Skip to main content
Top
Published in: Journal of Gastroenterology 9/2017

01-09-2017 | Review

Celiac disease: from etiological factors to evolving diagnostic approaches

Authors: Anantdeep Kaur, Olga Shimoni, Michael Wallach

Published in: Journal of Gastroenterology | Issue 9/2017

Login to get access

Abstract

Celiac disease has advanced from a medical rarity to a highly prevalent disorder. Patients with the disease show varying degrees of chronic inflammation within the small intestine due to an aberrant immune response to the digestion of gliadin found in wheat. As a result, cytokines and antibodies are produced in celiac patients that can be used as specific biomarkers for developing diagnostic tests. This review paper describes celiac disease in terms of its etiological cause, pathological effects, current diagnostic tests based on mucosal biopsy, and the genetic basis for the disease. In addition, it discusses the use of gliadin-induced cytokines, antibodies and autoantibodies as a diagnostic tool for celiac disease. Despite good initial results in terms of sensitivity and specificity, when these immunological tests were used on a large scale, even in combination with genetic testing, the results showed lower predictive value. This review addresses that issue and ends with an outlook on future work required to develop diagnostic tests with greater accuracy in predicting celiac disease in the general public, thus avoiding the need for endoscopy and mucosal biopsy.
Literature
1.
go back to reference Ludvigsson JF, Leffler DA, Bai JC, et al. The Oslo definitions for celiac disease and related terms. Gut. 2013;62:43–52.PubMedCrossRef Ludvigsson JF, Leffler DA, Bai JC, et al. The Oslo definitions for celiac disease and related terms. Gut. 2013;62:43–52.PubMedCrossRef
2.
go back to reference Kang JY, Kang AHY, Green A, et al. Systematic review: worldwide variation in the frequency of celiac disease and changes over time. Aliment Pharmacol Ther. 2013;38:226–45.PubMedCrossRef Kang JY, Kang AHY, Green A, et al. Systematic review: worldwide variation in the frequency of celiac disease and changes over time. Aliment Pharmacol Ther. 2013;38:226–45.PubMedCrossRef
3.
go back to reference Lionetti E, Gatti S, Pulvirenti A, et al. Celiac disease from a global perspective. Best Pract Res Clin Gastroenterol. 2015;29:365–79.PubMedCrossRef Lionetti E, Gatti S, Pulvirenti A, et al. Celiac disease from a global perspective. Best Pract Res Clin Gastroenterol. 2015;29:365–79.PubMedCrossRef
4.
go back to reference Hovell CJ, Collett JA, Vautier G, et al. High prevalence of celiac disease in a population-based study from Western Australia: a case for screening? Med J Aust. 2001;175:247–50.PubMed Hovell CJ, Collett JA, Vautier G, et al. High prevalence of celiac disease in a population-based study from Western Australia: a case for screening? Med J Aust. 2001;175:247–50.PubMed
5.
go back to reference Rubio-Tapia A, Ludvigsson JF, Brantner TL, et al. The prevalence of celiac disease in the United States. Am J Gastroenetrol. 2012;107:1538–44.CrossRef Rubio-Tapia A, Ludvigsson JF, Brantner TL, et al. The prevalence of celiac disease in the United States. Am J Gastroenetrol. 2012;107:1538–44.CrossRef
6.
go back to reference Woychik JH, Boundy JA, Dimler RJ. Starch gel-electrophoresis of wheat gluten proteins with concentrated urea. Arch Biochem Biophys. 1961;94:477–82.PubMedCrossRef Woychik JH, Boundy JA, Dimler RJ. Starch gel-electrophoresis of wheat gluten proteins with concentrated urea. Arch Biochem Biophys. 1961;94:477–82.PubMedCrossRef
7.
go back to reference Bushuk W, Zillman RR. Wheat cultivar identification by gliadin electrophoregrams: 1. Apparatus, method, and nomenclature. Can J Plant Sci. 1978;58:505–15.CrossRef Bushuk W, Zillman RR. Wheat cultivar identification by gliadin electrophoregrams: 1. Apparatus, method, and nomenclature. Can J Plant Sci. 1978;58:505–15.CrossRef
8.
go back to reference Metakovsky EV, Branlard G, Chernakov VM, et al. Recombinant mapping of some chromosome 1A-, 1B-, 1D-, and 6B-controlled gliadins and low molecular weight glutenin subunits in common wheat. Theor Appl Genet. 1997;94:788–95.CrossRef Metakovsky EV, Branlard G, Chernakov VM, et al. Recombinant mapping of some chromosome 1A-, 1B-, 1D-, and 6B-controlled gliadins and low molecular weight glutenin subunits in common wheat. Theor Appl Genet. 1997;94:788–95.CrossRef
9.
go back to reference Anderson OD, Dong L, Huo N, et al. A new class of wheat gliadin genes and proteins. PLoS ONE. 2012;7:12. Anderson OD, Dong L, Huo N, et al. A new class of wheat gliadin genes and proteins. PLoS ONE. 2012;7:12.
10.
go back to reference Kasarda DD, Okita TW, Bernardin JE, et al. Nucleic acid (cDNA) and amino acid sequences of α-type gliadins from wheat (Triticum aestivum). Proc Natl Acad Sci. 1984;81:4712–6.PubMedPubMedCentralCrossRef Kasarda DD, Okita TW, Bernardin JE, et al. Nucleic acid (cDNA) and amino acid sequences of α-type gliadins from wheat (Triticum aestivum). Proc Natl Acad Sci. 1984;81:4712–6.PubMedPubMedCentralCrossRef
11.
go back to reference Anderson OD, Greene FC. The α-gliadin gene family. DNA and protein sequence variation, subfamily structure, and origins of pseudogenes. Theor Appl Genet. 1997;95:59–65.CrossRef Anderson OD, Greene FC. The α-gliadin gene family. DNA and protein sequence variation, subfamily structure, and origins of pseudogenes. Theor Appl Genet. 1997;95:59–65.CrossRef
12.
go back to reference Noma S, Kawaura K, Hayakawa K, et al. Comprehensive molecular characterization of the α/β gliadin multigene family in hexaploid wheat. Mol Genet Genom. 2016;291:65–77.CrossRef Noma S, Kawaura K, Hayakawa K, et al. Comprehensive molecular characterization of the α/β gliadin multigene family in hexaploid wheat. Mol Genet Genom. 2016;291:65–77.CrossRef
13.
go back to reference Bronstein HD, Haeffner LJ, Kowlessar OD. Enzymatic digestion of gliadin: the effect of the resultant peptides in adult celiac disease. Clin Chim Acta. 1966;14:141–55.PubMedCrossRef Bronstein HD, Haeffner LJ, Kowlessar OD. Enzymatic digestion of gliadin: the effect of the resultant peptides in adult celiac disease. Clin Chim Acta. 1966;14:141–55.PubMedCrossRef
14.
go back to reference Weiser H. Relation between gliadin structure and celiac toxicity. Acta Paediatr. 1996;412:3–9.CrossRef Weiser H. Relation between gliadin structure and celiac toxicity. Acta Paediatr. 1996;412:3–9.CrossRef
15.
go back to reference Shan L, Molberg O, Parrot I, et al. Structural basis for gluten intolerance in celiac disease. Science. 2002;297:2275–9.PubMedCrossRef Shan L, Molberg O, Parrot I, et al. Structural basis for gluten intolerance in celiac disease. Science. 2002;297:2275–9.PubMedCrossRef
16.
go back to reference Qi PF, Chen Q, Oullet T, et al. The molecular diversity of α-gliadin genes in the tribe Triticea. Genetica. 2013;141:303–10.PubMedCrossRef Qi PF, Chen Q, Oullet T, et al. The molecular diversity of α-gliadin genes in the tribe Triticea. Genetica. 2013;141:303–10.PubMedCrossRef
17.
go back to reference Oberhuber G, Granditsch G, Vogelsang H. The histopathology of celiac disease: time for a standardised report scheme for pathologists. Eur J Gastroentrol. 1999;11:1185–94.CrossRef Oberhuber G, Granditsch G, Vogelsang H. The histopathology of celiac disease: time for a standardised report scheme for pathologists. Eur J Gastroentrol. 1999;11:1185–94.CrossRef
18.
go back to reference Marsh MN, Vincenzo V, Srivastava A. Histology of gluten related disorders. Gastroenterol Hepatol Bed Bench. 2015;8:171–7. Marsh MN, Vincenzo V, Srivastava A. Histology of gluten related disorders. Gastroenterol Hepatol Bed Bench. 2015;8:171–7.
20.
go back to reference Bao F, Green PH, Bhagat G. An update on celiac disease histopathology and the road ahead. Arch Pathol Lab Med. 2012;136:735–45.PubMedCrossRef Bao F, Green PH, Bhagat G. An update on celiac disease histopathology and the road ahead. Arch Pathol Lab Med. 2012;136:735–45.PubMedCrossRef
21.
go back to reference Castillo NE, Theethira TG, Leffler DA. The present and the future in diagnosis and management of celiac disease. Gastroenterol Rep. 2015;3(1):3–11.CrossRef Castillo NE, Theethira TG, Leffler DA. The present and the future in diagnosis and management of celiac disease. Gastroenterol Rep. 2015;3(1):3–11.CrossRef
22.
24.
go back to reference Howell MD, Austin RK, Kelleher D, et al. An HLA-D region restriction length polymorphism associated with celiac disease. J Exp Med. 1986;169:345–50. Howell MD, Austin RK, Kelleher D, et al. An HLA-D region restriction length polymorphism associated with celiac disease. J Exp Med. 1986;169:345–50.
25.
go back to reference Olivares M, Neef A, Castillejo G, et al. The HLA-DQ2 genotype selects for early intestinal microbiota composition in infants at high risk of developing celiac disease. Gut. 2015;64:406–17.PubMedCrossRef Olivares M, Neef A, Castillejo G, et al. The HLA-DQ2 genotype selects for early intestinal microbiota composition in infants at high risk of developing celiac disease. Gut. 2015;64:406–17.PubMedCrossRef
26.
go back to reference Lundin KEA, Scott H, Hansen T, et al. Gliadin-specific, HLA-DQ (alpha1*0501, beta1*0201) restricted T cells isolated from the small intestinal mucosa of celiac disease patients. J Exp Med. 1993;178:187–96.PubMedCrossRef Lundin KEA, Scott H, Hansen T, et al. Gliadin-specific, HLA-DQ (alpha1*0501, beta1*0201) restricted T cells isolated from the small intestinal mucosa of celiac disease patients. J Exp Med. 1993;178:187–96.PubMedCrossRef
27.
go back to reference Costantini S, Rossi M, Colonna G, et al. Modelling of HLA-DQ2 and its interaction with gluten peptides to explain molecular recognition in celiac disease. J Mol Graph Model. 2005;23:419–31.PubMedCrossRef Costantini S, Rossi M, Colonna G, et al. Modelling of HLA-DQ2 and its interaction with gluten peptides to explain molecular recognition in celiac disease. J Mol Graph Model. 2005;23:419–31.PubMedCrossRef
28.
go back to reference Henderson KN, Tye-Din JA, Reid H, et al. A structural and immunological basis for the role of human leukocyte antigen DQ8 in celiac disease. Immunity. 2007;27:23–34.PubMedCrossRef Henderson KN, Tye-Din JA, Reid H, et al. A structural and immunological basis for the role of human leukocyte antigen DQ8 in celiac disease. Immunity. 2007;27:23–34.PubMedCrossRef
29.
go back to reference Broughton SE, Petersen J, Theodossis A, et al. Biased T cell receptor usage directed against human leukocyte antigen DQ8-restricted gliadin peptides is associated with celiac disease. Immunity. 2012;37:611–21.PubMedCrossRef Broughton SE, Petersen J, Theodossis A, et al. Biased T cell receptor usage directed against human leukocyte antigen DQ8-restricted gliadin peptides is associated with celiac disease. Immunity. 2012;37:611–21.PubMedCrossRef
30.
go back to reference Hadithi M, von Blomberg BM, Crusius JB, et al. Accuracy of serologic tests and HLA-DQ typing for diagnosing celiac disease. Ann Intern Med. 2007;147:294–302.PubMedCrossRef Hadithi M, von Blomberg BM, Crusius JB, et al. Accuracy of serologic tests and HLA-DQ typing for diagnosing celiac disease. Ann Intern Med. 2007;147:294–302.PubMedCrossRef
31.
go back to reference Husby S, Koletzko S, Korponay-Szabó IR, et al. European Society for Pediatric Gastroenterology, Hepatology, and Nutrition guidelines for the diagnosis of celiac disease. J Pediatr Gastroenterol Nutr. 2012;54:136–60.PubMedCrossRef Husby S, Koletzko S, Korponay-Szabó IR, et al. European Society for Pediatric Gastroenterology, Hepatology, and Nutrition guidelines for the diagnosis of celiac disease. J Pediatr Gastroenterol Nutr. 2012;54:136–60.PubMedCrossRef
33.
34.
go back to reference Abraham G, Rohmer A, Tye-Din JA, et al. Genomic prediction of celiac disease targeting HLA-positive individuals. Genome Med. 2015;7:3–11.CrossRef Abraham G, Rohmer A, Tye-Din JA, et al. Genomic prediction of celiac disease targeting HLA-positive individuals. Genome Med. 2015;7:3–11.CrossRef
35.
go back to reference Kruppa K. Endomysial antibodies predict celiac disease irrespective of the titers or clinical presentation. World J Gastroenterol. 2012;18:2511–6.CrossRef Kruppa K. Endomysial antibodies predict celiac disease irrespective of the titers or clinical presentation. World J Gastroenterol. 2012;18:2511–6.CrossRef
36.
go back to reference Fasano A. Zonulin and its regulation of intestinal barrier function: the biological door to inflammation, autoimmunity, and cancer. Physiol Rev. 2011;91:151–75.PubMedCrossRef Fasano A. Zonulin and its regulation of intestinal barrier function: the biological door to inflammation, autoimmunity, and cancer. Physiol Rev. 2011;91:151–75.PubMedCrossRef
37.
go back to reference Lammers KM, Lu R, Brownley J, et al. Gliadin induces an increase in intestinal permeability and zonulin release by binding to the chemokine receptor CXCR3. Gastroenterology. 2008;135:194–204.PubMedPubMedCentralCrossRef Lammers KM, Lu R, Brownley J, et al. Gliadin induces an increase in intestinal permeability and zonulin release by binding to the chemokine receptor CXCR3. Gastroenterology. 2008;135:194–204.PubMedPubMedCentralCrossRef
38.
go back to reference Schumann M, Richter JF, Wedell I, et al. Mechanisms of epithelial translocation of the alpha(2)-gliadin-33 mer in celiac sprue. Gut. 2008;57:747–54.PubMedCrossRef Schumann M, Richter JF, Wedell I, et al. Mechanisms of epithelial translocation of the alpha(2)-gliadin-33 mer in celiac sprue. Gut. 2008;57:747–54.PubMedCrossRef
39.
go back to reference Matysiak-Budnik T, Moura IC, Arcos-Fajardo M, et al. Secretory IgA mediates retrotranscytosis of intact gliadin peptides via the transferrin receptor in celiac disease. J Exp Med. 2008;205:143–54.PubMedPubMedCentralCrossRef Matysiak-Budnik T, Moura IC, Arcos-Fajardo M, et al. Secretory IgA mediates retrotranscytosis of intact gliadin peptides via the transferrin receptor in celiac disease. J Exp Med. 2008;205:143–54.PubMedPubMedCentralCrossRef
40.
go back to reference Bodd M, Ráki 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.PubMedCrossRef Bodd M, Ráki 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.PubMedCrossRef
41.
go back to reference Peluso I, Fantini MC, Fina D, et al. IL-21 counteracts the regulatory T cell-mediated suppression of human CD4+ T lymphocytes. J Immunol. 2007;178:732–9.PubMedCrossRef Peluso I, Fantini MC, Fina D, et al. IL-21 counteracts the regulatory T cell-mediated suppression of human CD4+ T lymphocytes. J Immunol. 2007;178:732–9.PubMedCrossRef
42.
go back to reference Hüe S, Mention J-J, Monteiro RC, et al. A direct role for NKG2D/MICA interaction in villous atrophy during celiac disease. Immunity. 2004;21:367–77.PubMedCrossRef Hüe S, Mention J-J, Monteiro RC, et al. A direct role for NKG2D/MICA interaction in villous atrophy during celiac disease. Immunity. 2004;21:367–77.PubMedCrossRef
43.
44.
go back to reference Sollid LM, Jabri B. Triggers and drivers of autoimmunity: lessons from celiac disease. Nat Rev Immunol. 2013;13:294–302.PubMedCrossRef Sollid LM, Jabri B. Triggers and drivers of autoimmunity: lessons from celiac disease. Nat Rev Immunol. 2013;13:294–302.PubMedCrossRef
45.
go back to reference Sabatino AD, Vanoli A, Giuffrida P, et al. The function of tissue transglutaminase in celiac disease. Autoimmun Rev. 2012;11:746–53.PubMedCrossRef Sabatino AD, Vanoli A, Giuffrida P, et al. The function of tissue transglutaminase in celiac disease. Autoimmun Rev. 2012;11:746–53.PubMedCrossRef
48.
go back to reference Du Pre’ MF, Sollid LM. T-cell and B-cell immunity in celiac disease. Best Pract Res Clin Gastroenterol. 2015;29:413–23.CrossRef Du Pre’ MF, Sollid LM. T-cell and B-cell immunity in celiac disease. Best Pract Res Clin Gastroenterol. 2015;29:413–23.CrossRef
49.
go back to reference Dieterich W, Laag E, Schopper H, et al. Autoantibodies to tissue transglutaminase as predictors of celiac disease. Gastroenterology. 1998;115:1317–21.PubMedCrossRef Dieterich W, Laag E, Schopper H, et al. Autoantibodies to tissue transglutaminase as predictors of celiac disease. Gastroenterology. 1998;115:1317–21.PubMedCrossRef
50.
go back to reference O’Farrelly C, Kelly J, Hekkens W, et al. Alpha gliadin antibody levels: a serological test for celiac disease. Br Med J (Clin Res Ed). 1983;25:2007–10.CrossRef O’Farrelly C, Kelly J, Hekkens W, et al. Alpha gliadin antibody levels: a serological test for celiac disease. Br Med J (Clin Res Ed). 1983;25:2007–10.CrossRef
51.
go back to reference Savilahti E, Viander M, Perkkio M, et al. IgA antigliadin antibodies: a marker of mucosal damage in childhood celiac disease. Lancet. 1983;1:320–2.PubMedCrossRef Savilahti E, Viander M, Perkkio M, et al. IgA antigliadin antibodies: a marker of mucosal damage in childhood celiac disease. Lancet. 1983;1:320–2.PubMedCrossRef
52.
go back to reference Tucker NT, Barghuthy FS, Prihoda TJ, et al. Antigliadin antibodies detected by enzyme-linked immunosorbent assay as a marker of childhood celiac disease. J Pediatr. 1988;113:286–9.PubMedCrossRef Tucker NT, Barghuthy FS, Prihoda TJ, et al. Antigliadin antibodies detected by enzyme-linked immunosorbent assay as a marker of childhood celiac disease. J Pediatr. 1988;113:286–9.PubMedCrossRef
53.
go back to reference Bodé S, Weile B, Krasilnikoff PA, et al. The diagnostic value of the gliadin antibody test in celiac disease in children: a prospective study. J Pediatr Gastroenterol Nutr. 1993;17:260–4.PubMedCrossRef Bodé S, Weile B, Krasilnikoff PA, et al. The diagnostic value of the gliadin antibody test in celiac disease in children: a prospective study. J Pediatr Gastroenterol Nutr. 1993;17:260–4.PubMedCrossRef
54.
go back to reference Lerner A, Kumar V, Iancu TC. Immunological diagnosis of childhood celiac disease: comparison between antigliadin, antireticulin and antiendomysial antibodies. Clin Exp Immunol. 1994;95:78–82.PubMedPubMedCentralCrossRef Lerner A, Kumar V, Iancu TC. Immunological diagnosis of childhood celiac disease: comparison between antigliadin, antireticulin and antiendomysial antibodies. Clin Exp Immunol. 1994;95:78–82.PubMedPubMedCentralCrossRef
55.
go back to reference Chartrand LJ, Aglunik J, Vanounou T, et al. Effectiveness of anti-Gliadin antibodies as a screening test for celiac disease in children. Can Med Assoc J. 1997;1997(157):527–33. Chartrand LJ, Aglunik J, Vanounou T, et al. Effectiveness of anti-Gliadin antibodies as a screening test for celiac disease in children. Can Med Assoc J. 1997;1997(157):527–33.
56.
go back to reference Lagerqvist C, Ingrid D, Tony H, et al. Antigliadin immunoglobulin A best in finding celiac disease in children younger than 18 months of age. J Pediatr Gastroenterol Nutr. 2008;47:428–35.PubMedCrossRef Lagerqvist C, Ingrid D, Tony H, et al. Antigliadin immunoglobulin A best in finding celiac disease in children younger than 18 months of age. J Pediatr Gastroenterol Nutr. 2008;47:428–35.PubMedCrossRef
57.
go back to reference Pereira SV, Raba J, Messina GA. IgG anti-Gliadin determination with an immunological microfluidic system applied to the automated diagnostic of the celiac disease. Anal Bioanal Chem. 2010;396:2921–7.PubMedCrossRef Pereira SV, Raba J, Messina GA. IgG anti-Gliadin determination with an immunological microfluidic system applied to the automated diagnostic of the celiac disease. Anal Bioanal Chem. 2010;396:2921–7.PubMedCrossRef
59.
go back to reference Picarelli A. 31-43 amino acid sequence of the a-Gliadin induces anti-Endomysial antibody production during in vitro challenge. Scand J Gastroenterol. 1999;34:1099–102.PubMedCrossRef Picarelli A. 31-43 amino acid sequence of the a-Gliadin induces anti-Endomysial antibody production during in vitro challenge. Scand J Gastroenterol. 1999;34:1099–102.PubMedCrossRef
60.
go back to reference Rostami K, Jo Kerckhaert, Tiemessen R, et al. Sensitivity of antiendomysium and antigliadin antibodies in untreated celiac disease: disappointing in clinical practice: sensitivity of IgA EMA and AGA in celiac disease. Am J Gastroenterol. 1994;94:888–94.CrossRef Rostami K, Jo Kerckhaert, Tiemessen R, et al. Sensitivity of antiendomysium and antigliadin antibodies in untreated celiac disease: disappointing in clinical practice: sensitivity of IgA EMA and AGA in celiac disease. Am J Gastroenterol. 1994;94:888–94.CrossRef
61.
go back to reference Leffler DA, Schuppan D. Update on serologic testing in celiac disease. Am J Gastroenterol. 2010;105:2520–4.PubMedCrossRef Leffler DA, Schuppan D. Update on serologic testing in celiac disease. Am J Gastroenterol. 2010;105:2520–4.PubMedCrossRef
62.
go back to reference Bruce SE, Bjarnason I, Peter I. Human jejunal transglutaminase: demonstration of activity, enzyme kinetics and substrate specificity with special relation to gliadin and celiac disease. Clin Sci. 1985;68:573–9.PubMedCrossRef Bruce SE, Bjarnason I, Peter I. Human jejunal transglutaminase: demonstration of activity, enzyme kinetics and substrate specificity with special relation to gliadin and celiac disease. Clin Sci. 1985;68:573–9.PubMedCrossRef
63.
go back to reference Hill PG, Holmes GK. Celiac disease: a biopsy is not always necessary for diagnosis. Aliment Pharmacol Ther. 2008;27:572–7.PubMedCrossRef Hill PG, Holmes GK. Celiac disease: a biopsy is not always necessary for diagnosis. Aliment Pharmacol Ther. 2008;27:572–7.PubMedCrossRef
64.
go back to reference Vivas S, Ruiz de Morales JG, Riestra S, et al. Duodenal biopsy may be avoided when high transglutaminase antibody titres are present. World J Gastroenterol. 2009;15:4775–80.PubMedPubMedCentralCrossRef Vivas S, Ruiz de Morales JG, Riestra S, et al. Duodenal biopsy may be avoided when high transglutaminase antibody titres are present. World J Gastroenterol. 2009;15:4775–80.PubMedPubMedCentralCrossRef
65.
go back to reference Tortara R, Imperatore N, Capone P, et al. The presence of anti-Endomysial antibodies and the level of anti-tissue transglutaminase can be used to diagnose adult celiac disease without duodenal biopsy. Aliment Pharmacol Ther. 2014;40:122–9. Tortara R, Imperatore N, Capone P, et al. The presence of anti-Endomysial antibodies and the level of anti-tissue transglutaminase can be used to diagnose adult celiac disease without duodenal biopsy. Aliment Pharmacol Ther. 2014;40:122–9.
66.
go back to reference Dahlbom I, Nyberg BI, Bernston L, et al. Simultaneous detection of IgA and IgG antibodies against tissue transglutaminase: the preferred pre-biopsy test in childhood celiac disease. Scand J Clin Lab Invest. 2016;76:208–16.PubMedCrossRef Dahlbom I, Nyberg BI, Bernston L, et al. Simultaneous detection of IgA and IgG antibodies against tissue transglutaminase: the preferred pre-biopsy test in childhood celiac disease. Scand J Clin Lab Invest. 2016;76:208–16.PubMedCrossRef
67.
go back to reference Sblattero D, Berti I, Trevisiol C, et al. Human recombinant tissue transglutaminase ELISA: an innovative diagnostic assay for celiac disease. Am J Gastroenterol. 2000;95:1253–7.PubMedCrossRef Sblattero D, Berti I, Trevisiol C, et al. Human recombinant tissue transglutaminase ELISA: an innovative diagnostic assay for celiac disease. Am J Gastroenterol. 2000;95:1253–7.PubMedCrossRef
68.
go back to reference Lock RJ, Gilmour JEM, Unsworth DJ. Anti-tissue transglutaminase, anti-endomysium and anti-R1-reticulin autoantibodies: the antibody trinity of celiac disease. Clin Exp Immunol. 1999;116:258–62.PubMedPubMedCentralCrossRef Lock RJ, Gilmour JEM, Unsworth DJ. Anti-tissue transglutaminase, anti-endomysium and anti-R1-reticulin autoantibodies: the antibody trinity of celiac disease. Clin Exp Immunol. 1999;116:258–62.PubMedPubMedCentralCrossRef
69.
go back to reference Lewis NR, Scott BB. Meta-analysis: deamidated gliadin peptide antibody and tissue transglutaminase antibody compared as screening tests for celiac disease. Aliment Pharmacol Ther. 2010;31:73–81.PubMedCrossRef Lewis NR, Scott BB. Meta-analysis: deamidated gliadin peptide antibody and tissue transglutaminase antibody compared as screening tests for celiac disease. Aliment Pharmacol Ther. 2010;31:73–81.PubMedCrossRef
70.
go back to reference Dahlbom I, Korponay-Szabo IR, Kovacs JB, et al. Prediction of clinical and mucosal severity of celiac disease and dermatitis herpetiformis by quantification of IgA/IgG serum antibodies to tissue transglutaminase. J Pediatr Gastroenterol Nutr. 2010;50:140–6.PubMedCrossRef Dahlbom I, Korponay-Szabo IR, Kovacs JB, et al. Prediction of clinical and mucosal severity of celiac disease and dermatitis herpetiformis by quantification of IgA/IgG serum antibodies to tissue transglutaminase. J Pediatr Gastroenterol Nutr. 2010;50:140–6.PubMedCrossRef
71.
go back to reference Volta U, Granito A, Fiorini E, et al. Usefulness of antibodies to deamidated gliadin peptides in celiac disease diagnosis and follow-up. Dig Dis Sci. 2008;53:1582–8.PubMedCrossRef Volta U, Granito A, Fiorini E, et al. Usefulness of antibodies to deamidated gliadin peptides in celiac disease diagnosis and follow-up. Dig Dis Sci. 2008;53:1582–8.PubMedCrossRef
72.
go back to reference Dahle C, Hagman A, Ignatova S, et al. Antibodies against deamidated gliadin peptides identify adult celiac disease patients negative for antibodies against endomysium and tissue transglutaminase. Aliment Pharmacol Ther. 2010;32:254–60.PubMedCrossRef Dahle C, Hagman A, Ignatova S, et al. Antibodies against deamidated gliadin peptides identify adult celiac disease patients negative for antibodies against endomysium and tissue transglutaminase. Aliment Pharmacol Ther. 2010;32:254–60.PubMedCrossRef
73.
go back to reference Barbato M, Maiellaa G, Camilloa CD, et al. The anti-deamidated gliadin peptide antibodies unmask celiac disease in small children with chronic diarrhoea. Dig Liver Dis. 2011;43:465–9.PubMedCrossRef Barbato M, Maiellaa G, Camilloa CD, et al. The anti-deamidated gliadin peptide antibodies unmask celiac disease in small children with chronic diarrhoea. Dig Liver Dis. 2011;43:465–9.PubMedCrossRef
74.
go back to reference Amarri S, Alvisi P, De Giorgio R, et al. Antibodies to deamidated gliadin peptides: an accurate predictor of celiac disease in infancy. J Clin Immunol. 2013;33:1027–30.PubMedCrossRef Amarri S, Alvisi P, De Giorgio R, et al. Antibodies to deamidated gliadin peptides: an accurate predictor of celiac disease in infancy. J Clin Immunol. 2013;33:1027–30.PubMedCrossRef
75.
go back to reference Schwertz E, Kahlberg F, Sack U, et al. Serologic assay based on gliadin-related nonapeptides as a highly sensitive and specific diagnostic aid in celiac disease. Clin Chem. 2004;50:2370–5.PubMedCrossRef Schwertz E, Kahlberg F, Sack U, et al. Serologic assay based on gliadin-related nonapeptides as a highly sensitive and specific diagnostic aid in celiac disease. Clin Chem. 2004;50:2370–5.PubMedCrossRef
76.
go back to reference Agardh A. Antibodies against synthetic deamidated gliadin peptides and tissue transglutaminase for the identification of childhood celiac disease. Clin Gastroenterol Hepatol. 2007;5:1276–81.PubMedCrossRef Agardh A. Antibodies against synthetic deamidated gliadin peptides and tissue transglutaminase for the identification of childhood celiac disease. Clin Gastroenterol Hepatol. 2007;5:1276–81.PubMedCrossRef
77.
go back to reference Ankelo M, Kleimola V, Simell S, et al. Antibody responses to deamidated gliadin peptide show high specificity and parallel antibodies to tissue transglutaminase in developing celiac disease. Clin Exp Immunol. 2007;150:285–93.PubMedPubMedCentralCrossRef Ankelo M, Kleimola V, Simell S, et al. Antibody responses to deamidated gliadin peptide show high specificity and parallel antibodies to tissue transglutaminase in developing celiac disease. Clin Exp Immunol. 2007;150:285–93.PubMedPubMedCentralCrossRef
78.
go back to reference Rashtak S, Ettore MW, Homburger HA, et al. Comparative usefulness of deamidated gliadin antibodies in the diagnosis of celiac disease. Clin Gastroenetrol Hepatol. 2008;6:426–32.CrossRef Rashtak S, Ettore MW, Homburger HA, et al. Comparative usefulness of deamidated gliadin antibodies in the diagnosis of celiac disease. Clin Gastroenetrol Hepatol. 2008;6:426–32.CrossRef
79.
go back to reference Basso D, Guariso G, Fogar P, et al. Antibodies against synthetic deamidated gliadin peptides for celiac disease diagnosis and follow-up in children. Clin Chem. 2008;55:150–7.PubMedCrossRef Basso D, Guariso G, Fogar P, et al. Antibodies against synthetic deamidated gliadin peptides for celiac disease diagnosis and follow-up in children. Clin Chem. 2008;55:150–7.PubMedCrossRef
80.
go back to reference Sugai E, Moreno ML, Hwang HJ, et al. Celiac disease serology in patients with different pretest probabilities: is biopsy avoidable? World J Gastroenterol. 2010;16:3144–52.PubMedPubMedCentralCrossRef Sugai E, Moreno ML, Hwang HJ, et al. Celiac disease serology in patients with different pretest probabilities: is biopsy avoidable? World J Gastroenterol. 2010;16:3144–52.PubMedPubMedCentralCrossRef
82.
go back to reference Brottveit M, Ráki M, Bergseng E, et al. Assessing possible celiac disease by an HLA-DQ2-gliadin tetramer test. Am J Gastroenterol. 2011;106:1318–24.PubMedCrossRef Brottveit M, Ráki M, Bergseng E, et al. Assessing possible celiac disease by an HLA-DQ2-gliadin tetramer test. Am J Gastroenterol. 2011;106:1318–24.PubMedCrossRef
83.
go back to reference Goletti D, Vincenti D, Carrara S, et al. Selected RD1 peptides for active tuberculosis diagnosis: comparison of a gamma interferon whole-blood enzyme-linked immunosorbent assay and an enzyme-linked immunospot assay. Clin Diagn Lab Immunol. 2005;12:1311–6.PubMedPubMedCentral Goletti D, Vincenti D, Carrara S, et al. Selected RD1 peptides for active tuberculosis diagnosis: comparison of a gamma interferon whole-blood enzyme-linked immunosorbent assay and an enzyme-linked immunospot assay. Clin Diagn Lab Immunol. 2005;12:1311–6.PubMedPubMedCentral
84.
go back to reference Ruhwald M, Bjerregaard-Anderson M, Rabna P, et al. CXCL10/IP-10 release is induced by incubation of whole blood from tuberculosis patients with ESAT-6, CFP10 and TB7.7. Microbes Infect. 2007;9:806–12.PubMedCrossRef Ruhwald M, Bjerregaard-Anderson M, Rabna P, et al. CXCL10/IP-10 release is induced by incubation of whole blood from tuberculosis patients with ESAT-6, CFP10 and TB7.7. Microbes Infect. 2007;9:806–12.PubMedCrossRef
85.
go back to reference Lalvani A, Meroni PL, Millington KA, et al. Recent advances in diagnostic technology: applications in autoimmune and infectious diseases. Clin Exp Rheumatol. 2008;26:S62–6.PubMed Lalvani A, Meroni PL, Millington KA, et al. Recent advances in diagnostic technology: applications in autoimmune and infectious diseases. Clin Exp Rheumatol. 2008;26:S62–6.PubMed
86.
go back to reference Anderson RP, Degano P, Godkin AJ, et al. 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.PubMedCrossRef Anderson RP, Degano P, Godkin AJ, et al. 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.PubMedCrossRef
87.
go back to reference Tye-Din JA, Stewart JA, Dromey JA, et al. Comprehensive, quantitative mapping of T cell epitopes in gluten in celiac disease. Sci Transl Med. 2010;2:41.CrossRef Tye-Din JA, Stewart JA, Dromey JA, et al. Comprehensive, quantitative mapping of T cell epitopes in gluten in celiac disease. Sci Transl Med. 2010;2:41.CrossRef
88.
go back to reference Ontiveros N, Tye-Din JA, Hardy MY, et al. Ex-vivo whole blood secretion of interferon (IFN)-γ and IFN-γ-inducible protein-10 measured by enzyme-linked immunosorbent assay are as sensitive as IFN-γ enzyme-linked immunospot for the detection of gluten-reactive T cells in human leucocyte antigen (HLA)-DQ2.5+-associated celiac disease. Clin Exp Immunol. 2014;175:305–15.PubMedPubMedCentralCrossRef Ontiveros N, Tye-Din JA, Hardy MY, et al. Ex-vivo whole blood secretion of interferon (IFN)-γ and IFN-γ-inducible protein-10 measured by enzyme-linked immunosorbent assay are as sensitive as IFN-γ enzyme-linked immunospot for the detection of gluten-reactive T cells in human leucocyte antigen (HLA)-DQ2.5+-associated celiac disease. Clin Exp Immunol. 2014;175:305–15.PubMedPubMedCentralCrossRef
89.
go back to reference Lehmann PV, Zhang W. Unique strengths of ELISPOT for T cell diagnostics. Methods Mol Biol. 2012;792:3–23.PubMedCrossRef Lehmann PV, Zhang W. Unique strengths of ELISPOT for T cell diagnostics. Methods Mol Biol. 2012;792:3–23.PubMedCrossRef
90.
go back to reference Salazer C, Nagadia R, Pandit P, et al. A novel saliva-based micro-RNA biomarker panel to detect head and neck cancers. Cell Oncol. 2014;37:331–8.CrossRef Salazer C, Nagadia R, Pandit P, et al. A novel saliva-based micro-RNA biomarker panel to detect head and neck cancers. Cell Oncol. 2014;37:331–8.CrossRef
91.
go back to reference Baldini C, Giusti L, Ciregia F, et al. Proteomic analysis of saliva: a unique tool to distinguish primary Sjogren’s syndrome from secondary Sjogren’s syndrome and other sicca syndromes. Arthritis Res Ther. 2011;13(6):R194.PubMedPubMedCentralCrossRef Baldini C, Giusti L, Ciregia F, et al. Proteomic analysis of saliva: a unique tool to distinguish primary Sjogren’s syndrome from secondary Sjogren’s syndrome and other sicca syndromes. Arthritis Res Ther. 2011;13(6):R194.PubMedPubMedCentralCrossRef
92.
go back to reference Jacobs R, Maasdorp E, Malherbe S, et al. Diagnostic potential of novel salivary host biomarkers as candidates for the immunological diagnosis of tuberculosis disease and monitoring of tuberculosis treatment response. PLoS ONE. 2016;12:1311–6. Jacobs R, Maasdorp E, Malherbe S, et al. Diagnostic potential of novel salivary host biomarkers as candidates for the immunological diagnosis of tuberculosis disease and monitoring of tuberculosis treatment response. PLoS ONE. 2016;12:1311–6.
93.
go back to reference Sueki A, Matsuda K, Yamaguchi A, et al. Evaluation of saliva as a diagnostic materials for influenza virus infection by PCR-based assay. Clin Chim Acta. 2016;453:71–4.PubMedCrossRef Sueki A, Matsuda K, Yamaguchi A, et al. Evaluation of saliva as a diagnostic materials for influenza virus infection by PCR-based assay. Clin Chim Acta. 2016;453:71–4.PubMedCrossRef
94.
95.
go back to reference Lenander-Lumikari Ihalin R, Lahteenoja H. Changes in whole saliva in patients with celiac disease. Arch Oral Biol. 2000;45(5):347–54.PubMedCrossRef Lenander-Lumikari Ihalin R, Lahteenoja H. Changes in whole saliva in patients with celiac disease. Arch Oral Biol. 2000;45(5):347–54.PubMedCrossRef
96.
go back to reference Bonamico M, Nenna R, Montuori M, et al. First salivary screening of celiac disease by detection of anti-transglutaminase autoantibody radioimmunoassay in 5000 Italian primary schoolchildren. J Pediatr Gastroenterol Nutr. 2011;52(1):17–20.PubMedCrossRef Bonamico M, Nenna R, Montuori M, et al. First salivary screening of celiac disease by detection of anti-transglutaminase autoantibody radioimmunoassay in 5000 Italian primary schoolchildren. J Pediatr Gastroenterol Nutr. 2011;52(1):17–20.PubMedCrossRef
97.
go back to reference Adornetto G, Fabiani L, Volpe G, et al. An electrochemical immunoassay for the screening of celiac disease in saliva samples. Anal Bioanal Chem. 2015;407:7189–96.PubMedCrossRef Adornetto G, Fabiani L, Volpe G, et al. An electrochemical immunoassay for the screening of celiac disease in saliva samples. Anal Bioanal Chem. 2015;407:7189–96.PubMedCrossRef
98.
go back to reference Actis AB, Perovic NR, Defagó D, et al. Fatty acid profile of human saliva: a possible indicator of dietary fat intake. Arch Oral Biol. 2005;50:1–6.PubMedCrossRef Actis AB, Perovic NR, Defagó D, et al. Fatty acid profile of human saliva: a possible indicator of dietary fat intake. Arch Oral Biol. 2005;50:1–6.PubMedCrossRef
99.
go back to reference Benkebil F, Combescure C, Anghel SI, et al. Diagnostic accuracy of a new point-of-care screening assay for celiac disease. World J Gastroenterol. 2013;19:5111–7.PubMedPubMedCentralCrossRef Benkebil F, Combescure C, Anghel SI, et al. Diagnostic accuracy of a new point-of-care screening assay for celiac disease. World J Gastroenterol. 2013;19:5111–7.PubMedPubMedCentralCrossRef
100.
go back to reference Bienvenu F, Duvanel CB, Seignovert C, et al. Evaluation of a point-of-care test based on deamidated gliadin peptides for celiac disease screening in a large pediatric population. Eur J Gastroenterol Hepatol. 2012. doi:10.1097/meg.0b013e3283582d95.PubMed Bienvenu F, Duvanel CB, Seignovert C, et al. Evaluation of a point-of-care test based on deamidated gliadin peptides for celiac disease screening in a large pediatric population. Eur J Gastroenterol Hepatol. 2012. doi:10.​1097/​meg.​0b013e3283582d95​.PubMed
101.
go back to reference Watanabe C, Komoto S, Hokari R, et al. Prevalence of serum celiac antibody in patients with IBD in Japan. J Gastroenterol. 2014;49:825–34.PubMedCrossRef Watanabe C, Komoto S, Hokari R, et al. Prevalence of serum celiac antibody in patients with IBD in Japan. J Gastroenterol. 2014;49:825–34.PubMedCrossRef
102.
go back to reference Wungjiranirun M, Kelly CP, Leffler DA. Current status of celiac disease drug development. Am J Gastroenterol. 2016;111:779–86.PubMedCrossRef Wungjiranirun M, Kelly CP, Leffler DA. Current status of celiac disease drug development. Am J Gastroenterol. 2016;111:779–86.PubMedCrossRef
103.
go back to reference Soler M, Estevez MC, Moreno Mde L, et al. Label-free SPR detection of gluten peptides in urine for non-invasive celiac disease follow-up. Biosens Bioelectron. 2016;79:158–64.PubMedCrossRef Soler M, Estevez MC, Moreno Mde L, et al. Label-free SPR detection of gluten peptides in urine for non-invasive celiac disease follow-up. Biosens Bioelectron. 2016;79:158–64.PubMedCrossRef
104.
go back to reference Comino I, Fernández-Bañares F, Esteve M, et al. Fecal gluten peptides reveal limitations of serological tests and food questionnaires for monitoring gluten-free diet in celiac disease patients. Am J Gastroenterol. 2016;111:1456–65.PubMedPubMedCentralCrossRef Comino I, Fernández-Bañares F, Esteve M, et al. Fecal gluten peptides reveal limitations of serological tests and food questionnaires for monitoring gluten-free diet in celiac disease patients. Am J Gastroenterol. 2016;111:1456–65.PubMedPubMedCentralCrossRef
105.
106.
go back to reference Fu E, Liang T, Spicar-Mihalic P, et al. Two-dimensional paper network format that enables simple multistep assays for use in low-resource settings in the context of malaria antigen detection. Anal Chem. 2012;84:4574–9.PubMedPubMedCentralCrossRef Fu E, Liang T, Spicar-Mihalic P, et al. Two-dimensional paper network format that enables simple multistep assays for use in low-resource settings in the context of malaria antigen detection. Anal Chem. 2012;84:4574–9.PubMedPubMedCentralCrossRef
107.
go back to reference Veigas B, Jacob JM, Costa MN, et al. Gold on paper–paper platform for Au-nanoprobe TB detection. Lab Chip. 2012;12:4802–8.PubMedCrossRef Veigas B, Jacob JM, Costa MN, et al. Gold on paper–paper platform for Au-nanoprobe TB detection. Lab Chip. 2012;12:4802–8.PubMedCrossRef
Metadata
Title
Celiac disease: from etiological factors to evolving diagnostic approaches
Authors
Anantdeep Kaur
Olga Shimoni
Michael Wallach
Publication date
01-09-2017
Publisher
Springer Japan
Published in
Journal of Gastroenterology / Issue 9/2017
Print ISSN: 0944-1174
Electronic ISSN: 1435-5922
DOI
https://doi.org/10.1007/s00535-017-1357-7

Other articles of this Issue 9/2017

Journal of Gastroenterology 9/2017 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine