germ layers, established through the co-culture of different cell types or differentiation of pluripotent stem cells. Induced pluripotent stem cell-derived intestinal organoids are an example.

Neural crest cells

(NCCs). Neural progenitor cells derived from the cranial and sacral neural crest which migrate to the gut and give rise to the submucosal and myenteric plexuses of the enteric nervous system.

Progenitor cells

A transitional cell type between stem and fully differentiated cell types, that has lost the ability for self-renewal but retains the capacity for differentiation.

Mesangioblasts

Blood vessel-associated multipotent progenitor cells with the capacity to differentiate into a variety of mesodermal cell types.

Hydrogels

3D structural networks composed of natural (for example, Matrigel) or synthetic (for example, polyglycolic acid) polymer units that can absorb large amounts of water relative to the dry weight of the component polymers.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tullie, L., Jones, B.C., De Coppi, P. et al. Building gut from scratch — progress and update of intestinal tissue engineering. Nat Rev Gastroenterol Hepatol 19, 417–431 (2022). https://doi.org/10.1038/s41575-022-00586-x

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41575-022-00586-x

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing