Intestinal Cell Plasticity: Comparison
Please note this is a comparison between Version 2 by Lily Guo and Version 1 by NATHALIE SPHYRIS.

Under constant barrage from chemical, pathogenic, and mechanical stresses, the intestinal epithelium is homeostatically replenished by a pool of Lgr5⁺ intestinal stem cells (ISCs), residing at the bottom of submucosal invaginations termed crypts. Decorated with the RSPO-receptor LGR5, which potentiates canonical Wnt/β-catenin signalling, these actively cycling cells can both self-renew and give rise to short-lived transit-amplifying cells. In turn, transit-amplifying cells undergo successive rounds of cell division and differentiation to generate the full gamut of terminally differentiated intestinal cell types tasked with performing pleiotropic absorptive, secretory, immune, and barrier functions. The self-renewal capabilities and multipotency of Lgr5⁺ ISCs are tightly controlled by instructive cues emanating from epithelial and stromal components of the ISC niche in the vicinity of the lower crypt. 

The intestinal epithelium displays a remarkable ability to regenerate following demise of homeostatic Lgr5⁺ ISCs post injury. Plasticity—the ability of lineage-restricted cells to regain self-renewal capacity and multi-lineage differentiation potential in response to environmental cues—is pervasive among multiple intestinal cell populations. Reserve stem-like cells, lineage-committed progenitors, and/or fully differentiated cell types can all contribute to regeneration and repair through dedifferentiation and reversion to an Lgr5⁺ stem-like state. In line with the pervasive plasticity of the intestinal epithelium, accumulating evidence supports both “bottom-up” and “top-down” histogenesis of colorectal tumours whereby the cells-of-origin comprise either ISCs at the crypt base or differentiated cells at the crypt apex, respectively. 

  • intestinal stem cells (ISCs)
  • plasticity
  • LGR5
  • intestinal epithelium
  • colorectal cancer
  • regeneration
  • YAP
  • cancer stem cells
Please wait, diff process is still running!

References

  1. Beumer, J.; Clevers, H. Cell fate specification and differentiation in the adult mammalian intestine. Nat. Rev. Mol. Cell Biol. 2021, 22, 39–53.
  2. Cheng, H.; Leblond, C.P. Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. V. Unitarian Theory of the origin of the four epithelial cell types. Am. J. Anat. 1974, 141, 537–561.
  3. Sasaki, N.; Sachs, N.; Wiebrands, K.; Ellenbroek, S.I.; Fumagalli, A.; Lyubimova, A.; Begthel, H.; van den Born, M.; van Es, J.H.; Karthaus, W.R.; et al. Reg4+ deep crypt secretory cells function as epithelial niche for Lgr5+ stem cells in colon. Proc. Natl. Acad. Sci. USA 2016, 113, E5399–E5407.
  4. De Lau, W.; Barker, N.; Low, T.Y.; Koo, B.K.; Li, V.S.; Teunissen, H.; Kujala, P.; Haegebarth, A.; Peters, P.J.; van de Wetering, M.; et al. Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. Nature 2011, 476, 293–297.
  5. Carmon, K.S.; Gong, X.; Lin, Q.; Thomas, A.; Liu, Q. R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/beta-catenin signaling. Proc. Natl. Acad. Sci. USA 2011, 108, 11452–11457.
  6. De Lau, W.; Peng, W.C.; Gros, P.; Clevers, H. The R-spondin/Lgr5/Rnf43 module: Regulator of Wnt signal strength. Genes Dev. 2014, 28, 305–316.
  7. Barker, N.; van Es, J.H.; Kuipers, J.; Kujala, P.; van den Born, M.; Cozijnsen, M.; Haegebarth, A.; Korving, J.; Begthel, H.; Peters, P.J.; et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 2007, 449, 1003–1007.
  8. Sato, T.; Vries, R.G.; Snippert, H.J.; van de Wetering, M.; Barker, N.; Stange, D.E.; van Es, J.H.; Abo, A.; Kujala, P.; Peters, P.J.; et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 2009, 459, 262–265.
  9. Tian, H.; Biehs, B.; Warming, S.; Leong, K.G.; Rangell, L.; Klein, O.D.; de Sauvage, F.J. A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature 2011, 478, 255–259.
  10. Metcalfe, C.; Kljavin, N.M.; Ybarra, R.; de Sauvage, F.J. Lgr5+ stem cells are indispensable for radiation-induced intestinal regeneration. Cell Stem Cell 2014, 14, 149–159.
  11. Potten, C.S.; Hume, W.J.; Reid, P.; Cairns, J. The segregation of DNA in epithelial stem cells. Cell 1978, 15, 899–906.
  12. Sangiorgi, E.; Capecchi, M.R. Bmi1 is expressed in vivo in intestinal stem cells. Nat. Genet. 2008, 40, 915–920.
  13. Montgomery, R.K.; Carlone, D.L.; Richmond, C.A.; Farilla, L.; Kranendonk, M.E.; Henderson, D.E.; Baffour-Awuah, N.Y.; Ambruzs, D.M.; Fogli, L.K.; Algra, S.; et al. Mouse telomerase reverse transcriptase (mTert) expression marks slowly cycling intestinal stem cells. Proc. Natl. Acad Sci. USA 2011, 108, 179–184.
  14. Takeda, N.; Jain, R.; LeBoeuf, M.R.; Wang, Q.; Lu, M.M.; Epstein, J.A. Interconversion between intestinal stem cell populations in distinct niches. Science 2011, 334, 1420–1424.
  15. Powell, A.E.; Wang, Y.; Li, Y.; Poulin, E.J.; Means, A.L.; Washington, M.K.; Higginbotham, J.N.; Juchheim, A.; Prasad, N.; Levy, S.E.; et al. The pan-ErbB negative regulator Lrig1 is an intestinal stem cell marker that functions as a tumor suppressor. Cell 2012, 149, 146–158.
  16. Li, N.; Yousefi, M.; Nakauka-Ddamba, A.; Jain, R.; Tobias, J.; Epstein, J.A.; Jensen, S.T.; Lengner, C.J. Single-cell analysis of proxy reporter allele-marked epithelial cells establishes intestinal stem cell hierarchy. Stem Cell Rep. 2014, 3, 876–891.
  17. Yan, K.S.; Chia, L.A.; Li, X.; Ootani, A.; Su, J.; Lee, J.Y.; Su, N.; Luo, Y.; Heilshorn, S.C.; Amieva, M.R.; et al. The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations. Proc. Natl. Acad. Sci. USA 2012, 109, 466–471.
  18. Asfaha, S.; Hayakawa, Y.; Muley, A.; Stokes, S.; Graham, T.A.; Ericksen, R.E.; Westphalen, C.B.; von Burstin, J.; Mastracci, T.L.; Worthley, D.L.; et al. Krt19(+)/Lgr5(-) cells are radioresistant cancer-initiating stem cells in the colon and intestine. Cell Stem Cell 2015, 16, 627–638.
  19. Bankaitis, E.D.; Ha, A.; Kuo, C.J.; Magness, S.T. Reserve stem cells in intestinal homeostasis and injury. Gastroenterology 2018, 155, 1348–1361.
  20. Tetteh, P.W.; Basak, O.; Farin, H.F.; Wiebrands, K.; Kretzschmar, K.; Begthel, H.; van den Born, M.; Korving, J.; de Sauvage, F.; van Es, J.H.; et al. Replacement of lost Lgr5-positive stem cells through plasticity of their enterocyte-lineage daughters. Cell Stem Cell 2016, 18, 203–213.
  21. Yan, K.S.; Gevaert, O.; Zheng, G.X.Y.; Anchang, B.; Probert, C.S.; Larkin, K.A.; Davies, P.S.; Cheng, Z.F.; Kaddis, J.S.; Han, A.; et al. Intestinal enteroendocrine lineage cells possess homeostatic and injury-inducible stem cell activity. Cell Stem Cell 2017, 21, 78–90.
  22. Schonhoff, S.E.; Giel-Moloney, M.; Leiter, A.B. Neurogenin 3-expressing progenitor cells in the gastrointestinal tract differentiate into both endocrine and non-endocrine cell types. Dev. Biol. 2004, 270, 443–454.
  23. Gross, S.; Balderes, D.; Liu, J.; Asfaha, S.; Gu, G.; Wang, T.C.; Sussel, L. Nkx2.2 is expressed in a subset of enteroendocrine cells with expanded lineage potential. Am. J. Physiol. Gastrointest. Liver Physiol. 2015, 309, G975–G987.
  24. Sei, Y.; Lu, X.; Liou, A.; Zhao, X.; Wank, S.A. A stem cell marker-expressing subset of enteroendocrine cells resides at the crypt base in the small intestine. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 300, G345–G356.
  25. Jadhav, U.; Saxena, M.; O’Neill, N.K.; Saadatpour, A.; Yuan, G.C.; Herbert, Z.; Murata, K.; Shivdasani, R.A. Dynamic reorganization of chromatin accessibility signatures during dedifferentiation of secretory precursors into Lgr5+ intestinal stem cells. Cell Stem Cell 2017, 21, 65–77.
  26. Van Es, J.H.; Sato, T.; van de Wetering, M.; Lyubimova, A.; Yee Nee, A.N.; Gregorieff, A.; Sasaki, N.; Zeinstra, L.; van den Born, M.; Korving, J.; et al. Dll1+ secretory progenitor cells revert to stem cells upon crypt damage. Nat. Cell Biol. 2012, 14, 1099–1104.
  27. Castillo-Azofeifa, D.; Fazio, E.N.; Nattiv, R.; Good, H.J.; Wald, T.; Pest, M.A.; de Sauvage, F.J.; Klein, O.D.; Asfaha, S. Atoh1(+) secretory progenitors possess renewal capacity independent of Lgr5(+) cells during colonic regeneration. EMBO J. 2019, 38, e99984.
  28. Ishibashi, F.; Shimizu, H.; Nakata, T.; Fujii, S.; Suzuki, K.; Kawamoto, A.; Anzai, S.; Kuno, R.; Nagata, S.; Ito, G.; et al. Contribution of ATOH1(+) cells to the homeostasis, repair, and tumorigenesis of the colonic epithelium. Stem Cell Rep. 2018, 10, 27–42.
  29. Tomic, G.; Morrissey, E.; Kozar, S.; Ben-Moshe, S.; Hoyle, A.; Azzarelli, R.; Kemp, R.; Chilamakuri, C.S.R.; Itzkovitz, S.; Philpott, A.; et al. Phospho-regulation of ATOH1 is required for plasticity of secretory progenitors and tissue regeneration. Cell Stem Cell 2018, 23, 436–443.
  30. Buczacki, S.J.; Zecchini, H.I.; Nicholson, A.M.; Russell, R.; Vermeulen, L.; Kemp, R.; Winton, D.J. Intestinal label-retaining cells are secretory precursors expressing Lgr5. Nature 2013, 495, 65–69.
  31. Harnack, C.; Berger, H.; Antanaviciute, A.; Vidal, R.; Sauer, S.; Simmons, A.; Meyer, T.F.; Sigal, M. R-spondin 3 promotes stem cell recovery and epithelial regeneration in the colon. Nat. Commun. 2019, 10, 4368.
  32. Westphalen, C.B.; Asfaha, S.; Hayakawa, Y.; Takemoto, Y.; Lukin, D.J.; Nuber, A.H.; Brandtner, A.; Setlik, W.; Remotti, H.; Muley, A.; et al. Long-lived intestinal tuft cells serve as colon cancer-initiating cells. J. Clin. Investig. 2014, 124, 1283–1295.
  33. Sei, Y.; Feng, J.; Samsel, L.; White, A.; Zhao, X.; Yun, S.; Citrin, D.; McCoy, J.P.; Sundaresan, S.; Hayes, M.M.; et al. Mature enteroendocrine cells contribute to basal and pathological stem cell dynamics in the small intestine. Am. J. Physiol. Gastrointest. Liver Physiol. 2018, 315, G495–G510.
  34. Schmitt, M.; Schewe, M.; Sacchetti, A.; Feijtel, D.; van de Geer, W.S.; Teeuwssen, M.; Sleddens, H.F.; Joosten, R.; van Royen, M.E.; van de Werken, H.J.G.; et al. Paneth cells respond to inflammation and contribute to tissue regeneration by acquiring stem-like features through SCF/c-Kit signaling. Cell Rep. 2018, 24, 2312–2328.
  35. Yu, S.; Tong, K.; Zhao, Y.; Balasubramanian, I.; Yap, G.S.; Ferraris, R.P.; Bonder, E.M.; Verzi, M.P.; Gao, N. Paneth Cell Multipotency Induced by Notch Activation following Injury. Cell Stem Cell 2018, 23, 46–59.
  36. Jones, J.C.; Brindley, C.D.; Elder, N.H.; Myers, M.G., Jr.; Rajala, M.W.; Dekaney, C.M.; McNamee, E.N.; Frey, M.R.; Shroyer, N.F.; Dempsey, P.J. Cellular plasticity of Defa4(Cre)-expressing Paneth cells in response to Notch activation and intestinal injury. Cell. Mol. Gastroenterol. Hepatol. 2019, 7, 533–554.
  37. Snippert, H.J.; van Es, J.H.; van den Born, M.; Begthel, H.; Stange, D.E.; Barker, N.; Clevers, H. Prominin-1/CD133 marks stem cells and early progenitors in mouse small intestine. Gastroenterology 2009, 136, 2187–2194.
  38. Itzkovitz, S.; Lyubimova, A.; Blat, I.C.; Maynard, M.; van Es, J.; Lees, J.; Jacks, T.; Clevers, H.; van Oudenaarden, A. Single-molecule transcript counting of stem-cell markers in the mouse intestine. Nat. Cell Biol. 2011, 14, 106–114.
  39. Munoz, J.; Stange, D.E.; Schepers, A.G.; van de Wetering, M.; Koo, B.K.; Itzkovitz, S.; Volckmann, R.; Kung, K.S.; Koster, J.; Radulescu, S.; et al. The Lgr5 intestinal stem cell signature: Robust expression of proposed quiescent ‘+4’ cell markers. EMBO J. 2012, 31, 3079–3091.
  40. Tao, S.; Tang, D.; Morita, Y.; Sperka, T.; Omrani, O.; Lechel, A.; Sakk, V.; Kraus, J.; Kestler, H.A.; Kuhl, M.; et al. Wnt activity and basal niche position sensitize intestinal stem and progenitor cells to DNA damage. EMBO J. 2015, 34, 624–640.
  41. Li, N.; Yousefi, M.; Nakauka-Ddamba, A.; Tobias, J.W.; Jensen, S.T.; Morrisey, E.E.; Lengner, C.J. Heterogeneity in readouts of canonical wnt pathway activity within intestinal crypts. Dev. Dyn. 2016, 245, 822–833.
  42. Van der Flier, L.G.; Sabates-Bellver, J.; Oving, I.; Haegebarth, A.; De Palo, M.; Anti, M.; Van Gijn, M.E.; Suijkerbuijk, S.; Van de Wetering, M.; Marra, G.; et al. The intestinal Wnt/TCF signature. Gastroenterology 2007, 132, 628–632.
  43. van der Flier, L.G.; van Gijn, M.E.; Hatzis, P.; Kujala, P.; Haegebarth, A.; Stange, D.E.; Begthel, H.; van den Born, M.; Guryev, V.; Oving, I.; et al. Transcription factor achaete scute-like 2 controls intestinal stem cell fate. Cell 2009, 136, 903–912.
  44. Schuijers, J.; Junker, J.P.; Mokry, M.; Hatzis, P.; Koo, B.K.; Sasselli, V.; van der Flier, L.G.; Cuppen, E.; van Oudenaarden, A.; Clevers, H. Ascl2 acts as an R-spondin/Wnt-responsive switch to control stemness in intestinal crypts. Cell Stem Cell 2015, 16, 158–170.
  45. Li, N.; Nakauka-Ddamba, A.; Tobias, J.; Jensen, S.T.; Lengner, C.J. Mouse Label-Retaining Cells Are Molecularly and Functionally Distinct from Reserve Intestinal Stem Cells. Gastroenterology 2016, 151, 298–310.
  46. Barriga, F.M.; Montagni, E.; Mana, M.; Mendez-Lago, M.; Hernando-Momblona, X.; Sevillano, M.; Guillaumet-Adkins, A.; Rodriguez-Esteban, G.; Buczacki, S.J.A.; Gut, M.; et al. Mex3a marks a slowly dividing subpopulation of Lgr5+ intestinal stem cells. Cell Stem Cell 2017, 20, 801–816.
  47. Giroux, V.; Stephan, J.; Chatterji, P.; Rhoades, B.; Wileyto, E.P.; Klein-Szanto, A.J.; Lengner, C.J.; Hamilton, K.E.; Rustgi, A.K. Mouse intestinal Krt15+ crypt cells are radio-resistant and tumor initiating. Stem Cell Rep. 2018, 10, 1947–1958.
  48. Sheng, X.; Lin, Z.; Lv, C.; Shao, C.; Bi, X.; Deng, M.; Xu, J.; Guerrero-Juarez, C.F.; Li, M.; Wu, X.; et al. Cycling Stem Cells Are Radioresistant and Regenerate the Intestine. Cell Rep. 2020, 32, 107952.
  49. Li, L.; Clevers, H. Coexistence of quiescent and active adult stem cells in mammals. Science 2010, 327, 542–545.
  50. Kim, T.H.; Saadatpour, A.; Guo, G.; Saxena, M.; Cavazza, A.; Desai, N.; Jadhav, U.; Jiang, L.; Rivera, M.N.; Orkin, S.H.; et al. Single-cell transcript profiles reveal multilineage priming in early progenitors derived from Lgr5(+) intestinal stem cells. Cell Rep. 2016, 16, 2053–2060.
  51. Baulies, A.; Angelis, N.; Foglizzo, V.; Danielsen, E.T.; Patel, H.; Novellasdemunt, L.; Kucharska, A.; Carvalho, J.; Nye, E.; De Coppi, P.; et al. The transcription co-repressors MTG8 and MTG16 regulate exit of intestinal stem cells from their niche and differentiation into enterocyte vs secretory lineages. Gastroenterology 2020, 159, 1328–1341.
  52. Leedham, S.J. Reserving the right to change the intestinal stem cell model. Cell Stem Cell 2020, 26, 301–302.
  53. Murata, K.; Jadhav, U.; Madha, S.; van Es, J.; Dean, J.; Cavazza, A.; Wucherpfennig, K.; Michor, F.; Clevers, H.; Shivdasani, R.A. Ascl2-dependent cell dedifferentiation drives regeneration of ablated intestinal stem cells. Cell Stem Cell 2020, 26, 377–390.
  54. Kim, T.H.; Li, F.; Ferreiro-Neira, I.; Ho, L.L.; Luyten, A.; Nalapareddy, K.; Long, H.; Verzi, M.; Shivdasani, R.A. Broadly permissive intestinal chromatin underlies lateral inhibition and cell plasticity. Nature 2014, 506, 511–515.
  55. Nusse, Y.M.; Savage, A.K.; Marangoni, P.; Rosendahl-Huber, A.K.M.; Landman, T.A.; de Sauvage, F.J.; Locksley, R.M.; Klein, O.D. Parasitic helminths induce fetal-like reversion in the intestinal stem cell niche. Nature 2018, 559, 109–113.
  56. Yui, S.; Azzolin, L.; Maimets, M.; Pedersen, M.T.; Fordham, R.P.; Hansen, S.L.; Larsen, H.L.; Guiu, J.; Alves, M.R.P.; Rundsten, C.F.; et al. YAP/TAZ-dependent reprogramming of colonic epithelium links ECM remodeling to tissue regeneration. Cell Stem Cell 2018, 22, 35–49.
  57. Mustata, R.C.; Vasile, G.; Fernandez-Vallone, V.; Strollo, S.; Lefort, A.; Libert, F.; Monteyne, D.; Perez-Morga, D.; Vassart, G.; Garcia, M.I. Identification of Lgr5-independent spheroid-generating progenitors of the mouse fetal intestinal epithelium. Cell Rep. 2013, 5, 421–432.
  58. Gregorieff, A.; Wrana, J.L. Hippo signalling in intestinal regeneration and cancer. Curr. Opin. Cell Biol. 2017, 48, 17–25.
  59. Gregorieff, A.; Liu, Y.; Inanlou, M.R.; Khomchuk, Y.; Wrana, J.L. Yap-dependent reprogramming of Lgr5(+) stem cells drives intestinal regeneration and cancer. Nature 2015, 526, 715–718.
  60. Barry, E.R.; Morikawa, T.; Butler, B.L.; Shrestha, K.; de la Rosa, R.; Yan, K.S.; Fuchs, C.S.; Magness, S.T.; Smits, R.; Ogino, S.; et al. Restriction of intestinal stem cell expansion and the regenerative response by YAP. Nature 2013, 493, 106–110.
  61. Li, Q.; Sun, Y.; Jarugumilli, G.K.; Liu, S.; Dang, K.; Cotton, J.L.; Xiol, J.; Chan, P.Y.; DeRan, M.; Ma, L.; et al. Lats1/2 sustain intestinal stem cells and Wnt activation through TEAD-dependent and independent transcription. Cell Stem Cell 2020, 26, 675–692.
  62. Cheung, P.; Xiol, J.; Dill, M.T.; Yuan, W.C.; Panero, R.; Roper, J.; Osorio, F.G.; Maglic, D.; Li, Q.; Gurung, B.; et al. Regenerative Reprogramming of the intestinal stem cell state via Hippo signaling suppresses metastatic colorectal cancer. Cell Stem Cell 2020, 27, 590–604.
  63. Ayyaz, A.; Kumar, S.; Sangiorgi, B.; Ghoshal, B.; Gosio, J.; Ouladan, S.; Fink, M.; Barutcu, S.; Trcka, D.; Shen, J.; et al. Single-cell transcriptomes of the regenerating intestine reveal a revival stem cell. Nature 2019, 569, 121–125.
  64. Bigaeva, E.; Uniken Venema, W.T.C.; Weersma, R.K.; Festen, E.A.M. Understanding human gut diseases at single-cell resolution. Hum. Mol. Genet. 2020, 29, R51–R58.
  65. Cai, J.; Maitra, A.; Anders, R.A.; Taketo, M.M.; Pan, D. β-Catenin destruction complex-independent regulation of Hippo-YAP signaling by APC in intestinal tumorigenesis. Genes Dev. 2015, 29, 1493–1506.
  66. Roulis, M.; Kaklamanos, A.; Schernthanner, M.; Bielecki, P.; Zhao, J.; Kaffe, E.; Frommelt, L.S.; Qu, R.; Knapp, M.S.; Henriques, A.; et al. Paracrine orchestration of intestinal tumorigenesis by a mesenchymal niche. Nature 2020, 580, 524–529.
  67. Zhou, D.; Zhang, Y.; Wu, H.; Barry, E.; Yin, Y.; Lawrence, E.; Dawson, D.; Willis, J.E.; Markowitz, S.D.; Camargo, F.D.; et al. Mst1 and Mst2 protein kinases restrain intestinal stem cell proliferation and colonic tumorigenesis by inhibition of Yes-associated protein (Yap) overabundance. Proc. Natl. Acad. Sci. USA 2011, 108, E1312–E1320.
  68. Rosenbluh, J.; Nijhawan, D.; Cox, A.G.; Li, X.; Neal, J.T.; Schafer, E.J.; Zack, T.I.; Wang, X.; Tsherniak, A.; Schinzel, A.C.; et al. β-Catenin-driven cancers require a YAP1 transcriptional complex for survival and tumorigenesis. Cell 2012, 151, 1457–1473.
  69. Reischmann, N.; Andrieux, G.; Griffin, R.; Reinheckel, T.; Boerries, M.; Brummer, T. BRAF(V600E) drives dedifferentiation in small intestinal and colonic organoids and cooperates with mutant p53 and Apc loss in transformation. Oncogene 2020, 39, 6053–6070.
  70. Han, T.; Goswami, S.; Hu, Y.; Tang, F.; Zafra, M.P.; Murphy, C.; Cao, Z.; Poirier, J.T.; Khurana, E.; Elemento, O.; et al. Lineage Reversion Drives WNT Independence in Intestinal Cancer. Cancer Discov. 2020, 10, 1590–1609.
  71. Kim, H.B.; Kim, M.; Park, Y.S.; Park, I.; Kim, T.; Yang, S.Y.; Cho, C.J.; Hwang, D.; Jung, J.H.; Markowitz, S.D.; et al. Prostaglandin E(2) activates YAP and a positive-signaling loop to promote colon regeneration after colitis but also carcinogenesis in mice. Gastroenterology 2017, 152, 616–630.
  72. Koulis, C.; Yap, R.; Engel, R.; Jardé, T.; Wilkins, S.; Solon, G.; Shapiro, J.D.; Abud, H.; McMurrick, P. Personalized medicine-current and emerging predictive and prognostic biomarkers in colorectal cancer. Cancers 2020, 12, 812.
  73. Ganesh, K.; Basnet, H.; Kaygusuz, Y.; Laughney, A.M.; He, L.; Sharma, R.; O’Rourke, K.P.; Reuter, V.P.; Huang, Y.H.; Turkekul, M.; et al. L1CAM defines the regenerative origin of metastasis-initiating cells in colorectal cancer. Nat. Cancer 2020, 1, 28–45.
  74. Preston, S.L.; Wong, W.M.; Chan, A.O.; Poulsom, R.; Jeffery, R.; Goodlad, R.A.; Mandir, N.; Elia, G.; Novelli, M.; Bodmer, W.F.; et al. Bottom-up histogenesis of colorectal adenomas: Origin in the monocryptal adenoma and initial expansion by crypt fission. Cancer Res. 2003, 63, 3819–3825.
  75. Shih, I.M.; Wang, T.L.; Traverso, G.; Romans, K.; Hamilton, S.R.; Ben-Sasson, S.; Kinzler, K.W.; Vogelstein, B. Top-down morphogenesis of colorectal tumors. Proc. Natl. Acad. Sci. USA 2001, 98, 2640–2645.
  76. Barker, N.; Ridgway, R.A.; van Es, J.H.; van de Wetering, M.; Begthel, H.; van den Born, M.; Danenberg, E.; Clarke, A.R.; Sansom, O.J.; Clevers, H. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature 2009, 457, 608–611.
  77. Hilkens, J.; Timmer, N.C.; Boer, M.; Ikink, G.J.; Schewe, M.; Sacchetti, A.; Koppens, M.A.J.; Song, J.Y.; Bakker, E.R.M. RSPO3 expands intestinal stem cell and niche compartments and drives tumorigenesis. Gut 2017, 66, 1095–1105.
  78. Zhu, L.; Gibson, P.; Currle, D.S.; Tong, Y.; Richardson, R.J.; Bayazitov, I.T.; Poppleton, H.; Zakharenko, S.; Ellison, D.W.; Gilbertson, R.J. Prominin 1 marks intestinal stem cells that are susceptible to neoplastic transformation. Nature 2009, 457, 603–607.
  79. Powell, A.E.; Vlacich, G.; Zhao, Z.Y.; McKinley, E.T.; Washington, M.K.; Manning, H.C.; Coffey, R.J. Inducible loss of one Apc allele in Lrig1-expressing progenitor cells results in multiple distal colonic tumors with features of familial adenomatous polyposis. Am. J. Physiol. Gastrointest. Liver Physiol. 2014, 307, G16–G23.
  80. Cammareri, P.; Vincent, D.F.; Hodder, M.C.; Ridgway, R.A.; Murgia, C.; Nobis, M.; Campbell, A.D.; Varga, J.; Huels, D.J.; Subramani, C.; et al. TGFbeta pathway limits dedifferentiation following WNT and MAPK pathway activation to suppress intestinal tumourigenesis. Cell Death Differ. 2017, 24, 1681–1693.
  81. Schwitalla, S.; Fingerle, A.A.; Cammareri, P.; Nebelsiek, T.; Goktuna, S.I.; Ziegler, P.K.; Canli, O.; Heijmans, J.; Huels, D.J.; Moreaux, G.; et al. Intestinal tumorigenesis initiated by dedifferentiation and acquisition of stem-cell-like properties. Cell 2013, 152, 25–38.
  82. Tetteh, P.W.; Kretzschmar, K.; Begthel, H.; van den Born, M.; Korving, J.; Morsink, F.; Farin, H.; van Es, J.H.; Offerhaus, G.J.; Clevers, H. Generation of an inducible colon-specific Cre enzyme mouse line for colon cancer research. Proc. Natl. Acad. Sci. USA 2016, 113, 11859–11864.
  83. Hayakawa, Y.; Tsuboi, M.; Asfaha, S.; Kinoshita, H.; Niikura, R.; Konishi, M.; Hata, M.; Oya, Y.; Kim, W.; Middelhoff, M.; et al. BHLHA15-positive secretory precursor cells can give rise to tumors in intestine and colon in mice. Gastroenterology 2019, 156, 1066–1081.
  84. Davis, H.; Irshad, S.; Bansal, M.; Rafferty, H.; Boitsova, T.; Bardella, C.; Jaeger, E.; Lewis, A.; Freeman-Mills, L.; Giner, F.C.; et al. Aberrant epithelial GREM1 expression initiates colonic tumorigenesis from cells outside the stem cell niche. Nat. Med. 2015, 21, 62–70.
  85. Herrinton, L.J.; Liu, L.; Levin, T.R.; Allison, J.E.; Lewis, J.D.; Velayos, F. Incidence and mortality of colorectal adenocarcinoma in persons with inflammatory bowel disease from 1998 to 2010. Gastroenterology 2012, 143, 382–389.
  86. Vermeulen, L.; Todaro, M.; de Sousa Mello, F.; Sprick, M.R.; Kemper, K.; Perez Alea, M.; Richel, D.J.; Stassi, G.; Medema, J.P. Single-cell cloning of colon cancer stem cells reveals a multi-lineage differentiation capacity. Proc. Natl. Acad. Sci. USA 2008, 105, 13427–13432.
  87. Junttila, M.R.; Mao, W.; Wang, X.; Wang, B.E.; Pham, T.; Flygare, J.; Yu, S.F.; Yee, S.; Goldenberg, D.; Fields, C.; et al. Targeting LGR5+ cells with an antibody-drug conjugate for the treatment of colon cancer. Sci. Transl. Med. 2015, 7, 314ra186.
  88. Kemper, K.; Prasetyanti, P.R.; De Lau, W.; Rodermond, H.; Clevers, H.; Medema, J.P. Monoclonal antibodies against Lgr5 identify human colorectal cancer stem cells. Stem Cells 2012, 30, 2378–2386.
  89. Schepers, A.G.; Snippert, H.J.; Stange, D.E.; van den Born, M.; van Es, J.H.; van de Wetering, M.; Clevers, H. Lineage tracing reveals Lgr5+ stem cell activity in mouse intestinal adenomas. Science 2012, 337, 730–735.
  90. Roper, J.; Tammela, T.; Cetinbas, N.M.; Akkad, A.; Roghanian, A.; Rickelt, S.; Almeqdadi, M.; Wu, K.; Oberli, M.A.; Sanchez-Rivera, F.J.; et al. In vivo genome editing and organoid transplantation models of colorectal cancer and metastasis. Nat. Biotechnol. 2017, 35, 569–576.
  91. de Sousa e Melo, F.; Kurtova, A.V.; Harnoss, J.M.; Kljavin, N.; Hoeck, J.D.; Hung, J.; Anderson, J.E.; Storm, E.E.; Modrusan, Z.; Koeppen, H.; et al. A distinct role for Lgr5(+) stem cells in primary and metastatic colon cancer. Nature 2017, 543, 676–680.
  92. Cortina, C.; Turon, G.; Stork, D.; Hernando-Momblona, X.; Sevillano, M.; Aguilera, M.; Tosi, S.; Merlos-Suarez, A.; Stephan-Otto Attolini, C.; Sancho, E.; et al. A genome editing approach to study cancer stem cells in human tumors. EMBO Mol. Med. 2017, 9, 869–879.
  93. Shimokawa, M.; Ohta, Y.; Nishikori, S.; Matano, M.; Takano, A.; Fujii, M.; Date, S.; Sugimoto, S.; Kanai, T.; Sato, T. Visualization and targeting of LGR5(+) human colon cancer stem cells. Nature 2017, 545, 187–192.
  94. Kobayashi, S.; Yamada-Okabe, H.; Suzuki, M.; Natori, O.; Kato, A.; Matsubara, K.; Jau Chen, Y.; Yamazaki, M.; Funahashi, S.; Yoshida, K.; et al. LGR5-positive colon cancer stem cells interconvert with drug-resistant LGR5-negative cells and are capable of tumor reconstitution. Stem Cells 2012, 30, 2631–2644.
  95. Fumagalli, A.; Suijkerbuijk, S.J.E.; Begthel, H.; Beerling, E.; Oost, K.C.; Snippert, H.J.; van Rheenen, J.; Drost, J. A surgical orthotopic organoid transplantation approach in mice to visualize and study colorectal cancer progression. Nat. Protoc. 2018, 13, 235–247.
  96. Fumagalli, A.; Oost, K.C.; Kester, L.; Morgner, J.; Bornes, L.; Bruens, L.; Spaargaren, L.; Azkanaz, M.; Schelfhorst, T.; Beerling, E.; et al. Plasticity of Lgr5-negative cancer cells drives metastasis in colorectal cancer. Cell Stem Cell 2020, 26, 569–578.
More
Video Production Service