Skip to main content
Erschienen in: Seminars in Immunopathology 4/2018

27.06.2018 | Review

AHR signaling in the development and function of intestinal immune cells and beyond

verfasst von: Luisa Cervantes-Barragan, Marco Colonna

Erschienen in: Seminars in Immunopathology | Ausgabe 4/2018

Einloggen, um Zugang zu erhalten

Abstract

The intestinal immune system is challenged daily with the task of recognizing and eliminating pathogens while simultaneously tolerating dietary and commensal antigens. All components must effectively coordinate to differentiate a continual barrage of environmental cues and mount appropriate responses dependent on the nature of the stimuli encountered. Playing a pivotal role, the aryl hydrocarbon receptor (AHR) is a chemical sensor that detects both dietary and microbial cues and is important for development, maintenance, and function of several types of intestinal immune cells, particularly innate lymphoid cells (ILCs) and T cells. In this review, we will highlight recent advances in our knowledge of the role of AHR signaling in ILCs, T cells, B cells, and dendritic cells.
Literatur
1.
Zurück zum Zitat Gu YZ, Hogenesch JB, Bradfield CA (2000) The PAS superfamily: sensors of environmental and developmental signals. Annu Rev Pharmacol Toxicol 40:519–561CrossRefPubMed Gu YZ, Hogenesch JB, Bradfield CA (2000) The PAS superfamily: sensors of environmental and developmental signals. Annu Rev Pharmacol Toxicol 40:519–561CrossRefPubMed
2.
Zurück zum Zitat McIntosh BE, Hogenesch JB, Bradfield CA (2010) Mammalian Per-Arnt-Sim proteins in environmental adaptation. Annu Rev Physiol 72:625–645CrossRefPubMed McIntosh BE, Hogenesch JB, Bradfield CA (2010) Mammalian Per-Arnt-Sim proteins in environmental adaptation. Annu Rev Physiol 72:625–645CrossRefPubMed
3.
Zurück zum Zitat Kewley RJ, Whitelaw ML, Chapman-Smith A (2004) The mammalian basic helix-loop-helix/PAS family of transcriptional regulators. Int J Biochem Cell Biol 36:189–204CrossRefPubMed Kewley RJ, Whitelaw ML, Chapman-Smith A (2004) The mammalian basic helix-loop-helix/PAS family of transcriptional regulators. Int J Biochem Cell Biol 36:189–204CrossRefPubMed
4.
Zurück zum Zitat Frericks M, Meissner M, Esser C (2007) Microarray analysis of the AHR system: tissue-specific flexibility in signal and target genes. Toxicol Appl Pharmacol 220:320–332CrossRefPubMed Frericks M, Meissner M, Esser C (2007) Microarray analysis of the AHR system: tissue-specific flexibility in signal and target genes. Toxicol Appl Pharmacol 220:320–332CrossRefPubMed
5.
Zurück zum Zitat Perdew GH (1988) Association of the Ah receptor with the 90-kDa heat shock protein. J Biol Chem 263:13802–13805PubMed Perdew GH (1988) Association of the Ah receptor with the 90-kDa heat shock protein. J Biol Chem 263:13802–13805PubMed
6.
Zurück zum Zitat Carver LA, Bradfield CA (1997) Ligand-dependent interaction of the aryl hydrocarbon receptor with a novel immunophilin homolog in vivo. J Biol Chem 272:11452–11456CrossRefPubMed Carver LA, Bradfield CA (1997) Ligand-dependent interaction of the aryl hydrocarbon receptor with a novel immunophilin homolog in vivo. J Biol Chem 272:11452–11456CrossRefPubMed
7.
Zurück zum Zitat Meyer BK, Pray-Grant MG, Vanden Heuvel JP, Perdew GH (1998) Hepatitis B virus X-associated protein 2 is a subunit of the unliganded aryl hydrocarbon receptor core complex and exhibits transcriptional enhancer activity. Mol Cell Biol 18:978–988CrossRefPubMedPubMedCentral Meyer BK, Pray-Grant MG, Vanden Heuvel JP, Perdew GH (1998) Hepatitis B virus X-associated protein 2 is a subunit of the unliganded aryl hydrocarbon receptor core complex and exhibits transcriptional enhancer activity. Mol Cell Biol 18:978–988CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Nair SC, Toran EJ, Rimerman RA, Hjermstad S, Smithgall TE, Smith DF (1996) A pathway of multi-chaperone interactions common to diverse regulatory proteins: estrogen receptor, Fes tyrosine kinase, heat shock transcription factor Hsf1, and the aryl hydrocarbon receptor. Cell Stress Chaperones 1:237–250CrossRefPubMedPubMedCentral Nair SC, Toran EJ, Rimerman RA, Hjermstad S, Smithgall TE, Smith DF (1996) A pathway of multi-chaperone interactions common to diverse regulatory proteins: estrogen receptor, Fes tyrosine kinase, heat shock transcription factor Hsf1, and the aryl hydrocarbon receptor. Cell Stress Chaperones 1:237–250CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat McGuire J, Whitelaw ML, Pongratz I, Gustafsson JA, Poellinger L (1994) A cellular factor stimulates ligand-dependent release of hsp90 from the basic helix-loop-helix dioxin receptor. Mol Cell Biol 14:2438–2446CrossRefPubMedPubMedCentral McGuire J, Whitelaw ML, Pongratz I, Gustafsson JA, Poellinger L (1994) A cellular factor stimulates ligand-dependent release of hsp90 from the basic helix-loop-helix dioxin receptor. Mol Cell Biol 14:2438–2446CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Fukunaga BN, Probst MR, Reisz-Porszasz S, Hankinson O (1995) Identification of functional domains of the aryl hydrocarbon receptor. J Biol Chem 270:29270–29278CrossRefPubMed Fukunaga BN, Probst MR, Reisz-Porszasz S, Hankinson O (1995) Identification of functional domains of the aryl hydrocarbon receptor. J Biol Chem 270:29270–29278CrossRefPubMed
11.
Zurück zum Zitat Schiering C, Vonk A, Das S, Stockinger B, Wincent E (2018) Cytochrome P4501-inhibiting chemicals amplify aryl hydrocarbon receptor activation and IL-22 production in T helper 17 cells. Biochem Pharmacol 151:47–58CrossRefPubMed Schiering C, Vonk A, Das S, Stockinger B, Wincent E (2018) Cytochrome P4501-inhibiting chemicals amplify aryl hydrocarbon receptor activation and IL-22 production in T helper 17 cells. Biochem Pharmacol 151:47–58CrossRefPubMed
12.
Zurück zum Zitat Schiering C, Wincent E, Metidji A, Iseppon A, Li Y, Potocnik AJ, Omenetti S, Henderson CJ, Wolf CR, Nebert DW, Stockinger B (2017) Feedback control of AHR signalling regulates intestinal immunity. Nature 542:242–245CrossRefPubMedPubMedCentral Schiering C, Wincent E, Metidji A, Iseppon A, Li Y, Potocnik AJ, Omenetti S, Henderson CJ, Wolf CR, Nebert DW, Stockinger B (2017) Feedback control of AHR signalling regulates intestinal immunity. Nature 542:242–245CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Stockinger B, Di Meglio P, Gialitakis M, Duarte JH (2014) The aryl hydrocarbon receptor: multitasking in the immune system. Annu Rev Immunol 32:403–432CrossRefPubMed Stockinger B, Di Meglio P, Gialitakis M, Duarte JH (2014) The aryl hydrocarbon receptor: multitasking in the immune system. Annu Rev Immunol 32:403–432CrossRefPubMed
14.
Zurück zum Zitat Mandal PK (2005) Dioxin: a review of its environmental effects and its aryl hydrocarbon receptor biology. J Comp Physiol B 175:221–230CrossRefPubMed Mandal PK (2005) Dioxin: a review of its environmental effects and its aryl hydrocarbon receptor biology. J Comp Physiol B 175:221–230CrossRefPubMed
15.
Zurück zum Zitat Nguyen LP, Bradfield CA (2008) The search for endogenous activators of the aryl hydrocarbon receptor. Chem Res Toxicol 21:102–116CrossRefPubMed Nguyen LP, Bradfield CA (2008) The search for endogenous activators of the aryl hydrocarbon receptor. Chem Res Toxicol 21:102–116CrossRefPubMed
16.
Zurück zum Zitat Denison MS, Nagy SR (2003) Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu Rev Pharmacol Toxicol 43:309–334CrossRefPubMed Denison MS, Nagy SR (2003) Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu Rev Pharmacol Toxicol 43:309–334CrossRefPubMed
17.
Zurück zum Zitat Bjeldanes LF, Kim JY, Grose KR, Bartholomew JC, Bradfield CA (1991) Aromatic hydrocarbon responsiveness-receptor agonists generated from indole-3-carbinol in vitro and in vivo: comparisons with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Proc Natl Acad Sci U S A 88:9543–9547CrossRefPubMedPubMedCentral Bjeldanes LF, Kim JY, Grose KR, Bartholomew JC, Bradfield CA (1991) Aromatic hydrocarbon responsiveness-receptor agonists generated from indole-3-carbinol in vitro and in vivo: comparisons with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Proc Natl Acad Sci U S A 88:9543–9547CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Zhang S, Qin C, Safe SH (2003) Flavonoids as aryl hydrocarbon receptor agonists/antagonists: effects of structure and cell context. Environ Health Perspect 111:1877–1882CrossRefPubMedPubMedCentral Zhang S, Qin C, Safe SH (2003) Flavonoids as aryl hydrocarbon receptor agonists/antagonists: effects of structure and cell context. Environ Health Perspect 111:1877–1882CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Takamura T, Harama D, Fukumoto S, Nakamura Y, Shimokawa N, Ishimaru K, Ikegami S, Makino S, Kitamura M, Nakao A (2011) Lactobacillus bulgaricus OLL1181 activates the aryl hydrocarbon receptor pathway and inhibits colitis. Immunol Cell Biol 89:817–822CrossRefPubMedPubMedCentral Takamura T, Harama D, Fukumoto S, Nakamura Y, Shimokawa N, Ishimaru K, Ikegami S, Makino S, Kitamura M, Nakao A (2011) Lactobacillus bulgaricus OLL1181 activates the aryl hydrocarbon receptor pathway and inhibits colitis. Immunol Cell Biol 89:817–822CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, Zecchi R, D'Angelo C, Massi-Benedetti C, Fallarino F, Carvalho A, Puccetti P, Romani L (2013) Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 39:372–385CrossRefPubMed Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, Zecchi R, D'Angelo C, Massi-Benedetti C, Fallarino F, Carvalho A, Puccetti P, Romani L (2013) Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 39:372–385CrossRefPubMed
21.
Zurück zum Zitat Cervantes-Barragan L, Chai JN, Tianero MD, Di Luccia B, Ahern PP, Merriman J, Cortez VS, Caparon MG, Donia MS, Gilfillan S, Cella M, Gordon JI, Hsieh CS, Colonna M (2017) Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science 357:806–810CrossRefPubMedPubMedCentral Cervantes-Barragan L, Chai JN, Tianero MD, Di Luccia B, Ahern PP, Merriman J, Cortez VS, Caparon MG, Donia MS, Gilfillan S, Cella M, Gordon JI, Hsieh CS, Colonna M (2017) Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science 357:806–810CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Lamas B, Richard ML, Leducq V, Pham HP, Michel ML, Da Costa G, Bridonneau C, Jegou S, Hoffmann TW, Natividad JM, Brot L, Taleb S, Couturier-Maillard A, Nion-Larmurier I, Merabtene F, Seksik P, Bourrier A, Cosnes J, Ryffel B, Beaugerie L, Launay JM, Langella P, Xavier RJ, Sokol H (2016) CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med 22:598–605CrossRefPubMedPubMedCentral Lamas B, Richard ML, Leducq V, Pham HP, Michel ML, Da Costa G, Bridonneau C, Jegou S, Hoffmann TW, Natividad JM, Brot L, Taleb S, Couturier-Maillard A, Nion-Larmurier I, Merabtene F, Seksik P, Bourrier A, Cosnes J, Ryffel B, Beaugerie L, Launay JM, Langella P, Xavier RJ, Sokol H (2016) CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med 22:598–605CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Sonowal R, Swimm A, Sahoo A, Luo L, Matsunaga Y, Wu Z, Bhingarde JA, Ejzak EA, Ranawade A, Qadota H, Powell DN, Capaldo CT, Flacker JM, Jones RM, Benian GM, Kalman D (2017) Indoles from commensal bacteria extend healthspan. Proc Natl Acad Sci U S A 114:E7506–E7E15CrossRefPubMedPubMedCentral Sonowal R, Swimm A, Sahoo A, Luo L, Matsunaga Y, Wu Z, Bhingarde JA, Ejzak EA, Ranawade A, Qadota H, Powell DN, Capaldo CT, Flacker JM, Jones RM, Benian GM, Kalman D (2017) Indoles from commensal bacteria extend healthspan. Proc Natl Acad Sci U S A 114:E7506–E7E15CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Moura-Alves P, Faé K, Houthuys E, Dorhoi A, Kreuchwig A, Furkert J, Barison N, Diehl A, Munder A, Constant P, Skrahina T, Guhlich-Bornhof U, Klemm M, Koehler AB, Bandermann S, Goosmann C, Mollenkopf HJ, Hurwitz R, Brinkmann V, Fillatreau S, Daffe M, Tümmler B, Kolbe M, Oschkinat H, Krause G, Kaufmann SH (2014) AhR sensing of bacterial pigments regulates antibacterial defence. Nature 512:387–392CrossRefPubMed Moura-Alves P, Faé K, Houthuys E, Dorhoi A, Kreuchwig A, Furkert J, Barison N, Diehl A, Munder A, Constant P, Skrahina T, Guhlich-Bornhof U, Klemm M, Koehler AB, Bandermann S, Goosmann C, Mollenkopf HJ, Hurwitz R, Brinkmann V, Fillatreau S, Daffe M, Tümmler B, Kolbe M, Oschkinat H, Krause G, Kaufmann SH (2014) AhR sensing of bacterial pigments regulates antibacterial defence. Nature 512:387–392CrossRefPubMed
25.
Zurück zum Zitat Gaitanis G, Magiatis P, Stathopoulou K, Bassukas ID, Alexopoulos EC, Velegraki A, Skaltsounis AL (2008) AhR ligands, malassezin, and indolo[3,2-b]carbazole are selectively produced by Malassezia furfur strains isolated from seborrheic dermatitis. J Invest Dermatol 128:1620–1625CrossRefPubMed Gaitanis G, Magiatis P, Stathopoulou K, Bassukas ID, Alexopoulos EC, Velegraki A, Skaltsounis AL (2008) AhR ligands, malassezin, and indolo[3,2-b]carbazole are selectively produced by Malassezia furfur strains isolated from seborrheic dermatitis. J Invest Dermatol 128:1620–1625CrossRefPubMed
26.
Zurück zum Zitat Magiatis P, Pappas P, Gaitanis G, Mexia N, Melliou E, Galanou M, Vlachos C, Stathopoulou K, Skaltsounis AL, Marselos M, Velegraki A, Denison MS, Bassukas ID (2013) Malassezia yeasts produce a collection of exceptionally potent activators of the Ah (dioxin) receptor detected in diseased human skin. J Invest Dermatol 133:2023–2030CrossRefPubMedPubMedCentral Magiatis P, Pappas P, Gaitanis G, Mexia N, Melliou E, Galanou M, Vlachos C, Stathopoulou K, Skaltsounis AL, Marselos M, Velegraki A, Denison MS, Bassukas ID (2013) Malassezia yeasts produce a collection of exceptionally potent activators of the Ah (dioxin) receptor detected in diseased human skin. J Invest Dermatol 133:2023–2030CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Mezrich JD, Fechner JH, Zhang X, Johnson BP, Burlingham WJ, Bradfield CA (2010) An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J Immunol 185:3190–3198CrossRefPubMedPubMedCentral Mezrich JD, Fechner JH, Zhang X, Johnson BP, Burlingham WJ, Bradfield CA (2010) An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J Immunol 185:3190–3198CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Seok SH, Ma ZX, Feltenberger JB, Chen H, Scarlett C, Lin Z, Satyshur KA, Cortopassi M, Jefcoate CR, Ge Y, Tang W, Bradfield CA, Xing Y (2018) Trace derivatives of kynurenine potently activate the aryl hydrocarbon receptor (AHR). J Biol Chem 293(6):1994–2005CrossRefPubMed Seok SH, Ma ZX, Feltenberger JB, Chen H, Scarlett C, Lin Z, Satyshur KA, Cortopassi M, Jefcoate CR, Ge Y, Tang W, Bradfield CA, Xing Y (2018) Trace derivatives of kynurenine potently activate the aryl hydrocarbon receptor (AHR). J Biol Chem 293(6):1994–2005CrossRefPubMed
29.
Zurück zum Zitat Rannug U, Rannug A, Sjöberg U, Li H, Westerholm R, Bergman J (1995) Structure elucidation of two tryptophan-derived, high affinity Ah receptor ligands. Chem Biol 2:841–845CrossRefPubMed Rannug U, Rannug A, Sjöberg U, Li H, Westerholm R, Bergman J (1995) Structure elucidation of two tryptophan-derived, high affinity Ah receptor ligands. Chem Biol 2:841–845CrossRefPubMed
30.
Zurück zum Zitat Smirnova A, Wincent E, Vikström Bergander L, Alsberg T, Bergman J, Rannug A, Rannug U (2016) Evidence for new light-independent pathways for generation of the endogenous aryl hydrocarbon receptor agonist FICZ. Chem Res Toxicol 29:75–86CrossRefPubMed Smirnova A, Wincent E, Vikström Bergander L, Alsberg T, Bergman J, Rannug A, Rannug U (2016) Evidence for new light-independent pathways for generation of the endogenous aryl hydrocarbon receptor agonist FICZ. Chem Res Toxicol 29:75–86CrossRefPubMed
32.
33.
34.
Zurück zum Zitat Ebihara T, Song C, Ryu SH, Plougastel-Douglas B, Yang L, Levanon D, Groner Y, Bern MD, Stappenbeck TS, Colonna M, Egawa T, Yokoyama WM (2015) Runx3 specifies lineage commitment of innate lymphoid cells. Nat Immunol 16:1124–1133CrossRefPubMedPubMedCentral Ebihara T, Song C, Ryu SH, Plougastel-Douglas B, Yang L, Levanon D, Groner Y, Bern MD, Stappenbeck TS, Colonna M, Egawa T, Yokoyama WM (2015) Runx3 specifies lineage commitment of innate lymphoid cells. Nat Immunol 16:1124–1133CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Li S, Heller JJ, Bostick JW, Lee A, Schjerven H, Kastner P, Chan S, Chen ZE, Zhou L (2016) Ikaros inhibits group 3 innate lymphoid cell development and function by suppressing the aryl hydrocarbon receptor pathway. Immunity 45:185–197CrossRefPubMedPubMedCentral Li S, Heller JJ, Bostick JW, Lee A, Schjerven H, Kastner P, Chan S, Chen ZE, Zhou L (2016) Ikaros inhibits group 3 innate lymphoid cell development and function by suppressing the aryl hydrocarbon receptor pathway. Immunity 45:185–197CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Lee JS, Cella M, McDonald KG, Garlanda C, Kennedy GD, Nukaya M, Mantovani A, Kopan R, Bradfield CA, Newberry RD, Colonna M (2011) AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch. Nat Immunol 13:144–151CrossRefPubMedPubMedCentral Lee JS, Cella M, McDonald KG, Garlanda C, Kennedy GD, Nukaya M, Mantovani A, Kopan R, Bradfield CA, Newberry RD, Colonna M (2011) AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch. Nat Immunol 13:144–151CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Kiss EA, Vonarbourg C, Kopfmann S, Hobeika E, Finke D, Esser C, Diefenbach A (2011) Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science 334:1561–1565CrossRefPubMed Kiss EA, Vonarbourg C, Kopfmann S, Hobeika E, Finke D, Esser C, Diefenbach A (2011) Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science 334:1561–1565CrossRefPubMed
38.
Zurück zum Zitat Hughes T, Briercheck EL, Freud AG, Trotta R, McClory S, Scoville SD, Keller K, Deng Y, Cole J, Harrison N, Mao C, Zhang J, Benson DM, Yu J, Caligiuri MA (2014) The transcription factor AHR prevents the differentiation of a stage 3 innate lymphoid cell subset to natural killer cells. Cell Rep 8:150–162CrossRefPubMedPubMedCentral Hughes T, Briercheck EL, Freud AG, Trotta R, McClory S, Scoville SD, Keller K, Deng Y, Cole J, Harrison N, Mao C, Zhang J, Benson DM, Yu J, Caligiuri MA (2014) The transcription factor AHR prevents the differentiation of a stage 3 innate lymphoid cell subset to natural killer cells. Cell Rep 8:150–162CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Shin JH, Zhang L, Murillo-Sauca O, Kim J, Kohrt HE, Bui JD, Sunwoo JB (2013) Modulation of natural killer cell antitumor activity by the aryl hydrocarbon receptor. Proc Natl Acad Sci U S A 110:12391–12396CrossRefPubMedPubMedCentral Shin JH, Zhang L, Murillo-Sauca O, Kim J, Kohrt HE, Bui JD, Sunwoo JB (2013) Modulation of natural killer cell antitumor activity by the aryl hydrocarbon receptor. Proc Natl Acad Sci U S A 110:12391–12396CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Wagage S, John B, Krock BL, Hall AO, Randall LM, Karp CL, Simon MC, Hunter CA (2014) The aryl hydrocarbon receptor promotes IL-10 production by NK cells. J Immunol 192:1661–1670CrossRefPubMedPubMedCentral Wagage S, John B, Krock BL, Hall AO, Randall LM, Karp CL, Simon MC, Hunter CA (2014) The aryl hydrocarbon receptor promotes IL-10 production by NK cells. J Immunol 192:1661–1670CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Qiu J, Heller JJ, Guo X, Chen ZM, Fish K, Fu YX, Zhou L (2012) The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity 36:92–104CrossRefPubMed Qiu J, Heller JJ, Guo X, Chen ZM, Fish K, Fu YX, Zhou L (2012) The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity 36:92–104CrossRefPubMed
42.
Zurück zum Zitat Qiu J, Guo X, Chen ZM, He L, Sonnenberg GF, Artis D, Fu YX, Zhou L (2013) Group 3 innate lymphoid cells inhibit T-cell-mediated intestinal inflammation through aryl hydrocarbon receptor signaling and regulation of microflora. Immunity 39:386–399CrossRefPubMed Qiu J, Guo X, Chen ZM, He L, Sonnenberg GF, Artis D, Fu YX, Zhou L (2013) Group 3 innate lymphoid cells inhibit T-cell-mediated intestinal inflammation through aryl hydrocarbon receptor signaling and regulation of microflora. Immunity 39:386–399CrossRefPubMed
43.
Zurück zum Zitat Wagage S, Harms Pritchard G, Dawson L, Buza EL, Sonnenberg GF, Hunter CA (2015) The group 3 innate lymphoid cell defect in aryl hydrocarbon receptor deficient mice is associated with T cell hyperactivation during intestinal infection. PLoS One 10:e0128335CrossRefPubMedPubMedCentral Wagage S, Harms Pritchard G, Dawson L, Buza EL, Sonnenberg GF, Hunter CA (2015) The group 3 innate lymphoid cell defect in aryl hydrocarbon receptor deficient mice is associated with T cell hyperactivation during intestinal infection. PLoS One 10:e0128335CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Funatake CJ, Marshall NB, Steppan LB, Mourich DV, Kerkvliet NI (2005) Cutting edge: activation of the aryl hydrocarbon receptor by 2,3,7,8-tetrachlorodibenzo-p-dioxin generates a population of CD4+ CD25+ cells with characteristics of regulatory T cells. J Immunol 175:4184–4188CrossRefPubMed Funatake CJ, Marshall NB, Steppan LB, Mourich DV, Kerkvliet NI (2005) Cutting edge: activation of the aryl hydrocarbon receptor by 2,3,7,8-tetrachlorodibenzo-p-dioxin generates a population of CD4+ CD25+ cells with characteristics of regulatory T cells. J Immunol 175:4184–4188CrossRefPubMed
45.
Zurück zum Zitat Apetoh L, Quintana FJ, Pot C, Joller N, Xiao S, Kumar D, Burns EJ, Sherr DH, Weiner HL, Kuchroo VK (2010) The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27. Nat Immunol 11:854–861CrossRefPubMedPubMedCentral Apetoh L, Quintana FJ, Pot C, Joller N, Xiao S, Kumar D, Burns EJ, Sherr DH, Weiner HL, Kuchroo VK (2010) The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27. Nat Immunol 11:854–861CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Mascanfroni ID, Takenaka MC, Yeste A, Patel B, Wu Y, Kenison JE, Siddiqui S, Basso AS, Otterbein LE, Pardoll DM, Pan F, Priel A, Clish CB, Robson SC, Quintana FJ (2015) Metabolic control of type 1 regulatory T cell differentiation by AHR and HIF1-α. Nat Med 21:638–646CrossRefPubMedPubMedCentral Mascanfroni ID, Takenaka MC, Yeste A, Patel B, Wu Y, Kenison JE, Siddiqui S, Basso AS, Otterbein LE, Pardoll DM, Pan F, Priel A, Clish CB, Robson SC, Quintana FJ (2015) Metabolic control of type 1 regulatory T cell differentiation by AHR and HIF1-α. Nat Med 21:638–646CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Ehrlich AK, Pennington JM, Tilton S, Wang X, Marshall NB, Rohlman D, Funatake C, Punj S, O'Donnell E, Yu Z, Kolluri SK, Kerkvliet NI (2017) AhR activation increases IL-2 production by alloreactive CD4+ T cells initiating the differentiation of mucosal-homing Tim3+ Lag3+ Tr1 cells. Eur J Immunol 47:1989–2001CrossRefPubMed Ehrlich AK, Pennington JM, Tilton S, Wang X, Marshall NB, Rohlman D, Funatake C, Punj S, O'Donnell E, Yu Z, Kolluri SK, Kerkvliet NI (2017) AhR activation increases IL-2 production by alloreactive CD4+ T cells initiating the differentiation of mucosal-homing Tim3+ Lag3+ Tr1 cells. Eur J Immunol 47:1989–2001CrossRefPubMed
48.
Zurück zum Zitat Veldhoen M, Hirota K, Westendorf AM, Buer J, Dumoutier L, Renauld JC, Stockinger B (2008) The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature 453:106–109CrossRefPubMed Veldhoen M, Hirota K, Westendorf AM, Buer J, Dumoutier L, Renauld JC, Stockinger B (2008) The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature 453:106–109CrossRefPubMed
49.
Zurück zum Zitat Quintana FJ, Basso AS, Iglesias AH, Korn T, Farez MF, Bettelli E, Caccamo M, Oukka M, Weiner HL (2008) Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor. Nature 453:65–71CrossRefPubMed Quintana FJ, Basso AS, Iglesias AH, Korn T, Farez MF, Bettelli E, Caccamo M, Oukka M, Weiner HL (2008) Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor. Nature 453:65–71CrossRefPubMed
50.
Zurück zum Zitat Veldhoen M, Hirota K, Christensen J, O'Garra A, Stockinger B (2009) Natural agonists for aryl hydrocarbon receptor in culture medium are essential for optimal differentiation of Th17 T cells. J Exp Med 206:43–49CrossRefPubMedPubMedCentral Veldhoen M, Hirota K, Christensen J, O'Garra A, Stockinger B (2009) Natural agonists for aryl hydrocarbon receptor in culture medium are essential for optimal differentiation of Th17 T cells. J Exp Med 206:43–49CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Kimura A, Naka T, Nakahama T, Chinen I, Masuda K, Nohara K, Fujii-Kuriyama Y, Kishimoto T (2009) Aryl hydrocarbon receptor in combination with Stat1 regulates LPS-induced inflammatory responses. J Exp Med 206:2027–2035CrossRefPubMedPubMedCentral Kimura A, Naka T, Nakahama T, Chinen I, Masuda K, Nohara K, Fujii-Kuriyama Y, Kishimoto T (2009) Aryl hydrocarbon receptor in combination with Stat1 regulates LPS-induced inflammatory responses. J Exp Med 206:2027–2035CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Rutz S, Noubade R, Eidenschenk C, Ota N, Zeng W, Zheng Y, Hackney J, Ding J, Singh H, Ouyang W (2011) Transcription factor c-Maf mediates the TGF-β-dependent suppression of IL-22 production in T(H)17 cells. Nat Immunol 12:1238–1245CrossRefPubMed Rutz S, Noubade R, Eidenschenk C, Ota N, Zeng W, Zheng Y, Hackney J, Ding J, Singh H, Ouyang W (2011) Transcription factor c-Maf mediates the TGF-β-dependent suppression of IL-22 production in T(H)17 cells. Nat Immunol 12:1238–1245CrossRefPubMed
53.
Zurück zum Zitat Ehrlich AK, Pennington JM, Bisson WH, Kolluri SK, Kerkvliet NI (2018) TCDD, FICZ, and other high affinity AhR ligands dose-dependently determine the fate of CD4+ T cell differentiation. Toxicol Sci 161(2):310–320CrossRefPubMed Ehrlich AK, Pennington JM, Bisson WH, Kolluri SK, Kerkvliet NI (2018) TCDD, FICZ, and other high affinity AhR ligands dose-dependently determine the fate of CD4+ T cell differentiation. Toxicol Sci 161(2):310–320CrossRefPubMed
54.
Zurück zum Zitat Li Y, Innocentin S, Withers DR, Roberts NA, Gallagher AR, Grigorieva EF, Wilhelm C, Veldhoen M (2011) Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation. Cell 147:629–640CrossRefPubMed Li Y, Innocentin S, Withers DR, Roberts NA, Gallagher AR, Grigorieva EF, Wilhelm C, Veldhoen M (2011) Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation. Cell 147:629–640CrossRefPubMed
55.
Zurück zum Zitat Konkel JE, Maruyama T, Carpenter AC, Xiong Y, Zamarron BF, Hall BE, Kulkarni AB, Zhang P, Bosselut R, Chen W (2011) Control of the development of CD8αα+ intestinal intraepithelial lymphocytes by TGF-β. Nat Immunol 12:312–319CrossRefPubMedPubMedCentral Konkel JE, Maruyama T, Carpenter AC, Xiong Y, Zamarron BF, Hall BE, Kulkarni AB, Zhang P, Bosselut R, Chen W (2011) Control of the development of CD8αα+ intestinal intraepithelial lymphocytes by TGF-β. Nat Immunol 12:312–319CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Mucida D, Husain MM, Muroi S, van Wijk F, Shinnakasu R, Naoe Y, Reis BS, Huang Y, Lambolez F, Docherty M, Attinger A, Shui JW, Kim G, Lena CJ, Sakaguchi S, Miyamoto C, Wang P, Atarashi K, Park Y, Nakayama T, Honda K, Ellmeier W, Kronenberg M, Taniuchi I, Cheroutre H (2013) Transcriptional reprogramming of mature CD4+ helper T cells generates distinct MHC class II-restricted cytotoxic T lymphocytes. Nat Immunol 14:281–289CrossRefPubMedPubMedCentral Mucida D, Husain MM, Muroi S, van Wijk F, Shinnakasu R, Naoe Y, Reis BS, Huang Y, Lambolez F, Docherty M, Attinger A, Shui JW, Kim G, Lena CJ, Sakaguchi S, Miyamoto C, Wang P, Atarashi K, Park Y, Nakayama T, Honda K, Ellmeier W, Kronenberg M, Taniuchi I, Cheroutre H (2013) Transcriptional reprogramming of mature CD4+ helper T cells generates distinct MHC class II-restricted cytotoxic T lymphocytes. Nat Immunol 14:281–289CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Reis BS, Rogoz A, Costa-Pinto FA, Taniuchi I, Mucida D (2013) Mutual expression of the transcription factors Runx3 and ThPOK regulates intestinal CD4+ T cell immunity. Nat Immunol 14:271–280CrossRefPubMedPubMedCentral Reis BS, Rogoz A, Costa-Pinto FA, Taniuchi I, Mucida D (2013) Mutual expression of the transcription factors Runx3 and ThPOK regulates intestinal CD4+ T cell immunity. Nat Immunol 14:271–280CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Hauben E, Gregori S, Draghici E, Migliavacca B, Olivieri S, Woisetschläger M, Roncarolo MG (2008) Activation of the aryl hydrocarbon receptor promotes allograft-specific tolerance through direct and dendritic cell-mediated effects on regulatory T cells. Blood 112:1214–1222CrossRefPubMed Hauben E, Gregori S, Draghici E, Migliavacca B, Olivieri S, Woisetschläger M, Roncarolo MG (2008) Activation of the aryl hydrocarbon receptor promotes allograft-specific tolerance through direct and dendritic cell-mediated effects on regulatory T cells. Blood 112:1214–1222CrossRefPubMed
59.
Zurück zum Zitat Lawrence BP, Denison MS, Novak H, Vorderstrasse BA, Harrer N, Neruda W, Reichel C, Woisetschläger M (2008) Activation of the aryl hydrocarbon receptor is essential for mediating the anti-inflammatory effects of a novel low-molecular-weight compound. Blood 112:1158–1165CrossRefPubMedPubMedCentral Lawrence BP, Denison MS, Novak H, Vorderstrasse BA, Harrer N, Neruda W, Reichel C, Woisetschläger M (2008) Activation of the aryl hydrocarbon receptor is essential for mediating the anti-inflammatory effects of a novel low-molecular-weight compound. Blood 112:1158–1165CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Jurado-Manzano BB, Zavala-Reyes D, Turrubiartes-Martínez EA, Portales-Pérez DP, González-Amaro R, Layseca-Espinosa E (2017) FICZ generates human tDCs that induce CD4+ CD25high Foxp3+ Treg-like cell differentiation. Immunol Lett 190:84–92CrossRefPubMed Jurado-Manzano BB, Zavala-Reyes D, Turrubiartes-Martínez EA, Portales-Pérez DP, González-Amaro R, Layseca-Espinosa E (2017) FICZ generates human tDCs that induce CD4+ CD25high Foxp3+ Treg-like cell differentiation. Immunol Lett 190:84–92CrossRefPubMed
61.
Zurück zum Zitat Kado S, Chang WLW, Chi AN, Wolny M, Shepherd DM, Vogel CFA (2017) Aryl hydrocarbon receptor signaling modifies Toll-like receptor-regulated responses in human dendritic cells. Arch Toxicol 91:2209–2221CrossRefPubMed Kado S, Chang WLW, Chi AN, Wolny M, Shepherd DM, Vogel CFA (2017) Aryl hydrocarbon receptor signaling modifies Toll-like receptor-regulated responses in human dendritic cells. Arch Toxicol 91:2209–2221CrossRefPubMed
62.
Zurück zum Zitat Platzer B, Richter S, Kneidinger D, Waltenberger D, Woisetschläger M, Strobl H (2009) Aryl hydrocarbon receptor activation inhibits in vitro differentiation of human monocytes and Langerhans dendritic cells. J Immunol 183:66–74CrossRefPubMed Platzer B, Richter S, Kneidinger D, Waltenberger D, Woisetschläger M, Strobl H (2009) Aryl hydrocarbon receptor activation inhibits in vitro differentiation of human monocytes and Langerhans dendritic cells. J Immunol 183:66–74CrossRefPubMed
63.
Zurück zum Zitat Boitano AE, Wang J, Romeo R, Bouchez LC, Parker AE, Sutton SE, Walker JR, Flaveny CA, Perdew GH, Denison MS, Schultz PG, Cooke MP (2010) Aryl hydrocarbon receptor antagonists promote the expansion of human hematopoietic stem cells. Science 329:1345–1348CrossRefPubMedPubMedCentral Boitano AE, Wang J, Romeo R, Bouchez LC, Parker AE, Sutton SE, Walker JR, Flaveny CA, Perdew GH, Denison MS, Schultz PG, Cooke MP (2010) Aryl hydrocarbon receptor antagonists promote the expansion of human hematopoietic stem cells. Science 329:1345–1348CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Thordardottir S, Hangalapura BN, Hutten T, Cossu M, Spanholtz J, Schaap N, Radstake TR, van der Voort R, Dolstra H (2014) The aryl hydrocarbon receptor antagonist StemRegenin 1 promotes human plasmacytoid and myeloid dendritic cell development from CD34+ hematopoietic progenitor cells. Stem Cells Dev 23:955–967CrossRefPubMed Thordardottir S, Hangalapura BN, Hutten T, Cossu M, Spanholtz J, Schaap N, Radstake TR, van der Voort R, Dolstra H (2014) The aryl hydrocarbon receptor antagonist StemRegenin 1 promotes human plasmacytoid and myeloid dendritic cell development from CD34+ hematopoietic progenitor cells. Stem Cells Dev 23:955–967CrossRefPubMed
65.
Zurück zum Zitat Goudot C, Coillard A, Villani AC, Gueguen P, Cros A, Sarkizova S, Tang-Huau TL, Bohec M, Baulande S, Hacohen N, Amigorena S, Segura E (2017) Aryl hydrocarbon receptor controls monocyte differentiation into dendritic cells versus macrophages. Immunity 47:582–96.e6 Goudot C, Coillard A, Villani AC, Gueguen P, Cros A, Sarkizova S, Tang-Huau TL, Bohec M, Baulande S, Hacohen N, Amigorena S, Segura E (2017) Aryl hydrocarbon receptor controls monocyte differentiation into dendritic cells versus macrophages. Immunity 47:582–96.e6
66.
Zurück zum Zitat Jux B, Kadow S, Esser C (2009) Langerhans cell maturation and contact hypersensitivity are impaired in aryl hydrocarbon receptor-null mice. J Immunol 182:6709–6717CrossRefPubMed Jux B, Kadow S, Esser C (2009) Langerhans cell maturation and contact hypersensitivity are impaired in aryl hydrocarbon receptor-null mice. J Immunol 182:6709–6717CrossRefPubMed
67.
Zurück zum Zitat Liu H, Ramachandran I, Gabrilovich DI (2014) Regulation of plasmacytoid dendritic cell development in mice by aryl hydrocarbon receptor. Immunol Cell Biol 92:200–203CrossRefPubMed Liu H, Ramachandran I, Gabrilovich DI (2014) Regulation of plasmacytoid dendritic cell development in mice by aryl hydrocarbon receptor. Immunol Cell Biol 92:200–203CrossRefPubMed
68.
Zurück zum Zitat Quintana FJ, Murugaiyan G, Farez MF, Mitsdoerffer M, Tukpah AM, Burns EJ, Weiner HL (2010) An endogenous aryl hydrocarbon receptor ligand acts on dendritic cells and T cells to suppress experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A 107:20768–20773CrossRefPubMedPubMedCentral Quintana FJ, Murugaiyan G, Farez MF, Mitsdoerffer M, Tukpah AM, Burns EJ, Weiner HL (2010) An endogenous aryl hydrocarbon receptor ligand acts on dendritic cells and T cells to suppress experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A 107:20768–20773CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Vogel CF, Goth SR, Dong B, Pessah IN, Matsumura F (2008) Aryl hydrocarbon receptor signaling mediates expression of indoleamine 2,3-dioxygenase. Biochem Biophys Res Commun 375:331–335CrossRefPubMedPubMedCentral Vogel CF, Goth SR, Dong B, Pessah IN, Matsumura F (2008) Aryl hydrocarbon receptor signaling mediates expression of indoleamine 2,3-dioxygenase. Biochem Biophys Res Commun 375:331–335CrossRefPubMedPubMedCentral
70.
Zurück zum Zitat Nguyen NT, Kimura A, Nakahama T, Chinen I, Masuda K, Nohara K, Fujii-Kuriyama Y, Kishimoto T (2010) Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism. Proc Natl Acad Sci U S A 107:19961–19966CrossRefPubMedPubMedCentral Nguyen NT, Kimura A, Nakahama T, Chinen I, Masuda K, Nohara K, Fujii-Kuriyama Y, Kishimoto T (2010) Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism. Proc Natl Acad Sci U S A 107:19961–19966CrossRefPubMedPubMedCentral
71.
Zurück zum Zitat Bessede A, Gargaro M, Pallotta MT, Matino D, Servillo G, Brunacci C, Bicciato S, Mazza EM, Macchiarulo A, Vacca C, Iannitti R, Tissi L, Volpi C, Belladonna ML, Orabona C, Bianchi R, Lanz TV, Platten M, Della Fazia MA, Piobbico D, Zelante T, Funakoshi H, Nakamura T, Gilot D, Denison MS, Guillemin GJ, DuHadaway JB, Prendergast GC, Metz R, Geffard M, Boon L, Pirro M, Iorio A, Veyret B, Romani L, Grohmann U, Fallarino F, Puccetti P (2014) Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature 511:184–190CrossRefPubMedPubMedCentral Bessede A, Gargaro M, Pallotta MT, Matino D, Servillo G, Brunacci C, Bicciato S, Mazza EM, Macchiarulo A, Vacca C, Iannitti R, Tissi L, Volpi C, Belladonna ML, Orabona C, Bianchi R, Lanz TV, Platten M, Della Fazia MA, Piobbico D, Zelante T, Funakoshi H, Nakamura T, Gilot D, Denison MS, Guillemin GJ, DuHadaway JB, Prendergast GC, Metz R, Geffard M, Boon L, Pirro M, Iorio A, Veyret B, Romani L, Grohmann U, Fallarino F, Puccetti P (2014) Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature 511:184–190CrossRefPubMedPubMedCentral
72.
Zurück zum Zitat Sulentic CE, Kaminski NE (2011) The long winding road toward understanding the molecular mechanisms for B-cell suppression by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Sci 120(Suppl 1):S171–S191CrossRefPubMed Sulentic CE, Kaminski NE (2011) The long winding road toward understanding the molecular mechanisms for B-cell suppression by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Sci 120(Suppl 1):S171–S191CrossRefPubMed
73.
Zurück zum Zitat Allan LL, Sherr DH (2005) Constitutive activation and environmental chemical induction of the aryl hydrocarbon receptor/transcription factor in activated human B lymphocytes. Mol Pharmacol 67:1740–1750CrossRefPubMed Allan LL, Sherr DH (2005) Constitutive activation and environmental chemical induction of the aryl hydrocarbon receptor/transcription factor in activated human B lymphocytes. Mol Pharmacol 67:1740–1750CrossRefPubMed
74.
Zurück zum Zitat Tanaka G, Kanaji S, Hirano A, Arima K, Shinagawa A, Goda C, Yasunaga S, Ikizawa K, Yanagihara Y, Kubo M, Kuriyama-Fujii Y, Sugita Y, Inokuchi A, Izuhara K (2005) Induction and activation of the aryl hydrocarbon receptor by IL-4 in B cells. Int Immunol 17:797–805CrossRefPubMed Tanaka G, Kanaji S, Hirano A, Arima K, Shinagawa A, Goda C, Yasunaga S, Ikizawa K, Yanagihara Y, Kubo M, Kuriyama-Fujii Y, Sugita Y, Inokuchi A, Izuhara K (2005) Induction and activation of the aryl hydrocarbon receptor by IL-4 in B cells. Int Immunol 17:797–805CrossRefPubMed
75.
Zurück zum Zitat Villa M, Gialitakis M, Tolaini M, Ahlfors H, Henderson CJ, Wolf CR, Brink R, Stockinger B (2017) Aryl hydrocarbon receptor is required for optimal B-cell proliferation. EMBO J 36:116–128CrossRefPubMed Villa M, Gialitakis M, Tolaini M, Ahlfors H, Henderson CJ, Wolf CR, Brink R, Stockinger B (2017) Aryl hydrocarbon receptor is required for optimal B-cell proliferation. EMBO J 36:116–128CrossRefPubMed
76.
Zurück zum Zitat Inoue H, Mishima K, Yamamoto-Yoshida S, Ushikoshi-Nakayama R, Nakagawa Y, Yamamoto K, Ryo K, Ide F, Saito I (2012) Aryl hydrocarbon receptor-mediated induction of EBV reactivation as a risk factor for Sjögren’s syndrome. J Immunol 188:4654–4662CrossRefPubMed Inoue H, Mishima K, Yamamoto-Yoshida S, Ushikoshi-Nakayama R, Nakagawa Y, Yamamoto K, Ryo K, Ide F, Saito I (2012) Aryl hydrocarbon receptor-mediated induction of EBV reactivation as a risk factor for Sjögren’s syndrome. J Immunol 188:4654–4662CrossRefPubMed
77.
Zurück zum Zitat Yoshida T, Katsuya K, Oka T, Koizumi S, Wakita D, Kitamura H, Nishimura T (2012) Effects of AhR ligands on the production of immunoglobulins in purified mouse B cells. Biomed Res 33:67–74CrossRefPubMed Yoshida T, Katsuya K, Oka T, Koizumi S, Wakita D, Kitamura H, Nishimura T (2012) Effects of AhR ligands on the production of immunoglobulins in purified mouse B cells. Biomed Res 33:67–74CrossRefPubMed
78.
Zurück zum Zitat Phadnis-Moghe AS, Li J, Crawford RB, Kaminski NE (2016) SHP-1 is directly activated by the aryl hydrocarbon receptor and regulates BCL-6 in the presence of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Toxicol Appl Pharmacol 310:41–50CrossRefPubMedPubMedCentral Phadnis-Moghe AS, Li J, Crawford RB, Kaminski NE (2016) SHP-1 is directly activated by the aryl hydrocarbon receptor and regulates BCL-6 in the presence of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Toxicol Appl Pharmacol 310:41–50CrossRefPubMedPubMedCentral
79.
Zurück zum Zitat Kovalova N, Manzan M, Crawford R, Kaminski N (2016) Role of aryl hydrocarbon receptor polymorphisms on TCDD-mediated CYP1B1 induction and IgM suppression by human B cells. Toxicol Appl Pharmacol 309:15–23CrossRefPubMedPubMedCentral Kovalova N, Manzan M, Crawford R, Kaminski N (2016) Role of aryl hydrocarbon receptor polymorphisms on TCDD-mediated CYP1B1 induction and IgM suppression by human B cells. Toxicol Appl Pharmacol 309:15–23CrossRefPubMedPubMedCentral
Metadaten
Titel
AHR signaling in the development and function of intestinal immune cells and beyond
verfasst von
Luisa Cervantes-Barragan
Marco Colonna
Publikationsdatum
27.06.2018
Verlag
Springer Berlin Heidelberg
Erschienen in
Seminars in Immunopathology / Ausgabe 4/2018
Print ISSN: 1863-2297
Elektronische ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-018-0694-9

Weitere Artikel der Ausgabe 4/2018

Seminars in Immunopathology 4/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.