Rapid accumulation of CD14+CD11c+ dendritic cells in gut mucosa of celiac disease after in vivo gluten challenge

Ann-Christin Røberg Beitnes, Melinda Ráki, Margit Brottveit, Knut Erik Aslaksen Lundin, Frode Lars Jahnsen, Ludvig Magne Sollid, Ann-Christin Røberg Beitnes, Melinda Ráki, Margit Brottveit, Knut Erik Aslaksen Lundin, Frode Lars Jahnsen, Ludvig Magne Sollid

Abstract

Background: Of antigen-presenting cells (APCs) expressing HLA-DQ molecules in the celiac disease (CD) lesion, CD11c(+) dendritic cells (DCs) co-expressing the monocyte marker CD14 are increased, whereas other DC subsets (CD1c(+) or CD103(+)) and CD163(+)CD11c(-) macrophages are all decreased. It is unclear whether these changes result from chronic inflammation or whether they represent early events in the gluten response. We have addressed this in a model of in vivo gluten challenge.

Methods: Treated HLA-DQ2(+) CD patients (n = 12) and HLA-DQ2(+) gluten-sensitive control subjects (n = 12) on a gluten-free diet (GFD) were orally challenged with gluten for three days. Duodenal biopsies obtained before and after gluten challenge were subjected to immunohistochemistry. Single cell digests of duodenal biopsies from healthy controls (n = 4), treated CD (n = 3) and untreated CD (n = 3) patients were analyzed by flow cytometry.

Results: In treated CD patients, the gluten challenge increased the density of CD14(+)CD11c(+) DCs, whereas the density of CD103(+)CD11c(+) DCs and CD163(+)CD11c(-) macrophages decreased, and the density of CD1c(+)CD11c(+) DCs remained unchanged. Most CD14(+)CD11c(+) DCs co-expressed CCR2. The density of neutrophils also increased in the challenged mucosa, but in most patients no architectural changes or increase of CD3(+) intraepithelial lymphocytes (IELs) were found. In control tissue no significant changes were observed.

Conclusions: Rapid accumulation of CD14(+)CD11c(+) DCs is specific to CD and precedes changes in mucosal architecture, indicating that this DC subset may be directly involved in the immunopathology of the disease. The expression of CCR2 and CD14 on the accumulating CD11c(+) DCs indicates that these cells are newly recruited monocytes.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. CD14 + CD11c + dendritic…
Figure 1. CD14+CD11c+ dendritic cells are selectively increased in duodenal mucosa of celiac disease patients after short-term gluten challenge.
Density of HLA-DQ+ antigen-presenting cell subsets in cryosections from celiac disease (CD) patients on gluten-free diet (GFD) before and after a three-day gluten challenge. The density of CD163+CD11c− macrophages is calculated by subtracting the number of CD163+CD11c+ cells from the total number of CD163+HLA-DQ+ cells. Paired data are connected by lines. ns = not significant (A). Three-color immunofluorescence staining for CD11c (green), CD14 (red) and cytokeratin (blue) in cryosection of duodenal mucosa from a CD patient on GFD before and after a three-day gluten challenge. Original magnification X 400 (B).
Figure 2. Expression of CCR2 on HLA-DR…
Figure 2. Expression of CCR2 on HLA-DR+ leukocytes in blood and duodenal mucosa.
Flow cytometric analysis of peripheral blood mononuclear cells from treated celiac disease (CD) patient (A) and viable single cells of duodenal mucosa from treated CD patient showing the expression of CCR2 (B) and DC-SIGN (C) on CD45+HLA-DR+ cells depending on the expression of CD11c and CD14. Dead cells were excluded by adding 0.2 µg/mL propidium iodide immediately before acquisition. The data are representative for two independent experiments.
Figure 3. CCR2 is highly expressed on…
Figure 3. CCR2 is highly expressed on CD14+CD11c+ dendritic cells in duodenal mucosa.
Percentage of CCR2+ cells in subsets of antigen-presenting cells from duodenal mucosa digests assessed by flow cytometry. Median is indicated by horizontal line. CD = celiac disease.
Figure 4. Neuthrophils are increased in duodenal…
Figure 4. Neuthrophils are increased in duodenal mucosa of celiac disease patients after short-term gluten challenge.
Density of CD3+ intraepithelial lymphocytes (IELs) per 100 epithelial cells (A); neutrophils per mm2 (B); and eosinophils per mm2 (C) in the lamina propria (LP) in sections of duodenal mucosa from celiac disease patients on gluten-free diet before and after a three-day gluten challenge. Paired data are connected by lines. ns = not significant.
Figure 5. Peripheral blood CD14 + monocytes…
Figure 5. Peripheral blood CD14+ monocytes efficiently present gluten to gluten-specific T cell clones.
Purity of CD14+CD11c+ monocytes isolated from peripheral blood mononuclear cells of two individuals are shown (A and B, upper panels). The monocytes were incubated with medium or two different gluten peptides ±100 U/ml IFN-γ for 24 hours, washed and incubated with a T-cell clone for 72 hours. The proliferative T-cell response (measured by thymindine-incorporation) is shown (A and B, lower panels). Experiments with IFN-γ are indicated (hatched columns).
Figure 6. Density of various leukocyte subsets…
Figure 6. Density of various leukocyte subsets remains unchanged in duodenal mucosa of gluten-sensitive control subjects after short-term gluten challenge.
Density of CD14+CD11c+ dendritic cells (DCs) per mm2 (A) and CD103+CD11c+ DCs per mm2 (B) in the lamina propria (LP); CD3+ intraepithelial lymphocytes (IELs) per 100 epithelial cells (C); neutrophils per mm2 (D) and eosinophils per mm2 (E) in the LP in sections of duodenal mucosa from gluten-sensitive control subjects on gluten-free diet before and after a three-day gluten challenge. Paired data are connected by lines. ns = not significant.

References

    1. Sollid LM. Coeliac disease: dissecting a complex inflammatory disorder. Nat Rev Immunol. 2002;2:647–655.
    1. Jabri B, Sollid LM. Tissue-mediated control of immunopathology in coeliac disease. Nat Rev Immunol. 2009;9:858–870.
    1. Walker MM, Murray JA. An update in the diagnosis of coeliac disease. Histopathology. 2011;59:166–179.
    1. Raki M, Tollefsen S, Molberg O, Lundin KE, Sollid LM, et al. A unique dendritic cell subset accumulates in the celiac lesion and efficiently activates gluten-reactive T cells. Gastroenterology. 2006;131:428–438.
    1. Beitnes AC, Raki M, Lundin KE, Jahnsen J, Sollid LM, et al. Density of CD163(+) CD11c(+) dendritic cells increases and CD103(+) dendritic cells decreases in the coeliac lesion. Scand J Immunol. 2011;74:186–194.
    1. Brottveit M, Raki M, Bergseng E, Fallang LE, Simonsen B, et al. Assessing possible celiac disease by an HLA-DQ2-gliadin Tetramer Test. Am J Gastroenterol. 2011;106:1318–1324.
    1. Oberhuber G, Granditsch G, Vogelsang H. The histopathology of coeliac disease: time for a standardized report scheme for pathologists. Eur J Gastroenterol Hepatol. 1999;11:1185–1194.
    1. Spits H, Borst J, Giphart M, Coligan J, Terhorst C, et al. HLA-DC antigens can serve as recognition elements for human cytotoxic T lymphocytes. Eur J Immunol. 1984;14:299–304.
    1. Micklem KJ, Dong Y, Willis A, Pulford KA, Visser L, et al. HML-1 antigen on mucosa-associated T cells, activated cells, and hairy leukemic cells is a new integrin containing the beta 7 subunit. Am J Pathol. 1991;139:1297–1301.
    1. Wang JE, Warris A, Ellingsen EA, Jorgensen PF, Flo TH, et al. Involvement of CD14 and toll-like receptors in activation of human monocytes by Aspergillus fumigatus hyphae. Infect Immun. 2001;69:2402–2406.
    1. Huitfeldt HS, Brandtzaeg P. Various keratin antibodies produce immunohistochemical staining of human myocardium and myometrium. Histochemistry. 1985;83:381–389.
    1. Jahnsen FL, Haraldsen G, Rugtveit J, Halstensen TS, Brandtzaeg P. Differential interference contrast microscopy combined with immunofluorescence: a new method to phenotype eosinophils in situ. J Immunol Methods. 1994;173:77–91.
    1. Arentz-Hansen H, Korner R, Molberg O, Quarsten H, Vader W, et al. The intestinal T cell response to alpha-gliadin in adult celiac disease is focused on a single deamidated glutamine targeted by tissue transglutaminase. J Exp Med. 2000;191:603–612.
    1. Shan L, Molberg O, Parrot I, Hausch F, Filiz F, et al. Structural basis for gluten intolerance in celiac sprue. Science. 2002;297:2275–2279.
    1. Molberg O, McAdam SN, Lundin KE, Sollid LM. Studies of gliadin-specific T-cells in celiac disease. Methods Mol Med. 2000;41:105–124.
    1. Wong KL, Tai JJ, Wong WC, Han H, Sem X, et al. Gene expression profiling reveals the defining features of the classical, intermediate, and nonclassical human monocyte subsets. Blood. 2011;118:e16–e31.
    1. Tallone T, Turconi G, Soldati G, Pedrazzini G, Moccetti T, et al. Heterogeneity of human monocytes: an optimized four-color flow cytometry protocol for analysis of monocyte subsets. J Cardiovasc Transl Res. 2011;4:211–219.
    1. Shantsila E, Wrigley B, Tapp L, Apostolakis S, Montoro-Garcia S, et al. Immunophenotypic characterisation of human monocyte subsets: Possible implications for cardiovascular disease pathophysiology. J Thromb Haemost. 2011;9:1056–1066.
    1. Leigh RJ, Marsh MN, Crowe P, Kelly C, Garner V, et al. Studies of intestinal lymphoid tissue. IX. Dose-dependent, gluten-induced lymphoid infiltration of coeliac jejunal epithelium. Scand J Gastroenterol. 1985;20:715–719.
    1. Marsh MN. Gluten, major histocompatibility complex, and the small intestine. A molecular and immunobiologic approach to the spectrum of gluten sensitivity (‘celiac sprue’). Gastroenterology. 1992;102:330–354.
    1. Dhesi I, Marsh MN, Kelly C, Crowe P. Morphometric analysis of small intestinal mucosa. II. Determination of lamina propria volumes; plasma cell and neutrophil populations within control and coeliac disease mucosae. Virchows Arch A Pathol Anat Histopathol. 1984;403:173–180.
    1. Marsh MN, Hinde J. Inflammatory component of celiac sprue mucosa. I. Mast cells, basophils, and eosinophils. Gastroenterology. 1985;89:92–101.
    1. Schwartzkopff F, Petersen F, Grimm TA, Brandt E. CXC chemokine ligand 4 (CXCL4) down-regulates CC chemokine receptor expression on human monocytes. Innate Immun 2010
    1. Cheong C, Matos I, Choi JH, Dandamudi DB, Shrestha E, et al. Microbial stimulation fully differentiates monocytes to DC-SIGN/CD209(+) dendritic cells for immune T cell areas. Cell. 2010;143:416–429.
    1. Schuppan D, Junker Y, Barisani D. Celiac disease: from pathogenesis to novel therapies. Gastroenterology. 2009;137:1912–1933.
    1. Troncone R, Jabri B. Coeliac disease and gluten sensitivity. J Intern Med. 2011;269:582–590.
    1. Rakhimova M, Esslinger B, Schulze-Krebs A, Hahn EG, Schuppan D, et al. In vitro differentiation of human monocytes into dendritic cells by peptic-tryptic digest of gliadin is independent of genetic predisposition and the presence of celiac disease. J Clin Immunol. 2009;29:29–37.
    1. Palova-Jelinkova L, Rozkova D, Pecharova B, Bartova J, Sediva A, et al. Gliadin fragments induce phenotypic and functional maturation of human dendritic cells. J Immunol. 2005;175:7038–7045.
    1. Maiuri L, Ciacci C, Ricciardelli I, Vacca L, Raia V, et al. Association between innate response to gliadin and activation of pathogenic T cells in coeliac disease. Lancet. 2003;362:30–37.
    1. Diosdado B, van BH, Strengman E, Franke L, van OE, et al. Neutrophil recruitment and barrier impairment in celiac disease: a genomic study. Clin Gastroenterol Hepatol. 2007;5:574–581.
    1. Halstensen TS, Scott H, Fausa O, Brandtzaeg P. Gluten stimulation of coeliac mucosa in vitro induces activation (CD25) of lamina propria CD4+ T cells and macrophages but no crypt-cell hyperplasia. Scand J Immunol. 1993;38:581–590.
    1. Raki M, Molberg O, Tollefsen S, Lundin KE, Sollid LM. The effects of atorvastatin on gluten-induced intestinal T cell responses in coeliac disease. Clin Exp Immunol. 2005;142:333–340.
    1. Bodd M, Raki M, Tollefsen S, Fallang LE, Bergseng E, et al. HLA-DQ2-restricted gluten-reactive T cells produce IL-21 but not IL-17 or IL-22. Mucosal Immunol. 2010;3:594–601.

Source: PubMed

3
Tilaa