The C-Type Lectin Receptor SIGNR3 Binds to Fungi Present in Commensal Microbiota and Influences Immune Regulation in Experimental Colitis

Magdalena Eriksson, Timo Johannssen, Dorthe von Smolinski, Achim D Gruber, Peter H Seeberger, Bernd Lepenies, Magdalena Eriksson, Timo Johannssen, Dorthe von Smolinski, Achim D Gruber, Peter H Seeberger, Bernd Lepenies

Abstract

Inflammatory bowel disease is a condition of acute and chronic inflammation of the gut. An important factor contributing to pathogenesis is a dysregulated mucosal immunity against commensal bacteria and fungi. Host pattern-recognition receptors (PRRs) sense commensals in the gut and are involved in maintaining the balance between controlled responses to pathogens and overwhelming innate immune activation. C-type lectin receptors (CLRs) are PRRs recognizing glycan structures on pathogens and self-antigens. Here we examined the role of the murine CLR specific intracellular adhesion molecule-3 grabbing non-integrin homolog-related 3 (SIGNR3) in the recognition of commensals and its involvement in intestinal immunity. SIGNR3 is the closest murine homolog of the human dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) receptor recognizing similar carbohydrate ligands such as terminal fucose or high-mannose glycans. We discovered that SIGNR3 recognizes fungi present in the commensal microbiota. To analyze whether this interaction impacts the intestinal immunity against microbiota, the dextran sulfate sodium-induced colitis model was employed. SIGNR3(-/-) mice exhibited an increased weight loss associated with more severe colitis symptoms compared to wild-type control mice. The increased inflammation in SIGNR3(-/-) mice was accompanied by a higher level of TNF-α in colon. Our findings demonstrate for the first time that SIGNR3 recognizes intestinal fungi and has an immune regulatory role in colitis.

Keywords: C-type lectin receptor; SIGNR3; carbohydrate recognition; colitis; fungi; host innate immunity; microbiota.

Figures

Figure 1
Figure 1
Binding of SIGNR3 to fungi present in commensal microbiota. (A) ELISA analysis of binding of CLR-hFc fusion proteins to heat inactivated intestinal microbiota demonstrated significant binding of SIGNR3-hFc. Binding of SIGNR3-hFc to zymosan, and MGL1-hFc to colon microbiota were used as positive controls. If indicated, incubation and washing was performed in 25 mM EDTA buffer to analyze Ca2+-dependency of the interaction. Results are representative of three independent experiments. (B) Representative dot plots of SytoRed stained gut microbes (APC+) incubated with hFc, SIGNR3-hFc, or DCAR-hFc. Detection with a PE-labeled goat anti-hFc antibody demonstrates binding of SIGNR3-hFc to gut microbes. (C) Corresponding statistical analysis of flow cytometry data representative from three independent experiments. (D) The interaction of SIGNR3-hFc and microbes was inhibited by preincubation with S. cerevisiae-derived mannan in the concentrations of 0.01–10 mg/ml. Data are representative of two independent experiments. (E) Staining with a FITC-labeled anti-C. albicans antibody reported to cross-react with other fungi revealed about 7% of the microbes were fungi. Staining of cells with fusion proteins demonstrated that SIGNR3-hFc recognizes>60% of the commensal fungi in the colon. (F) Statistical analysis of commensal fungi detected by SIGNR3- and DCAR-hFc as determined by flow cytometry. Shown are data and gating representative for three independent experiments. (A–F) Data are presented as mean + SEM. The p-values were determined using unpaired Student’s t-test. **p < 0.01, ***p < 0.001, ****p < 0.0001.
Figure 2
Figure 2
Wild-type and SIGNR3−/− mice were fed 3% DSS supplemented to the drinking water for seven consecutive days (n = 31 for SIGNR3−/− mice, n = 30 for wild-type mice). (A) Weight was recorded daily. On day 7, disease activity index (B), stool score (C), and colon length (D) were measured. SIGNR3−/− mice displayed an increased susceptibility to DSS colitis as indicated by a more severe weight loss and exacerbated colitis symptoms. A summary of four independent experiments is shown. Data are expressed as mean + SEM. The p-values were determined using a mixed linear model (A) and unpaired Student’s t-test (B–D). Significance is indicated by asterisks (*), *p < 0.05, **p < 0.01, ***p < 0.001.
Figure 3
Figure 3
After 7 days of DSS treatment, mice were sacrificed and paraffin sections of the colon of each mouse were prepared and stained with hematoxylin and eosin (H&E). A histopathological evaluation of each section was performed in a blinded manner. (A) H&E stained sections from colon of C57BL/6 wild-type, and SIGNR3−/− mice at day 7, and untreated control mice. (B) Each colon was divided into three segments of identical length (oral, middle, rectal) and each segment of the colon was analyzed separately. The degree of infiltration of inflammatory cells and mucosal erosion/ulceration was graded from none (score 0) to mild (score 1), moderate (score 3), or severe (score 4) (n = 5). Data are expressed as mean + SEM. The p-values were determined using unpaired Student’s t-test. Arrows show examples of mucosal ulceration. Significance is indicated by asterisks (*), *p < 0.05.
Figure 4
Figure 4
Cytokines in colon homogenates from wild-type and SIGNR3−/− mice fed with 3% DSS were analyzed by cytometric bead array. The levels of TNF-α (A), IL-6 (B), and IL-1β (C) were measured (n = 8 for DSS-treated SIGNR3−/− and wild-type mice, n = 5 for untreated wild-type mice). Data are expressed as mean + SEM. The p-values were determined using unpaired Student’s t-test. Significance is indicated by asterisks (*), *p < 0.05.

References

    1. Bouma G, Strober W. The immunological and genetic basis of inflammatory bowel disease. Nat Rev Immunol (2003) 3:521–3310.1038/nri1132
    1. Wirtz S, Neufert C, Weigmann B, Neurath MF. Chemically induced mouse models of intestinal inflammation. Nat Protoc (2007) 2:541–610.1038/nprot.2007.41
    1. Underhill D, Braun J. Current understanding of fungal microflora in inflammatory bowel disease pathogenesis. Inflamm Bowel Dis (2008) 14:1147–5310.1002/ibd.20402
    1. Maloy KJ, Powrie F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature (2011) 474:298–30610.1038/nature10208
    1. Fraser JD, Irving BA, Crabtree GR, Weiss A. Regulation of interleukin-2 gene enhancer activity by the T cell accessory molecule CD28. Science (1991) 251:313–610.1126/science.1846244
    1. Denning TL, Kim G, Kronenberg M. Cutting edge: CD4+CD25+ regulatory T cells impaired for intestinal homing can prevent colitis. J Immunol (2005) 174:7487–91
    1. Kim G, Turovskaya O, Levin M, Byrne FR, Whoriskey JS, McCabe JG, et al. Spontaneous colitis occurrence in transgenic mice with altered B7-mediated costimulation. J Immunol (2008) 181:5278–88
    1. Shui JW, Larange A, Kim G, Vela JL, Zahner S, Cheroutre H, et al. HVEM signalling at mucosal barriers provides host defence against pathogenic bacteria. Nature (2012) 488:222–510.1038/nature11242
    1. Steinberg MW, Turovskaya O, Shaikh RB, Kim G, McCole DF, Pfeffer K, et al. A crucial role for HVEM and BTLA in preventing intestinal inflammation. J Exp Med (2008) 205:1463–7610.1084/jem.20071160
    1. Chen GY, Nunez G. Gut Immunity: a NOD to the commensals. Curr Biol (2009) 19:R171–410.1016/j.cub.2008.12.027
    1. Rubino SJ, Selvanantham T, Girardin SE, Philpott DJ. Nod-like receptors in the control of intestinal inflammation. Curr Opin Immunol (2012) 24:398–40410.1016/j.coi.2012.04.010
    1. Stagg AJ, Hart AL, Knight SC, Kamm MA. The dendritic cell: its role in intestinal inflammation and relationship with gut bacteria. Gut (2003) 52:1522–910.1136/gut.52.10.1522
    1. Hart AL, Lammers K, Brigidi P, Vitali B, Rizzello F, Gionchetti P, et al. Modulation of human dendritic cell phenotype and function by probiotic bacteria. Gut (2004) 53:1602–910.1136/gut.2003.037325
    1. Ng SC, Benjamin JL, McCarthy NE, Hedin CR, Koutsoumpas A, Plamondon S, et al. Relationship between human intestinal dendritic cells, gut microbiota, and disease activity in Crohn’s disease. Inflamm Bowel Dis (2011) 17:2027–3710.1002/ibd.21590
    1. McGreal EP, Miller JL, Gordon S. Ligand recognition by antigen-presenting cell C-type lectin receptors. Curr Opin Immunol (2005) 17:18–2410.1016/j.coi.2004.12.001
    1. Kanazawa N. Dendritic cell immunoreceptors: C-type lectin receptors for pattern-recognition and signaling on antigen-presenting cells. J Dermatol Sci (2007) 45:77–8610.1016/j.jdermsci.2006.09.001
    1. Abreu MT. Toll-like receptor signalling in the intestinal epithelium: how bacterial recognition shapes intestinal function. Nat Rev Immunol (2010) 10:131–4410.1038/nri2707
    1. Hart AL, Al-Hassi HO, Rigby RJ, Bell SJ, Emmanuel AV, Knight SC, et al. Characteristics of intestinal dendritic cells in inflammatory bowel diseases. Gastroenterology (2005) 129:50–6510.1053/j.gastro.2005.05.013
    1. Cario E, Podolsky DK. Differential alteration in intestinal epithelial cell expression of Toll-like receptor 3 (TLR3) and TLR4 in inflammatory bowel disease. Infect Immun (2000) 68:7010–710.1128/IAI.68.12.7010-7017.2000
    1. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by Toll-like receptors is required for intestinal homeostasis. Cell (2004) 118:229–4110.1016/j.cell.2004.07.002
    1. Fukata M, Michelsen KS, Eri R, Thomas LS, Hu B, Lukasek K, et al. Toll-like receptor-4 is required for intestinal response to epithelial injury and limiting bacterial translocation in a murine model of acute colitis. Am J Physiol Gastrointest Liver Physiol (2005) 288:G1055–6510.1152/ajpgi.00328.2004
    1. Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos R, et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature (2001) 411:603–610.1038/35079114
    1. Watanabe T, Kitani A, Murray PJ, Wakatsuki Y, Fuss IJ, Strober W. Nucleotide binding oligomerization domain 2 deficiency leads to dysregulated TLR2 signaling and induction of antigen-specific colitis. Immunity (2006) 25:473–8510.1016/j.immuni.2006.06.018
    1. Sancho D, Reis E, Sousa C. Signaling by myeloid C-type lectin receptors in immunity and homeostasis. Annu Rev Immunol (2012) 30:491–52910.1146/annurev-immunol-031210-101352
    1. Saba K, Denda-Nagai K, Irimura T. A C-type lectin MGL1/CD301a plays an anti-inflammatory role in murine experimental colitis. Am J Pathol (2009) 174:144–5210.2353/ajpath.2009.080235
    1. Müller S, Schaffer T, Flogerzi B, Seibold-Schmid B, Schnider J, Takahashi K, et al. Mannan-binding lectin deficiency results in unusual antibody production and excessive experimental colitis in response to mannose-expressing mild gut pathogens. Gut (2010) 59:1493–50010.1136/gut.2010.208348
    1. Saunders SP, Barlow JL, Walsh CM, Bellsoi A, Smith P, McKenzie AN, et al. C-type lectin SIGN-R1 has a role in experimental colitis and responsiveness to lipopolysaccharide. J Immunol (2010) 184:2627–3710.4049/jimmunol.0901970
    1. Iliev ID, Funari VA, Taylor KD, Nguyen Q, Reyes CN, Strom SP, et al. Interactions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis. Science (2012) 336:1314–710.1126/science.1221789
    1. Geijtenbeek TB, Torensma R, van Vliet SJ, van Duijnhoven GC, Adema GJ, van Kooyk Y, et al. Identification of DC-SIGN, a novel dendritic cell-specific ICAM-3 receptor that supports primary immune responses. Cell (2000) 100:575–8510.1016/S0092-8674(00)80693-5
    1. Geijtenbeek TB, Van Vliet SJ, Koppel EA, Sanchez-Hernandez M, Vandenbroucke-Grauls CM, Appelmelk B, et al. Mycobacteria target DC-SIGN to suppress dendritic cell function. J Exp Med (2003) 197:7–1710.1084/jem.20021229
    1. Park CG, Takahara K, Umemoto E, Yashima Y, Matsubara K, Matsuda Y, et al. Five mouse homologues of the human dendritic cell C-type lectin, DC-SIGN. Int Immunol (2001) 13:1283–9010.1093/intimm/13.10.1283
    1. Powlesland AS, Ward EM, Sadhu SK, Guo Y, Taylor ME, Drickamer K. Widely divergent biochemical properties of the complete set of mouse DC-SIGN-related proteins. J Biol Chem (2006) 281:20440–910.1074/jbc.M601925200
    1. Galustian C, Park CG, Chai W, Kiso M, Bruening SA, Kang YS, et al. High and low affinity carbohydrate ligands revealed for murine SIGN-R1 by carbohydrate array and cell binding approaches, and differing specificities for SIGN-R3 and langerin. Int Immunol (2004) 16:853–6610.1093/intimm/dxh089
    1. Takahara K, Yashima Y, Omatsu Y, Yoshida H, Kimura Y, Kang YS, et al. Functional comparison of the mouse DC-SIGN, SIGNR1, SIGNR3 and Langerin, C-type lectins. Int Immunol (2004) 16:819–2910.1093/intimm/dxh084
    1. Tanne A, Ma B, Boudou F, Tailleux L, Botella H, Badell E, et al. A murine DC-SIGN homologue contributes to early host defense against Mycobacterium tuberculosis. J Exp Med (2009) 206:2205–2010.1084/jem.20090188
    1. Lefevre L, Lugo-Villarino G, Meunier E, Valentin A, Olagnier D, Authier H, et al. The C-type lectin receptors Dectin-1, MR, and SIGNR3 contribute both positively and negatively to the macrophage response to Leishmania infantum. Immunity (2013) 38:1038–4910.1016/j.immuni.2013.04.010
    1. Cooper HS, Murthy SN, Shah RS, Sedergran DJ. Clinicopathologic study of dextran sulfate sodium experimental murine colitis. Lab Invest (1993) 69:238–49
    1. Levitz SM. Innate recognition of fungal cell walls. PLoS Pathog (2010) 6:e1000758.10.1371/journal.ppat.1000758
    1. Bourgeois C, Kuchler K. Fungal pathogens-a sweet and sour treat for Toll-like receptors. Front Cell Infect Microbiol (2012) 2:142.10.3389/fcimb.2012.00142
    1. Hardison SE, Brown GD. C-type lectin receptors orchestrate antifungal immunity. Nat Immunol (2012) 13:817–2210.1038/ni.2369
    1. Cambi A, Gijzen K, de Vries LJ, Torensma R, Joosten B, Adema GJ, et al. The C-type lectin DC-SIGN (CD209) is an antigen-uptake receptor for Candida albicans on dendritic cells. Eur J Immunol (2003) 33:532–810.1002/immu.200310029
    1. Geijtenbeek TB, den Dunnen J, Gringhuis SI. Pathogen recognition by DC-SIGN shapes adaptive immunity. Future Microbiol (2009) 4:879–9010.2217/fmb.09.51
    1. Rizzetto L, Kuka M, De Filippo C, Cambi A, Netea MG, Beltrame L, et al. Differential IL-17 production and mannan recognition contribute to fungal pathogenicity and commensalism. J Immunol (2010) 184:4258–6810.4049/jimmunol.0902972
    1. Gringhuis SI, den Dunnen J, Litjens M, van Het Hof B, van Kooyk Y, Geijtenbeek TB. C-type lectin DC-SIGN modulates Toll-like receptor signaling via Raf-1 kinase-dependent acetylation of transcription factor NF-kappaB. Immunity (2007) 26:605–1610.1016/j.immuni.2007.03.012
    1. te Velde AA, van Kooyk Y, Braat H, Hommes DW, Dellemijn TA, Slors JF, et al. Increased expression of DC-SIGN+IL-12+IL-18+ and CD83+IL-12-IL-18- dendritic cell populations in the colonic mucosa of patients with Crohn’s disease. Eur J Immunol (2003) 33:143–5110.1002/immu.200390017
    1. Yamasaki S, Matsumoto M, Takeuchi O, Matsuzawa T, Ishikawa E, Sakuma M, et al. C-type lectin Mincle is an activating receptor for pathogenic fungus, Malassezia. Proc Natl Acad Sci U S A (2009) 106:1897–90210.1073/pnas.0805177106
    1. Ishikawa T, Itoh F, Yoshida S, Saijo S, Matsuzawa T, Gonoi T, et al. Identification of distinct ligands for the C-type lectin receptors mincle and Dectin-2 in the pathogenic fungus Malassezia. Cell Host Microbe (2013) 13:477–8810.1016/j.chom.2013.03.008
    1. Johansson ME, Gustafsson JK, Sjoberg KE, Petersson J, Holm L, Sjovall H, et al. Bacteria penetrate the inner mucus layer before inflammation in the dextran sulfate colitis model. PLoS ONE (2010) 5:e12238.10.1371/journal.pone.0012238
    1. Johansson ME, Ambort D, Pelaseyed T, Schutte A, Gustafsson JK, Ermund A, et al. Composition and functional role of the mucus layers in the intestine. Cell Mol Life Sci (2011) 68:3635–4110.1007/s00018-011-0822-3
    1. Hans W, Schölmerich J, Gross V, Falk W. The role of the resident intestinal flora in acute and chronic dextran sulfate sodium-induced colitis in mice. Eur J Gastroenterol Hepatol (2000) 12:267–7310.1097/00042737-200012030-00002
    1. Watanabe T, Kitani A, Murray PJ, Strober W. NOD2 is a negative regulator of Toll-like receptor 2-mediated T helper type 1 responses. Nat Immunol (2004) 5:800–810.1038/ni1092
    1. Inohara N, Ogura Y, Chen FF, Muto A, Nunez G. Human Nod1 confers responsiveness to bacterial lipopolysaccharides. J Biol Chem (2001) 276:2551–410.1074/jbc.M009728200
    1. Macho Fernandez E, Valenti V, Rockel C, Hermann C, Pot B, Boneca IG, et al. Anti-inflammatory capacity of selected lactobacilli in experimental colitis is driven by NOD2-mediated recognition of a specific peptidoglycan-derived muropeptide. Gut (2011) 60:1050–910.1136/gut.2010.232918
    1. Neurath MF, Fuss I, Pasparakis M, Alexopoulou L, Haralambous S, Meyer zum Büschenfelde KH, et al. Predominant pathogenic role of tumor necrosis factor in experimental colitis in mice. Eur J Immunol (1997) 27:1743–5010.1002/eji.1830270722
    1. Apostolaki M, Armaka M, Victoratos P, Kollias G. Cellular mechanisms of TNF function in models of inflammation and autoimmunity. Curr Dir Autoimmun (2010) 11:1–2610.1159/000289195
    1. Engel MA, Neurath MF. New pathophysiological insights and modern treatment of IBD. J Gastroenterol (2010) 45:571–8310.1007/s00535-010-0219-3
    1. Lepenies B, Lee J, Sonkaria S. Targeting C-type lectin receptors with multi-valent carbohydrate ligands. Adv Drug Deliv Rev (2013).10.1016/j.addr.2013.05.007

Source: PubMed

3
Tilaa