Skip to main content
Top
Published in: Clinical Reviews in Allergy & Immunology 1/2019

01-08-2019

Transglutaminase 2 and Transglutaminase 2 Autoantibodies in Celiac Disease: a Review

Authors: Tiina Rauhavirta, Minna Hietikko, Teea Salmi, Katri Lindfors

Published in: Clinical Reviews in Allergy & Immunology | Issue 1/2019

Login to get access

Abstract

Celiac disease is a common inflammatory disorder with a prevalence of 1–2 % in which a distinct dietary wheat, rye, and barley component, gluten, induces small-bowel mucosal villous atrophy, crypt hyperplasia, and inflammation. The small-bowel mucosal damage can be reversed by a strict lifelong gluten-free diet, which is currently the only effective treatment for the condition. A key player in the pathogenetic process leading to the enteropathy is played by a protein called transglutaminase 2 (TG2), which is able to enzymatically modify gluten-derived gliadin peptides. The TG2-catalyzed deamidation of the gliadin peptides results in their increased binding affinity to the disease-predisposing human leukocyte antigen (HLA) DQ2 and DQ8 molecules, thus enabling a strong immune response to be launched. Blocking the enzymatic activity of TG2 has thus been suggested as a suitable novel pharmacological approach to treat celiac disease. By virtue of its transamidation capacity, TG2 is also able to cross-link gliadin peptides to itself, this resulting in the generation of TG2-gliadin peptide complexes whose presence might provide an explanation for the generation of the TG2 autoantibodies characteristic of celiac disease. Due to their excellent specificity for the disorder, the TG2-targeted autoantibodies are widely used in the diagnostics as a first-line test to select patients for gastrointestinal endoscopy. More recently, it has come to be appreciated that these autoantibodies and also the TG2-specific B cells might play an active role in the disease pathogenesis. In this review, we assess the role of TG2, TG2-specific B cells, and autoantibodies in celiac disease.
Literature
1.
go back to reference Lorand L, Graham R (2003) Transglutaminases: crosslinking enzymes with pleiotropic functions. Nat Rev Mol Cell Biol 4:140–156CrossRefPubMed Lorand L, Graham R (2003) Transglutaminases: crosslinking enzymes with pleiotropic functions. Nat Rev Mol Cell Biol 4:140–156CrossRefPubMed
3.
go back to reference Sarkar NK, Clarke DD, Waelsch H (1957) An enzymically catalyzed incorporation of amines into proteins. Biochim Biophys Acta 25:451–452CrossRefPubMed Sarkar NK, Clarke DD, Waelsch H (1957) An enzymically catalyzed incorporation of amines into proteins. Biochim Biophys Acta 25:451–452CrossRefPubMed
4.
go back to reference Folk JEFJ (1977) The epsilon-(gamma-glutamyl)lysine crosslink and the catalytic role of transglutaminases. Adv Protein Chem 31:1–133CrossRefPubMed Folk JEFJ (1977) The epsilon-(gamma-glutamyl)lysine crosslink and the catalytic role of transglutaminases. Adv Protein Chem 31:1–133CrossRefPubMed
5.
go back to reference Gentile V, Davies P, Baldini A (1994) The human tissue transglutaminase gene maps on chromosome 20q12 by in situ fluorescence hybridization. Genomics 20:295–297CrossRefPubMed Gentile V, Davies P, Baldini A (1994) The human tissue transglutaminase gene maps on chromosome 20q12 by in situ fluorescence hybridization. Genomics 20:295–297CrossRefPubMed
6.
go back to reference Liu S, Cerione R, Clardy J (2002) Structural basis for the guanine nucleotide-binding activity of tissue transglutaminase and its regulation of transamidation activity. Proc Natl Acad Sci U S A 99:2743–2747CrossRefPubMedPubMedCentral Liu S, Cerione R, Clardy J (2002) Structural basis for the guanine nucleotide-binding activity of tissue transglutaminase and its regulation of transamidation activity. Proc Natl Acad Sci U S A 99:2743–2747CrossRefPubMedPubMedCentral
7.
go back to reference Pinkas DM, Strop P, Brunger AT, Khosla C (2007) Transglutaminase 2 undergoes a large conformational change upon activation. PLoS Biol 5:2788–2796CrossRef Pinkas DM, Strop P, Brunger AT, Khosla C (2007) Transglutaminase 2 undergoes a large conformational change upon activation. PLoS Biol 5:2788–2796CrossRef
8.
go back to reference Lai T, Greenberg CS (2013) TGM2 and implications for human disease: role of alternative splicing. Front Biosci 18:504–519CrossRef Lai T, Greenberg CS (2013) TGM2 and implications for human disease: role of alternative splicing. Front Biosci 18:504–519CrossRef
10.
go back to reference Davies PJ, Murtaugh MP, Moore WT Jr, Johnson GS, Lucas D (1985) Retinoic acid-induced expression of tissue transglutaminase in human promyelocytic leukemia (HL-60) cells. J Biol Chem 260:5166–5174PubMed Davies PJ, Murtaugh MP, Moore WT Jr, Johnson GS, Lucas D (1985) Retinoic acid-induced expression of tissue transglutaminase in human promyelocytic leukemia (HL-60) cells. J Biol Chem 260:5166–5174PubMed
11.
go back to reference Park D, Choi SS, Ha K (2010) Transglutaminase 2: a multi-functional protein in multiple subcellular compartments. Amino Acids 39:619–631CrossRefPubMed Park D, Choi SS, Ha K (2010) Transglutaminase 2: a multi-functional protein in multiple subcellular compartments. Amino Acids 39:619–631CrossRefPubMed
12.
go back to reference Zemskov EA, Mikhailenko I, Hsia R, Zaritskaya L, Belkin AM (2011) Unconventional secretion of tissue transglutaminase involves phospholipid-dependent delivery into recycling endosomes. FEBS J 278:96–96 Zemskov EA, Mikhailenko I, Hsia R, Zaritskaya L, Belkin AM (2011) Unconventional secretion of tissue transglutaminase involves phospholipid-dependent delivery into recycling endosomes. FEBS J 278:96–96
13.
go back to reference Achyuthan KE, Greenberg CS (1987) Identification of a guanosine triphosphate-binding site on guinea pig liver transglutaminase. Role of GTP and calcium ions in modulating activity. J Biol Chem 262:1901–1906PubMed Achyuthan KE, Greenberg CS (1987) Identification of a guanosine triphosphate-binding site on guinea pig liver transglutaminase. Role of GTP and calcium ions in modulating activity. J Biol Chem 262:1901–1906PubMed
14.
15.
go back to reference Siegel M, Strnad P, Watts RE, Choi K, Jabri B, Omary MB, Khosla C (2008) Extracellular transglutaminase 2 is catalytically inactive, but is transiently activated upon tissue injury. PLoS One 3:e1861CrossRefPubMedPubMedCentral Siegel M, Strnad P, Watts RE, Choi K, Jabri B, Omary MB, Khosla C (2008) Extracellular transglutaminase 2 is catalytically inactive, but is transiently activated upon tissue injury. PLoS One 3:e1861CrossRefPubMedPubMedCentral
16.
go back to reference Jin X, Stamnaes J, Klock C, DiRaimondo TR, Sollid LM, Khosla C (2011) Activation of extracellular transglutaminase 2 by thioredoxin. J Biol Chem 286:37866–37873CrossRefPubMedPubMedCentral Jin X, Stamnaes J, Klock C, DiRaimondo TR, Sollid LM, Khosla C (2011) Activation of extracellular transglutaminase 2 by thioredoxin. J Biol Chem 286:37866–37873CrossRefPubMedPubMedCentral
17.
go back to reference Lai T, Hausladen A, Slaughter T, Eu J, Stamler J, Greenberg C (2001) Calcium regulates S-nitrosylation, denitrosylation, and activity of tissue transglutaminase. Biotechnology (N Y) 40:4904–4910 Lai T, Hausladen A, Slaughter T, Eu J, Stamler J, Greenberg C (2001) Calcium regulates S-nitrosylation, denitrosylation, and activity of tissue transglutaminase. Biotechnology (N Y) 40:4904–4910
18.
go back to reference Mishra S, Murphy L (2004) Tissue transglutaminase has intrinsic kinase activity—identification of transglutaminase 2 as an insulin-like growth factor-binding protein-3 kinase. J Biol Chem 279:23863–23868CrossRefPubMed Mishra S, Murphy L (2004) Tissue transglutaminase has intrinsic kinase activity—identification of transglutaminase 2 as an insulin-like growth factor-binding protein-3 kinase. J Biol Chem 279:23863–23868CrossRefPubMed
19.
go back to reference Mishra S, Melino G, Murphy LJ (2007) Transglutaminase 2 kinase activity facilitates protein kinase A-induced phosphorylation of retinoblastoma protein. J Biol Chem 282:18108–18115CrossRefPubMed Mishra S, Melino G, Murphy LJ (2007) Transglutaminase 2 kinase activity facilitates protein kinase A-induced phosphorylation of retinoblastoma protein. J Biol Chem 282:18108–18115CrossRefPubMed
20.
go back to reference Wang Z, Griffin M (2012) TG2, a novel extracellular protein with multiple functions. Amino Acids 42:939–949CrossRefPubMed Wang Z, Griffin M (2012) TG2, a novel extracellular protein with multiple functions. Amino Acids 42:939–949CrossRefPubMed
21.
go back to reference Esposito C, Caputo I (2005) Mammalian transglutaminases—identification of substrates as a key to physiological function and physiopathological relevance. FEBS J 272:615–631CrossRefPubMed Esposito C, Caputo I (2005) Mammalian transglutaminases—identification of substrates as a key to physiological function and physiopathological relevance. FEBS J 272:615–631CrossRefPubMed
22.
go back to reference Kanchan K, Fuxreiter M, Fésüs L (2015) Physiological, pathological, and structural implications of non-enzymatic protein–protein interactions of the multifunctional human transglutaminase 2. Cell Mol Life Sci 72:3009–3035CrossRefPubMed Kanchan K, Fuxreiter M, Fésüs L (2015) Physiological, pathological, and structural implications of non-enzymatic protein–protein interactions of the multifunctional human transglutaminase 2. Cell Mol Life Sci 72:3009–3035CrossRefPubMed
23.
go back to reference Fleckenstein B, Molberg Y, Qiao S, Schmid D, von der Mullbe F, Elgstoen K, Jung G, Sollid L (2002) Gliadin T cell epitope selection by tissue transglutaminase in celiac disease—role of enzyme specificity and pH influence on the transamidation versus deamidation reactions. J Biol Chem 277:34109–34116CrossRefPubMed Fleckenstein B, Molberg Y, Qiao S, Schmid D, von der Mullbe F, Elgstoen K, Jung G, Sollid L (2002) Gliadin T cell epitope selection by tissue transglutaminase in celiac disease—role of enzyme specificity and pH influence on the transamidation versus deamidation reactions. J Biol Chem 277:34109–34116CrossRefPubMed
24.
go back to reference Nakaoka H, Perez D, Baek K, Das T, Husain A, Misono K, Im M, Graham R (1994) G(h)—a Gtp-binding protein with transglutaminase activity and receptor signaling function. Science 264:1593–1596CrossRefPubMed Nakaoka H, Perez D, Baek K, Das T, Husain A, Misono K, Im M, Graham R (1994) G(h)—a Gtp-binding protein with transglutaminase activity and receptor signaling function. Science 264:1593–1596CrossRefPubMed
25.
go back to reference Iismaa SE, Chung L, Wu M, Teller DC, Yee VC, Graham RM (1997) The core domain of the tissue transglutaminase Gh hydrolyzes GTP and ATP. Biochemistry 36:11655–1166 4CrossRefPubMed Iismaa SE, Chung L, Wu M, Teller DC, Yee VC, Graham RM (1997) The core domain of the tissue transglutaminase Gh hydrolyzes GTP and ATP. Biochemistry 36:11655–1166 4CrossRefPubMed
26.
go back to reference Hasegawa G, Suwa M, Ichikawa Y, Ohtsuka T, Kumagai S, Kikuchi M, Sato Y, Saito Y (2003) A novel function of tissue-type transglutaminase: protein disulphide isomerase. Biochem J 373:793–803CrossRefPubMedPubMedCentral Hasegawa G, Suwa M, Ichikawa Y, Ohtsuka T, Kumagai S, Kikuchi M, Sato Y, Saito Y (2003) A novel function of tissue-type transglutaminase: protein disulphide isomerase. Biochem J 373:793–803CrossRefPubMedPubMedCentral
27.
go back to reference Mishra S, Saleh A, Espino PS, Davie JR, Murphy LJ (2006) Phosphorylation of histones by tissue transglutaminase. J Biol Chem 281:5532–5538CrossRefPubMed Mishra S, Saleh A, Espino PS, Davie JR, Murphy LJ (2006) Phosphorylation of histones by tissue transglutaminase. J Biol Chem 281:5532–5538CrossRefPubMed
28.
go back to reference Akimov S, Krylov D, Fleischman L, Belkin A (2000) Tissue transglutaminase is an integrin-binding adhesion coreceptor for fibronectin. J Cell Biol 148:825–838CrossRefPubMedPubMedCentral Akimov S, Krylov D, Fleischman L, Belkin A (2000) Tissue transglutaminase is an integrin-binding adhesion coreceptor for fibronectin. J Cell Biol 148:825–838CrossRefPubMedPubMedCentral
29.
go back to reference Akimov S, Belkin A (2001) Cell surface tissue transglutaminase is involved in adhesion and migration of monocytic cells on fibronectin. Blood 98:1567–1576CrossRefPubMed Akimov S, Belkin A (2001) Cell surface tissue transglutaminase is involved in adhesion and migration of monocytic cells on fibronectin. Blood 98:1567–1576CrossRefPubMed
30.
go back to reference Iismaa SE, Mearns BM, Lorand L, Graham RM (2009) Transglutaminases and disease: lessons from genetically engineered mouse models and inherited disorders. Physiol Rev 89:991–1023CrossRefPubMed Iismaa SE, Mearns BM, Lorand L, Graham RM (2009) Transglutaminases and disease: lessons from genetically engineered mouse models and inherited disorders. Physiol Rev 89:991–1023CrossRefPubMed
31.
go back to reference Sulkanen S, Halttunen T, Laurila K, Kolho K, Korponay-Szabó IR, Sarnesto A, Savilahti E, Collin P, Mäki M (1998) Tissue transglutaminase autoantibody enzyme-linked immunosorbent assay in detecting celiac disease. Gastroenterology 115:1322–1328CrossRefPubMed Sulkanen S, Halttunen T, Laurila K, Kolho K, Korponay-Szabó IR, Sarnesto A, Savilahti E, Collin P, Mäki M (1998) Tissue transglutaminase autoantibody enzyme-linked immunosorbent assay in detecting celiac disease. Gastroenterology 115:1322–1328CrossRefPubMed
32.
go back to reference Chorzelski TP, Sulej J, Tchorzewska H, Jablonska S, Beutner EH, Kumar V (1983) IgA class endomysium antibodies in dermatitis herpetiformis and coeliac disease a. Ann N Y Acad Sci 420:325–334CrossRefPubMed Chorzelski TP, Sulej J, Tchorzewska H, Jablonska S, Beutner EH, Kumar V (1983) IgA class endomysium antibodies in dermatitis herpetiformis and coeliac disease a. Ann N Y Acad Sci 420:325–334CrossRefPubMed
33.
go back to reference Rashtak S, Ettore MW, Homburger HA, Murray JA (2008) Comparative usefulness of deamidated gliadin antibodies in the diagnosis of celiac disease. Clin Gastroenterol Hepatol 6:426–432CrossRefPubMedPubMedCentral Rashtak S, Ettore MW, Homburger HA, Murray JA (2008) Comparative usefulness of deamidated gliadin antibodies in the diagnosis of celiac disease. Clin Gastroenterol Hepatol 6:426–432CrossRefPubMedPubMedCentral
34.
go back to reference Mustalahti K, Catassi C, Reunanen A, Fabiani E, Heier M, McMillan S, Murray L, Metzger M, Gasparin M, Bravi E (2010) The prevalence of celiac disease in Europe: results of a centralized, international mass screening project. Ann Med 42:587–595CrossRefPubMed Mustalahti K, Catassi C, Reunanen A, Fabiani E, Heier M, McMillan S, Murray L, Metzger M, Gasparin M, Bravi E (2010) The prevalence of celiac disease in Europe: results of a centralized, international mass screening project. Ann Med 42:587–595CrossRefPubMed
35.
go back to reference Fasano A, Berti I, Gerarduzzi T, Not T, Colletti RB, Drago S, Elitsur Y, Green PH, Guandalini S, Hill ID (2003) Prevalence of celiac disease in at-risk and not-at-risk groups in the United States: a large multicenter study. Arch Intern Med 163:286–292CrossRefPubMed Fasano A, Berti I, Gerarduzzi T, Not T, Colletti RB, Drago S, Elitsur Y, Green PH, Guandalini S, Hill ID (2003) Prevalence of celiac disease in at-risk and not-at-risk groups in the United States: a large multicenter study. Arch Intern Med 163:286–292CrossRefPubMed
36.
go back to reference Lohi S, Mustalahti K, Kaukinen K, Laurila K, Collin P, Rissanen H, Lohi O, Bravi E, Gasparin M, Reunanen A (2007) Increasing prevalence of coeliac disease over time. Aliment Pharmacol Ther 26:1217–1225CrossRefPubMed Lohi S, Mustalahti K, Kaukinen K, Laurila K, Collin P, Rissanen H, Lohi O, Bravi E, Gasparin M, Reunanen A (2007) Increasing prevalence of coeliac disease over time. Aliment Pharmacol Ther 26:1217–1225CrossRefPubMed
37.
38.
go back to reference Mäki M, Mustalahti K, Kokkonen J, Kulmala P, Haapalahti M, Karttunen T, Ilonen J, Laurila K, Dahlbom I, Hansson T (2003) Prevalence of celiac disease among children in Finland. N Engl J Med 348:2517–2524CrossRefPubMed Mäki M, Mustalahti K, Kokkonen J, Kulmala P, Haapalahti M, Karttunen T, Ilonen J, Laurila K, Dahlbom I, Hansson T (2003) Prevalence of celiac disease among children in Finland. N Engl J Med 348:2517–2524CrossRefPubMed
39.
go back to reference Vilppula A, Kaukinen K, Luostarinen L, Krekelä I, Patrikainen H, Valve R, Mäki M, Collin P (2009) Increasing prevalence and high incidence of celiac disease in elderly people: a population-based study. BMC Gastroenterol 9:1CrossRef Vilppula A, Kaukinen K, Luostarinen L, Krekelä I, Patrikainen H, Valve R, Mäki M, Collin P (2009) Increasing prevalence and high incidence of celiac disease in elderly people: a population-based study. BMC Gastroenterol 9:1CrossRef
40.
go back to reference Tack GJ, Verbeek WH, Schreurs MW, Mulder CJ (2010) The spectrum of celiac disease: epidemiology, clinical aspects and treatment. Nat Rev Gastroenterol Hepatol 7:204–213CrossRefPubMed Tack GJ, Verbeek WH, Schreurs MW, Mulder CJ (2010) The spectrum of celiac disease: epidemiology, clinical aspects and treatment. Nat Rev Gastroenterol Hepatol 7:204–213CrossRefPubMed
41.
go back to reference Sollid LM (2004) Intraepithelial lymphocytes in celiac disease: license to kill revealed. Immunity 21:303–304PubMed Sollid LM (2004) Intraepithelial lymphocytes in celiac disease: license to kill revealed. Immunity 21:303–304PubMed
42.
go back to reference Akobeng AK, Ramanan AV, Buchan I, Heller RF (2006) Effect of breast feeding on risk of coeliac disease: a systematic review and meta-analysis of observational studies. Arch Dis Child 91:39–43CrossRefPubMed Akobeng AK, Ramanan AV, Buchan I, Heller RF (2006) Effect of breast feeding on risk of coeliac disease: a systematic review and meta-analysis of observational studies. Arch Dis Child 91:39–43CrossRefPubMed
43.
go back to reference Silano M, Agostoni C, Guandalini S (2010) Effect of the timing of gluten introduction on the development of celiac disease. World J Gastroenterol 16:1939–1942CrossRefPubMedPubMedCentral Silano M, Agostoni C, Guandalini S (2010) Effect of the timing of gluten introduction on the development of celiac disease. World J Gastroenterol 16:1939–1942CrossRefPubMedPubMedCentral
44.
go back to reference Vriezinga SL, Auricchio R, Bravi E, Castillejo G, Chmielewska A, Crespo Escobar P, Kolaček S, Koletzko S, Korponay-Szabo IR, Mummert E (2014) Randomized feeding intervention in infants at high risk for celiac disease. N Engl J Med 371:1304–1315CrossRefPubMed Vriezinga SL, Auricchio R, Bravi E, Castillejo G, Chmielewska A, Crespo Escobar P, Kolaček S, Koletzko S, Korponay-Szabo IR, Mummert E (2014) Randomized feeding intervention in infants at high risk for celiac disease. N Engl J Med 371:1304–1315CrossRefPubMed
45.
go back to reference Silano M, Agostoni C, Sanz Y, Guandalini S (2016) Infant feeding and risk of developing celiac disease: a systematic review. BMJ Open 6:e009163–2015-009163CrossRef Silano M, Agostoni C, Sanz Y, Guandalini S (2016) Infant feeding and risk of developing celiac disease: a systematic review. BMJ Open 6:e009163–2015-009163CrossRef
46.
go back to reference Mårild K, Kahrs CR, Tapia G, Stene LC, Størdal K (2015) Infections and risk of celiac disease in childhood: a prospective nationwide cohort study. Am J Gastroenterol 110:1475–1484CrossRefPubMed Mårild K, Kahrs CR, Tapia G, Stene LC, Størdal K (2015) Infections and risk of celiac disease in childhood: a prospective nationwide cohort study. Am J Gastroenterol 110:1475–1484CrossRefPubMed
47.
go back to reference Stene LC, Honeyman MC, Hoffenberg EJ, Haas JE, Sokol RJ, Emery L, Taki I, Norris JM, Erlich HA, Eisenbarth GS (2006) Rotavirus infection frequency and risk of celiac disease autoimmunity in early childhood: a longitudinal study. Am J Gastroenterol 101:2333–2340CrossRefPubMed Stene LC, Honeyman MC, Hoffenberg EJ, Haas JE, Sokol RJ, Emery L, Taki I, Norris JM, Erlich HA, Eisenbarth GS (2006) Rotavirus infection frequency and risk of celiac disease autoimmunity in early childhood: a longitudinal study. Am J Gastroenterol 101:2333–2340CrossRefPubMed
48.
go back to reference Plot L, Amital H, Barzilai O, Ram M, Nicola B, Shoenfeld Y (2009) Infections may have a protective role in the etiopathogenesis of celiac disease. Ann N Y Acad Sci 1173:670–674CrossRefPubMed Plot L, Amital H, Barzilai O, Ram M, Nicola B, Shoenfeld Y (2009) Infections may have a protective role in the etiopathogenesis of celiac disease. Ann N Y Acad Sci 1173:670–674CrossRefPubMed
49.
go back to reference Kondrashova A, Mustalahti K, Kaukinen K, Viskari H, Volodicheva V, Haapala A, Ilonen J, Knip M, Mäki M, Hyöty H (2008) Lower economic status and inferior hygienic environment may protect against celiac disease. Ann Med 40:223–231CrossRefPubMed Kondrashova A, Mustalahti K, Kaukinen K, Viskari H, Volodicheva V, Haapala A, Ilonen J, Knip M, Mäki M, Hyöty H (2008) Lower economic status and inferior hygienic environment may protect against celiac disease. Ann Med 40:223–231CrossRefPubMed
50.
go back to reference Dieli-Crimi R, Cénit MC, Núñez C (2015) The genetics of celiac disease: a comprehensive review of clinical implications. J Autoimmun 64:26–41CrossRefPubMed Dieli-Crimi R, Cénit MC, Núñez C (2015) The genetics of celiac disease: a comprehensive review of clinical implications. J Autoimmun 64:26–41CrossRefPubMed
51.
go back to reference Hadithi M, Von Blomberg B, Mary E, Crusius JBA, Bloemena E, Kostense PJ, Meijer JW, Mulder CJ, Stehouwer CD (2007) Accuracy of serologic tests and HLA-DQ typing for diagnosing celiac disease. Ann Intern Med 147:294–302CrossRefPubMed Hadithi M, Von Blomberg B, Mary E, Crusius JBA, Bloemena E, Kostense PJ, Meijer JW, Mulder CJ, Stehouwer CD (2007) Accuracy of serologic tests and HLA-DQ typing for diagnosing celiac disease. Ann Intern Med 147:294–302CrossRefPubMed
52.
go back to reference Romanos J, Rosen A, Kumar V, Trynka G, Franke L, Szperl A, Gutierrez-Achury J, van Diemen CC, Kanninga R, SA J, Steck A, Eisenbarth G, van Heel DA, Cukrowska B, Bruno V, Mazzilli MC, Nunez C, Bilbao JR, Mearin ML, Barisani D, Rewers M, Norris JM, Ivarsson A, Boezen HM, Liu E, Wijmenga C, Prevent CD Group (2014) Improving coeliac disease risk prediction by testing non-HLA variants additional to HLA variants. Gut 63:415–422CrossRefPubMed Romanos J, Rosen A, Kumar V, Trynka G, Franke L, Szperl A, Gutierrez-Achury J, van Diemen CC, Kanninga R, SA J, Steck A, Eisenbarth G, van Heel DA, Cukrowska B, Bruno V, Mazzilli MC, Nunez C, Bilbao JR, Mearin ML, Barisani D, Rewers M, Norris JM, Ivarsson A, Boezen HM, Liu E, Wijmenga C, Prevent CD Group (2014) Improving coeliac disease risk prediction by testing non-HLA variants additional to HLA variants. Gut 63:415–422CrossRefPubMed
53.
go back to reference Kivelä L, Kaukinen K, Lähdeaho M, Huhtala H, Ashorn M, Ruuska T, Hiltunen P, Visakorpi J, Mäki M, Kurppa K (2015) Presentation of celiac disease in Finnish children is no longer changing: a 50-year perspective. J Pediatr 167:1109–1115CrossRefPubMed Kivelä L, Kaukinen K, Lähdeaho M, Huhtala H, Ashorn M, Ruuska T, Hiltunen P, Visakorpi J, Mäki M, Kurppa K (2015) Presentation of celiac disease in Finnish children is no longer changing: a 50-year perspective. J Pediatr 167:1109–1115CrossRefPubMed
54.
go back to reference Garampazzi A, Rapa A, Mura S, Capelli A, Valori A, Boldorini R, Oderda G (2007) Clinical pattern of celiac disease is still changing. J Pediatr Gastroenterol Nutr 45:611–614CrossRefPubMed Garampazzi A, Rapa A, Mura S, Capelli A, Valori A, Boldorini R, Oderda G (2007) Clinical pattern of celiac disease is still changing. J Pediatr Gastroenterol Nutr 45:611–614CrossRefPubMed
55.
go back to reference Leffler DA, Green PH, Fasano A (2015) Extraintestinal manifestations of coeliac disease. Nat Rev Gastroenterol Hepatol 12:561–571CrossRefPubMed Leffler DA, Green PH, Fasano A (2015) Extraintestinal manifestations of coeliac disease. Nat Rev Gastroenterol Hepatol 12:561–571CrossRefPubMed
56.
go back to reference Reunala T, Salmi TT, Hervonen K (2015) Dermatitis herpetiformis: pathognomonic transglutaminase IgA deposits in the skin and excellent prognosis on a gluten-free diet. Acta Derm Venereol 95:917–922CrossRefPubMed Reunala T, Salmi TT, Hervonen K (2015) Dermatitis herpetiformis: pathognomonic transglutaminase IgA deposits in the skin and excellent prognosis on a gluten-free diet. Acta Derm Venereol 95:917–922CrossRefPubMed
57.
go back to reference Dieterich W, Laag E, Bruckner-Tuderman L, Reunala T, Kárpáti S, Zágoni T, Riecken EO, Schuppan D (1999) Antibodies to tissue transglutaminase as serologic markers in patients with dermatitis herpetiformis. J Investig Dermatol 113:133–136CrossRefPubMed Dieterich W, Laag E, Bruckner-Tuderman L, Reunala T, Kárpáti S, Zágoni T, Riecken EO, Schuppan D (1999) Antibodies to tissue transglutaminase as serologic markers in patients with dermatitis herpetiformis. J Investig Dermatol 113:133–136CrossRefPubMed
58.
go back to reference Salmi TT, Hervonen K, Laurila K, Collin P, MäKI M, Koskinen O, Huhtala H, Kaukinen K, Reunala T (2014) Small bowel transglutaminase 2-specific IgA deposits in dermatitis herpetiformis. Acta Derm Venereol 94:393–397CrossRefPubMed Salmi TT, Hervonen K, Laurila K, Collin P, MäKI M, Koskinen O, Huhtala H, Kaukinen K, Reunala T (2014) Small bowel transglutaminase 2-specific IgA deposits in dermatitis herpetiformis. Acta Derm Venereol 94:393–397CrossRefPubMed
59.
go back to reference Sardy M, Karpati S, Merkl B, Paulsson M, Smyth N (2002) Epidermal transglutaminase (TGase 3) is the autoantigen of dermatitis herpetiformis. J Exp Med 195:747–757CrossRefPubMedPubMedCentral Sardy M, Karpati S, Merkl B, Paulsson M, Smyth N (2002) Epidermal transglutaminase (TGase 3) is the autoantigen of dermatitis herpetiformis. J Exp Med 195:747–757CrossRefPubMedPubMedCentral
60.
go back to reference Hadjivassiliou M, Aeschlimann P, Strigun A, Sanders DS, Woodroofe N, Aeschlimann D (2008) Autoantibodies in gluten ataxia recognize a novel neuronal transglutaminase. Ann Neurol 64:332–343CrossRefPubMed Hadjivassiliou M, Aeschlimann P, Strigun A, Sanders DS, Woodroofe N, Aeschlimann D (2008) Autoantibodies in gluten ataxia recognize a novel neuronal transglutaminase. Ann Neurol 64:332–343CrossRefPubMed
61.
go back to reference Hadjivassiliou M, Sanders DS, Grünewald RA, Woodroofe N, Boscolo S, Aeschlimann D (2010) Gluten sensitivity: from gut to brain. Lancet Neurol 9:318–330CrossRefPubMed Hadjivassiliou M, Sanders DS, Grünewald RA, Woodroofe N, Boscolo S, Aeschlimann D (2010) Gluten sensitivity: from gut to brain. Lancet Neurol 9:318–330CrossRefPubMed
63.
go back to reference Bardella MT, Vecchi M, Conte D, Del Ninno E, Fraquelli M, Pacchetti S, Minola E, Landoni M, Cesana BM, De Franchis R (1999) Chronic unexplained hypertransaminasemia may be caused by occult celiac disease. Hepatology 29:654–657CrossRefPubMed Bardella MT, Vecchi M, Conte D, Del Ninno E, Fraquelli M, Pacchetti S, Minola E, Landoni M, Cesana BM, De Franchis R (1999) Chronic unexplained hypertransaminasemia may be caused by occult celiac disease. Hepatology 29:654–657CrossRefPubMed
64.
go back to reference Kaukinen K, Halme L, Collin P, Färkkilä M, Mäki M, Vehmanen P, Partanen J, Höckerstedt K (2002) Celiac disease in patients with severe liver disease: gluten-free diet may reverse hepatic failure. Gastroenterology 122:881–888CrossRefPubMed Kaukinen K, Halme L, Collin P, Färkkilä M, Mäki M, Vehmanen P, Partanen J, Höckerstedt K (2002) Celiac disease in patients with severe liver disease: gluten-free diet may reverse hepatic failure. Gastroenterology 122:881–888CrossRefPubMed
65.
go back to reference Mazure R, Vazquez H, Gonzalez D, Mautalen C, Pedreira S, Boerr L, Bai JC (1994) Bone mineral affection in asymptomatic adult patients with celiac disease. Am J Gastroenterol 89:2130–2134PubMed Mazure R, Vazquez H, Gonzalez D, Mautalen C, Pedreira S, Boerr L, Bai JC (1994) Bone mineral affection in asymptomatic adult patients with celiac disease. Am J Gastroenterol 89:2130–2134PubMed
66.
go back to reference Vazquez H, Mazure R, Gonzalez D, Flores D, Pedreira S, Niveloni S, Smecuol E, Mauriño E, Bai JC (2000) Risk of fractures in celiac disease patients: a cross-sectional, case-control study. Am J Gastroenterol 95:183–189CrossRef Vazquez H, Mazure R, Gonzalez D, Flores D, Pedreira S, Niveloni S, Smecuol E, Mauriño E, Bai JC (2000) Risk of fractures in celiac disease patients: a cross-sectional, case-control study. Am J Gastroenterol 95:183–189CrossRef
67.
go back to reference Saccone G, Berghella V, Sarno L, Maruotti GM, Cetin I, Greco L, Khashan AS, McCarthy F, Martinelli D, Fortunato F (2015) Celiac disease and obstetric complications: a systematic review and metaanalysis. Obstet Gynecol 4:225–234 Saccone G, Berghella V, Sarno L, Maruotti GM, Cetin I, Greco L, Khashan AS, McCarthy F, Martinelli D, Fortunato F (2015) Celiac disease and obstetric complications: a systematic review and metaanalysis. Obstet Gynecol 4:225–234
68.
go back to reference Ventura A, Magazzù G, Greco L (1999) Duration of exposure to gluten and risk for autoimmune disorders in patients with celiac disease. Gastroenterology 117:297–303CrossRefPubMed Ventura A, Magazzù G, Greco L (1999) Duration of exposure to gluten and risk for autoimmune disorders in patients with celiac disease. Gastroenterology 117:297–303CrossRefPubMed
69.
go back to reference Viljamaa M, Kaukinen K, Huhtala H, Kyrönpalo S, Rasmussen M, Collin P (2005) Coeliac disease, autoimmune diseases and gluten exposure. Scand J Gastroenterol 40:437–443CrossRefPubMed Viljamaa M, Kaukinen K, Huhtala H, Kyrönpalo S, Rasmussen M, Collin P (2005) Coeliac disease, autoimmune diseases and gluten exposure. Scand J Gastroenterol 40:437–443CrossRefPubMed
70.
go back to reference Mårild K, Stephansson O, Grahnquist L, Cnattingius S, Söderman G, Ludvigsson JF (2013) Down syndrome is associated with elevated risk of celiac disease: a nationwide case-control study. J Pediatr 163:237–242CrossRefPubMed Mårild K, Stephansson O, Grahnquist L, Cnattingius S, Söderman G, Ludvigsson JF (2013) Down syndrome is associated with elevated risk of celiac disease: a nationwide case-control study. J Pediatr 163:237–242CrossRefPubMed
71.
go back to reference Frost AR, Band MM, Conway GS (2009) Serological screening for coeliac disease in adults with Turner’s syndrome: prevalence and clinical significance of endomysium antibody positivity. Eur J Endocrinol 160:675–679CrossRefPubMed Frost AR, Band MM, Conway GS (2009) Serological screening for coeliac disease in adults with Turner’s syndrome: prevalence and clinical significance of endomysium antibody positivity. Eur J Endocrinol 160:675–679CrossRefPubMed
72.
go back to reference Walker-Smith J, Guandalini S, Schmitz J, Shmerling D, Visakorpi J (1990) Revised criteria for diagnosis of coeliac disease. Arch Dis Child 65:909–911CrossRef Walker-Smith J, Guandalini S, Schmitz J, Shmerling D, Visakorpi J (1990) Revised criteria for diagnosis of coeliac disease. Arch Dis Child 65:909–911CrossRef
73.
go back to reference Taavela J, Koskinen O, Huhtala H, Lähdeaho M, Popp A, Laurila K, Collin P, Kaukinen K, Kurppa K, Mäki M (2013) Validation of morphometric analyses of small-intestinal biopsy readouts in celiac disease. PLoS One 8:e76163CrossRefPubMedPubMedCentral Taavela J, Koskinen O, Huhtala H, Lähdeaho M, Popp A, Laurila K, Collin P, Kaukinen K, Kurppa K, Mäki M (2013) Validation of morphometric analyses of small-intestinal biopsy readouts in celiac disease. PLoS One 8:e76163CrossRefPubMedPubMedCentral
74.
go back to reference Salmi T, Collin P, Reunala T, Mäki M, Kaukinen K (2010) Diagnostic methods beyond conventional histology in coeliac disease diagnosis. Dig Liver Dis 42:28–32CrossRefPubMed Salmi T, Collin P, Reunala T, Mäki M, Kaukinen K (2010) Diagnostic methods beyond conventional histology in coeliac disease diagnosis. Dig Liver Dis 42:28–32CrossRefPubMed
75.
go back to reference Freeman HJ (2004) REVIEW: adult celiac disease and the severe “flat” small bowel biopsy lesion. Dig Dis Sci 49:535–545CrossRefPubMed Freeman HJ (2004) REVIEW: adult celiac disease and the severe “flat” small bowel biopsy lesion. Dig Dis Sci 49:535–545CrossRefPubMed
76.
go back to reference Marsh MN (1992) Gluten, major histocompatibility complex, and the small intestine. A molecular and immunobiologic approach to the spectrum of gluten sensitivity (“celiac sprue”). Gastroenterology 102:330–354CrossRefPubMed Marsh MN (1992) Gluten, major histocompatibility complex, and the small intestine. A molecular and immunobiologic approach to the spectrum of gluten sensitivity (“celiac sprue”). Gastroenterology 102:330–354CrossRefPubMed
77.
go back to reference Kurppa K, Collin P, Viljamaa M, Haimila K, Saavalainen P, Partanen J, Laurila K, Huhtala H, Paasikivi K, Mäki M (2009) Diagnosing mild enteropathy celiac disease: a randomized, controlled clinical study. Gastroenterology 136:816–823CrossRefPubMed Kurppa K, Collin P, Viljamaa M, Haimila K, Saavalainen P, Partanen J, Laurila K, Huhtala H, Paasikivi K, Mäki M (2009) Diagnosing mild enteropathy celiac disease: a randomized, controlled clinical study. Gastroenterology 136:816–823CrossRefPubMed
78.
go back to reference Kaukinen K, Lindfors K, Collin P, Koskinen O, Maki M (2010) Coeliac disease—a diagnostic and therapeutic challenge. Clin Chem Lab Med 48:1205–1216CrossRefPubMed Kaukinen K, Lindfors K, Collin P, Koskinen O, Maki M (2010) Coeliac disease—a diagnostic and therapeutic challenge. Clin Chem Lab Med 48:1205–1216CrossRefPubMed
79.
go back to reference Walker MM, Murray JA, Ronkainen J, Aro P, Storskrubb T, D'Amato M, Lahr B, Talley NJ, Agreus L (2010) Detection of celiac disease and lymphocytic enteropathy by parallel serology and histopathology in a population-based study. Gastroenterology 139:112–119CrossRefPubMedPubMedCentral Walker MM, Murray JA, Ronkainen J, Aro P, Storskrubb T, D'Amato M, Lahr B, Talley NJ, Agreus L (2010) Detection of celiac disease and lymphocytic enteropathy by parallel serology and histopathology in a population-based study. Gastroenterology 139:112–119CrossRefPubMedPubMedCentral
80.
go back to reference Järvinen TT, Kaukinen K, Laurila K, Kyrönpalo S, Rasmussen M, Mäki M, Korhonen H, Reunala T, Collin P (2003) Intraepithelial lymphocytes in celiac disease. Am J Gastroenterol 98:1332–1337CrossRefPubMed Järvinen TT, Kaukinen K, Laurila K, Kyrönpalo S, Rasmussen M, Mäki M, Korhonen H, Reunala T, Collin P (2003) Intraepithelial lymphocytes in celiac disease. Am J Gastroenterol 98:1332–1337CrossRefPubMed
81.
go back to reference Leffler DA, Schuppan D (2010) Update on serologic testing in celiac disease. Am J Gastroenterol 105:2520–2524CrossRefPubMed Leffler DA, Schuppan D (2010) Update on serologic testing in celiac disease. Am J Gastroenterol 105:2520–2524CrossRefPubMed
82.
go back to reference Kaukinen K, Collin P, Laurila K, Kaartinen T, Partanen J, Mäki M (2007) Resurrection of gliadin antibodies in coeliac disease. Deamidated gliadin peptide antibody test provides additional diagnostic benefit. Scand J Gastroenterol 42:1428–1433CrossRefPubMed Kaukinen K, Collin P, Laurila K, Kaartinen T, Partanen J, Mäki M (2007) Resurrection of gliadin antibodies in coeliac disease. Deamidated gliadin peptide antibody test provides additional diagnostic benefit. Scand J Gastroenterol 42:1428–1433CrossRefPubMed
83.
go back to reference Kurppa K, Lindfors K, Collin P, Saavalainen P, Partanen J, Haimila K, Huhtala H, Laurila K, Maki M, Kaukinen K (2011) Antibodies against deamidated gliadin peptides in early-stage celiac disease. J Clin Gastroenterol 45:673–678CrossRefPubMed Kurppa K, Lindfors K, Collin P, Saavalainen P, Partanen J, Haimila K, Huhtala H, Laurila K, Maki M, Kaukinen K (2011) Antibodies against deamidated gliadin peptides in early-stage celiac disease. J Clin Gastroenterol 45:673–678CrossRefPubMed
84.
go back to reference Dahle C, Hagman A, Ignatova S, Ström M (2010) Antibodies against deamidated gliadin peptides identify adult coeliac disease patients negative for antibodies against endomysium and tissue transglutaminase. Aliment Pharmacol Ther 32:254–260CrossRefPubMed Dahle C, Hagman A, Ignatova S, Ström M (2010) Antibodies against deamidated gliadin peptides identify adult coeliac disease patients negative for antibodies against endomysium and tissue transglutaminase. Aliment Pharmacol Ther 32:254–260CrossRefPubMed
85.
go back to reference Seah P, Fry L, Rossiter M, Hopfbrand A, Holborow E (1971) Anti-reticulin antibodies in childhood coeliac disease. Lancet 298:681–682CrossRef Seah P, Fry L, Rossiter M, Hopfbrand A, Holborow E (1971) Anti-reticulin antibodies in childhood coeliac disease. Lancet 298:681–682CrossRef
86.
go back to reference Chorzelski T, Beutner E, Sulej J, Tchorzewska H, Jablonska S, Kumar V, Kapuscinska A (1984) IgA anti-endomysium antibody. A new immunological marker of dermatitis herpetiformis and coeliac disease. Br J Dermatol 111:395–402CrossRefPubMed Chorzelski T, Beutner E, Sulej J, Tchorzewska H, Jablonska S, Kumar V, Kapuscinska A (1984) IgA anti-endomysium antibody. A new immunological marker of dermatitis herpetiformis and coeliac disease. Br J Dermatol 111:395–402CrossRefPubMed
88.
go back to reference Dieterich W, Ehnis T, Bauer M, Donner P, Volta U, Riecken E, Schuppan D (1997) Identification of tissue transglutaminase as the autoantigen of celiac disease. Nat Med 3:797–801CrossRefPubMed Dieterich W, Ehnis T, Bauer M, Donner P, Volta U, Riecken E, Schuppan D (1997) Identification of tissue transglutaminase as the autoantigen of celiac disease. Nat Med 3:797–801CrossRefPubMed
89.
go back to reference Maki M, Holm K, Hallstrom O, Collin P, Viander M, Savilahti E, Lipsanen V, Koskimies S (1991) Serological markers and HLA genes among healthy first-degree relatives of patients with coeliac disease. Lancet 338:1350–1353CrossRefPubMed Maki M, Holm K, Hallstrom O, Collin P, Viander M, Savilahti E, Lipsanen V, Koskimies S (1991) Serological markers and HLA genes among healthy first-degree relatives of patients with coeliac disease. Lancet 338:1350–1353CrossRefPubMed
90.
go back to reference Taavela J, Kurppa K, Collin P, Lähdeaho M, Salmi T, Saavalainen P, Haimila K, Huhtala H, Laurila K, Sievänen H (2013) Degree of damage to the small bowel and serum antibody titers correlate with clinical presentation of patients with celiac disease. Clin Gastroenterol Hepatol 11:166–171CrossRefPubMed Taavela J, Kurppa K, Collin P, Lähdeaho M, Salmi T, Saavalainen P, Haimila K, Huhtala H, Laurila K, Sievänen H (2013) Degree of damage to the small bowel and serum antibody titers correlate with clinical presentation of patients with celiac disease. Clin Gastroenterol Hepatol 11:166–171CrossRefPubMed
91.
go back to reference Husby S, Koletzko S, Korponay-Szabo IR, Mearin ML, Phillips A, Shamir R, Troncone R, Giersiepen K, Branski D, Catassi C, Lelgeman M, Maki M, Ribes-Koninckx C, Ventura A, Zimmer KP, ESPGHAN Working Group on Coeliac Disease Diagnosis, ESPGHAN Gastroenterology Committee & European Society for Pediatric Gastroenterology, Hepatology, and Nutrition (2012) European Society for Pediatric Gastroenterology, hepatology, and nutrition guidelines for the diagnosis of coeliac disease. J Pediatr Gastroenterol Nutr 54:136–160CrossRefPubMed Husby S, Koletzko S, Korponay-Szabo IR, Mearin ML, Phillips A, Shamir R, Troncone R, Giersiepen K, Branski D, Catassi C, Lelgeman M, Maki M, Ribes-Koninckx C, Ventura A, Zimmer KP, ESPGHAN Working Group on Coeliac Disease Diagnosis, ESPGHAN Gastroenterology Committee & European Society for Pediatric Gastroenterology, Hepatology, and Nutrition (2012) European Society for Pediatric Gastroenterology, hepatology, and nutrition guidelines for the diagnosis of coeliac disease. J Pediatr Gastroenterol Nutr 54:136–160CrossRefPubMed
92.
go back to reference Nemec G, Ventura A, Stefano M, Di Leo G, Baldas V, Tommasini A, Ferrara F, Taddio A, Citta A, Sblattero D (2006) Looking for celiac disease: diagnostic accuracy of two rapid commercial assays. Am J Gastroenterol 101:1597–1600CrossRefPubMed Nemec G, Ventura A, Stefano M, Di Leo G, Baldas V, Tommasini A, Ferrara F, Taddio A, Citta A, Sblattero D (2006) Looking for celiac disease: diagnostic accuracy of two rapid commercial assays. Am J Gastroenterol 101:1597–1600CrossRefPubMed
93.
go back to reference Popp A, Jinga M, Jurcut C, Balaban V, Bardas C, Laurila K, Vasilescu F, Ene A, Anca I, Maki M (2013) Fingertip rapid point-of-care test in adult case-finding in coeliac disease. BMC Gastroenterol 13:115CrossRefPubMedPubMedCentral Popp A, Jinga M, Jurcut C, Balaban V, Bardas C, Laurila K, Vasilescu F, Ene A, Anca I, Maki M (2013) Fingertip rapid point-of-care test in adult case-finding in coeliac disease. BMC Gastroenterol 13:115CrossRefPubMedPubMedCentral
94.
go back to reference Mooney PD, Wong SH, Johnston AJ, Kurien M, Avgerinos A, Sanders DS (2015) Increased detection of celiac disease with measurement of deamidated gliadin peptide antibody before endoscopy. Clin Gastroenterol Hepatol 13:1278–1284CrossRefPubMed Mooney PD, Wong SH, Johnston AJ, Kurien M, Avgerinos A, Sanders DS (2015) Increased detection of celiac disease with measurement of deamidated gliadin peptide antibody before endoscopy. Clin Gastroenterol Hepatol 13:1278–1284CrossRefPubMed
95.
go back to reference Marzari R, Sblattero D, Florian F, Tongiorgi E, Not T, Tommasini A, Ventura A, Bradbury A (2001) Molecular dissection of the tissue transglutaminase antoantibody response in celiac disease. J Immunol 166:4170–4176CrossRefPubMed Marzari R, Sblattero D, Florian F, Tongiorgi E, Not T, Tommasini A, Ventura A, Bradbury A (2001) Molecular dissection of the tissue transglutaminase antoantibody response in celiac disease. J Immunol 166:4170–4176CrossRefPubMed
96.
go back to reference Sblattero D, Ventura A, Tommasini A, Cattin L, Martelossi S, Florian F, Marzari R, Bradbury A, Not T (2006) Cryptic gluten intolerance in type 1 diabetes: identifying suitable candidates for a gluten free diet. Gut 55:133–134CrossRefPubMedPubMedCentral Sblattero D, Ventura A, Tommasini A, Cattin L, Martelossi S, Florian F, Marzari R, Bradbury A, Not T (2006) Cryptic gluten intolerance in type 1 diabetes: identifying suitable candidates for a gluten free diet. Gut 55:133–134CrossRefPubMedPubMedCentral
97.
go back to reference Shiner M, Ballard J (1972) Antigen-antibody reactions in jejunal mucosa in childhood coeliac disease after gluten challenge. Lancet 299:1202–1205CrossRef Shiner M, Ballard J (1972) Antigen-antibody reactions in jejunal mucosa in childhood coeliac disease after gluten challenge. Lancet 299:1202–1205CrossRef
98.
go back to reference Kárpáti S, Kósnai I, Török É, Kovács JB (1988) Immunoglobulin a deposition in jejunal mucosa of children with dermatitis herpetiformis. J Investig Dermatol 91:336–339CrossRefPubMed Kárpáti S, Kósnai I, Török É, Kovács JB (1988) Immunoglobulin a deposition in jejunal mucosa of children with dermatitis herpetiformis. J Investig Dermatol 91:336–339CrossRefPubMed
99.
go back to reference Korponay-Szabo I, Halttunen T, Szalai Z, Laurila K, Kiraly R, Kovacs J, Fesus L, Maki M (2004) In vivo targeting of intestinal and extraintestinal transglutaminase 2 by coeliac autoantibodies. Gut 53:641–648CrossRefPubMedPubMedCentral Korponay-Szabo I, Halttunen T, Szalai Z, Laurila K, Kiraly R, Kovacs J, Fesus L, Maki M (2004) In vivo targeting of intestinal and extraintestinal transglutaminase 2 by coeliac autoantibodies. Gut 53:641–648CrossRefPubMedPubMedCentral
100.
go back to reference Kaukinen K, Peräaho M, Collin P, Partanen J, Woolley N, Kaartinen T, Nuutinen T, Halttunen T, Mäki M, Korponay-Szabo I (2005) Small-bowel mucosal transglutaminase 2-specific IgA deposits in coeliac disease without villous atrophy: a prospective and randomized clinical study. Scand J Gastroenterol 40:564–572CrossRefPubMed Kaukinen K, Peräaho M, Collin P, Partanen J, Woolley N, Kaartinen T, Nuutinen T, Halttunen T, Mäki M, Korponay-Szabo I (2005) Small-bowel mucosal transglutaminase 2-specific IgA deposits in coeliac disease without villous atrophy: a prospective and randomized clinical study. Scand J Gastroenterol 40:564–572CrossRefPubMed
101.
go back to reference Salmi TT, Collin P, Korponay-Szabo IR, Laurila K, Partanen J, Huhtala H, Kiraly R, Lorand L, Reunala T, Maki M, Kaukinen K (2006) Endomysial antibody-negative coeliac disease: clinical characteristics and intestinal autoantibody deposits. Gut 55:1746–1753CrossRefPubMedPubMedCentral Salmi TT, Collin P, Korponay-Szabo IR, Laurila K, Partanen J, Huhtala H, Kiraly R, Lorand L, Reunala T, Maki M, Kaukinen K (2006) Endomysial antibody-negative coeliac disease: clinical characteristics and intestinal autoantibody deposits. Gut 55:1746–1753CrossRefPubMedPubMedCentral
102.
go back to reference Jv M (1969) Granular deposits of immunoglobulins in the skin of patients with dermatitis herpetiformis. An immunofluorescent study. Br J Dermatol 81:493–503CrossRef Jv M (1969) Granular deposits of immunoglobulins in the skin of patients with dermatitis herpetiformis. An immunofluorescent study. Br J Dermatol 81:493–503CrossRef
103.
go back to reference Heil PM, Volc-Platzer B, Karlhofer F, Gebhart W, Huber W, Benesch T, Vogelsang H, Stingl G (2005) Transglutaminases as diagnostically relevant autoantigens in patients with gluten sensitivity. J Dtsch Dermatol Ges 3:974–978CrossRefPubMed Heil PM, Volc-Platzer B, Karlhofer F, Gebhart W, Huber W, Benesch T, Vogelsang H, Stingl G (2005) Transglutaminases as diagnostically relevant autoantigens in patients with gluten sensitivity. J Dtsch Dermatol Ges 3:974–978CrossRefPubMed
104.
go back to reference Marietta EV, Camilleri MJ, Castro LA, Krause PK, Pittelkow MR, Murray JA (2008) Transglutaminase autoantibodies in dermatitis herpetiformis and celiac sprue. J Investig Dermatol 128:332–335CrossRefPubMed Marietta EV, Camilleri MJ, Castro LA, Krause PK, Pittelkow MR, Murray JA (2008) Transglutaminase autoantibodies in dermatitis herpetiformis and celiac sprue. J Investig Dermatol 128:332–335CrossRefPubMed
105.
go back to reference Hull CM, Liddle M, Hansen N, Meyer L, Schmidt L, Taylor T, Jaskowski T, Hill H, Zone J (2008) Elevation of IgA anti-epidermal transglutaminase antibodies in dermatitis herpetiformis. Br J Dermatol 159:120–124CrossRefPubMed Hull CM, Liddle M, Hansen N, Meyer L, Schmidt L, Taylor T, Jaskowski T, Hill H, Zone J (2008) Elevation of IgA anti-epidermal transglutaminase antibodies in dermatitis herpetiformis. Br J Dermatol 159:120–124CrossRefPubMed
106.
go back to reference Borroni G, Biagi F, Ciocca O, Vassallo C, Carugno A, Cananzi R, Campanella J, Bianchi P, Brazzelli V, Corazza G (2013) IgA anti-epidermal transglutaminase autoantibodies: a sensible and sensitive marker for diagnosis of dermatitis herpetiformis in adult patients. J Eur Acad Dermatol Venereol 27:836–841CrossRefPubMed Borroni G, Biagi F, Ciocca O, Vassallo C, Carugno A, Cananzi R, Campanella J, Bianchi P, Brazzelli V, Corazza G (2013) IgA anti-epidermal transglutaminase autoantibodies: a sensible and sensitive marker for diagnosis of dermatitis herpetiformis in adult patients. J Eur Acad Dermatol Venereol 27:836–841CrossRefPubMed
107.
go back to reference Jaskowski TD, Hamblin T, Wilson AR, Hill HR, Book LS, Meyer LJ, Zone JJ, Hull CM (2009) IgA anti-epidermal transglutaminase antibodies in dermatitis herpetiformis and pediatric celiac disease. J Investig Dermatol 129:2728–2730CrossRefPubMed Jaskowski TD, Hamblin T, Wilson AR, Hill HR, Book LS, Meyer LJ, Zone JJ, Hull CM (2009) IgA anti-epidermal transglutaminase antibodies in dermatitis herpetiformis and pediatric celiac disease. J Investig Dermatol 129:2728–2730CrossRefPubMed
108.
go back to reference Janatuinen EK, Kemppainen TA, Julkunen RJ, Kosma VM, Maki M, Heikkinen M, Uusitupa MI (2002) No harm from five year ingestion of oats in coeliac disease. Gut 50:332–335CrossRefPubMedPubMedCentral Janatuinen EK, Kemppainen TA, Julkunen RJ, Kosma VM, Maki M, Heikkinen M, Uusitupa MI (2002) No harm from five year ingestion of oats in coeliac disease. Gut 50:332–335CrossRefPubMedPubMedCentral
110.
go back to reference Hopman EG, von Blomberg ME, Batstra MR, Morreau H, Dekker FW, Koning F, Lamers CB, Mearin ML (2008) Gluten tolerance in adult patients with celiac disease 20 years after diagnosis? Eur J Gastroenterol Hepatol 20:423–429CrossRefPubMed Hopman EG, von Blomberg ME, Batstra MR, Morreau H, Dekker FW, Koning F, Lamers CB, Mearin ML (2008) Gluten tolerance in adult patients with celiac disease 20 years after diagnosis? Eur J Gastroenterol Hepatol 20:423–429CrossRefPubMed
111.
go back to reference Bardella M, Fredella C, Trovato C, Ermacora E, Cavalli R, Saladino V, Prampolini L (2003) Long-term remission in patients with dermatitis herpetiformis on a normal diet. Br J Dermatol 149:968–971CrossRefPubMed Bardella M, Fredella C, Trovato C, Ermacora E, Cavalli R, Saladino V, Prampolini L (2003) Long-term remission in patients with dermatitis herpetiformis on a normal diet. Br J Dermatol 149:968–971CrossRefPubMed
112.
go back to reference Paek SY, Steinberg SM, Katz SI (2011) Remission in dermatitis herpetiformis: a cohort study. Arch Dermatol 147:301–305CrossRefPubMed Paek SY, Steinberg SM, Katz SI (2011) Remission in dermatitis herpetiformis: a cohort study. Arch Dermatol 147:301–305CrossRefPubMed
113.
go back to reference Tio M, Cox M, Eslick G (2012) Meta-analysis: coeliac disease and the risk of all-cause mortality, any malignancy and lymphoid malignancy. Aliment Pharmacol Ther 35:540–551CrossRefPubMed Tio M, Cox M, Eslick G (2012) Meta-analysis: coeliac disease and the risk of all-cause mortality, any malignancy and lymphoid malignancy. Aliment Pharmacol Ther 35:540–551CrossRefPubMed
114.
go back to reference Hervonen K, Vornanen M, Kautiainen H, Collin P, Reunala T (2005) Lymphoma in patients with dermatitis herpetiformis and their first-degree relatives. Br J Dermatol 152:82–86CrossRefPubMed Hervonen K, Vornanen M, Kautiainen H, Collin P, Reunala T (2005) Lymphoma in patients with dermatitis herpetiformis and their first-degree relatives. Br J Dermatol 152:82–86CrossRefPubMed
115.
go back to reference Blazina Š, Bratanič N, Čampa AŠ (2010) Bone mineral density and importance of strict gluten-free diet in children and adolescents with celiac disease. Bone 47:598–603CrossRefPubMed Blazina Š, Bratanič N, Čampa AŠ (2010) Bone mineral density and importance of strict gluten-free diet in children and adolescents with celiac disease. Bone 47:598–603CrossRefPubMed
116.
go back to reference See JA, Kaukinen K, Makharia GK, Gibson PR, Murray JA (2015) Practical insights into gluten-free diets. Nat Rev Gastroenterol Hepatol 12:580–591CrossRefPubMed See JA, Kaukinen K, Makharia GK, Gibson PR, Murray JA (2015) Practical insights into gluten-free diets. Nat Rev Gastroenterol Hepatol 12:580–591CrossRefPubMed
117.
go back to reference van Gils T, Nijeboer P, van Wanrooij RL, Bouma G, Mulder CJ (2015) Mechanisms and management of refractory coeliac disease. Nat Rev Gastroenterol Hepatol 12:572–579CrossRefPubMed van Gils T, Nijeboer P, van Wanrooij RL, Bouma G, Mulder CJ (2015) Mechanisms and management of refractory coeliac disease. Nat Rev Gastroenterol Hepatol 12:572–579CrossRefPubMed
118.
go back to reference Ilus T, Kaukinen K, Virta L, Huhtala H, Mäki M, Kurppa K, Heikkinen M, Heikura M, Hirsi E, Jantunen K (2014) Refractory coeliac disease in a country with a high prevalence of clinically-diagnosed coeliac disease. Aliment Pharmacol Ther 39:418–425CrossRefPubMed Ilus T, Kaukinen K, Virta L, Huhtala H, Mäki M, Kurppa K, Heikkinen M, Heikura M, Hirsi E, Jantunen K (2014) Refractory coeliac disease in a country with a high prevalence of clinically-diagnosed coeliac disease. Aliment Pharmacol Ther 39:418–425CrossRefPubMed
119.
go back to reference Sanchez M, Mohaidle A, Baistrocchi A, Matoso D, Vázquez H, González A, Mazure R, Maffei E, Ferrari G, Smecuol E (2011) Risk of fracture in celiac disease: gender, dietary compliance, or both. World J Gastroenterol 17:3035–3042CrossRefPubMedPubMedCentral Sanchez M, Mohaidle A, Baistrocchi A, Matoso D, Vázquez H, González A, Mazure R, Maffei E, Ferrari G, Smecuol E (2011) Risk of fracture in celiac disease: gender, dietary compliance, or both. World J Gastroenterol 17:3035–3042CrossRefPubMedPubMedCentral
120.
go back to reference Norström F, Sandström O, Lindholm L, Ivarsson A (2012) A gluten-free diet effectively reduces symptoms and health care consumption in a Swedish celiac disease population. BMC Gastroenterol 12:1CrossRef Norström F, Sandström O, Lindholm L, Ivarsson A (2012) A gluten-free diet effectively reduces symptoms and health care consumption in a Swedish celiac disease population. BMC Gastroenterol 12:1CrossRef
121.
122.
go back to reference Sulic A, Kurppa K, Rauhavirta T, Kaukinen K, Lindfors K (2015) Transglutaminase as a therapeutic target for celiac disease. Expert Opin Ther Targets 19:335–348CrossRefPubMed Sulic A, Kurppa K, Rauhavirta T, Kaukinen K, Lindfors K (2015) Transglutaminase as a therapeutic target for celiac disease. Expert Opin Ther Targets 19:335–348CrossRefPubMed
123.
go back to reference Shan L, Molberg O, Parrot I, Hausch F, Filiz F, Gray G, Sollid L, Khosla C (2002) Structural basis for gluten intolerance in celiac sprue. Science 297:2275–2279CrossRefPubMed Shan L, Molberg O, Parrot I, Hausch F, Filiz F, Gray G, Sollid L, Khosla C (2002) Structural basis for gluten intolerance in celiac sprue. Science 297:2275–2279CrossRefPubMed
124.
go back to reference Quarsten H, Molberg Ø, Fugger L, McAdam SN, Sollid LM (1999) HLA binding and T cell recognition of a tissue transglutaminase-modified gliadin epitope. Eur J Immunol 29:2506–2514CrossRefPubMed Quarsten H, Molberg Ø, Fugger L, McAdam SN, Sollid LM (1999) HLA binding and T cell recognition of a tissue transglutaminase-modified gliadin epitope. Eur J Immunol 29:2506–2514CrossRefPubMed
125.
go back to reference Vader L, de Ru A, van der Wal Y, Kooy Y, Benckhuijsen W, Mearin M, Drijfhout J, van Veelen P, Koning F (2002) Specificity of tissue transglutaminase explains cereal toxicity in celiac disease. J Exp Med 195:643–649CrossRefPubMedPubMedCentral Vader L, de Ru A, van der Wal Y, Kooy Y, Benckhuijsen W, Mearin M, Drijfhout J, van Veelen P, Koning F (2002) Specificity of tissue transglutaminase explains cereal toxicity in celiac disease. J Exp Med 195:643–649CrossRefPubMedPubMedCentral
126.
go back to reference Molberg O, Mcadam S, Korner R, Quarsten H, Kristiansen C, Madsen L, Fugger L, Scott H, Noren O, Roepstorff P, Lundin K, Sjostrom H, Sollid L (1998) Tissue transglutaminase selectively modifies gliadin peptides that are recognized by gut-derived T cells in celiac disease RID G-8565-2011. Nat Med 4:713–717CrossRefPubMed Molberg O, Mcadam S, Korner R, Quarsten H, Kristiansen C, Madsen L, Fugger L, Scott H, Noren O, Roepstorff P, Lundin K, Sjostrom H, Sollid L (1998) Tissue transglutaminase selectively modifies gliadin peptides that are recognized by gut-derived T cells in celiac disease RID G-8565-2011. Nat Med 4:713–717CrossRefPubMed
127.
go back to reference Dahal-Koirala S, Risnes L, Christophersen A, Sarna V, Lundin KE, Sollid L, Qiao S (2016) TCR sequencing of single cells reactive to DQ2. 5-glia-α2 and DQ2. 5-glia-ω2 reveals clonal expansion and epitope-specific V-gene usage. Mucosal Immunol. doi:10.1038/mi.2015.147 CrossRefPubMed Dahal-Koirala S, Risnes L, Christophersen A, Sarna V, Lundin KE, Sollid L, Qiao S (2016) TCR sequencing of single cells reactive to DQ2. 5-glia-α2 and DQ2. 5-glia-ω2 reveals clonal expansion and epitope-specific V-gene usage. Mucosal Immunol. doi:10.​1038/​mi.​2015.​147 CrossRefPubMed
128.
go back to reference Qiao SW, Christophersen A, Lundin KE, Sollid LM (2014) Biased usage and preferred pairing of alpha- and beta-chains of TCRs specific for an immunodominant gluten epitope in coeliac disease. Int Immunol 26:13–19CrossRefPubMed Qiao SW, Christophersen A, Lundin KE, Sollid LM (2014) Biased usage and preferred pairing of alpha- and beta-chains of TCRs specific for an immunodominant gluten epitope in coeliac disease. Int Immunol 26:13–19CrossRefPubMed
129.
go back to reference Qiao SW, Raki M, Gunnarsen KS, Loset GA, Lundin KE, Sandlie I, Sollid LM (2011) Posttranslational modification of gluten shapes TCR usage in celiac disease. J Immunol 187:3064–3071CrossRefPubMed Qiao SW, Raki M, Gunnarsen KS, Loset GA, Lundin KE, Sandlie I, Sollid LM (2011) Posttranslational modification of gluten shapes TCR usage in celiac disease. J Immunol 187:3064–3071CrossRefPubMed
130.
go back to reference Petersen J, Montserrat V, Mujico JR, Loh KL, Beringer DX, van Lummel M, Thompson A, Mearin ML, Schweizer J, Kooy-Winkelaar Y (2014) T-cell receptor recognition of HLA-DQ2–gliadin complexes associated with celiac disease. Nat Struct Mol Biol 21:480–488CrossRefPubMed Petersen J, Montserrat V, Mujico JR, Loh KL, Beringer DX, van Lummel M, Thompson A, Mearin ML, Schweizer J, Kooy-Winkelaar Y (2014) T-cell receptor recognition of HLA-DQ2–gliadin complexes associated with celiac disease. Nat Struct Mol Biol 21:480–488CrossRefPubMed
131.
go back to reference Fleckenstein B, Qiao SW, Larsen MR, Jung G, Roepstorff P, Sollid LM (2004) Molecular characterization of covalent complexes between tissue transglutaminase and gliadin peptides. J Biol Chem 279:17607–17616CrossRefPubMed Fleckenstein B, Qiao SW, Larsen MR, Jung G, Roepstorff P, Sollid LM (2004) Molecular characterization of covalent complexes between tissue transglutaminase and gliadin peptides. J Biol Chem 279:17607–17616CrossRefPubMed
132.
go back to reference du Pré MF, Sollid LM (2015) T-cell and B-cell immunity in celiac disease. Best Pract Res Clin Gastroenterol 29:413–423CrossRefPubMed du Pré MF, Sollid LM (2015) T-cell and B-cell immunity in celiac disease. Best Pract Res Clin Gastroenterol 29:413–423CrossRefPubMed
133.
go back to reference Di Niro R, Mesin L, Zheng N, Stamnaes J, Morrissey M, Lee J, Huang M, Iversen R, du Pre MF, Qiao S, Lundin KEA, Wilson PC, Sollid LM (2012) High abundance of plasma cells secreting transglutaminase 2-specific IgA autoantibodies with limited somatic hypermutation in celiac disease intestinal lesions. Nat Med 18:441–445CrossRefPubMedPubMedCentral Di Niro R, Mesin L, Zheng N, Stamnaes J, Morrissey M, Lee J, Huang M, Iversen R, du Pre MF, Qiao S, Lundin KEA, Wilson PC, Sollid LM (2012) High abundance of plasma cells secreting transglutaminase 2-specific IgA autoantibodies with limited somatic hypermutation in celiac disease intestinal lesions. Nat Med 18:441–445CrossRefPubMedPubMedCentral
134.
go back to reference Villanacci V, Not T, Sblattero D, Gaiotto T, Chirdo F, Galletti A, Bassotti G (2009) Mucosal tissue transglutaminase expression in celiac disease. J Cell Mol Med 13:334–340CrossRefPubMed Villanacci V, Not T, Sblattero D, Gaiotto T, Chirdo F, Galletti A, Bassotti G (2009) Mucosal tissue transglutaminase expression in celiac disease. J Cell Mol Med 13:334–340CrossRefPubMed
135.
go back to reference Evans DF, Pye G, Bramley R, Clark AG, Dyson TJ, Hardcastle JD (1988) Measurement of gastrointestinal pH profiles in normal ambulant human subjects. Gut 29:1035–1041CrossRefPubMedPubMedCentral Evans DF, Pye G, Bramley R, Clark AG, Dyson TJ, Hardcastle JD (1988) Measurement of gastrointestinal pH profiles in normal ambulant human subjects. Gut 29:1035–1041CrossRefPubMedPubMedCentral
136.
go back to reference Rauhavirta T, Qiao S, Jiang Z, Myrsky E, Loponen J, Korponay-Szabo IR, Salovaara H, Garcia-Horsman JA, Venalainen J, Mannisto PT, Collighan R, Mongeot A, Griffin M, Maki M, Kaukinen K, Lindfors K (2011) Epithelial transport and deamidation of gliadin peptides: a role for coeliac disease patient immunoglobulin a. Clin Exp Immunol 164:127–136CrossRefPubMedPubMedCentral Rauhavirta T, Qiao S, Jiang Z, Myrsky E, Loponen J, Korponay-Szabo IR, Salovaara H, Garcia-Horsman JA, Venalainen J, Mannisto PT, Collighan R, Mongeot A, Griffin M, Maki M, Kaukinen K, Lindfors K (2011) Epithelial transport and deamidation of gliadin peptides: a role for coeliac disease patient immunoglobulin a. Clin Exp Immunol 164:127–136CrossRefPubMedPubMedCentral
137.
go back to reference Thomazy VA, Vega F, Medeiros LJ, Davies PJ, Jones D (2003) Phenotypic modulation of the stromal reticular network in normal and neoplastic lymph nodes: tissue transglutaminase reveals coordinate regulation of multiple cell types. Am J Pathol 163:165–174CrossRefPubMedPubMedCentral Thomazy VA, Vega F, Medeiros LJ, Davies PJ, Jones D (2003) Phenotypic modulation of the stromal reticular network in normal and neoplastic lymph nodes: tissue transglutaminase reveals coordinate regulation of multiple cell types. Am J Pathol 163:165–174CrossRefPubMedPubMedCentral
138.
go back to reference Mention J, Ahmed MB, Bègue B, Barbe U, Verkarre V, Asnafi V, Colombel J, Cugnenc P, Ruemmele FM, Mcintyre E (2003) Interleukin 15: a key to disrupted intraepithelial lymphocyte homeostasis and lymphomagenesis in celiac disease. Gastroenterology 125:730–745CrossRefPubMed Mention J, Ahmed MB, Bègue B, Barbe U, Verkarre V, Asnafi V, Colombel J, Cugnenc P, Ruemmele FM, Mcintyre E (2003) Interleukin 15: a key to disrupted intraepithelial lymphocyte homeostasis and lymphomagenesis in celiac disease. Gastroenterology 125:730–745CrossRefPubMed
139.
go back to reference Barone MV, Zanzi D, Maglio M, Nanayakkara M, Santagata S, Lania G, Miele E, Ribecco MTS, Maurano F, Auricchio R (2011) Gliadin-mediated proliferation and innate immune activation in celiac disease are due to alterations in vesicular trafficking. PLoS One 6:e17039CrossRefPubMedPubMedCentral Barone MV, Zanzi D, Maglio M, Nanayakkara M, Santagata S, Lania G, Miele E, Ribecco MTS, Maurano F, Auricchio R (2011) Gliadin-mediated proliferation and innate immune activation in celiac disease are due to alterations in vesicular trafficking. PLoS One 6:e17039CrossRefPubMedPubMedCentral
140.
go back to reference Hue S, Mention J, Monteiro R, Zhang S, Cellier C, Schmitz J, Verkarre V, Fodil N, Bahram S, Cerf-Bensussan N, Caillat-Zucman S (2004) A direct role for NKG2D/MICA interaction in villous atrophy during celiac disease. Immunity 21:367–377CrossRefPubMed Hue S, Mention J, Monteiro R, Zhang S, Cellier C, Schmitz J, Verkarre V, Fodil N, Bahram S, Cerf-Bensussan N, Caillat-Zucman S (2004) A direct role for NKG2D/MICA interaction in villous atrophy during celiac disease. Immunity 21:367–377CrossRefPubMed
141.
go back to reference Roberts AI, Lee L, Schwarz E, Groh V, Spies T, Ebert EC, Jabri B (2001) NKG2D receptors induced by IL-15 costimulate CD28-negative effector CTL in the tissue microenvironment. J Immunol 167:5527–5530CrossRefPubMed Roberts AI, Lee L, Schwarz E, Groh V, Spies T, Ebert EC, Jabri B (2001) NKG2D receptors induced by IL-15 costimulate CD28-negative effector CTL in the tissue microenvironment. J Immunol 167:5527–5530CrossRefPubMed
142.
go back to reference Setty M, Discepolo V, Abadie V, Kamhawi S, Mayassi T, Kent A, Ciszewski C, Maglio M, Kistner E, Bhagat G (2015) Distinct and synergistic contributions of epithelial stress and adaptive immunity to functions of intraepithelial killer cells and active celiac disease. Gastroenterology 149:681–691CrossRefPubMedPubMedCentral Setty M, Discepolo V, Abadie V, Kamhawi S, Mayassi T, Kent A, Ciszewski C, Maglio M, Kistner E, Bhagat G (2015) Distinct and synergistic contributions of epithelial stress and adaptive immunity to functions of intraepithelial killer cells and active celiac disease. Gastroenterology 149:681–691CrossRefPubMedPubMedCentral
143.
go back to reference Meresse B, Curran SA, Ciszewski C, Orbelyan G, Setty M, Bhagat G, Lee L, Tretiakova M, Semrad C, Kistner E, Winchester RJ, Braud V, Lanier LL, Geraghty DE, Green PH, Guandalini S, Jabri B (2006) Reprogramming of CTLs into natural killer-like cells in celiac disease. J Exp Med 203:1343–1355CrossRefPubMedPubMedCentral Meresse B, Curran SA, Ciszewski C, Orbelyan G, Setty M, Bhagat G, Lee L, Tretiakova M, Semrad C, Kistner E, Winchester RJ, Braud V, Lanier LL, Geraghty DE, Green PH, Guandalini S, Jabri B (2006) Reprogramming of CTLs into natural killer-like cells in celiac disease. J Exp Med 203:1343–1355CrossRefPubMedPubMedCentral
144.
go back to reference Steinsbø Ø, Dunand CJH, Huang M, Mesin L, Salgado-Ferrer M, Lundin KE, Jahnsen J, Wilson PC, Sollid LM (2014) Restricted VH/VL usage and limited mutations in gluten-specific IgA of coeliac disease lesion plasma cells. Nat Commun 5:4041CrossRefPubMedPubMedCentral Steinsbø Ø, Dunand CJH, Huang M, Mesin L, Salgado-Ferrer M, Lundin KE, Jahnsen J, Wilson PC, Sollid LM (2014) Restricted VH/VL usage and limited mutations in gluten-specific IgA of coeliac disease lesion plasma cells. Nat Commun 5:4041CrossRefPubMedPubMedCentral
145.
go back to reference Sblattero D, Florian F, Azzoni E, Zyla T, Park M, Baldas V, Not T, Ventura A, Bradbury A, Marzari R (2002) The analysis of the fine specificity of celiac disease antibodies using tissue transglutaminase fragments. Eur J Biochem 269:5175–5181CrossRefPubMed Sblattero D, Florian F, Azzoni E, Zyla T, Park M, Baldas V, Not T, Ventura A, Bradbury A, Marzari R (2002) The analysis of the fine specificity of celiac disease antibodies using tissue transglutaminase fragments. Eur J Biochem 269:5175–5181CrossRefPubMed
146.
go back to reference Comerford R, Byrne G, Feighery C, Kelly J (2012) Binding of autoantibodies to the core region of tissue transglutaminase is a feature of paediatric coeliac disease. J Pediatr Gastroenterol Nutr 55:445–450CrossRefPubMed Comerford R, Byrne G, Feighery C, Kelly J (2012) Binding of autoantibodies to the core region of tissue transglutaminase is a feature of paediatric coeliac disease. J Pediatr Gastroenterol Nutr 55:445–450CrossRefPubMed
147.
go back to reference Simon-Vecsei Z, Kiraly R, Bagossi P, Toth B, Dahlbom I, Caja S, Csosz E, Lindfors K, Sblattero D, Nemes E, Maki M, Fesus L, Korponay-Szabo IR (2012) A single conformational transglutaminase 2 epitope contributed by three domains is critical for celiac antibody binding and effects. Proc Natl Acad Sci U S A 109:431–436CrossRefPubMed Simon-Vecsei Z, Kiraly R, Bagossi P, Toth B, Dahlbom I, Caja S, Csosz E, Lindfors K, Sblattero D, Nemes E, Maki M, Fesus L, Korponay-Szabo IR (2012) A single conformational transglutaminase 2 epitope contributed by three domains is critical for celiac antibody binding and effects. Proc Natl Acad Sci U S A 109:431–436CrossRefPubMed
148.
go back to reference Iversen R, Di Niro R, Stamnaes J, Lundin KE, Wilson PC, Sollid LM (2013) Transglutaminase 2-specific autoantibodies in celiac disease target clustered, N-terminal epitopes not displayed on the surface of cells. J Immunol 190:5981–5991CrossRefPubMedPubMedCentral Iversen R, Di Niro R, Stamnaes J, Lundin KE, Wilson PC, Sollid LM (2013) Transglutaminase 2-specific autoantibodies in celiac disease target clustered, N-terminal epitopes not displayed on the surface of cells. J Immunol 190:5981–5991CrossRefPubMedPubMedCentral
149.
go back to reference Farrace MG, Picarelli A, Di Tola M, Sabbatella L, Marchione OP, Ippolito G, Piacentini M (2001) Presence of anti-“tissue” transglutaminase antibodies in inflammatory intestinal diseases: an apoptosis-associated event? Cell Death Differ 8:767–770CrossRefPubMed Farrace MG, Picarelli A, Di Tola M, Sabbatella L, Marchione OP, Ippolito G, Piacentini M (2001) Presence of anti-“tissue” transglutaminase antibodies in inflammatory intestinal diseases: an apoptosis-associated event? Cell Death Differ 8:767–770CrossRefPubMed
150.
go back to reference Lidar M, Langevitz P, Barzilai O, Ram M, Porat-Katz B, Bizzaro N, Tonutti E, Maieron R, Chowers Y, Bar-Meir S (2009) Infectious serologies and autoantibodies in inflammatory bowel disease. Ann N Y Acad Sci 1173:640–648CrossRefPubMed Lidar M, Langevitz P, Barzilai O, Ram M, Porat-Katz B, Bizzaro N, Tonutti E, Maieron R, Chowers Y, Bar-Meir S (2009) Infectious serologies and autoantibodies in inflammatory bowel disease. Ann N Y Acad Sci 1173:640–648CrossRefPubMed
151.
go back to reference Pereda I, Bartolomé-Pacheco MJ, Martín M, López-Escribano H, Echevarría S, López-Hoyos M (2001) Antitissue transglutaminase antibodies in HIV infection and effect of highly active antiretroviral therapy. J Acquir Immune Defic Syndr 27:507–508CrossRefPubMed Pereda I, Bartolomé-Pacheco MJ, Martín M, López-Escribano H, Echevarría S, López-Hoyos M (2001) Antitissue transglutaminase antibodies in HIV infection and effect of highly active antiretroviral therapy. J Acquir Immune Defic Syndr 27:507–508CrossRefPubMed
152.
go back to reference Peracchi M, Trovato C, Longhi M, Gasparin M, Conte D, Tarantino C, Prati D, Bardella MT (2002) Tissue transglutaminase antibodies in patients with end-stage heart failure. Am J Gastroenterol 97:2850–2854CrossRefPubMed Peracchi M, Trovato C, Longhi M, Gasparin M, Conte D, Tarantino C, Prati D, Bardella MT (2002) Tissue transglutaminase antibodies in patients with end-stage heart failure. Am J Gastroenterol 97:2850–2854CrossRefPubMed
153.
go back to reference Sárdy M, Csikós M, Geisen C, Preisz K, Kornseé Z, Tomsits E, Töx U, Hunzelmann N, Wieslander J, Kárpáti S (2007) Tissue transglutaminase ELISA positivity in autoimmune disease independent of gluten-sensitive disease. Clin Chim Acta 376:126–135CrossRefPubMed Sárdy M, Csikós M, Geisen C, Preisz K, Kornseé Z, Tomsits E, Töx U, Hunzelmann N, Wieslander J, Kárpáti S (2007) Tissue transglutaminase ELISA positivity in autoimmune disease independent of gluten-sensitive disease. Clin Chim Acta 376:126–135CrossRefPubMed
154.
go back to reference Kiraly R, Vecsei Z, Demenyi T, Korponay-Szabo I, Fesus L (2006) Coeliac autoantibodies can enhance transamidating and inhibit GTPase activity of tissue transglutaminase: dependence on reaction environment and enzyme fitness. J Autoimmun 26:278–287CrossRefPubMed Kiraly R, Vecsei Z, Demenyi T, Korponay-Szabo I, Fesus L (2006) Coeliac autoantibodies can enhance transamidating and inhibit GTPase activity of tissue transglutaminase: dependence on reaction environment and enzyme fitness. J Autoimmun 26:278–287CrossRefPubMed
155.
go back to reference Myrsky E, Caja S, Simon-Vecsei Z, Korponay-Szabo IR, Nadalutti C, Collighan R, Mongeot A, Griffin M, Maki M, Kaukinen K, Lindfors K (2009) Celiac disease IgA modulates vascular permeability in vitro through the activity of transglutaminase 2 and RhoA. Cell Mol Life Sci 66:3375–3385CrossRefPubMed Myrsky E, Caja S, Simon-Vecsei Z, Korponay-Szabo IR, Nadalutti C, Collighan R, Mongeot A, Griffin M, Maki M, Kaukinen K, Lindfors K (2009) Celiac disease IgA modulates vascular permeability in vitro through the activity of transglutaminase 2 and RhoA. Cell Mol Life Sci 66:3375–3385CrossRefPubMed
156.
go back to reference Dieterich W, Trapp D, Esslinger B, Leidenberger M, Piper J, Hahn E, Schuppan D (2003) Autoantibodies of patients with coeliac disease are insufficient to block tissue transglutaminase activity. Gut 52:1562–1566CrossRefPubMedPubMedCentral Dieterich W, Trapp D, Esslinger B, Leidenberger M, Piper J, Hahn E, Schuppan D (2003) Autoantibodies of patients with coeliac disease are insufficient to block tissue transglutaminase activity. Gut 52:1562–1566CrossRefPubMedPubMedCentral
157.
go back to reference Byrne G, Feighery C, Jackson J, Kelly J (2010) Coeliac disease autoantibodies mediate significant inhibition of tissue transglutaminase. Clin Immunol 136:426–431CrossRefPubMed Byrne G, Feighery C, Jackson J, Kelly J (2010) Coeliac disease autoantibodies mediate significant inhibition of tissue transglutaminase. Clin Immunol 136:426–431CrossRefPubMed
158.
go back to reference Esposito C, Paparo F, Caputo I, Rossi M, Maglio M, Sblattero D, Not T, Porta R, Auricchio S, Marzari R, Troncone R (2002) Anti-tissue transglutaminase antibodies from coeliac patients inhibit transglutaminase activity both in vitro and in situ. Gut 51:177–181CrossRefPubMedPubMedCentral Esposito C, Paparo F, Caputo I, Rossi M, Maglio M, Sblattero D, Not T, Porta R, Auricchio S, Marzari R, Troncone R (2002) Anti-tissue transglutaminase antibodies from coeliac patients inhibit transglutaminase activity both in vitro and in situ. Gut 51:177–181CrossRefPubMedPubMedCentral
159.
go back to reference Halttunen T, Maki M (1999) Serum immunoglobulin a from patients with celiac disease inhibits human T84 intestinal crypt epithelial cell differentiation. Gastroenterology 116:566–572CrossRefPubMed Halttunen T, Maki M (1999) Serum immunoglobulin a from patients with celiac disease inhibits human T84 intestinal crypt epithelial cell differentiation. Gastroenterology 116:566–572CrossRefPubMed
160.
go back to reference Barone MV, Caputo I, Ribecco MT, Maglio M, Marzari R, Sblattero D, Troncone R, Auricchio S, Esposito C (2007) Humoral immune response to tissue transglutaminase is related to epithelial cell proliferation in celiac disease. Gastroenterology 132:1245–1253CrossRefPubMed Barone MV, Caputo I, Ribecco MT, Maglio M, Marzari R, Sblattero D, Troncone R, Auricchio S, Esposito C (2007) Humoral immune response to tissue transglutaminase is related to epithelial cell proliferation in celiac disease. Gastroenterology 132:1245–1253CrossRefPubMed
161.
go back to reference Teesalu K, Panarina M, Uibo O, Uibo R, Utt M (2012) Autoantibodies from patients with celiac disease inhibit transglutaminase 2 binding to heparin/heparan sulfate and interfere with intestinal epithelial cell adhesion. Amino Acids 42:1055–1064CrossRefPubMed Teesalu K, Panarina M, Uibo O, Uibo R, Utt M (2012) Autoantibodies from patients with celiac disease inhibit transglutaminase 2 binding to heparin/heparan sulfate and interfere with intestinal epithelial cell adhesion. Amino Acids 42:1055–1064CrossRefPubMed
162.
go back to reference Zanoni G, Navone R, Lunardi C, Tridente G, Bason C, Sivori S, Beri R, Dolcino M, Valletta E, Corrocher R, Puccetti A (2006) In celiac disease, a subset of autoantibodies against transglutaminase binds toll-like receptor 4 and induces activation of monocytes. PLoS Med 3:1637–1653CrossRef Zanoni G, Navone R, Lunardi C, Tridente G, Bason C, Sivori S, Beri R, Dolcino M, Valletta E, Corrocher R, Puccetti A (2006) In celiac disease, a subset of autoantibodies against transglutaminase binds toll-like receptor 4 and induces activation of monocytes. PLoS Med 3:1637–1653CrossRef
163.
go back to reference Lebreton C, Ménard S, Abed J, Moura IC, Coppo R, Dugave C, Monteiro RC, Fricot A, Traore MG, Griffin M, Cellier C, Malamut G, Cerf-Bensussan N, Heyman M (2012) Interactions among secretory immunoglobulin a, CD71, and transglutaminase-2 affect permeability of intestinal epithelial cells to gliadin peptides. Gastroenterology 143(3):698–707CrossRefPubMed Lebreton C, Ménard S, Abed J, Moura IC, Coppo R, Dugave C, Monteiro RC, Fricot A, Traore MG, Griffin M, Cellier C, Malamut G, Cerf-Bensussan N, Heyman M (2012) Interactions among secretory immunoglobulin a, CD71, and transglutaminase-2 affect permeability of intestinal epithelial cells to gliadin peptides. Gastroenterology 143(3):698–707CrossRefPubMed
164.
go back to reference Ménard S, Lebreton C, Schumann M, Matysiak-Budnik T, Dugave C, Bouhnik Y, Malamut G, Cellier C, Allez M, Crenn P (2012) Paracellular versus transcellular intestinal permeability to gliadin peptides in active celiac disease. Am J Pathol 180:608–615CrossRefPubMed Ménard S, Lebreton C, Schumann M, Matysiak-Budnik T, Dugave C, Bouhnik Y, Malamut G, Cellier C, Allez M, Crenn P (2012) Paracellular versus transcellular intestinal permeability to gliadin peptides in active celiac disease. Am J Pathol 180:608–615CrossRefPubMed
165.
go back to reference Matysiak-Budnik T, Moura IC, Arcos-Fajardo M, Lebreton C, Menard S, Candalh C, 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, Heyman M (2008) Secretory IgA mediates retrotranscytosis of intact gliadin peptides via the transferrin receptor in celiac disease. J Exp Med 205:143–154CrossRefPubMedPubMedCentral Matysiak-Budnik T, Moura IC, Arcos-Fajardo M, Lebreton C, Menard S, Candalh C, 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, Heyman M (2008) Secretory IgA mediates retrotranscytosis of intact gliadin peptides via the transferrin receptor in celiac disease. J Exp Med 205:143–154CrossRefPubMedPubMedCentral
166.
go back to reference Nadalutti CA, Korponay-Szabo IR, Kaukinen K, Griffin M, Mäki M, Lindfors K (2014) Celiac disease patient IgA antibodies induce endothelial adhesion and cell polarization defects via extracellular transglutaminase 2. Cell Mol Life Sci 71:1315–1326CrossRefPubMed Nadalutti CA, Korponay-Szabo IR, Kaukinen K, Griffin M, Mäki M, Lindfors K (2014) Celiac disease patient IgA antibodies induce endothelial adhesion and cell polarization defects via extracellular transglutaminase 2. Cell Mol Life Sci 71:1315–1326CrossRefPubMed
167.
go back to reference Kalliokoski S, Sulic A, Korponay-Szabó IR, Szondy Z, Frias R, Perez MA, Martucciello S, Roivainen A, Pelliniemi LJ, Esposito C (2013) Celiac disease-specific TG2-targeted autoantibodies inhibit angiogenesis ex vivo and in vivo in mice by interfering with endothelial cell dynamics. PLoS One 8:e65887CrossRefPubMedPubMedCentral Kalliokoski S, Sulic A, Korponay-Szabó IR, Szondy Z, Frias R, Perez MA, Martucciello S, Roivainen A, Pelliniemi LJ, Esposito C (2013) Celiac disease-specific TG2-targeted autoantibodies inhibit angiogenesis ex vivo and in vivo in mice by interfering with endothelial cell dynamics. PLoS One 8:e65887CrossRefPubMedPubMedCentral
168.
go back to reference Myrsky E, Kaukinen K, Syrjanen M, Korponay-Szabo IR, Maki M, Lindfors K (2008) Coeliac disease-specific autoantibodies targeted against transglutaminase 2 disturb angiogenesis. Clin Exp Immunol 152:111–119CrossRefPubMedPubMedCentral Myrsky E, Kaukinen K, Syrjanen M, Korponay-Szabo IR, Maki M, Lindfors K (2008) Coeliac disease-specific autoantibodies targeted against transglutaminase 2 disturb angiogenesis. Clin Exp Immunol 152:111–119CrossRefPubMedPubMedCentral
169.
go back to reference Cooke WT, Holmes GKT (1984) Coeliac disease. Churchill Livingstone, London Cooke WT, Holmes GKT (1984) Coeliac disease. Churchill Livingstone, London
170.
go back to reference Freitag T, Schulze-Koops H, Niedobitek G, Melino G, Schuppan D (2004) The role of the immune response against tissue transglutaminase in the pathogenesis of coeliac disease. Autoimmun Rev 3:13–20CrossRefPubMed Freitag T, Schulze-Koops H, Niedobitek G, Melino G, Schuppan D (2004) The role of the immune response against tissue transglutaminase in the pathogenesis of coeliac disease. Autoimmun Rev 3:13–20CrossRefPubMed
171.
go back to reference Di Niro R, Sblattero D, Florian F, Stebel M, Zentilin L, Giacca M, Villanacci V, Galletti A, Not T, Ventura A (2008) Anti-idiotypic response in mice expressing human autoantibodies. Mol Immunol 45:1782–1791CrossRefPubMed Di Niro R, Sblattero D, Florian F, Stebel M, Zentilin L, Giacca M, Villanacci V, Galletti A, Not T, Ventura A (2008) Anti-idiotypic response in mice expressing human autoantibodies. Mol Immunol 45:1782–1791CrossRefPubMed
172.
go back to reference Kalliokoski S, Caja S, Frias R, Laurila K, Koskinen O, Niemelä O, Mäki M, Kaukinen K, Korponay-Szabó IR, Lindfors K (2015) Injection of celiac disease patient sera or immunoglobulins to mice reproduces a condition mimicking early developing celiac disease. J Mol Med 93:51–62CrossRefPubMed Kalliokoski S, Caja S, Frias R, Laurila K, Koskinen O, Niemelä O, Mäki M, Kaukinen K, Korponay-Szabó IR, Lindfors K (2015) Injection of celiac disease patient sera or immunoglobulins to mice reproduces a condition mimicking early developing celiac disease. J Mol Med 93:51–62CrossRefPubMed
173.
go back to reference Hadjivassiliou M, Maki M, Sanders D, Williamson C, Grunewald R, Woodroofe N, Korponay-Szabo I (2006) Autoantibody targeting of brain and intestinal transglutaminase in gluten ataxia. Neurology 66:373–377CrossRefPubMed Hadjivassiliou M, Maki M, Sanders D, Williamson C, Grunewald R, Woodroofe N, Korponay-Szabo I (2006) Autoantibody targeting of brain and intestinal transglutaminase in gluten ataxia. Neurology 66:373–377CrossRefPubMed
174.
go back to reference Boscolo S, Lorenzon A, Sblattero D, Florian F, Stebel M, Marzari R, Not T, Aeschlimann D, Ventura A, Hadjivassiliou M (2010) Anti transglutaminase antibodies cause ataxia in mice. PLoS One 5:e9698CrossRefPubMedPubMedCentral Boscolo S, Lorenzon A, Sblattero D, Florian F, Stebel M, Marzari R, Not T, Aeschlimann D, Ventura A, Hadjivassiliou M (2010) Anti transglutaminase antibodies cause ataxia in mice. PLoS One 5:e9698CrossRefPubMedPubMedCentral
175.
go back to reference Smecuol E, Mauriño E, Vazquez H, Pedreira S, Niveloni S, Mazure R, Boerr L, Bai JC (1996) Gynaecological and obstetric disorders in coeliac disease: frequent clinical onset during pregnancy or the puerperium. Eur J Gastroenterol Hepatol 8:63–68CrossRefPubMed Smecuol E, Mauriño E, Vazquez H, Pedreira S, Niveloni S, Mazure R, Boerr L, Bai JC (1996) Gynaecological and obstetric disorders in coeliac disease: frequent clinical onset during pregnancy or the puerperium. Eur J Gastroenterol Hepatol 8:63–68CrossRefPubMed
176.
go back to reference Lasa JS, Zubiaurre I, Soifer LO (2014) Risk of infertility in patients with celiac disease: a meta-analysis of observational studies. Arq Gastroenterol 51:144–150CrossRefPubMed Lasa JS, Zubiaurre I, Soifer LO (2014) Risk of infertility in patients with celiac disease: a meta-analysis of observational studies. Arq Gastroenterol 51:144–150CrossRefPubMed
177.
go back to reference Anjum N, Baker PN, Robinson NJ, Aplin JD (2009) Maternal celiac disease autoantibodies bind directly to syncytiotrophoblast and inhibit placental tissue transglutaminase activity. Reprod Biol Endocrinol 7:16CrossRefPubMedPubMedCentral Anjum N, Baker PN, Robinson NJ, Aplin JD (2009) Maternal celiac disease autoantibodies bind directly to syncytiotrophoblast and inhibit placental tissue transglutaminase activity. Reprod Biol Endocrinol 7:16CrossRefPubMedPubMedCentral
178.
go back to reference Sóñora C, Calo G, Fraccaroli L, Pérez-Leirós C, Hernández A, Ramhorst R (2014) Tissue transglutaminase on trophoblast cells as a possible target of autoantibodies contributing to pregnancy complications in celiac patients. Am J Reprod Immunol 72:485–495CrossRefPubMed Sóñora C, Calo G, Fraccaroli L, Pérez-Leirós C, Hernández A, Ramhorst R (2014) Tissue transglutaminase on trophoblast cells as a possible target of autoantibodies contributing to pregnancy complications in celiac patients. Am J Reprod Immunol 72:485–495CrossRefPubMed
Metadata
Title
Transglutaminase 2 and Transglutaminase 2 Autoantibodies in Celiac Disease: a Review
Authors
Tiina Rauhavirta
Minna Hietikko
Teea Salmi
Katri Lindfors
Publication date
01-08-2019
Publisher
Springer US
Published in
Clinical Reviews in Allergy & Immunology / Issue 1/2019
Print ISSN: 1080-0549
Electronic ISSN: 1559-0267
DOI
https://doi.org/10.1007/s12016-016-8557-4

Other articles of this Issue 1/2019

Clinical Reviews in Allergy & Immunology 1/2019 Go to the issue
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

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.