19 Detection and Control of Gluten as a Food Allergen 2015 Handbook of Food Allergen Detection and...

11
Handbook of Food Allergen Detection and Control. http://dx.doi.org/10.1533/9781782420217.3.367 Copyright © 2015 Elsevier Ltd. All rights reserved. Detection and control of gluten as a food allergen I. Nasr 1 , J. Messing 2 , I. H. Nasr 3 , P. J. Ciclitira 4 1 Guys and St Thomas’ NHS Trust, London, UK; 2 Rutgers University, Piscataway, NJ, USA; 3 Barts Health NHS Trust, London, UK; 4 King’s College London, London, UK 19 19.1 Introduction Celiac disease (CD) is a chronic, systemic, autoimmune disorder in genetically predisposed individuals. Gluten proteins and related prolamins found in wheat, barley, and rye trigger an autoimmune injury to the gut, skin, liver, joints, uterus, and other organs. 1 In the gut, the resulting injury leads to atrophy of the mucosal villous lining. The villous atrophy is identified on small bowel biopsy, which is considered the gold standard for diagnosing this condition. False negative small bowel histology can be expected due to patchy small bowel mucosal changes. In the past, the classical presentation of CD was symptomatic of gastrointestinal disorders including diarrhoea and weight loss. Since the 1980s, fewer patients have presented with symptomatic CD and there has been a significant shift toward more patients presenting as asymptomatic adults detected at screening. 2 CD affects on average 1 % of the population. Milder clinical features and use of serology have led to an increased detection rate. 3 The age of presentation varies. It can affect children and elderly alike. The peak age of presentation is in childhood as the presenting symptoms are usually malabsorption and failure to thrive. The other peak is in women of childbearing age who present with anemia. The third peak is in the fourth to fifth decade who present with osteoporosis. Untreated CD is associated with significant mortality. Undiagnosed CD is associated with a nearly four-fold increased risk of death. 4 With increased disease awareness, newer trends for detection of the disease are important. 19.1.1 Etiology of celiac disease (CD) Celiac disease (CD) is an intestinal disorder with multifactorial etiology. Genetic and environmental factors are recognized to be involved in the process. This complex interplay between intrinsic and extrinsic factors could explain the range of clinical presentation from asymptomatic individuals to severe malabsorption. 5 HLA and non-HLA genes together with gluten and possibly additional environmental factors predispose to CD. 6 There is a high prevalence of CD among first-degree relatives of celiac patients. 7 It has been reported that high frequency of rotavirus infections may

description

glutenul din cereale

Transcript of 19 Detection and Control of Gluten as a Food Allergen 2015 Handbook of Food Allergen Detection and...

  • Handbook of Food Allergen Detection and Control. http://dx.doi.org/10.1533/9781782420217.3.367Copyright 2015 Elsevier Ltd. All rights reserved.

    Detection and control of gluten as a food allergen I. Nasr 1 , J. Messing 2 , I. H. Nasr 3 , P. J. Ciclitira 4 1 Guys and St Thomas NHS Trust , London, UK ; 2 Rutgers University , Piscataway, NJ, USA ; 3 Barts Health NHS Trust , London, UK ; 4 King s College London , London, UK

    19

    19.1 Introduction

    Celiac disease (CD) is a chronic, systemic, autoimmune disorder in genetically predisposed individuals. Gluten proteins and related prolamins found in wheat, barley, and rye trigger an autoimmune injury to the gut, skin, liver, joints, uterus, and other organs. 1 In the gut, the resulting injury leads to atrophy of the mucosal villous lining. The villous atrophy is identifi ed on small bowel biopsy, which is considered the gold standard for diagnosing this condition. False negative small bowel histology can be expected due to patchy small bowel mucosal changes.

    In the past, the classical presentation of CD was symptomatic of gastrointestinal disorders including diarrhoea and weight loss. Since the 1980s, fewer patients have presented with symptomatic CD and there has been a signifi cant shift toward more patients presenting as asymptomatic adults detected at screening. 2 CD affects on average 1 % of the population. Milder clinical features and use of serology have led to an increased detection rate. 3 The age of presentation varies. It can affect children and elderly alike. The peak age of presentation is in childhood as the presenting symptoms are usually malabsorption and failure to thrive. The other peak is in women of childbearing age who present with anemia. The third peak is in the fourth to fi fth decade who present with osteoporosis. Untreated CD is associated with signifi cant mortality. Undiagnosed CD is associated with a nearly four-fold increased risk of death. 4 With increased disease awareness, newer trends for detection of the disease are important.

    19.1.1 Etiology of celiac disease (CD)

    Celiac disease (CD) is an intestinal disorder with multifactorial etiology. Genetic and environmental factors are recognized to be involved in the process. This complex interplay between intrinsic and extrinsic factors could explain the range of clinical presentation from asymptomatic individuals to severe malabsorption. 5 HLA and non-HLA genes together with gluten and possibly additional environmental factors predispose to CD. 6 There is a high prevalence of CD among fi rst-degree relatives of celiac patients. 7 It has been reported that high frequency of rotavirus infections may

  • 368 Handbook of Food Allergen Detection and Control

    increase the risk of CD autoimmunity in childhood in genetically predisposed individuals. 8 CD, like type 1 diabetes, rheumatoid arthritis, and multiple sclerosis, has a chronic nature where particular HLA alleles are over-represented among the patients. 9 Patients go into complete remission when they are put on a gluten-free diet, and they relapse when gluten is reintroduced into the diet. CD is in this respect unique among the chronic infl ammatory HLA-associated diseases in that a critical environmental factor has been identifi ed. 6

    19.1.2 Gluten toxicity

    Gluten toxicity in CD indicates a small bowel infl ammatory response. CD is confi rmed when the intestinal epithelium demonstrates villous atrophy, blunting or at least an epithelial infi ltrate of T-cells. Other signs of toxicity may be exhibited outside the intestinal territory, such as the characteristic skin rash dermatitis herpetiformis. The toxicity effects on the small bowel epithelium are potentially reversible once toxic gluten is eliminated from the diet resulting in better nutrient absorption and preventing complications such as osteoporosis 10 and small bowel lymphoma. 11

    19.1.3 Classifi cation of cereal proteins involved

    Wheat grains contain the outer husk (bran), the germ and the endosperm (white fl our), which accounts for approximately 70 % of the whole grain and contains the toxic components. 12 The storage proteins of cereals constitute the ethanol soluble fraction, known as prolamin because of preponderance of proline and glutamine. 13 Prolamin is known as gluten for wheat, secalins for rye, hordeins for barley, avenins for oats and zeins for maize. Gluten protein is divided according to solubility into gliadins, which are soluble in 4090 % ethanol, and glutenins, which are insoluble. Each wheat variety expresses a large number of related proteins through repeated gene duplications and mutations making biochemical studies and toxicity mapping very diffi cult. 14 All four gliadins sub-fractions ( , , and u) were shown to exacerbate CD both in vitro and in vivo . 15 Several recent studies have indicated that glutenins may be involved in the CD disease process. 1619

    19.2 Pathogenesis of celiac disease (CD)

    CD is strongly associated with human leukocyte antigen (HLA) class II gene (DR3, DR5/DR7 or DR4) known as HLA-DQ2 and HLA-DQ8 located on chromosome 6p21. 20 It is associated to a larger extent with class II HLA-DQ2 (haplotypes DR-17 or DR5/7) and, to a lesser extent, HLA-DQ8 (haplotype DR-4). 21 These haplotypes are located in antigen-presenting cells of the mucosa. Approximately 95 % of the individuals with CD express HLA-DQ2 heterodimer and the remainder express HLA-DQ8. 22 The absence of these molecules has a negative predictive value for CD close to 100 %. 23

  • Detection and control of gluten as a food allergen 369

    19.2.1 Adaptive immune response

    Individuals with CD, in contrast to the healthy population, exhibit a T-cell response to gluten in the small intestine, which is restricted to either HLA-DQ2 or -DQ8. 24 Gluten is the single major environmental factor that triggers CD. The requirement for DQ2 or DQ8 is a major factor in the genetic predisposition to CD. However, most people who demonstrate a positive DQ2 or DQ8 do not develop CD despite daily exposure to gluten in their diet. This corroborates the fact that additional environmental or genetic factors are required for loss of immune tolerance to dietary gluten. HLA-DQ2 or HLA-DQ8 is required to present gluten peptides to the gluten-specifi c T-cells that are present in the gut of CD sufferers. As a result, antigen-specifi c T-cell response is activated. 25 The enzyme tissue transglutaminase (tTG) has been identifi ed as the autoantigen against which the abnormal immune response is directed.

    In CD, gliadin peptides resist human proteases and remain intact in the intestinal lumen and have an increased content of proline and glutamine residues. 2628 These peptides gain access to the lamina propria probably due to faulty tight junctions or endothelial cell transcytosis. 29,30 Gluten peptides are de-aminated in the submucosa by tissue transglutaminase (tTG). This creates a high-affi nity binding between these peptides and either HLA-DQ2 or HLA-DQ8 that is in the antigen-presenting cells and also activates helper T-cells. 31 A gliadin 33-mer peptide has several characteristics, suggesting it is the primary initiator of the infl ammatory response to gluten in celiac patients. 32 It stimulates interleukin-15 by dendritic cells and macrophages, which then stimulate intra-epithelial lymphocytes leading to epithelial damage. 33,34

    19.2.2 Innate immune response

    The innate immune response has an important role in the pathogenesis of CD and in some of its related complications. In particular, an increase in the number of intraepithelial lymphocytes (IELs) in the mucosa of the small intestine is a characteristic feature of CD, and those cells are likely to be important for the on-going pathogenesis of CD. 3539 In vitro studies of intestinal organ cultures, primary antigen-presenting cells, and epithelial and monocytic cell lines support the concept that gluten affects the innate immune system. 40 The IELs have a signifi cant role in the pathogenesis of CD complications, in particular refractory CD and the development of enteropathy-associated T-cell lymphoma (EATL). 4143 The exposure to toxic gluten in CD results in activation of / T-cell IELs. 14 Various mechanisms including the release of tumour necrosis factor (TNF- ), 44 interferon (IFN- ), 4547 and Fas ligand (FasL) system 48 are involved in mediating mucosal damage in T-cell-dependent intestinal diseases. Natural killer group 2, member D/major histocompatibility complex class I chain-related gene A (NKG2D/MICA) pathway, 27 perform with subsequent granzyme induction 49 and matrix metalloproteinase tissue remodeling 50 may also contribute. Interleukin (IL)15 also has a vital role the innate immune response. Upregulation of IL-15 expression by epithelial cells or macrophages increases MICA ligand expression on epithelial cells to facilitate the NKG2D receptor signaling pathway. 27,37

  • 370 Handbook of Food Allergen Detection and Control

    19.3 Testing of gluten toxicity

    Due to the complex heterogenicity of gluten, it is quite challenging to have a gold standard for testing. Immunological techniques such as enzyme-linked immunosorbent assay (ELISA) offer a sensitive and simple way of detecting antibodies against whole gluten digest 51 or a single peptide. 52 These techniques may miss other toxic cereals depending on the antibodies used. In addition, T cell epitopes are known to participate in disease mechanism which can be missed because antiserum recognizes B cell epitomes. Spaenij-Dekking has raised monoclonal antibodies to the proposed immunodominant epitope, in order to address this problem. 53 In addition, the majority of the immunological methods available use antibodies directed at the gliadins and miss some of the glutenins proteins that are now being implicated in disease pathogenesis 16,19 and will need to be considered when selecting reagents for measurement of gluten.

    19.3.1 In vitro testing

    Biopsies obtained from patients with CD are tested for the response to gluten toxicity. The small intestinal epithelium morphology from the untreated patients improves when their small intestinal biopsies are placed in an organ culture chamber for up to 24 hours in the absence of gluten. 54 This improvement can be altered by the addition of gluten. 55 Peptides can be tested in organ culture for evaluation of toxicity in CD; however, clinical in vivo testing is mandatory to confi rm the toxicity of these. 14

    19.3.2 In vivo testing

    A baseline small bowel epithelial biopsy is assessed for villus height to crypt depth (VH:CD), enterocyte height (ECH) and IEL count. A gluten challenge is introduced in vivo , usually with bread over a four-week period, may be indicated when there is doubt regarding the diagnosis.

    19.4 Current EU labeling and codex guidelines

    Tolerability of gluten varies amongst individuals. Western European dietary products labeled as gluten-free accept some amount of gluten. This is usually to improve baking qualities and palatability. Patients with CD generally show good histological and clinical response despite consuming such a wheat starch-containing diet. 56,57 However, in other parts of the world, such as Australia and the North American continent, a gluten-free diet should contain no gluten and a naturally gluten-free diet is used instead. If some gluten is included in the product, then foods are labeled as low gluten. 58 In 2012, the new European regulation for labeling products in relation to the gluten content took effect. The gluten content threshold was set in accordance

  • Detection and control of gluten as a food allergen 371

    to the Codex Alimentarius. Regulation (EC) No 41/2009 applies to all foodstuffs with the exception of infant and follow-on formulae.

    The WHO Codex Alimentarius standard proposes a gluten limit of 0.02 % (200 ppm) for gluten-free food and 0.002 % (20 ppm) for naturally gluten-free food. The Regulation (EC) 41/2009 states that gluten-free products should contain less than 20 mg/kg of gluten in the fi nished product. Very low gluten-containing products should contain less than 100 mg/kg of gluten in the fi nished product. The difference in the acceptable levels refl ects the fact that the exact tolerable limit for gluten is unknown. 59 Catassi et al . showed that less than 50 mg gluten per day is safe; however, some individuals relapsed when ingesting gluten at a much lower dose. 60 The safe maximum level of gluten remains debatable. The normal average intake of gluten by healthy individuals is approximately 13 g per day. 61

    19.5 Methods for measurement of gluten contamination in foods

    Gluten contamination in gluten-free products cannot be absolutely avoided and the safe threshold for gluten remains debatable. One way of detecting the toxic gluten epitopes is through ELISA, a method which uses monoclonal antibodies (MAB) specifi c for the immunotoxic peptides present in food. It is highly sensitive, specifi c and reproducible. The experience with this, however, depends on the monoclonal antibody used and whether it targets a single peptide or the whole protein. Some commercially available kits use antibodies against u-gliadin, which recognizes wheat and rye but not barley. 62 Valdes et al . 63 used an antibody that was able to target wheat, barley and rye prolamins. In this study, they were able to quantify levels as low as 1.56 ppm gliadins and 3.2 ppm gluten. Other kits can detect gliadins, hordeins and secalins to the same extent using a combination of three MABs. 64 Another issue is the fact that MABs target B-cell epitopes. As CD is a result of infl ammatory T-cell response, Spaenij-Dekking et al . 53 developed antisera that are able to detect T-cell eptitopes of the - and -gliadin. Gluten quantifi cation may not be completely accurate as the methods used rely on what is targeted and which toxic epitope is present in the food tested. Methods to test glutenins will also need to be established.

    19.5.1 Malted cereals

    Despite breaking down the cereals, there is no evidence that toxic gluten is disintegrated. It is known now that certain, signifi cantly cell-reactive areas of gliadin are highly resistant to enzymatic degradation. 26,28 Gliadin-like epitopes have been found in beer and malt. 65,66 Experts describe encountering patients with CD who are symptomatic following consumption of large amounts of malted products. Therefore, the question on whether malted breakfast cereals are consumed should be considered when dealing with patients with CD, who are symptomatic despite consuming a gluten-free diet.

  • 372 Handbook of Food Allergen Detection and Control

    19.5.2 Communion wafers

    Wheat fl our is used for the making of these wafers. It has been demonstrated that, in addition to an otherwise gluten-free diet, consumption of a milligram of gluten from wheat fl our-based wafers leads to failed mucosal recovery after 18 months challenge. 67 Gluten-free communion wafers containing Codex wheat starch are commercially available. A list of products and suppliers is available on Coeliac UK website.

    19.5.3 Oats

    Early feeding experiments suggested that oats are toxic to individuals with CD and have been excluded from the gluten-free diet. The possible harmful effect of oats has been revised and subsequently studied. Janatuinen et al . 68 in a Finnish randomized trial of 92 patients with CD compared the effects of gluten-free diets with and without oats. Eleven patients withdrew from the study. Duodenal biopsies revealed no effect on the mucosa in either group. Serological normalization was not delayed. The 35 patties from the oats study groups were re-evaluated after fi ve years. 69 Twenty-three of them were consuming oats regularly with no harmful effects detected. Other smaller studies were conducted. Some concluded that oats are safe and well tolerated. 70,71 Other published data suggest the possibility that a small group of individuals with CD is intolerant to oats. 72,73

    It is diffi cult to conclude whether oats are safe to use in CD. The studies show that intolerance to oats is not very frequent, but toxicity can still occur. Some patients withdrew from some of the studies, which raises the question of intolerance to oats. Patients with CD eating oats were found to have more abdominal complaints than those not eating oats. 74 Commercially available oats are frequently contaminated with cereals containing toxic gluten and therefore are not suitable to be included as part of a strict gluten-free diet. 75

    19.6 Future trends and summary

    The standard methods for diagnosing CD included serology testing, histological confi rmation and identifying the HLA status. In recent years, studies have looked into the use of a fi nger prick-based kit for self-testing of fi rst- and second-degree relatives of CD patients. 76 However, so far the gold standard for diagnosing CD is by histological analysis of the small bowel biopsy. Further research is needed to identify an alternative reliable method which is easily accessible to all.

    CD is a gluten-sensitive small intestinal enteropathy. Treatments involve a gluten-free diet omitting wheat, rye, barley and possibly oats. In vitro and in vivo testing of sequenced gluten peptides have identifi ed a variety of cereal epitopes that exacerbate the condition. Debate continues regarding the maximum permitted amount of gluten that patients with CD can safely tolerate. Research is required to increase our knowledge of the pathogenesis of the condition and to improve therapeutic options.

  • Detection and control of gluten as a food allergen 373

    References

    1. Rewers MJ . Epidemiology of Celiac Disease: What Are the Prevalence, Incidence, and Progression of Celiac Disease ? National Institutes of Health , Bethesda, MD , 2004 .

    2. Rampertab SD , Pooran N , Brar P , Singh P and Green PHR . Trends in the presentation of celiac disease . Am J Med , 355.e9 355.e14 , 2006 .

    3. Murray JA , Van Dyke C , Plevak MF , Dierkhising RA , Zinsmeister AR and Melton LJ 3rd. Trends in the identifi cation and clinical features of celiac disease in a North American community, 19502001 . Clin Gastroenterol Hepatol , 1 ( 1 ), 19 27 , 2003 .

    4. Rubio-Tapia A , Kyle RA , Kaplan EL , Johnson DR , Page W , Erdtmann F , Brantner TL , Kim WR , Phelps TK , Lahr BD , Zinsmeister AR , Melton LJ and Murray JA . Increased prevalence and mortality in undiagnosed celiac disease . Gastroenterology , 137 ( 1 ), 88 93 , 2009 .

    5. Schuppan D . Current concepts of celiac disease pathogenesis . Gastroenterology , 119 ( 1 ), 234 242 , 2000 .

    6. Sollid LM . Molecular basis of celiac disease . Ann Rev Immunol , 18 , 53 81 , 2000 . 7. Hogberg L , Falth-Magnusson K , Grodzinsky E , et al . Familial prevalence of coeliac

    disease: a twenty-year follow-up study . Scand J Gastroenterol , 38 , 61 65 , 2003 . 8. Stene LC , Honeyman MC , Hoffenberg EJ , Haas JE , Sokol RJ , Emery L , Taki I , Norris

    JM , Erlich HA , Eisenbarth GS and Rewers M . Rotavirus infection frequency and risk of celiac disease autoimmunity in early childhood: a longitudinal study . Am J Gastroenterol , 101 ( 10 ), 2333 2340 , 2006 .

    9. Thorsby E . Invited anniversary review: HLA associated diseases . Hum Immunol , 53 , 1 11 , 1997 .

    10. Scott EM , Gaywood I and Scott BB . Guidelines for osteoporosis in coeliac disease and infl ammatory bowel disease. British Society of Gastroenterology . Gut , 46 ( suppl 1 ), i1 8 , 2000 .

    11. Holmes GK , Prior P , Lane M and Allan RN . Malignancy in coeliac disease effect of a gluten free diet . Gut , 30 , 333 338 , 1989 .

    12. Ciclitira PJ , Ellis HJ and Lundin KEA . Gluten-free diet what is toxic? Best Pract Res Clin Gastroenterol , 19 ( 3 ), 359 371 , 2005 .

    13. Wieser H . The precipitating factor in coeliac disease . Bailliere s Clin Gastroenterol , 9 , 191 207 , 1995 .

    14. Buri C , Burri P , Bahler P. et al . Cytotoxic T cells are preferentially activated in the duodenal epithelium from patients with fl orid coeliac disease . J Pathol , 206 ( 2 ), 178 185 , 2005 .

    15. Ciclitira PJ , Evans DJ , Fagg NL et al . Clinical testing of gliadin fractions in coeliac patients . Clin Sci (Lond) , 66 , 357 364 , 1984 .

    16. Van De Wal Y , Kooy YM , Van Veelen P et al . Glutenin is involved in the gluten-driven mucosal T cell response . Eur J Immunol , 29 , 3133 3139 , 1999 .

    17. Vader W , Kooy Y , Van Veelen P et al . The gluten response in children with celiac disease is directed toward multiple gliadin and glutenin peptides . Gastroenterology , 122 , 17291737 , 2002 .

    18. Molberg O , Solheim Flaete N , Jensen T et al . Intestinal T-cell responses to high-molecular-weight glutenins in coeliac disease . Gastroenterology , 125 , 337 344 , 2003 .

    19. Dewar DH , Pollock EL and Engel W . The immunogenicity and in vivo toxicity of HMW glutenins in celiac disease . Gastroenterology , 126 ( suppl 2 ), A - 290 , 2004 .

  • 374 Handbook of Food Allergen Detection and Control

    20. Zhernakova A and Wijmenga C . HLA and non-HLA genes in celiac disease . In Fasano A , Troncone R and Branski D (eds), Frontiers in Celiac Disease . Pediatr Adolesc Med . Karger, Basel , 12 , 32 45 , 2008 .

    21. Tollefsen S , Arentz-Hansen H , Fleckenstein B , Molberg , Rki M , Kwok WW , Jung G , Lundin KEA and Sollid LM . HLA-DQ2 and -DQ8 signatures of gluten T cell epitopes in celiac disease . J Clin Invest , 116 ( 8 ), 2226 2236 , 2006 .

    22. Guandalini S and Setty M . Celiac disease . Curr Opin Gastroenterol , 24 ( 6 ), 707 712 , 2008 .

    23. Wolters VM and Wijmenga C . Genetic background of celiac disease and its clinical implications . Am J Gastroenterol , 103 ( 1 ), 190 195 , 2008 .

    24. Lundin KEA , Scott H , Hansen T et al . Gliadin specifi c, HLA-DQ (a1*0501, b1*0201) restricted T-cells isolated from the small intestinal mucosa of patients with coeliac disease . J Exp Med , 178 , 187 196 , 1993 .

    25. Arentz-Hansen H , Krner R , Molberg , Quarsten H , Vader W , Kooy YMC , Lundin KEA , Koning F , Roepstorff P , Sollid LM and McAdam SN . The intestinal T cell response to -gliadin in adult celiac disease is focused on a single deamidated glutamine targeted by tissue transglutaminase , J Exp Med , 191 ( 4 ), 603 612 , 2000 .

    26. Arentz-Hansen H , McAdam SN , Molberg et al . Celiac lesion T-cells recognize epitopes that cluster in regions rich in proline residues . Gastroenterology , 123 , 803 809 , 2002 .

    27. Vader LW , de Ru A , van der Wal Y et al . Specifi city of tissue transglutaminase explains cereal toxicity in celiac disease . J Exp Med , 195 , 643 649 , 2002 .

    28. Shan L , Molberg , Parrot I et al . Structural basis for gluten intolerance in celiac sprue . Science , 297 , 2275 2279 , 2002 .

    29. Visser J , Rozing J , Sapone A , Lammers K and Fasano A . Tight junctions, intestinal permeability, and autoimmunity: celiac disease and type 1 diabetes paradigms . Ann N Y Acad Sci , 1165 , 195 205 , 2009 .

    30. Matysiak-Budnik T , Moura IC , Arcos-Fajardo M , Lebreton C , Mnard S , Candalh C , Chaussade S , Malamut G , Cellier C , Cerf-Bensussan N , Monteiro RC , Heyman M , Matysiak-Budnik T , Moura IC , Arcos-Fajardo M , Ben-Khalifa K , Dugave C , Tamouza H , van Niel G , Bouhnik Y , Lamarque D , Chaussade S , Malamut G , Cellier C , Cerf-Bensussan N , Monteiro RC and Heyman M . Secretory IgA mediates retrotranscytosis of intact gliadin peptides via the transferrin receptor in celiac disease . J Exp Med , 205 ( 1 ), 143 154 , 2008 .

    31. Dieterich W , Ehnis T , Bauer M , et al . Identifi cation of tissue transglutaminase as the autoantigen of celiac disease . Nat Med , 3 , 797 801 , 1997 .

    32. Qiao S-W , Bergseng E , Molberg , Xia J , Fleckenstein B , Khosla C and Sollid LM . Antigen presentation to celiac lesion-derived T cells of a 33-mer gliadin peptide naturally formed by gastrointestinal digestion . J Immunol , 173 ( 3 ), 17571762 , 2004 .

    33. Rki M , Tollefsen S , Molberg , Lundin KE , Sollid LM and Jahnsen FL . A unique dendritic cell subset accumulates in the celiac lesion and effi ciently activates gluten-reactive T cells . Gastroenterology , 131 ( 2 ), 428 438 , 2006 .

    34. Mention JJ , Ben Ahmed M , Bgue B , Barbe U , Verkarre V , Asnafi V , Colombel JF , Cugnenc PH , Ruemmele FM , McIntyre E , Brousse N , Cellier C and Cerf-Bensussan N . Interleukin 15: a key to disrupted intraepithelial lymphocyte homeostasis and lymphomagenesis in celiac disease . Gastroenterology , 125 ( 3 ), 730 745 , 2003 .

    35. Hue S et al . A direct role for NKG2D/MICA interaction in villous atrophy during celiac disease . Immunity , 21 , 367 377 , 2004 .

    36. Jabri B et al . Selective expansion of intraepithelial lymphocytes expressing the HLA-E-specifi c natural killer receptor CD94 in celiac disease . Gastroenterology , 118 , 867 879 , 2000 .

  • Detection and control of gluten as a food allergen 375

    37. Meresse B et al . Coordinated induction by IL15 of a TCR-independent NKG2D signaling pathway converts CTL into lymphokine-activated killer cells in celiac disease . Immunity , 21 , 357 366 , 2004 .

    38. Meresse B et al . Reprogramming of CTLs into natural killer-like cells in celiac disease . J Exp Med , 203 , 1343 1355 , 2006 .

    39. Barone MV et al . Growth factor-like activity of gliadin, an alimentary protein: implications for coeliac disease . Gut , 56 , 480 488 , 2007 .

    40. Kagnoff MF . Celiac disease: pathogenesis of a model immunogenetic disease . J Clin Invest , 117 ( 1 ), 41 49 , 2007 .

    41. Cellier C et al . Refractory sprue, coeliac disease, and enteropathy-associated T-cell lymphoma. French Coeliac Disease Study Group . Lancet , 356 , 203 208 , 2000 .

    42. Tack GJ , van Wanrooij RL , Langerak AW , Tjon JM , von Blomberg BM , Heideman DA , van Bergen J , Koning F , Bouma G , Mulder CJ and Schreurs MW. Origin and immunophenotype of aberrant IEL in RCDII patients . Mol Immunol , 50 ( 4 ), 262 270 , 2012 .

    43. Bagdi E , Diss TC , Munson P and Isaacson PG . Mucosal intra-epithelial lymphocytes in enteropathy-associated T-cell lymphoma, ulcerative jejunitis, and refractory celiac disease constitute a neoplastic population . Blood , 94 ( 1 ), 260 264 , 1999 .

    44. Brown GR , Lindberg G , Meddings J et al . Tumor necrosis factor inhibitor ameliorates murine intestinal graft-versus-host disease . Gastroenterology , 116 , 593 601 , 1999 .

    45. Guy-Grand D , DiSanto JP , Henchoz P et al . Small bowel enteropathy: role of intraepithelial lymphocytes and of cytokines (IL-12, IFN-gamma, TNF) in the induction of epithelial cell death and renewal . Eur J Immunol , 28 , 730 744 , 1998 .

    46. Deem RL , Shanahan F and Targan SR . Triggered human mucosal T cells release tumour necrosis factor-alpha and interferon-gamma which kill human colonic epithelial cells . Clin Exp Immunol , 83 , 79 84 , 1991 .

    47. Abreu-Martin MT , Palladino AA , Faris M et al . Fas activates the JNK pathway in human colonic epithelial cells: lack of a direct role in apoptosis . Am J Physiol , 276 , G599 605 , 1999 .

    48. Ciccocioppo R , Di Sabatino A , Parroni R , Muzi P , DAl S , Ventura T , Pistoia MA , Cifone MG and Corazza GR . Increased enterocyte apoptosis and fas-fas ligand system in celiac disease . Am J Clin Pathol , 115 , 494 503 , 2001 .

    49. Ciccocioppo R , Di Sabatino A , Parroni R et al . Cytolytic mechanisms of intraepithelial lymphocytes in coeliac disease (CoD) . Clin Exp Immunol , 120 , 235 240 , 2000 .

    50. Ciccocioppo R , Di Sabatino A , Bauer M , Della Riccia DN , Bizzini F , Biagi F , Cifone MG , Corazza GR and Schuppan D . Matrix metalloproteinase pattern in celiac duodenal mucosa . Lab Invest , 85 ( 3 ), 397 407 , 2005 .

    51. Ellis HJ , Freedman AR and Ciclitira PJ . The production and characterisation of monoclonal antibodies to wheat gliadin peptides . J Immunol Methods , 120 , 17 22 , 1989 .

    52. Ellis HJ , Rosen-Bronson S , O Reilly N and Ciclitira PJ . Measurement of gluten using a monoclonal antibody to a coeliac-toxic peptide of A-gliadin . Gut , 43 , 190 195 , 1998 .

    53. Spaenij-Dekking EH , Kooy-Winkelaar EM , Nieuwenhuizen WF et al . A novel and sensitive method for the detection of T cell stimulatory epitopes of alpha/beta- and gamma-gliadin . Gut , 53 , 1267 1273 , 2004 .

    54. Browning TH and Trier JS . Organ culture of mucosal biopsies of human small intestine . J Clin Invest , 48 , 1423 1432 , 1969 .

    55. Townley RR , Cornell HJ , Bhathal PS and Mitchell JD . Toxicity of wheat gliadin fractions in coeliac disease . Lancet , 1 , 1363 1364 , 1973 .

  • 376 Handbook of Food Allergen Detection and Control

    56. Peraho M , Kaukinen K , Paasikivi K et al . Wheat-starch-based gluten-free products in the treatment of newly detected coeliac disease: prospective and randomized study . Aliment Pharmacol Ther , 17 , 587 594 , 2003 .

    57. Collin P , Thorell L , Kaukinen K and Mki M . The safe threshold for gluten contamination in gluten-free products. Can trace amounts be accepted in treatment of coeliac disease? Aliment Pharmacol Ther , 19 , 1277 1283 , 2004 .

    58. Fasano A and Catassi C . Current approaches to diagnosis and treatment of celiac disease: an evolving spectrum . Gastroenterology , 120 , 636 651 , 2001 .

    59. Stern M , Ciclitira PJ , van Eckert R et al . Analysis and clinical effects of gluten in coeliac disease . Eur J Gastroenterol Hepatol , 13 , 741 747 , 2001 .

    60. Catassi C , Fabiani E , Iacono G , DAgate C , Francavilla R , Biagi F , Volta U , Accomando S , Picarelli A , De Vitis I , Pianelli G , Gesuita R , Carle F , Mandolesi A , Bearzi I and Fasano A . A prospective, double-blind, placebo-controlled trial to establish a safe gluten threshold for patients with celiac disease . Am J Clin Nutr , 85 ( 1 ), 160 166 , 2007 .

    61. van Overbeek FM , Uil-Dieterman IG , Mol IW et al . The daily gluten intake in relatives of patients with coeliac disease compared with that of the general Dutch population . Eur J Gastroenterol Hepatol , 9 , 1097 1099 , 1997 .

    62. Skerritt JH and Hill AS . Enzyme immunoassay for determination of gluten in foods: collaborative study . J Assoc Off Anal Chem , 74 , 257 264 , 1991 .

    63. Valds I , Garca E , Llorente M and Mndez E . Innovative approach to low-level gluten determination in foods using a novel sandwich enzyme-linked immunosorbent assay protocol . Eur J Gastroenterol Hepatol , 15 ( 5 ), 465 747 , 2003 .

    64. Sorellb L , Lpeza JA , Valdsa I , Alfonsoa P , Camafeitaa E , Acevedob B , Chirdoc F , Gavilondob J and Mndeza E . An innovative sandwich ELISA system based on an antibody cocktail for gluten analysis . FEBS Lett , 439 ( 12 ), 46 50 , 1998 .

    65. Ellis HJ , Freedman AR and 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 , 189 , 123 130 , 1990 .

    66. Ellis HJ , Doyle AP , Day P et al . Demonstration of the presence of coeliac-activating gliadin-like epitopes in malted barley . Int Arch Allergy Immunol , 104 , 308 310 , 1994 .

    67. Biagi F , Campanella J , Martucci S et al . A milligram gluten a day keeps the mucosal recovery away: a case report . Nutr Rev , 62 , 360 363 , 2004 .

    68. Janatuinen EK , Pikkarainen PH , Kemppainen TA et al . A comparison of diets with and without oats in adults with coeliac disease . N Engl J Med , 333 , 1033 1037 , 1995 .

    69. Janatuinen EK , Kemppainen TA , Julkunen RJ et al . No harm from fi ve year ingestion of oats in celiac disease . Gut , 50 , 332 335 , 2002 .

    70. Srinivasan U , Leonard N , Jones E et al . Absence of oats toxicity in adult coeliac disease . BMJ , 313 , 1300 1301 , 1996 .

    71. Storsrud S , Olsson M , Arvidsson Lenner R et al . Adult coeliac patients do tolerate large amounts of oats . Eur J Clin Nutr , 57 , 163 169 , 2003 .

    72. Lundin KEA , Nilsen EM , Scott HG et al . Oats induced villous atrophy in coeliac disease . Gut , 52 , 16491652 , 2003 .

    73. Arentz-Hansen H , Fleckenstein B , Molberg et al . The molecular basis for oat intolerance in patients with coeliac disease . PloS Med , 1 , 84 92 , 2004 .

    74. Peraho M , Kaukinen K , Mustalahti K et al . Effect of an oats-containing gluten-free diet on symptoms and quality of life in coeliac disease. A randomized study . Scand J Gastroenterol , 39 , 27 31 , 2004 .

  • Detection and control of gluten as a food allergen 377

    75. Thompson T . Gluten contamination of commercial oat products in the United States . N Eng J Med , 351 , 2021 2022 , 2004 .

    76. Pichler J , Zilbauer M , Torrente F , Heuschkel R , Phillips A and Salvestrini C . Feasibility of a fi nger prick-based self-testing kit in fi rst- and second-degree relatives of children with coeliac disease . World J Gastroenterol , 17 ( 14 ), 18401843 , 2011.