Skip to main content
Top
Published in: Clinical and Translational Allergy 1/2018

Open Access 01-12-2018 | Research

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

Authors: 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

Published in: Clinical and Translational Allergy | Issue 1/2018

Login to get access

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.
Appendix
Available only for authorised users
Literature
1.
go back to reference Nowak-Wegrzyn A, Katz Y, Mehr SS, Koletzko S. Non-IgE-mediated gastrointestinal food allergy. J Allergy Clin Immunol. 2015;135(5):1114–24.CrossRefPubMed Nowak-Wegrzyn A, Katz Y, Mehr SS, Koletzko S. Non-IgE-mediated gastrointestinal food allergy. J Allergy Clin Immunol. 2015;135(5):1114–24.CrossRefPubMed
2.
go back to reference Nowak-Wegrzyn A, Jarocka-Cyrta E, Moschione CA. Food protein-induced enterocolitis syndrome. J Investig Allergol Clin Immunol. 2017;27(1):1–18.CrossRefPubMed Nowak-Wegrzyn A, Jarocka-Cyrta E, Moschione CA. Food protein-induced enterocolitis syndrome. J Investig Allergol Clin Immunol. 2017;27(1):1–18.CrossRefPubMed
3.
go back to reference 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.CrossRefPubMed 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.CrossRefPubMed
4.
go back to reference 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):647–53.CrossRefPubMed 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):647–53.CrossRefPubMed
5.
go back to reference 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):382–9.CrossRefPubMed 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):382–9.CrossRefPubMed
7.
go back to reference Feuille E, Nowak-Wegrzyn A. Food protein-induced enterocolitis syndrome, allergic proctocolitis, and enteropathy. Curr Allergy Asthma Rep. 2015;15(8):50.CrossRefPubMed Feuille E, Nowak-Wegrzyn A. Food protein-induced enterocolitis syndrome, allergic proctocolitis, and enteropathy. Curr Allergy Asthma Rep. 2015;15(8):50.CrossRefPubMed
9.
go back to reference 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):1885–96.CrossRefPubMedPubMedCentral 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):1885–96.CrossRefPubMedPubMedCentral
10.
go back to reference 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):572–83.CrossRefPubMed 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):572–83.CrossRefPubMed
11.
go back to reference Ghesquiere B, Wong BW, Kuchnio A, Carmeliet P. Metabolism of stromal and immune cells in health and disease. Nature. 2014;511(7508):167–76.CrossRefPubMed Ghesquiere B, Wong BW, Kuchnio A, Carmeliet P. Metabolism of stromal and immune cells in health and disease. Nature. 2014;511(7508):167–76.CrossRefPubMed
12.
go back to reference Mills EL, Kelly B, O’Neill LAJ. Mitochondria are the powerhouses of immunity. Nat Immunol. 2017;18(5):488–98.CrossRefPubMed Mills EL, Kelly B, O’Neill LAJ. Mitochondria are the powerhouses of immunity. Nat Immunol. 2017;18(5):488–98.CrossRefPubMed
13.
go back to reference 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):221–9.CrossRefPubMed 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):221–9.CrossRefPubMed
14.
go back to reference 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):235–44.CrossRefPubMed 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):235–44.CrossRefPubMed
15.
go back to reference 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):S166–75.PubMed 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):S166–75.PubMed
16.
go back to reference 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:879–81.PubMed 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:879–81.PubMed
17.
go back to reference 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):3394–404.CrossRefPubMed 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):3394–404.CrossRefPubMed
18.
go back to reference 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):1285–92.CrossRefPubMed 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):1285–92.CrossRefPubMed
19.
go back to reference 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):808–22.CrossRefPubMed 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):808–22.CrossRefPubMed
20.
go back to reference Pradelles P, Grassi J, Maclouf J. Enzyme immunoassays of eicosanoids using acetylcholine esterase as label: an alternative to radioimmunoassay. Anal Chem. 1985;57(7):1170–3.CrossRefPubMed Pradelles P, Grassi J, Maclouf J. Enzyme immunoassays of eicosanoids using acetylcholine esterase as label: an alternative to radioimmunoassay. Anal Chem. 1985;57(7):1170–3.CrossRefPubMed
21.
go back to reference 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):139–46.CrossRefPubMed 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):139–46.CrossRefPubMed
22.
go back to reference 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):372–81.CrossRefPubMed 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):372–81.CrossRefPubMed
23.
go back to reference 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):3222–31.CrossRefPubMed 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):3222–31.CrossRefPubMed
24.
go back to reference 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):590–2.CrossRefPubMed 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):590–2.CrossRefPubMed
25.
go back to reference 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):400–6.CrossRefPubMed 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):400–6.CrossRefPubMed
26.
go back to reference 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.CrossRefPubMed 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.CrossRefPubMed
27.
go back to reference 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):1493–5.CrossRefPubMed 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):1493–5.CrossRefPubMed
28.
go back to reference 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):6429–37.CrossRefPubMed 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):6429–37.CrossRefPubMed
30.
go back to reference 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):D521–6.CrossRefPubMedPubMedCentral 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):D521–6.CrossRefPubMedPubMedCentral
31.
go back to reference 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):747–51.CrossRefPubMed 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):747–51.CrossRefPubMed
32.
go back to reference 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):211–21.CrossRefPubMedPubMedCentral 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):211–21.CrossRefPubMedPubMedCentral
33.
go back to reference 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):912–9.CrossRefPubMed 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):912–9.CrossRefPubMed
34.
go back to reference 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):651–6.CrossRefPubMedPubMedCentral 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):651–6.CrossRefPubMedPubMedCentral
35.
go back to reference Powell GK. Milk- and soy-induced enterocolitis of infancy. Clinical features and standardization of challenge. J Pediatr. 1978;93(4):553–60.CrossRefPubMed Powell GK. Milk- and soy-induced enterocolitis of infancy. Clinical features and standardization of challenge. J Pediatr. 1978;93(4):553–60.CrossRefPubMed
37.
go back to reference 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):617–9.PubMed 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):617–9.PubMed
38.
go back to reference 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):464–70.CrossRefPubMed 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):464–70.CrossRefPubMed
39.
go back to reference Gronke K, Kofoed-Nielsen M, Diefenbach A. Innate lymphoid cells, precursors and plasticity. Immunol Lett. 2016;179:9–18.CrossRefPubMed Gronke K, Kofoed-Nielsen M, Diefenbach A. Innate lymphoid cells, precursors and plasticity. Immunol Lett. 2016;179:9–18.CrossRefPubMed
40.
go back to reference Almeida L, Lochner M, Berod L, Sparwasser T. Metabolic pathways in T cell activation and lineage differentiation. Semin Immunol. 2016;28(5):514–24.CrossRefPubMed Almeida L, Lochner M, Berod L, Sparwasser T. Metabolic pathways in T cell activation and lineage differentiation. Semin Immunol. 2016;28(5):514–24.CrossRefPubMed
41.
go back to reference 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):1327–33.CrossRefPubMed 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):1327–33.CrossRefPubMed
42.
go back to reference Lochner M, Berod L, Sparwasser T. Fatty acid metabolism in the regulation of T cell function. Trends Immunol. 2015;36(2):81–91.CrossRefPubMed Lochner M, Berod L, Sparwasser T. Fatty acid metabolism in the regulation of T cell function. Trends Immunol. 2015;36(2):81–91.CrossRefPubMed
43.
go back to reference 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):3536–43.CrossRefPubMed 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):3536–43.CrossRefPubMed
44.
go back to reference 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):4228–43.CrossRefPubMed 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):4228–43.CrossRefPubMed
Metadata
Title
Deep analysis of immune response and metabolic signature in children with food protein induced enterocolitis to cow’s milk
Authors
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
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Clinical and Translational Allergy / Issue 1/2018
Electronic ISSN: 2045-7022
DOI
https://doi.org/10.1186/s13601-018-0224-9

Other articles of this Issue 1/2018

Clinical and Translational Allergy 1/2018 Go to the issue