Toward the assessment of food toxicity for celiac patients: characterization of monoclonal antibodies to a main immunogenic gluten peptide

Belén Morón, Michael T Bethune, Isabel Comino, Hamid Manyani, Marina Ferragud, Manuel Carlos López, Angel Cebolla, Chaitan Khosla, Carolina Sousa, Belén Morón, Michael T Bethune, Isabel Comino, Hamid Manyani, Marina Ferragud, Manuel Carlos López, Angel Cebolla, Chaitan Khosla, Carolina Sousa

Abstract

Background and aims: Celiac disease is a permanent intolerance to gluten prolamins from wheat, barley, rye and, in some patients, oats. Partially digested gluten peptides produced in the digestive tract cause inflammation of the small intestine. High throughput, immune-based assays using monoclonal antibodies specific for these immunotoxic peptides would facilitate their detection in food and enable monitoring of their enzymatic detoxification. Two monoclonal antibodies, G12 and A1, were developed against a highly immunotoxic 33-mer peptide. The potential of each antibody for quantifying food toxicity for celiac patients was studied.

Methods: Epitope preferences of G12 and A1 antibodies were determined by ELISA with gluten-derived peptide variants of recombinant, synthetic or enzymatic origin.

Results: The recognition sequences of G12 and A1 antibodies were hexameric and heptameric epitopes, respectively. Although G12 affinity for the 33-mer was superior to A1, the sensitivity for gluten detection was higher for A1. This observation correlated to the higher number of A1 epitopes found in prolamins than G12 epitopes. Activation of T cell from gluten digested by glutenases decreased equivalently to the detection of intact peptides by A1 antibody. Peptide recognition of A1 included gliadin peptides involved in the both the adaptive and innate immunological response in celiac disease.

Conclusions: The sensitivity and epitope preferences of the A1 antibody resulted to be useful to detect gluten relevant peptides to infer the potential toxicity of food for celiac patients as well as to monitor peptide modifications by transglutaminase 2 or glutenases.

Conflict of interest statement

Competing Interests: Ángel Cebolla is a founder and stockholder in Biomedal S.L., which is developing the moAbs described in this article to detect gluten in food. Marina Ferragud is a staff scientist in Biomedal S.L. The rest of the authors don't have a personal or financial conflict of interest.

Figures

Figure 1. Standard curve of the detection…
Figure 1. Standard curve of the detection of C-LYTAG-33-mer polypeptide (A) and Sigma gliadin (B) by indirect ELISA with use of moAbs G12 (black) and A1 (white).
Each point of the curve represents the mean±standard deviation of n = 4 assays. IC50 values of the moAbs to both antigens are indicated.
Figure 2. Comparative reactivity of prolamins from…
Figure 2. Comparative reactivity of prolamins from wheat, barley, rye, oats, corn and rice from indirect ELISAs using moAbs G12 (black) and A1 (white).
Each point of the curve shows the mean of n = 3 assays. IC50 and CR values of the moAbs to prolamins are indicated. N.A.: Not applicable.
Figure 3. Analysis of anti-33-mer moAbs recognition…
Figure 3. Analysis of anti-33-mer moAbs recognition regions in recombinant 33-mer peptide fragments expressed in E. coli.
A. Nucleotide sequences and the deduced amino acid sequences of the encoded peptide fusions to C-LYTAG. B. Detection of C-LYTAG-peptide fusions by an indirect ELISA with the use of moAbs G12, A1 and 6B5L1.
Figure 4. Relative affinity of moAb G12…
Figure 4. Relative affinity of moAb G12 for different peptide variants derived from its recognition region (QPQLPY).
A. Amino acid sequences of the peptides. The G12 recognition sequence in the 33-mer peptide is in bold face. IC50 and CR values of the moAb G12 to peptides are indicated. B. Competition assay measuring the affinity of the moAb G12 for the peptides. Two separate assays were performed with the antibody, each with three repetitions. C. Localization of the peptides in the α-gliadin (accession number: JQ1047), γ-secalin (accession number: ABO32294.1) and C-hordein (accession number: AAA92333.1) sequences. The same color code for labelling the peptides has been used in A, B and C.
Figure 5. Relative affinity of moAb A1…
Figure 5. Relative affinity of moAb A1 for different peptide variants derived from its recognition region (QLPYPQP).
A. Amino acid sequences of the peptides. The A1 recognition sequence in the 33-mer peptide is in red. B, C, D and E. Competition assay for detection of the affinity of the moAb A1 for the peptides and their localization in α-gliadin (B; accession number: JQ1047), γ-secalin (C; accession number: ABO32294.1), C-hordein (D; accession number: AAA92333.1) and avenin (E; accession number: AAA32716.1). Two separate assays were performed with the moAb, each with three repetitions. IC50 values of the moAb A1 to peptides are indicated. N.A.: Not applicable. The color code for labelling the peptides is the same as that used in A.
Figure 6. Relative affinity of moAb A1…
Figure 6. Relative affinity of moAb A1 for peptide variants of the 33-mer recognition region (QLPYPQP) featuring single amino acid sustitutions.
A. Amino acid sequences of the peptides. The A1 recognition sequence in the 33-mer peptide is in bold face. IC50 and CR values of the moAb A1 to peptides are indicated. B. Competition assay for the detection of the affinity of the moAb A1 for the peptides. Two separate assays were performed with the antibody, each with three repetitions. The color code for labelling the peptides is the same as that used in A.
Figure 7. Indirect competitive ELISA using moAb…
Figure 7. Indirect competitive ELISA using moAb A1 to test whole-wheat bread digests for 33-mer content.
A. Concentration of 33-mer (µg/mL) in whole-wheat bread digests containing 0.6 mg/mL pepsin supplemented with specified concentrations of recombinant proEP-B2 (U/mg gluten). B. Concentration of 33-mer (µg/mL) in whole-wheat bread digests containing 0.6 mg/mL pepsin and 32 U/mg EP-B2 supplemented with specified concentrations of recombinant SC PEP (U/mg gluten). The concentration of 33-mer in each digest was determined by comparison to a synthetic 33-mer standard curve. Two separate assays were performed with the antibody, each with three repetitions.
Figure 8. Relative affinity of the anti-33-mer…
Figure 8. Relative affinity of the anti-33-mer moAbs for different peptides involved in the immune responses to gluten in CD patients.
A. Competition assay for detection of the affinity of G12 and A1 moAbs for a peptide containing recognition common regions (QPQLPYPQP) to the two moAbs and its deamidated counterpart (QPELPYPQP). IC50 and CR values of the anti-33-mer moAbs to peptides are indicated. B. Competition assay for detection of the affinity of the moAbs G12 and A1 for the innate peptide, p31-49. The 33-mer and the p31-49 peptide sequences are underlined and in bold face, respectively. Wheat alpha-gliadin (α-gliadin, accession number: JQ1047). Two separate assays were performed with the antibodies, each with three repetitions (A and B).

References

    1. Alaedini A, Green PHR. Narrative review: celiac disease: understanding a complex autoimmune disorder. Ann Int Med. 2005;142:289–299.
    1. Robins G, Howdle PD. Advances in celiac disease. Curr Opin Gastroenterol. 2005;21:152–161.
    1. Logan RF, Tucker G, Rifkind EA, Heading RC, Ferguson A. Changes in clinical features of coeliac disease in adults in Edinburgh and the Lothians 1960-79. BMJ. 1983;286:95–97.
    1. Fasano A, Catassi C. Current approaches to diagnosis and treatment of celiac disease: an evolving spectrum. Gastroenterology. 2001;120:636–651.
    1. Kumar V, Rajadhyaksha M, Wortsman J. Celiac disease associated autoimmune endocrinopathies. Clin Diagn Lab Immunol. 2001;8:678–685.
    1. Arentz-Hansen H, Fleckenstein B, Molberg φ, Scott H, Koning F, et al. The molecular basis for oat intolerance in patients with celiac disease. PLoS Medic. 2004;1:84–92.
    1. Kagnoff MF. Celiac disease: pathogenesis of a model immunogenetic disease. J Clin Invest. 2007;117:41–49.
    1. Wieser H. The precipitating factor in Celiac-Disease. Baillieres Clin Gastroenterol. 1995;9:191–207.
    1. Molberg φ, McAdam SN, Korner R, Quarsten H, Kristiansen C, et al. Tissue transglutaminase selectively modifies gliadin peptides that are recognized by gut-derived T cells in celiac disease. Nat Med. 1998;4:713–717.
    1. van de Wal Y, Kooy Y, van Veelen P, Peña S, Mearin L, et al. Selective deamidation by tissue transglutaminase strongly enhances gliadin-specific T cell-reactivity. J Immunol. 1998;161:1585–1588.
    1. Anderson RP, Degano P, Godkin AJ, Jewell DP, Hill AVS. In vivo antigen challenge in celiac disease identifies a single transglutaminase–modified peptide as the dominant A-gliadin T-cell epitope. Nat Med. 2000;6:337–342.
    1. Arentz-Hansen H, Korner R, Molberg φ, 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. Stern M, Ciclitira PJ, van Eckert R, Feighery C, Janssen FW, et al. Analysis and clinical effects of gluten in coeliac disease. Eur J Gastroenterol Hepatol. 2001;13:741–747.
    1. Shan L, Molberg φ, Parrot I, Hausch F, Filiz F, et al. Structural basis for gluten intolerance in celiac sprue. Science. 2002;297:2275–2279.
    1. Shan L, Marti T, Sollid LM, Gray GM, Khosla C. Comparative biochemical analysis of three bacterial prolyl endopeptidases: implications for coeliac sprue. Biochem J. 2004;383:311–318.
    1. Sollid LM, Khosla C. Future therapeutic options for celiac disease. Nat Clin Pract Gastroenterol Hepatol. 2005;2:140–147.
    1. Bethune MT, Strop P, Tang Y, Sollid LM, Khosla C. Heterologous expression, purification, refolding, and structural-functional, characterization of EP-B2, a self-activating barley cysteine endoprotease. Chem Biol. 2006;13:637–647.
    1. Gass J, Vora H, Bethune MT, Gray GM, Khosla C. Effect of barley endoprotease EB-P2 on gluten digestion in the intact rat. J Pharmacol Exp Ther. 2006;318:1178–1186.
    1. Siegel M, Bethune MT, Gass J, Ehren J, Xia J, et al. Rational design of combination enzyme therapy for celiac sprue. Chem Biol. 2006;13:649–658.
    1. Gass J, Bethune MT, Siegel M, Spencer A, Khosla C. Combination enzyme therapy for gastric digestion of dietary gluten in celiac sprue patients. Gastroenterology. 2007;133:472–480.
    1. Collin P, Thorell L, Kaukien K, Mäki M. Aliment Pharmacol Ther. 2004;19:1277–1283.
    1. Khosla C, Gray GM, Sollid LM. Putative efficacy and dosage of prolyl endopeptidase for digesting and detoxifying gliadin peptides. Gastroenterology. 2005;129:1362–1363.
    1. Morón B, Cebolla A, Manyani H, Álvarez-Maqueda M, Megías M, et al. Sensitive detection of cereal fractions that are toxic to celiac disease patients by using monoclonal antibodies to a main immunogenic wheat peptide. Am J Clin Nutr. 2008;87:405–414.
    1. Shewry PR, Tatham AS, Kasarda DD. Cereal proteins and coeliac disease. In: Marsch M, editor. Coeliac disease. Oxford: Blackwell Scientific Publications; 1992. pp. 305–348.
    1. Ellis HJ, Freedman AR, Ciclitira PJ. Detection and estimation of the barley prolamin content of beer and malt to assess their suitability for patients with coeliac disease. Clin Chim Acta. 1990;189:123–130.
    1. Ellis HJ, Doyle AP, Day P, Wieser H, Ciclitira PJ. Demostration of the presence of coeliac-activating gliadin-like epitopes in malted barley. Int Arch Allergy Inmunol. 1994;104:308–310.
    1. Stepniak D, Koning F. Celiac disease-sandwiched between innate and adaptive immunity. Human Immunol. 2006;67:460–468.
    1. Cerf-Bensussan N, Matysiak-Budnik T, Cellier C, Heyman M. Oral proteases: a new approach to managing coeliac disease. Gut. 2007;56:157–160.
    1. Skovbjerg H, Koch C, Anthonsen D, Sjöström H. Deamidation and cross-linking of gliadin peptides by transglutaminases and the relation to celiac disease. Biochim Biophys Acta. 2004;1690:220–230.
    1. Fehniger TA, Caligiuri MA. Interleukin 15: biology and relevance to human disease. Blood. 2001;97:14–32.
    1. Bernardo D, Garrote JA, Fernández-Salazar L, Riestra S, Arranz E. Is gliadin really safe for non-celiac individuals? Production of interleukin 15 in biopsy culture from non-celiac individuals challenged with gliadin peptides. Gut. 2007;56:889–890.
    1. Valdés I, García E, Llorente M, Méndez E. Innovative approach to low-level gluten determination in foods using a novel sandwich enzyme-linked immunosorbent assay protocol. Eur J Gastroenterol Hepatol. 2003;15:465–474.
    1. Spaenij-Dekking EHA, Kooy-Winkelaar EMC, Nieuwenhuizen WF, Drijfhout JW, Koning F. A novel and sensitive method for the delection of T cell stimulatory epitopes of α/β- and γ-gliadin. Gut. 2004;53:1267–1273.
    1. Gianfrani C, Siciliano RA, Facchiano AM, Camarca A, Mazzeo MF, et al. Transamidation of wheat flour inhibits the response to gliadin of intestinal T cells in celiac disease. Gastroenterology. 2007;133:780–789.
    1. Tepnel Biosystems. Gluten assay kit, for the quantitative determination of gluten in food products by enzyme immunoassay. Tepnel Biosystems, UK.
    1. Hill ID, Dirks MH, Liptak GS, Colleti RB, Fasano A, et al. Guideline for the diagnosis and treatment of celiac disease in children: recommendations of the North American Society for Pediatric Gastroenterology, Hepatology and Nutrition. J Ped Gastroenterol Nutr. 2005;40:1–19.
    1. Catassi C, Fabiani E, Iacono G, D'Agate C, Francavilla R, et al. A prospective, double-bind, placebo-controlled trial to establish a safe gluten threshold for patients with celiac disease. Am J Clin Nutr. 2007;85:160–166.
    1. Xia J, Sollid LM, Khosla C. Equilibrium and kinetic analysis of the unusual binding behavior of a highly immunogenic gluten peptide to HLA-DQ2. Biochem. 2005;44:4442–4449.
    1. Vora H, McIntire J, Kumar P, Deshpande M, Khosla C. A scaleable manufacturing process for pro-EP-B2, a cysteine protease from barley indicated for celiac sprue. Biotechnol Bioeng. 2007;98:177–185.
    1. Cebolla A, Sousa C, de Lorenzo V. Rational design of a bacterial transcriptional cascade for amplifying gene expression capacity. Nucl Acids Res. 2001;29:759–766.
    1. Cebolla A, Royo JL, de Lorenzo V, Santero E. Improvement of recombinant protein yield by a combination of transcriptional amplification and stabilization of gene expression. App Environ Microbiol. 2002;68:5034–5041.

Source: PubMed

3
Prenumerera