Adaptation of the human aryl hydrocarbon receptor to sense microbiota-derived indoles

Troy D Hubbard, Iain A Murray, William H Bisson, Tejas S Lahoti, Krishne Gowda, Shantu G Amin, Andrew D Patterson, Gary H Perdew, Troy D Hubbard, Iain A Murray, William H Bisson, Tejas S Lahoti, Krishne Gowda, Shantu G Amin, Andrew D Patterson, Gary H Perdew

Abstract

Ligand activation of the aryl hydrocarbon (AHR) has profound effects upon the immunological status of the gastrointestinal tract, establishing and maintaining signaling networks, which facilitate host-microbe homeostasis at the mucosal interface. However, the identity of the ligand(s) responsible for such AHR-mediated activation within the gut remains to be firmly established. Here, we combine in vitro ligand binding, quantitative gene expression, protein-DNA interaction and ligand structure activity analyses together with in silico modeling of the AHR ligand binding domain to identify indole, a microbial tryptophan metabolite, as a human-AHR selective agonist. Human AHR, acting as a host indole receptor may exhibit a unique bimolecular (2:1) binding stoichiometry not observed with typical AHR ligands. Such bimolecular indole-mediated activation of the human AHR within the gastrointestinal tract may provide a foundation for inter-kingdom signaling between the enteric microflora and the immune system to promote commensalism within the gut.

Figures

Figure 1. Indole dose-response assessment of AHR-dependent…
Figure 1. Indole dose-response assessment of AHR-dependent activity.
(A) HepG2 (40/6) cells and (B) Hepa 1.1 cells were treated as indicated for 4 h; cells were lysed, and luciferase activity was measured.
Figure 2. Indole stimulates AHR-target gene expression.
Figure 2. Indole stimulates AHR-target gene expression.
(A) Expression of AHR-responsive CYP1A1, (B) CYP1B1, and (C) AHR within Caco2 cells was determined through qPCR analysis following 4 h of treatment with vehicle, TCDD (10 nM), or indole (IND) at the indicated dose. (D) The mean CYP1A1 enzymatic activity was measured in Caco2 cells following 12 h treatment with DMSO, TCDD (10 nM), or Indole (100 μM) and 3 h incubation with luciferin-CEE reagent. (E) IL6 expression within Caco2 cells was determined by qPCR following 4 h treatment with indole (20 μM) with or without the addition of IL1B (10 ng/mL), AHR dependence was evaluated by 1 h antagonist pretreatment using GNF 351 (200 nM). (F) IL6 secretion by Caco2 cells was determined by ELISA following 24 h treatment with vehicle, TCDD (10 nM) or Indole (100 μM), with or without the addition of IL1B (10 ng/mL). (G) Cyp1a1 gene expression within isolated peritoneal macrophages from C57BL6 and AHR humanized mice were evaluated by qPCR following indicated treatment of 4 h.
Figure 3. Indole is a human specific…
Figure 3. Indole is a human specific AHR ligand.
Photoaffinity ligand binding competition assay in which increasing amounts of βNF and indole were added to hAHR or mAHR liver cytosol in combination with a fixed amount of the photoaffinity ligand to evaluate relative competition of indole within the ligand binding pocket of AHR between species. Higher concentrations of competing ligand were not tested as concentrations above 10 μM can yield non-specific competition.
Figure 4. Indole facilitates human specific AHR…
Figure 4. Indole facilitates human specific AHR nuclear localization and DRE binding capacity.
(A) Nuclear translocation of AHR was determined following indicated treatment (1 h) in HepG2 (human) and Hepa1 (mouse) cell lines via western blot analysis. Relative quantification of AHR (normalized to β-actin or Lamin A/C) was determined via Phosphoimager and OptiQuant software, and presented as digitized light units (DLU). (B) In vitro translated hAHR/ARNT gel shift assay displaying treatment capacity to transform hAHR or mAHR to AHR/ARNT/DNA complex.
Figure 5. Ligand specificity of hAHR for…
Figure 5. Ligand specificity of hAHR for microbiota-derived indoles.
HepG2 (40/6) cells were treated as indicated for 4 h; cells were lysed, and luciferase activity was measured.
Figure 6. Methyl-indole isomers exhibit differential capacity…
Figure 6. Methyl-indole isomers exhibit differential capacity to mediate AHR activity.
(A) HepG2 (40/6) cells were treated as indicated for 4 h; cells were lysed, and luciferase activity was measured. (B) Expression of AHR-responsive CYP1A1 and CYP1B1 within Caco2 cells was determined through qPCR analysis following 4 h of treatment with vehicle, indole (IND), 3-methyl indole (3-MI), 2-methyl indole (2-MI), or 1-methyl indole (1-MI) at a concentration of 20 μM. (C) The mean CYP1A1 enzymatic activity was measured in Caco2 cells following 12 h treatment with DMSO, TCDD (10 nM), or indole/methyl indole isomers (100 μM) and 3 h incubation with luciferin-CEE reagent. (D) Synergistic IL6 expression within Caco2 cells was determined by qPCR following 4 h treatment with vehicle, TCDD (10 nM), or indole/methyl indole isomers (20 μM) with or without the addition of IL1B (10 ng/mL). IL6 secretion by Caco2 cells was determined by ELISA following 24 h treatment with vehicle, TCDD (10 nM) or indole/ methyl indole isomers (100 μM) with or without the addition of IL1B (10 ng/mL). (E) In vitro translated AHR/ARNT gel shift assay displaying treatment capacity to transform human AHR to AHR/ARNT/DNA complex.
Figure 7. In silico modeling of AHR…
Figure 7. In silico modeling of AHR ligand binding domain.
Homology modeling of indirubin optimized ligand binding in (A) hAHR and (B) mAHR. The predicted two indole-binding model in (C) hAHR and (D) mAHR ligand binding domain. The predicted two 3-methyl indole-binding models in (E) hAHR and (F) mAHR ligand binding domain. Blue shading indicates the space-filling volume of the ligand binding pocket.

References

    1. Hirakawa H., Kodama T., Takumi-Kobayashi A., Honda T. & Yamaguchi A. Secreted indole serves as a signal for expression of type III secretion system translocators in enterohaemorrhagic Escherichia coli O157:H7. Microbiol (Reading, England) 155, 541–550 (2009).
    1. Lee J., Jayaraman A. & Wood T. K. Indole is an inter-species biofilm signal mediated by SdiA. BMC Microbiol 7, 42 (2007).
    1. Lee J. H. & Lee J. Indole as an intercellular signal in microbial communities. FEMS Microbiol Rev 34, 426–444 (2010).
    1. Martino P. D., Fursy R., Bret L., Sundararaju B. & Phillips R. S. Indole can act as an extracellular signal to regulate biofilm formation of Escherichia coli and other indole-producing bacteria. Can J Microbiol 49, 443–449 (2003).
    1. Newton W. A. & Snell E. E. Formation and interrelationships of tryptophanase and tryptophan synthetases in Escherichia Coli. J Bacteriol 89, 355–364 (1965).
    1. Smith T. A. Modification of the method for determining the production of indole by bacteria. J Exp Med 2, 543–547 (1897).
    1. Deeley M. C. & Yanofsky C. Nucleotide sequence of the structural gene for tryptophanase of Escherichia coli K-12. J Bacteriol 147, 787–796 (1981).
    1. Karlin D. A., Mastromarino A. J., Jones R. D., Stroehlein J. R. & Lorentz O. Fecal skatole and indole and breath methane and hydrogen in patients with large bowel polyps or cancer. J Cancer Res & Clinical Oncology 109, 135–141 (1985).
    1. Bansal T., Alaniz R. C., Wood T. K. & Jayaraman A. The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation. Proc Natl Acad Sci USA 107, 228–233 (2010).
    1. Hughes D. T. & Sperandio V. Inter-kingdom signalling: communication between bacteria and their hosts. Nature Rev 6, 111–120 (2008).
    1. Shimada Y. et al. Commensal bacteria-dependent indole production enhances epithelial barrier function in the colon. PloS One 8, e80604 (2013).
    1. DiNatale B. C. et al. Kynurenic acid is a potent endogenous aryl hydrocarbon receptor ligand that synergistically induces interleukin-6 in the presence of inflammatory signaling. Toxicol Sci 115, 89–97 (2010).
    1. Schroeder J. C. et al. The uremic toxin 3-indoxyl sulfate is a potent endogenous agonist for the human aryl hydrocarbon receptor. Biochem 49, 393–400 (2010).
    1. Oberg M., Bergander L., Hakansson H., Rannug U. & Rannug A. Identification of the tryptophan photoproduct 6-formylindolo[3,2-b]carbazole, in cell culture medium, as a factor that controls the background aryl hydrocarbon receptor activity. Toxicol Sci 85, 935–943 (2005).
    1. Miller C. A. 3rd. Expression of the human aryl hydrocarbon receptor complex in yeast. Activation of transcription by indole compounds. J Biol Chem 272, 32824–32829 (1997).
    1. Opitz C. A. et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 478, 197–203 (2011).
    1. Jin U. H. et al. Microbiome-derived tryptophan metabolites and their aryl hydrocarbon receptor-dependent agonist and antagonist activities. Mol Pharmacol 85, 777–788 (2014).
    1. Li Y. et al. Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation. Cell 147, 629–640 (2011).
    1. Takamura T. et al. Lactobacillus bulgaricus OLL1181 activates the aryl hydrocarbon receptor pathway and inhibits colitis. Immunology Cell Biol 89, 817–822 (2011).
    1. Takamura T. et al. Activation of the aryl hydrocarbon receptor pathway may ameliorate dextran sodium sulfate-induced colitis in mice. Immunol Cell Biol 88, 685–689 (2010).
    1. Qiu J. et al. The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity 36, 92–104 (2012).
    1. Arsenescu R. et al. Role of the xenobiotic receptor in inflammatory bowel disease. Inflammatory Bowel Dis 17, 1149–1162 (2011).
    1. Fukumoto S. et al. Identification of a probiotic bacteria-derived activator of the aryl hydrocarbon receptor that inhibits colitis. Immunology and cell biology 92, 460–465 (2014).
    1. Smith K. J. et al. Identification of a high-affinity ligand that exhibits complete aryl hydrocarbon receptor antagonism. J Pharmacology Exp Ther 338, 318–327 (2011).
    1. DiNatale B. C., Schroeder J. C., Francey L. J., Kusnadi A. & Perdew G. H. Mechanistic insights into the events that lead to synergistic induction of interleukin 6 transcription upon activation of the aryl hydrocarbon receptor and inflammatory signaling. J Biol Chem 285, 24388–24397 (2010).
    1. Flaveny C. A., Murray I. A., Chiaro C. R. & Perdew G. H. Ligand selectivity and gene regulation by the human aryl hydrocarbon receptor in transgenic mice. Mol Pharmacol 75, 1412–1420 (2009).
    1. Denison M. S. & Nagy S. R. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Ann Rev Pharmacol Toxicol 43, 309–334 (2003).
    1. Denison M. S., Soshilov A. A., He G., DeGroot D. E. & Zhao B. Exactly the same but different: promiscuity and diversity in the molecular mechanisms of action of the aryl hydrocarbon (dioxin) receptor. Toxicol Sci 124, 1–22 (2011).
    1. Ramadoss P. & Perdew G. H. Use of 2-azido-3-[125I]iodo-7,8-dibromodibenzo-p-dioxin as a probe to determine the relative ligand affinity of human versus mouse aryl hydrocarbon receptor in cultured cells. Mol Pharmacol 66, 129–136 (2004).
    1. Ciolino H. P., Daschner P. J. & Yeh G. C. Dietary flavonols quercetin and kaempferol are ligands of the aryl hydrocarbon receptor that affect CYP1A1 transcription differentially. Biochem J 340, 715–722 (1999).
    1. Heath-Pagliuso S. et al. Activation of the Ah receptor by tryptophan and tryptophan metabolites. Biochem 37, 11508–11515 (1998).
    1. Lijinsky W. & Shubik P. Benzo(a)Pyrene and other polynuclear hydrocarbons in charcoal-broiled meat. Science 145, 53–55 (1964).
    1. Miele L. et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatol 49, 1877–1887 (2009).
    1. Turner J. R. Intestinal mucosal barrier function in health and disease. Nat Rev Immunol 9, 799–809 (2009).
    1. Xavier R. J. & Podolsky D. K. Unravelling the pathogenesis of inflammatory bowel disease. Nature 448, 427–434 (2007).
    1. Mezrich J. D. et al. An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J Immunol 185, 3190–3198 (2010).
    1. Gaitanis G. et al. AhR ligands, malassezin, and indolo[3,2-b]carbazole are selectively produced by Malassezia furfur strains isolated from seborrheic dermatitis. J Invest Dermatol 128, 1620–1625 (2008).
    1. Wille G. et al. Malassezin—A novel agonist of the arylhydrocarbon receptor from the yeast Malassezia furfur. Bioorganic & Med Chem 9, 955–960 (2001).
    1. Rera M., Clark R. I. & Walker D. W. Intestinal barrier dysfunction links metabolic and inflammatory markers of aging to death in Drosophila. Proc Natl Acad Sci USA 109, 21528–21533 (2012).
    1. Guo L., Karpac J., Tran S. L., & Jasper H. PGRP-SC2 promotes gut immune homeostasis to limit commensal dysbiosis and extend lifespan. Cell 156, 109–122 (2014).
    1. Zhang X., Goncalves R. & Mosser D. M. The isolation and characterization of murine macrophages. Current Protoc Immunol Chapter 14, Unit 14.1 (2008).
    1. Singh S. S., Hord N. G., & Perdew G. H. Characterization of the activated form of the aryl hydrocarbon receptor in the nucleus of HeLa cells in the absence of exogenous ligand. Arch Biochem Biophys 329, 47–55 (1996).
    1. Chiaro C. R., Patel R. D. & Perdew G. H. 12(R)-Hydroxy-5(Z),8(Z),10(E),14(Z)-eicosatetraenoic acid [12(R)-HETE], an arachidonic acid derivative, is an activator of the aryl hydrocarbon receptor. Mol Pharmacol 74, 1649–1656 (2008).
    1. Perkins A. et al. A structural switch between agonist and antagonist bound conformations for a ligand-optimized model of the human Aryl hydrocarbon receptor ligand binding domain. Biol (Basel) 3, 645–669 (2014).
    1. Rajendran P. et al. HDAC turnover, CtIP acetylation and dysregulated DNA damage signaling in colon cancer cells treated with sulforaphane and related dietary isothiocyanates. Epigenetics 8, 612–623 (2013).
    1. Neves M. A., Totrov M. & Abagyan R. Docking and scoring with ICM: the benchmarking results and strategies for improvement. J Comput Aided Mol Des 26, 675–686 (2012).
    1. Abagyan R. & Totrov M. Biased probability Monte Carlo conformational searches and electrostatic calculations for peptides and proteins. J Mol Biol 235, 983–1002 (1994).
    1. Abagyan R., Tortrov M. & Kuznetsov D. ICM-A new method for protein modeling and design:applications to docking and structure prediction from distorted native conformation. J Comput Chem 15, 488–506 (1994).
    1. Wesson L. & Eisenberg D. Atomic solvation parameters applied to molecular dynamics of proteins in solution. Protein Sci 1, 227–235 (1992).
    1. Shrake A. & Rupley J. A. Environment and exposure to solvent of protein atoms. Lysozyme and insulin. J Mol Biol 79, 351–371 (1973).

Source: PubMed

3
Prenumerera