Serum cytokines elevated during gluten-mediated cytokine release in coeliac disease

G Goel, A J M Daveson, C E Hooi, J A Tye-Din, S Wang, E Szymczak, L J Williams, J L Dzuris, K M Neff, K E Truitt, R P Anderson, G Goel, A J M Daveson, C E Hooi, J A Tye-Din, S Wang, E Szymczak, L J Williams, J L Dzuris, K M Neff, K E Truitt, R P Anderson

Abstract

Cytokines have been extensively studied in coeliac disease, but cytokine release related to exposure to gluten and associated symptoms has only recently been described. Prominent, early elevations in serum interleukin (IL)-2 after gluten support a central role for T cell activation in the clinical reactions to gluten in coeliac disease. The aim of this study was to establish a quantitative hierarchy of serum cytokines and their relation to symptoms in patients with coeliac disease during gluten-mediated cytokine release reactions. Sera were analyzed from coeliac disease patients on a gluten free-diet (n = 25) and from a parallel cohort of healthy volunteers (n = 25) who underwent an unmasked gluten challenge. Sera were collected at baseline and 2, 4 and 6 h after consuming 10 g vital wheat gluten flour; 187 cytokines were assessed. Confirmatory analyses were performed by high-sensitivity electrochemiluminescence immunoassay. Cytokine elevations were correlated with symptoms. Cytokine release following gluten challenge in coeliac disease patients included significant elevations of IL-2, chemokine (C-C motif) ligand 20 (CCL20), IL-6, chemokine (C-X-C motif) ligand (CXCL)9, CXCL8, interferon (IFN)-γ, IL-10, IL-22, IL-17A, tumour necrosis factor (TNF)-α, CCL2 and amphiregulin. IL-2 and IL-17A were earliest to rise. Peak levels of cytokines were generally at 4 h. IL-2 increased most (median 57-fold), then CCL20 (median 10-fold). Cytokine changes were strongly correlated with one another, and the most severely symptomatic patients had the highest elevations. Early elevations of IL-2, IL-17A, IL-22 and IFN-γ after gluten in patients with coeliac disease implicates rapidly activated T cells as their probable source. Cytokine release after gluten could aid in monitoring experimental treatments and support diagnosis.

Keywords: IL-17; IL-2; coeliac disease; cytokine release syndrome; cytokines; gluten.

Conflict of interest statement

H. E. has no conflicts of interest to disclose. G. G., S. W., E. S., L. J. W., J. L. D., K. M. N., K. E. T. and R. P. A. are employees of ImmusanT, Inc. J. A. T.‐D. and A. J. M. D. have served as advisers to ImmusanT, Inc. R. P. A. is inventor of Patents, owned or licensed by ImmusanT, Inc., relating to the diagnostic application of gluten challenge. H. C. E. has no conflicts to disclose.

© 2019 British Society for Immunology.

Figures

Figure 1
Figure 1
Study design.
Figure 2
Figure 2
Heat‐map of serum cytokine elevations after gluten challenge. Peak fold change in cytokine levels were normalized by dividing by the sum of the mean plus 3× (standard deviations) of fold change observed in the unaffected cohort.
Figure 3
Figure 3
Serum cytokine profiles after gluten challenge and self‐rated severity assessments. Coeliac disease participants (n = 25) were grouped according to their overall severity of digestive symptoms, and compared to healthy volunteers (n = 25).

References

    1. Goel G, Tye‐Din JA, Qiao S‐W et al Cytokine release and gastrointestinal symptoms after gluten challenge in celiac disease. Science. Advances 2019; 5:aaw7756.
    1. Anderson RP, Degano P, Godkin AJ, Jewell DP, Hill AV. In vivo antigen challenge in celiac disease identifies a single transglutaminase‐modified peptide as the dominant A‐gliadin T‐cell epitope. Nat Med 2000; 6:337–42.
    1. Raki M, Fallang LE, Brottveit M et al Tetramer visualization of gut‐homing gluten‐specific T cells in the peripheral blood of celiac disease patients. Proc Natl Acad Sci USA 2007; 104:2831–6.
    1. Ontiveros N, Tye‐Din JA, Hardy MY, Anderson RP. Ex‐vivo whole blood secretion of interferon (IFN)‐gamma and IFN‐gamma‐inducible protein‐10 measured by enzyme‐linked immunosorbent assay are as sensitive as IFN‐gamma enzyme‐linked immunospot for the detection of gluten‐reactive T cells in human leucocyte antigen (HLA)‐DQ2.5(+) ‐associated coeliac disease. Clin Exp Immunol 2014; 175:305–15.
    1. Arentz‐Hansen H, Korner R, Molberg O 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; 191:603–12.
    1. Osman AA, Gunnel T, Dietl A et al B cell epitopes of gliadin. Clin Exp Immunol 2000; 121:248–54.
    1. Hoydahl LS, Richter L, Frick R et al Plasma cells are the most abundant gluten peptide MHC‐expressing cells in inflamed intestinal tissues from patients with celiac disease. Gastroenterology 2019; 156:1428–39 e10.
    1. Jelinkova L, Tuckova L, Cinova J, Flegelova Z, Tlaskalova‐Hogenova H. Gliadin stimulates human monocytes to production of IL‐8 and TNF‐alpha through a mechanism involving NF‐kappaB. FEBS Lett 2004; 571:81–5.
    1. Barone MV, Troncone R, Auricchio S. Gliadin peptides as triggers of the proliferative and stress/innate immune response of the celiac small intestinal mucosa. Int J Mol Sci 2014; 15:20518–37.
    1. Nilsen EM, Lundin KE, Krajci P, Scott H, Sollid LM, Brandtzaeg P. Gluten specific, HLA‐DQ restricted T cells from coeliac mucosa produce cytokines with Th1 or Th0 profile dominated by interferon gamma. Gut 1995; 37:766–76.
    1. Baumgart DC, Lowder JN, Targan SR, Sandborn WJ, Frankel MB. Transient cytokine‐induced liver injury following administration of the humanized anti‐CD3 antibody visilizumab (HuM291) in Crohn’s disease. Am J Gastroenterol 2009; 104:868–76.
    1. Suntharalingam G, Perry MR, Ward S et al Cytokine storm in a phase 1 trial of the anti‐CD28 monoclonal antibody TGN1412. N Engl J Med 2006;355: 1018–28.
    1. Winkler U, Jensen M, Manzke O, Schulz H, Diehl V, Engert A. Cytokine‐release syndrome in patients with B‐cell chronic lymphocytic leukemia and high lymphocyte counts after treatment with an anti‐CD20 monoclonal antibody (rituximab, IDEC‐C2B8). Blood 1999; 94:2217–24.
    1. Truitt KE, Daveson AJM, Ee HC et al A randomized, placebo‐controlled clinical trial of subcutaneous or intradermal Nexvax2, an investigational immunomodulatory peptide therapy for coeliac disease. Aliment Pharmacol Ther 2019; 50:547‐55.
    1. Tye‐Din JA, Daveson AJM, Ee HC et al Systemic interleukin‐2 release after gluten is sensitive and specific for coeliac disease, and correlates with symptoms. Aliment Pharmacol Ther 2019. doi:10.1111/apt.15477.
    1. Zaiss DMW, Gause WC, Osborne LC, Artis D. Emerging functions of amphiregulin in orchestrating immunity, inflammation, and tissue repair. Immunity 2015; 42:216–26.
    1. Abadie V, Jabri B. IL‐15: a central regulator of celiac disease immunopathology. Immunol Rev 2014; 260:221–34.
    1. Yoosuf S, Makharia GK. Evolving therapy for celiac disease. Front Pediatr 2019; 7:193.
    1. Sarna VK, Lundin KEA, Morkrid L, Qiao SW, Sollid LM, Christophersen A. HLA‐DQ‐gluten tetramer blood test accurately identifies patients with and without celiac disease in absence of gluten consumption. Gastroenterology 2018; 154:886–96.e6.
    1. Lee DW, Santomasso BD, Locke FL et al ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant 2019; 25:625–38.
    1. National Cancer Institute (NCI ) . Common Terminology Criteria For Adverse Events (CTCAE), version 5.0. National Cancer Institute July 2019. Available at: .
    1. Ludvigsson JF, Ciacci C, Green PH et al Outcome measures in coeliac disease trials: the Tampere recommendations. Gut 2018; 67:1410–24.
    1. Molina‐Infante J, Carroccio A. Suspected nonceliac gluten sensitivity confirmed in few patients after gluten challenge in double‐blind, placebo‐controlled trials . Clin Gastroenterol Hepatol 2017; 15: 339–48.
    1. Bruins MJ. The clinical response to gluten challenge: a review of the literature. Nutrients 2013; 5:4614–41.
    1. Kasarda DD. Can an increase in celiac disease be attributed to an increase in the gluten content of wheat as a consequence of wheat breeding? J Agric Food Chem 2013; 61:1155–9.
    1. Hoppe C, Gobel R, Kristensen M et al Intake and sources of gluten in 20‐ to 75‐year‐old Danish adults: a national dietary survey. Eur J Nutr 2017; 56:107–17.
    1. Goel G, King T, Daveson AJ et al Epitope‐specific immunotherapy targeting CD4‐positive T cells in coeliac disease: two randomised, double‐blind, placebo‐controlled phase 1 studies. Lancet Gastroenterol Hepatol 2017; 2:479–93.
    1. Abramowicz D, Schandene L, Goldman M et al Release of tumor necrosis factor, interleukin‐2, and gamma‐interferon in serum after injection of OKT3 monoclonal antibody in kidney transplant recipients. Transplantation 1989; 47:606–8.
    1. Chatenoud L, Ferran C, Legendre C et al In vivo cell activation following OKT3 administration. Systemic cytokine release and modulation by corticosteroids. Transplantation 1990; 49:697–702.
    1. Kooy‐Winkelaar YM, Bouwer D, Janssen GM et al CD4 T‐cell cytokines synergize to induce proliferation of malignant and nonmalignant innate intraepithelial lymphocytes. Proc Natl Acad Sci USA 2017; 114:E980–E989.
    1. Korneychuk N, Ramiro‐Puig E, Ettersperger J et al Interleukin 15 and CD4(+) T cells cooperate to promote small intestinal enteropathy in response to dietary antigen. Gastroenterology 2014; 146:1017–27.
    1. Bodd M, Raki M, Tollefsen S 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.
    1. Fernandez S, Molina IJ, Romero P et al Characterization of gliadin‐specific Th17 cells from the mucosa of celiac disease patients. Am J Gastroenterol 2011; 106:528–38.
    1. Fina D, Sarra M, Caruso R et al Interleukin 21 contributes to the mucosal T helper cell type 1 response in coeliac disease. Gut 2008; 57:887–92.
    1. Monteleone I, Sarra M, Del Vecchio Blanco G et al Characterization of IL‐17A‐producing cells in celiac disease mucosa. J Immunol 2010; 184:2211–8.
    1. Faghih M, Rostami‐Nejad M, Amani D et al Analysis of IL17A and IL21 expression in the small intestine of celiac disease patients and correlation with circulating thioredoxin level. Genet Test Mol Biomark 2018; 22:518–25.
    1. Lahdenpera AI, Falth‐Magnusson K, Hogberg L, Ludvigsson J, Vaarala O. Expression pattern of T‐helper 17 cell signaling pathway and mucosal inflammation in celiac disease. Scand J Gastroenterol 2014; 49:145–56.
    1. Bondar C, Araya RE, Guzman L, Rua EC, Chopita N, Chirdo FG. Role of CXCR3/CXCL10 axis in immune cell recruitment into the small intestine in celiac disease. PLoS ONE 2014; 9:e89068.
    1. Sato N, Patel HJ, Waldmann TA, Tagaya Y. The IL‐15/IL‐15Ralpha on cell surfaces enables sustained IL‐15 activity and contributes to the long survival of CD8 memory T cells. Proc Natl Acad Sci USA 2007; 104:588–93.
    1. van de Veen W, Wirz OF, Globinska A, Akdis M. Novel mechanisms in immune tolerance to allergens during natural allergen exposure and allergen‐specific immunotherapy. Curr Opin Immunol 2017; 48:74–81.
    1. Marquez A, Kerick M, Zhernakova A et al Meta‐analysis of Immunochip data of four autoimmune diseases reveals novel single‐disease and cross‐phenotype associations. Genome Med 2018; 10:97.
    1. Hunt KA, Zhernakova A, Turner G et al Newly identified genetic risk variants for celiac disease related to the immune response. Nat Genet 2008; 40:395–402.
    1. Crotty B, Rosenberg WM, Aronson JK, Jewell DP. Inhibition of binding of interferon‐gamma to its receptor by salicylates used in inflammatory bowel disease. Gut 1992; 33:1353–7.
    1. Deisseroth A, Ko CW, Nie L et al FDA approval: siltuximab for the treatment of patients with multicentric Castleman disease. Clin Cancer Res 2015; 21:950–4.
    1. Fragoulis GE, Siebert S, McInnes IB. Therapeutic targeting of IL‐17 and IL‐23 cytokines in immune‐mediated diseases. Annu Rev Med 2016; 67:337–53.
    1. Yao W, Ba Q, Li X et al A Natural CCR2 antagonist relieves tumor‐associated macrophage‐mediated immunosuppression to produce a therapeutic effect for liver cancer. EBioMedicine 2017; 22:58–67.
    1. Burton BR, Britton GJ, Fang H et al Sequential transcriptional changes dictate safe and effective antigen‐specific immunotherapy. Nat Commun 2014; 5:4741.
    1. Campbell JD, Buckland KF, McMillan SJ et al Peptide immunotherapy in allergic asthma generates IL‐10‐dependent immunological tolerance associated with linked epitope suppression. J Exp Med 2009; 206:1535–47.
    1. Salvati VM, Mazzarella G, Gianfrani C et al Recombinant human interleukin 10 suppresses gliadin dependent T cell activation in ex vivo cultured coeliac intestinal mucosa. Gut 2005; 54:46–53.
    1. Gianfrani C, Levings MK, Sartirana C et al Gliadin‐specific type 1 regulatory T cells from the intestinal mucosa of treated celiac patients inhibit pathogenic T cells. J Immunol 2006; 177:4178–86.
    1. Du Pre MF, Kozijn AE, van Berkel LA et al Tolerance to ingested deamidated gliadin in mice is maintained by splenic, type 1 regulatory T cells. Gastroenterology 2011; 141:610–20, 20 e1–2.
    1. Lundin KE, Scott H, Hansen T et al Gliadin‐specific, HLA‐DQ(alpha 1*0501, beta 1*0201) restricted T cells isolated from the small intestinal mucosa of celiac disease patients. J Exp Med 1993; 178:187–96.

Source: PubMed

3
Tilaa