Deep analysis of immune response and metabolic signature in children with food protein induced enterocolitis to cow's milk

Karine Adel-Patient, Guillaume Lezmi, Florence Anne Castelli, Sibylle Blanc, Hervé Bernard, Pascale Soulaines, Pascale Dumond, Sandrine Ah-Leung, Florence Lageix, Delphine de Boissieu, Naima Cortes-Perez, Stéphane Hazebrouck, François Fenaille, Christophe Junot, Christophe Dupont, Karine Adel-Patient, Guillaume Lezmi, Florence Anne Castelli, Sibylle Blanc, Hervé Bernard, Pascale Soulaines, Pascale Dumond, Sandrine Ah-Leung, Florence Lageix, Delphine de Boissieu, Naima Cortes-Perez, Stéphane Hazebrouck, François Fenaille, Christophe Junot, Christophe Dupont

Abstract

Background: Food Protein-Induced Enterocolitis Syndrome (FPIES) is considered to be a non-IgE mediated food allergy. However, its pathogenesis remains poorly understood and biomarkers are lacking. We aimed to perform in-depth characterization of humoral and cellular immune responses in children with cow's milk (CM)-FPIES and investigated whether there is a FPIES metabolomic signature.

Methods: Children with CM-FPIES and control subjects with an IgE-mediated CM allergy (IgE-CMA), both avoiding CM, were recruited on the day of an oral food challenge. Blood samples were collected before the challenge. Total and specific levels of IgE, IgG1-4, IgA, IgM and IgD to various whey and casein allergens and to their gastroduodenal digestion products were measured in plasma, using plasma from CM-tolerant peanut allergic patients (IgE-PA, not avoiding CM) as additional controls. Cytokine secretion and cellular proliferation were analyzed after stimulation of PBMC with different CM allergens. Metabolomic profiles were obtained for plasma samples using liquid chromatography coupled to high-resolution mass spectrometry.

Results: Nine children with CM-FPIES and 12 control subjects (6 IgE-CMA and 6 IgE-PA) were included. In children with CM-FPIES, total Ig concentrations were lower than in control subjects, specific Ig against CM components were weak to undetectable, and no specific IgE against CM digestion products were detected. Moreover, in CM-FPIES patients, we did not find any Th cell proliferation or associated cytokine secretion after allergen reactivation, whereas such responses were clearly found in children with IgE-CMA. Plasma metabolic profiles were different between CM allergic patients, with significantly lower concentrations of various fatty acids and higher concentrations of primary metabolites such as amino acids in CM-FPIES compared to IgE-CMA patients.

Conclusions: In CM-FPIES, both humoral and cellular specific immune responses are weak or absent, and this is not related to CM avoidance. A metabolomic signature was identified in patients with CM-FPIES that may be useful for the diagnosis and management of this disease.

Keywords: Cellular immunity; Cow’s milk; FPIES; Food allergy; Humoral immunity; Mechanisms; Metabolomics; Non-IgE.

Figures

Fig. 1
Fig. 1
Total IgE, IgGs, IgM and IgA (ng/mL) in children with CM-FPIES (red), IgE-CMA (blue) or IgE-PNA (green). *p < 0.05, **p < 0.01, ***p < 0.001 using Kruskal–Wallis and Dunn’s multiple comparison test
Fig. 2
Fig. 2
a Specific IgE (IU/mL) in patients with CM-FPIES (red), IgE-CMA (blue) or IgE-PA (green) assayed using immunoenzymatic assays. *p < 0.05, **p < 0.01, ***p < 0.001 using the Kruskal–Wallis test and Dunn’s multiple comparison test. b Representative IgE immunoblot of CM proteins using plasma from children with IgE-CMA (left) or FPIES-CMA (right). Lane 1: MW markers, Lane 2: CM proteins
Fig. 3
Fig. 3
Specific IgG1 (A), IgG2 (B), IgG3 (C), IgG4 (D) and IgA (E) in patients suffering from CM-FPIES (red), IgE-CMA (blue) or IgE-PNA (green). Values are given as µg/mL except for IgA where values are given as milli-Absorbance Unit at 414 nm. Bars indicate medians. Statistics *p < 0.05, **p < 0.01, ***p < 0.001 using Mann–Whitney test. “a” indicates a statistical difference using the Kruskal–Wallis test and Dunn’s multiple comparison test
Fig. 4
Fig. 4
Specific IgG4 against gastroduodenal digestion products. a Electrophoresis of gastroduodenal CM protein digestion products: CM was digested for 0 to 120 min under physiological conditions using pepsin (T0 to T120, gastric digestion). Digestion products obtained after 60 min of pepsin digestion (T60 gastric, TO’) were then submitted to trypsin/chymotrypsin physiological digestion for 1 to 30 min (T1′ to T30′, gastroduodenal digestion). b Undigested CM proteins (T0), gastric digestion products obtained at 5 (T5) and 60 min (T60) and gastroduodenal digestion products obtained after 60 min of gastric digestion and 30 min of duodenal digestion (T30′) were immobilized on plates, and specific IgG4 were assayed using individual plasma (FPIES-CMA: red, IgE-CMA: blue, IgE-PNA: green). Absorbance obtained at T0 was used as an internal reference for each patient (100%). Statistics: bars and asterisks indicate statistical differences between specified time points and T0 in one group of patients (*p < 0.05, **p < 0.01; Wilcoxon sign rank test) or significant differences between groups at a given time of digestion (*p < 0.05, **p < 0.01; Mann–Whitney t test)
Fig. 5
Fig. 5
IL-13 (a), IFNγ (b) and IL-6 (c) secreted after specific reactivation of PBMC from IgE-CMA (blue) or FPIES-CMA (red) patients. PBMC were stimulated for 6 days with 50 µg/mL of purified allergens and cytokines were assayed in supernatants. Results are expressed as individual values and medians (bar) obtained for the PBMC from IgE-CMA (n = 6) or FPIES-CMA (n = 9) patients. * indicates a difference between control (PBS and/or Ara h 2) and stimulated PBMC in the same group of patients (p < 0.05 using Wilcoxon sign rank test); Significant difference or trends between IgE-CMA and FPIES-CMA patients are also mentioned with the associated p value (Mann–Whitney t test)
Fig. 6
Fig. 6
Analysis of proliferative T cells in CMA patients after allergen reactivation. PBMC from IgE-CMA (a) or FPIES-CMA (b) patients were labelled with CFSE and then cultured for 6 days with PBS or allergens purified from cow’s milk. Cells were then recovered and labelled with anti-human CD4. Among SSC-FSC gated cells, single cells were selected and analyzed for CD4 expression and CFSE signal. Proliferative T cells are defined as CD4+CFSElow cells within a selected population (red square). Proliferative cells after reactivation with PBS, BLG, caseins or α-lact are shown
Fig. 7
Fig. 7
Cellular analysis in intestinal biopsies and PBMC from active versus controlled FPIES. Cellular analysis was performed on cells extracted from sigmoid biopsies obtained from children presenting controlled FPIES (non-active) or active FPIES. Cells were labelled and analyzed by flow cytometry using a NovoCyte flow cytometer and analysis was performed using NovoExpress™ software (Version 1.2.1; ACEA Biosciences, Inc.). Percentages of ILC1; ILC2, or ILC3 cells among parent cells (CD45+CD127+) and grandparent cells (Lin−SSClow) are indicated in colour and in brackets, respectively. Lin: mix of labelled anti-human CD3, CD11c, CD14, CD16, CD19, CD56, FcεRIα, CD1a, CD123
Fig. 8
Fig. 8
Plasma metabolites in samples from CM-FPIES, IgE-CMA or resolved IgE-CMA patients. a Discriminant metabolites between CM-FPIES (red symbols), IgE-CMA (dark blue symbols) and IgE-resolved (light blue symbols). a Fatty acids, b amino acids and their derivatives, c purine metabolites or vitamins. The p values of nonparametric Mann–Whitney statistical test are indicated

References

    1. Nowak-Wegrzyn A, Katz Y, Mehr SS, Koletzko S. Non-IgE-mediated gastrointestinal food allergy. J Allergy Clin Immunol. 2015;135(5):1114–1124. doi: 10.1016/j.jaci.2015.03.025.
    1. Nowak-Wegrzyn A, Jarocka-Cyrta E, Moschione CA. Food protein-induced enterocolitis syndrome. J Investig Allergol Clin Immunol. 2017;27(1):1–18. doi: 10.18176/jiaci.0135.
    1. Caubet JC, Szajewska H, Shamir R, Nowak-Wegrzyn A. Non-IgE-mediated gastrointestinal food allergies in children. Pediatr Allergy Immunol. 2017;28(1):6–17. doi: 10.1111/pai.12659.
    1. Katz Y, Goldberg MR, Rajuan N, Cohen A, Leshno M. The prevalence and natural course of food protein-induced enterocolitis syndrome to cow’s milk: a large-scale, prospective population-based study. J Allergy Clin Immunol. 2011;127(3):647–653. doi: 10.1016/j.jaci.2010.12.1105.
    1. Caubet JC, Ford LS, Sickles L, Jarvinen KM, Sicherer SH, Sampson HA, Nowak-Wegrzyn A. Clinical features and resolution of food protein-induced enterocolitis syndrome: 10-year experience. J Allergy Clin Immunol. 2014;134(2):382–389. doi: 10.1016/j.jaci.2014.04.008.
    1. Katz Y, Goldberg MR. Natural history of food protein-induced enterocolitis syndrome. Curr Opin Allergy Clin Immunol. 2014;14(3):229–239. doi: 10.1097/ACI.0000000000000053.
    1. Feuille E, Nowak-Wegrzyn A. Food protein-induced enterocolitis syndrome, allergic proctocolitis, and enteropathy. Curr Allergy Asthma Rep. 2015;15(8):50. doi: 10.1007/s11882-015-0546-9.
    1. Berin MC. Immunopathophysiology of food protein-induced enterocolitis syndrome. J Allergy Clin Immunol. 2015;135(5):1108–1113. doi: 10.1016/j.jaci.2014.12.1948.
    1. Goswami R, Blazquez AB, Kosoy R, Rahman A, Nowak-Wegrzyn A, Berin MC. Systemic innate immune activation in food protein induced enterocolitis syndrome (FPIES) J Allergy Clin Immunol. 2017;139(6):1885–1896. doi: 10.1016/j.jaci.2016.12.971.
    1. Caubet JC, Bencharitiwong R, Ross A, Sampson HA, Berin MC, Nowak-Wegrzyn A. Humoral and cellular responses to casein in patients with food protein-induced enterocolitis to cow’s milk. J Allergy Clin Immunol. 2017;139(2):572–583. doi: 10.1016/j.jaci.2016.02.047.
    1. Ghesquiere B, Wong BW, Kuchnio A, Carmeliet P. Metabolism of stromal and immune cells in health and disease. Nature. 2014;511(7508):167–176. doi: 10.1038/nature13312.
    1. Mills EL, Kelly B, O’Neill LAJ. Mitochondria are the powerhouses of immunity. Nat Immunol. 2017;18(5):488–498. doi: 10.1038/ni.3704.
    1. Koletzko S, Niggemann B, Arato A, Dias JA, Heuschkel R, Husby S, Mearin ML, Papadopoulou A, Ruemmele FM, Staiano A, et al. Diagnostic approach and management of cow’s-milk protein allergy in infants and children: ESPGHAN GI Committee practical guidelines. J Pediatr Gastroenterol Nutr. 2012;55(2):221–229. doi: 10.1097/MPG.0b013e31825c9482.
    1. Bernard H, Creminon C, Yvon M, Wal JM. Specificity of the human IgE response to the different purified caseins in allergy to cow’s milk proteins. Int Arch Allergy Immunol. 1998;115(3):235–244. doi: 10.1159/000023906.
    1. Blanc F, Bernard H, Alessandri S, Bublin M, Paty E, Leung SA, Patient KA, Wal JM. Update on optimized purification and characterization of natural milk allergens. Mol Nutr Food Res. 2008;52(Suppl 2):S166–S175.
    1. Wal JM, Bernard H, Creminon C, Hamberger C, David B, Peltre G. Cow’s milk allergy: the humoral immune response to eight purified allergens. Adv Exp Med Biol. 1995;371B:879–881.
    1. Charcosset A. Assessment of IgE and IgG4 binding capacities of cow’s milk proteins selectively altered by proteases. J Agric Food Chem. 2016;64(17):3394–3404. doi: 10.1021/acs.jafc.5b01782.
    1. Bernard H, Paty E, Mondoulet L, Burks AW, Bannon GA, Wal JM, Scheinmann P. Serological characteristics of peanut allergy in children. Allergy. 2003;58(12):1285–1292. doi: 10.1046/j.1398-9995.2003.00300.x.
    1. Grassi J, Didierlaurent A, Stadler BM. Quantitative determination of total and specific human IgE with the use of monoclonal antibodies. J Allergy Clin Immunol. 1986;77(6):808–822. doi: 10.1016/0091-6749(86)90378-7.
    1. Pradelles P, Grassi J, Maclouf J. Enzyme immunoassays of eicosanoids using acetylcholine esterase as label: an alternative to radioimmunoassay. Anal Chem. 1985;57(7):1170–1173. doi: 10.1021/ac00284a003.
    1. Bernard H, Drumare MF, Guillon B, Paty E, Scheinmann P, Wal JM. Immunochemical characterisation of structure and allergenicity of peanut 2S albumins using different formats of immunoassays. Anal Bioanal Chem. 2009;395(1):139–146. doi: 10.1007/s00216-009-2842-5.
    1. Mandalari G, del-Patient K, Barkholt V, Baro C, Bennett L, Bublin M, Gaier S, Graser G, Ladics GS, Mierzejewska D, et al. In vitro digestibility of beta-casein and beta-lactoglobulin under simulated human gastric and duodenal conditions: a multi-laboratory evaluation. Regul Toxicol Pharmacol. 2009;55(3):372–381. doi: 10.1016/j.yrtph.2009.08.010.
    1. Guimaraes V, Drumare MF, Lereclus D, Gohar M, Lamourette P, Nevers MC, Vaisanen-Tunkelrott ML, Bernard H, Guillon B, Creminon C, et al. In vitro digestion of Cry1Ab proteins and analysis of the impact on their immunoreactivity. J Agric Food Chem. 2010;58(5):3222–3231. doi: 10.1021/jf903189j.
    1. Morita H, Nomura I, Orihara K, Yoshida K, Akasawa A, Tachimoto H, Ohtsuka Y, Namai Y, Futamura M, Shoda T, et al. Antigen-specific T-cell responses in patients with non-IgE-mediated gastrointestinal food allergy are predominantly skewed to T(H)2. J Allergy Clin Immunol. 2013;131(2):590–592. doi: 10.1016/j.jaci.2012.09.005.
    1. Becattini S, Latorre D, Mele F, Foglierini M, De GC, Cassotta A, Fernandez B, Kelderman S, Schumacher TN, Corti D, et al. T cell immunity. Functional heterogeneity of human memory CD4(+) T cell clones primed by pathogens or vaccines. Science. 2015;347(6220):400–406. doi: 10.1126/science.1260668.
    1. Boudah S, Olivier MF, ros-Calt S, Oliveira L, Fenaille F, Tabet JC, Junot C. Annotation of the human serum metabolome by coupling three liquid chromatography methods to high-resolution mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci. 2014;966:34–47. doi: 10.1016/j.jchromb.2014.04.025.
    1. Giacomoni F, Le CG, Monsoor M, Landi M, Pericard P, Petera M, Duperier C, Tremblay-Franco M, Martin JF, Jacob D, et al. Workflow4Metabolomics: a collaborative research infrastructure for computational metabolomics. Bioinformatics. 2015;31(9):1493–1495. doi: 10.1093/bioinformatics/btu813.
    1. Roux A, Xu Y, Heilier JF, Olivier MF, Ezan E, Tabet JC, Junot C. Annotation of the human adult urinary metabolome and metabolite identification using ultra high performance liquid chromatography coupled to a linear quadrupole ion trap-Orbitrap mass spectrometer. Anal Chem. 2012;84(15):6429–6437. doi: 10.1021/ac300829f.
    1. Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30. doi: 10.1093/nar/28.1.27.
    1. Wishart DS, Tzur D, Knox C, Eisner R, Guo AC, Young N, Cheng D, Jewell K, Arndt D, Sawhney S, et al. HMDB: the human metabolome database. Nucleic Acids Res. 2007;35(Database issue):D521–D526. doi: 10.1093/nar/gkl923.
    1. Smith CA, O’Maille G, Want EJ, Qin C, Trauger SA, Brandon TR, Custodio DE, Abagyan R, Siuzdak G. METLIN: a metabolite mass spectral database. Ther Drug Monit. 2005;27(6):747–751. doi: 10.1097/01.ftd.0000179845.53213.39.
    1. Sumner LW, Amberg A, Barrett D, Beale MH, Beger R, Daykin CA, Fan TW, Fiehn O, Goodacre R, Griffin JL, et al. Proposed minimum reporting standards for chemical analysis Chemical Analysis Working Group (CAWG) Metabolomics Standards Initiative (MSI) Metabolomics. 2007;3(3):211–221. doi: 10.1007/s11306-007-0082-2.
    1. Shek LP, Bardina L, Castro R, Sampson HA, Beyer K. Humoral and cellular responses to cow milk proteins in patients with milk-induced IgE-mediated and non-IgE-mediated disorders. Allergy. 2005;60(7):912–919. doi: 10.1111/j.1398-9995.2005.00705.x.
    1. Konstantinou GN, Bencharitiwong R, Grishin A, Caubet JC, Bardina L, Sicherer SH, Sampson HA, Nowak-Wegrzyn A. The role of casein-specific IgA and TGF-beta in children with food protein-induced enterocolitis syndrome to milk. Pediatr Allergy Immunol. 2014;25(7):651–656. doi: 10.1111/pai.12288.
    1. Powell GK. Milk- and soy-induced enterocolitis of infancy. Clinical features and standardization of challenge. J Pediatr. 1978;93(4):553–560. doi: 10.1016/S0022-3476(78)80887-7.
    1. Savilahti E. Immunochemical study of the malabsorption syndrome with cow’s milk intolerance. Gut. 1973;14(6):491–501. doi: 10.1136/gut.14.6.491.
    1. Wada T, Toma T, Muraoka M, Matsuda Y, Yachie A. Elevation of fecal eosinophil-derived neurotoxin in infants with food protein-induced enterocolitis syndrome. Pediatr Allergy Immunol. 2014;25(6):617–619.
    1. Pecora V, Prencipe G, Valluzzi R, Dahdah L, Insalaco A, Cianferoni A, De BF, Fiocchi A. Inflammatory events during food protein-induced enterocolitis syndrome reactions. Pediatr Allergy Immunol. 2017;28(5):464–470. doi: 10.1111/pai.12723.
    1. Gronke K, Kofoed-Nielsen M, Diefenbach A. Innate lymphoid cells, precursors and plasticity. Immunol Lett. 2016;179:9–18. doi: 10.1016/j.imlet.2016.07.004.
    1. Almeida L, Lochner M, Berod L, Sparwasser T. Metabolic pathways in T cell activation and lineage differentiation. Semin Immunol. 2016;28(5):514–524. doi: 10.1016/j.smim.2016.10.009.
    1. Berod L, Friedrich C, Nandan A, Freitag J, Hagemann S, Harmrolfs K, Sandouk A, Hesse C, Castro CN, Bahre H, et al. De novo fatty acid synthesis controls the fate between regulatory T and T helper 17 cells. Nat Med. 2014;20(11):1327–1333. doi: 10.1038/nm.3704.
    1. Lochner M, Berod L, Sparwasser T. Fatty acid metabolism in the regulation of T cell function. Trends Immunol. 2015;36(2):81–91. doi: 10.1016/j.it.2014.12.005.
    1. Rubio I, Grund S, Song SP, Biskup C, Bandemer S, Fricke M, Forster M, Graziani A, Wittig U, Kliche S. TCR-induced activation of Ras proceeds at the plasma membrane and requires palmitoylation of N-Ras. J Immunol. 2010;185(6):3536–3543. doi: 10.4049/jimmunol.1000334.
    1. Rampoldi F, Bonrouhi M, Boehm ME, Lehmann WD, Popovic ZV, Kaden S, Federico G, Brunk F, Grone HJ, Porubsky S. Immunosuppression and Aberrant T Cell Development in the Absence of N-Myristoylation. J Immunol. 2015;195(9):4228–4243. doi: 10.4049/jimmunol.1500622.

Source: PubMed

3
구독하다