Browse > Article
http://dx.doi.org/10.14348/molcells.2018.2167

Indole-3-Carbinol Promotes Goblet-Cell Differentiation Regulating Wnt and Notch Signaling Pathways AhR-Dependently  

Park, Joo-Hung (Department of Biology, Changwon National University)
Lee, Jeong-Min (Department of Biology, Changwon National University)
Lee, Eun-Jin (Department of Biology, Changwon National University)
Hwang, Won-Bhin (Department of Biology, Changwon National University)
Kim, Da-Jeong (Department of Biology, Changwon National University)
Abstract
Using an in vitro model of intestinal organoids derived from intestinal crypts, we examined effects of indole-3-carbinol (I3C), a phytochemical that has anticancer and aryl hydrocarbon receptor (AhR)-activating abilities and thus is sold as a dietary supplement, on the development of intestinal organoids and investigated the underlying mechanisms. I3C inhibited the in vitro development of mouse intestinal organoids. Addition of ${\alpha}$-naphthoflavone, an AhR antagonist or AhR siRNA transfection, suppressed I3C function, suggesting that I3C-mediated interference with organoid development is AhR-dependent. I3C increased the expression of Muc2 and lysozyme, lineage-specific genes for goblet cells and Paneth cells, respectively, but inhibits the expression of IAP, a marker gene for enterocytes. In the intestines of mice treated with I3C, the number of goblet cells was reduced, but the number of Paneth cells and the depth and length of crypts and villi were not changed. I3C increased the level of active nonphosphorylated ${\beta}$-catenin, but suppressed the Notch signal. As a result, expression of Hes1, a Notch target gene and a transcriptional repressor that plays a key role in enterocyte differentiation, was reduced, whereas expression of Math1, involved in the differentiation of secretory lineages, was increased. These results provide direct evidence for the role of AhR in the regulation of the development of intestinal stem cells and indicate that such regulation is likely mediated by regulation of Wnt and Notch signals.
Keywords
AhR; goblet; intestinal organoid; I3C; Notch;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Wincent, E., Amini, N., Luecke, S., Glatt, H., Bergman, J., Crescenzi, C., Rannug, A., and Rannug, U. (2009). The suggested physiologic aryl hydrocarbon receptor activator and cytochrome P4501 substrate 6-formylindolo[3,2-b]carbazole is present in humans. J. Biol. Chem. 284, 2690-2696.   DOI
2 Yaktine, A.L., Harrison, G.G., and Lawrence, R.S. (2006). Reducing exposure to dioxins and related compounds through foods in the next generation. Nutr. Rev. 64, 403-409.   DOI
3 Yang, Q., Bermingham, N.A., Finegold, M.J., and Zoghbi, H.Y. (2001). Requirement of Math1 for secretory cell lineage commitment in the mouse intestine. Science 294, 2155-2158.   DOI
4 Hankinson, O. (2005). Role of coactivators in transcriptional activation by the aryl hydrocarbon receptor. Arch. Biochem. Biophys. 433, 379-386.   DOI
5 Heuberger, J., Kosel, F., Qi, J., Grossmann, K.S., Rajewsky, K., and Birchmeier, W. (2014). Shp2/MAPK signaling controls goblet/paneth cell fate decisions in the intestine. Proc. Natl. Acad. Sci. USA 111, 3472-3477.   DOI
6 Jensen, J., Pedersen, E.E., Galante, P., Hald, J., Heller, R.S., Ishibashi, M., Kageyama, R., Guillemot, F., Serup, P., and Madsen, O.D. (2000). Control of endodermal endocrine development by Hes-1. Nat. Genet. 24, 36-44.   DOI
7 Kiss, E.A., Vonarbourg, C., Kopfmann, S., Hobeika, E., Finke, D., Esser, C., and Diefenbach, A. (2011). Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science 334, 1561-1565.   DOI
8 Kopan, R., and Ilagan, M.X. (2009). The canonical Notch signaling pathway: unfolding the activation mechanism. Cell 137, 216-233.   DOI
9 Lasorella, A., Benezra, R., and Iavarone, A. (2014). The ID proteins: master regulators of cancer stem cells and tumour aggressiveness. Nat. Rev. Cancer 14, 77-91.   DOI
10 Lee, D.M., Lee, S.H., Jeong, K.T., Hwang, S.J., and Park, J.H. (2012). SDS3 interacts with ARNT in an AhR ligand-specific manner regulating expression of cKrox and S100A4 in CD4+CD8+ DPK thymocytes differentiation. Environ. Toxicol. Pharmacol. 34, 858-868.   DOI
11 Tung, E.K., Wong, B.Y., Yau, T.O., and Ng, I.O. (2012). Upregulation of the Wnt co-receptor LRP6 promotes hepatocarcinogenesis and enhances cell invasion. PLoS One 7, e36565.   DOI
12 Yang, L., Tang, H., Kong, Y., Xie, X., Chen, J., Song, C., Liu, X., Ye, F, Li, N., Wang, N., et al. (2015). LGR5 promotes breast cancer progression and maintains stem-like cells through activation of Wnt/${\beta}$-catenin signaling. Stem Cells 33, 2913-2924.   DOI
13 Zhang, N., Yantiss, R.K., Nam, H.S., Chin, Y., Zhou, X.K., Scherl, E.J., Bosworth, B.P., Reya, T., and Clevers, H. (2005). Wnt signalling in stem cells and cancer. Nature 434, 843-850.   DOI
14 Simon-Assmann, P., Turck, N., Sidhoum-Jenny, M., Gradwohl, G., and Kedinger, M. (2007). In vitro models of intestinal epithelial cell differentiation. Cell Biol Toxicol. 23, 241-256.   DOI
15 Veldhoen, M., Hirota, K., Westendorf, A.M., Buer, J., Dumoutier L, Renauld, J.C., and Stockinger, B. (2008). The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature 453, 106-109.   DOI
16 Li, Y., Innocentin, S., Withers, D.R., Roberts, N.A., Gallagher, A.R., Grigorieva, E.F., Wilhelm, C., and Veldhoen, M. (2011). Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation. Cell 147, 629-640.   DOI
17 Li, Y., Kong, D., Ahmad, A., Bao, B., and Sarkar, F.H. (2013). Antioxidant function of isoflavone and 3,3'-diindolylmethane: are they important for cancer prevention and therapy? Antioxid. Redox. Signal. 19, 139-150.   DOI
18 van der Flier, L.G., and Clevers, H. (2009). Stem cells, self-renewal, and differentiation in the intestinal epithelium. Ann. Rev. Physiol. 71, 241-260.   DOI
19 van Es, J.H., Jay, P., Gregorieff, A., van Gijn, M.E., Jonkheer, S., Hatzis, P., Thiele, A., van den Born, M., Begthel, H., Brabletz, T., et al. (2005a). Wnt signalling induces maturation of Paneth cells in intestinal crypts. Nat. Cell Biol. 7, 381-386.   DOI
20 van Es, J.H., van Gijn, M.E., Riccio, O., van den Born, M., Vooijs, M., Begthel, H., Cozijnsen, M., Robine, S., Winton, D.J., Radtke, F., et al. (2005b). Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature 435, 959-963.   DOI
21 Mustata, R.C., Van Loy, T., Lefort, A., Libert, F., Strollo, S., Vassart, G., and Garcia, M.I. (2011). Lgr4 is required for Paneth cell differentiation and maintenance of intestinal stem cells ex vivo. EMBO Rep. 12, 558-564.   DOI
22 Maruthanila, V.L., Poornima, J., and Mirunalini, S. (2014). Attenuation of carcinogenesis and the mechanism underlying by the influence of indole-3-carbinol and its metabolite 3,3'-diindolylmethane: a therapeutic marvel. Adv. Pharmacol. Sci. 2014, 832161.
23 McManus, E.J., Sakamoto, K., Armit, L.J., Ronaldson, L., Shpiro, N., Marquez, R., and Alessi, D.R. (2005). Role that phosphorylation of GSK3 plays in insulin and Wnt signalling defined by knockin analysis. EMBO J. 24, 1571-1583.   DOI
24 Medema, J.P., and Vermeulen, L. (2011). Microenvironmental regulation of stem cells in intestinal homeostasis and cancer. Nature 474, 318-326.   DOI
25 Ohtsuka, T., Ishibashi, M., Gradwohl, G., Nakanishi, S., Guillemot, F., and Kageyama, R. (1999). Hes1 and Hes5 as notch effectors in mammalian neuronal differentiation. EMBO J. 18, 2196-2207.   DOI
26 Wang, H.C., Zhou, Y., and Huang, S.K. (2017). SHP-2 phosphatase controls aryl hydrocarbon receptor-mediated ER stress response in mast cells. Arch. Toxicol. 91, 1739-1748.   DOI
27 Weng, J.R., Tsai, C.H., Kulp, S.K., and Chen, C.S. (2008). Indole-3-carbinol as a chemopreventive and anti-cancer agent. Cancer Lett. 262, 153-163.   DOI
28 Nguyen, L.P., and Bradfield, C.A. (2008). The search for endogenous activators of the aryl hydrocarbon receptor. Chem. Res. Toxicol. 21, 102-116.   DOI
29 Park, S.L., Justiniano, R., Williams, J.D., Cabello, C.M., Qiao, S., and Wondrak, G.T. (2015). The tryptophan-derived endogenous Aryl hydrocarbon receptor ligand 6-formylindolo[3,2-b]carbazole is a nanomolar UVA photosensitizer in epidermal keratinocytes. J. Invest. Dermatol. 135, 1649-1658.   DOI
30 MacDonald, B.T., Tamai, K., and He, X. (2009). Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev. Cell 17, 9-26.   DOI
31 Park, J.H., Choi, A.J., Kim, S.J., Cheong, S.W., and Jeong, S.Y. (2016). AhR activation by 6-formylindolo[3,2-b]carbazole and 2,3,7,8-tetrachlorodibenzo-p-dioxin inhibit the development of mouse intestinal epithelial cells. Environ. Toxicol. Pharmacol. 43, 44-53.   DOI
32 Perdew, G.H., and Babbs, C.F. (1991). Production of Ah receptor ligands in rat fecal suspensions containing tryptophan or indole-3-carbinol. Nutr. Cancer 16, 209-218.   DOI
33 Schneider, A.J., Branam, A.M., P and eterson, R.E. (2014). Intersection of AHR and Wnt signaling in development, health, and disease. Int. J. Mol. Sci. 15, 17852-17885.   DOI
34 Peterson, L.W., and Artis, D. (2014). Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat. Rev. Immunol. 14, 141-153.   DOI
35 Qiu, J., Heller, J.J., Guo, X., Chen, Z.M., Fish, K., Fu, Y.X., and Zhou, L. (2012). The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity 36, 92-104.   DOI
36 Quintana, F.J., Basso, A.S., Iglesias, A.H., Korn, T., Farez, M.F., Bettelli, E., Caccamo, M., Oukka, M., and Weiner, H.L. (2008). Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor. Nature 453, 65-71.   DOI
37 Abbott, B.D., Birnbaum, L.S., and Perdew, G.H. (1995). Developmental expression of two members of a new class of transcription factors: I. Expression of aryl hydrocarbon receptor in the C57BL/6N mouse embryo. Dev. Dyn. 204, 133-143.   DOI
38 Barker, N., van de Wetering, M., and Clevers, H. (2008). The intestinal stem cell. Genes Dev. 22, 1856-1864.   DOI
39 Ahmad, A., Sakr, W.A., and Rahman, K.M. (2010). Anticancer properties of indole compounds: mechanism of apoptosis induction and role in chemotherapy. Curr Drug Targets 11, 652-666.   DOI
40 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. (2007). Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003-1007.   DOI
41 Barouki, R., Coumoul, X., and Fernandez-Salguero, P.M. (2007). The aryl hydrocarbon receptor, more than a xenobiotic-interacting protein. FEBS Lett. 581, 3608-3615.   DOI
42 Bjeldanes, L.F., Kim, J.Y., Grose, K.R., Bartholomew, J.C., and Bradfield, C.A. (1991). Aromatic hydrocarbon responsivenessreceptor agonists generated from indole-3-carbinol in vitro and in vivo: comparisons with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Proc. Natl. Acad. Sci. USA 88, 9543-9547.   DOI
43 Bjerknes, M., and Cheng, H. (1981). The stem-cell zone of the small intestinal epithelium. I. Evidence from Paneth cells in the adult mouse. Am. J. Anat. 160, 51-63.   DOI
44 Bray, S.J. (2006). Notch signalling: a simple pathway becomes complex. Nat. Rev. Mol. Cell Biol. 7, 678-689.   DOI
45 Burbach, K.M., Poland, A., and Bradfield, C.A. (1992). Cloning of the Ah-receptor cDNA reveals a distinctive ligand-activated transcription factor. Proc. Natl. Acad. Sci. USA 89, 8185-8189.   DOI
46 Celloto, V.R., Oliveira, A.J., Goncalves, J.E., Watanabe, C.S., Matioli, G., and Goncalves, R.A. (2012). Biosynthesis of indole-3-acetic acid by new Klebsiella oxytoca free and immobilized cells on inorganic matrices. ScientificWorldJournal 2012, 1-7.
47 Denison, M.S., and Nagy, S.R. (2003). Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Ann. Rev. Pharmacol. Toxicol. 43, 309-334.   DOI
48 Cheng, H., and Leblond, C.P. (1974). Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. I. Columnar cell. Am. J. Anat. 141, 461-479.   DOI
49 Chmill, S., Kadow, S., Winter, M., Weighardt, H., and Esser, C. (2010). 2,3,7,8,-tetrachlorodibenzo-p-dioxin impairs stable establishment of oral tolerance in mice. Toxicol. Sci. 118, 98-107.   DOI
50 Coant, N., Ben Mkaddem, S., Pedruzzi, E., Guichard, C., Treton, X., Ducroc, R., Freund, J.N., Cazals-Hatem, D., Bouhnik, Y., Woerther, P.L., et al. (2010). NADPH oxidase 1 modulates WNT and NOTCH1 signaling to control the fate of proliferative progenitor cells in the colon. Mol. Cell Biol. 30, 2636-2650.   DOI
51 Fre, S., Hannezo, E., Sale, S., Huyghe, M., Lafkas, D., Kissel, H., Louvi, A., Greve, J., Louvard, D., and Artavanis-Tsakonas, S. (2011). Notch lineages and activity in intestinal stem cells determined by a new set of knock-in mice. PLoS One 6, e25785.   DOI
52 Fre, S., Huyghe, M., Mourikis, P., Robine, S., Louvard, D., and Artavanis-Tsakonas, S. (2005). Notch signals control the fate of immature progenitor cells in the intestine. Nature 435, 964-968.   DOI
53 Gandarillas, A., and Watt, F.M. (1997). c-Myc promotes differentiation of human epidermal stem cells. Genes Dev. 11, 2869-2882.   DOI
54 Hammerschmidt-Kamper, C., Biljes, D., Merches, K., Steiner, I., Daldrup, T., Bol-Schoenmakers, M., Pieters, R.H.H., and Esser, C. (2017). Indole-3-carbinol, a plant nutrient and AhR-Ligand precursor, supports oral tolerance against OVA and improves peanut allergy symptoms in mice. PLoS One 12, e0180321.   DOI