Skip to main content
Top
Published in: BMC Immunology 1/2018

Open Access 01-12-2018 | Research article

Small-intestinal TG2-specific plasma cells at different stages of coeliac disease

Authors: Minna Hietikko, Outi Koskinen, Kalle Kurppa, Kaija Laurila, Päivi Saavalainen, Teea Salmi, Tuire Ilus, Heini Huhtala, Katri Kaukinen, Katri Lindfors

Published in: BMC Immunology | Issue 1/2018

Login to get access

Abstract

Background

In coeliac disease, ingestion of gluten induces the production of transglutaminase 2 (TG2)-targeted autoantibodies by TG2-specific plasma cells present at high frequency in the small intestinal mucosa in untreated disease. During treatment with a gluten-free diet (GFD), the number of these cells decreases considerably. It has not been previously investigated whether the cells are also present prior to development of villous atrophy, or in non-responsive patients and those with dietary lapses. We aimed to define the frequency of small bowel mucosal TG2-specific plasma cells in coeliac disease patients with varying disease activity, and to investigate whether the frequency correlates with serum and small intestinal TG2-targeting antibodies as well as mucosal morphology and the number of intraepithelial lymphocytes.

Results

Mucosal TG2-specific plasma cells were found in 79% of patients prior to development of mucosal damage, in all patients with villous atrophy, and in 63% of the patients after 1 year on GFD. In these disease stages, TG2-specific plasma cells accounted for median of 2.3, 4.3, and 0.7% of all mucosal plasma cells, respectively. After long-term treatment, the cells were present in 20% of the patients in clinical remission (median 0%) and in 60% of the patients with poor dietary adherence (median 5.8%). In patients with non-responsive coeliac disease despite strict GFD, the cells were found in only one (9%) subject; the cells accounted for 2.4% of all plasma cells. A positive correlation between the percentage of TG2-specific plasma cells and serum TG2 antibody levels (rS = 0.69, P < 0.001) and the intensity of mucosal TG2-targeting IgA deposits (rS = 0.43, P < 0.001) was observed.

Conclusions

Our results show that TG2-specific plasma cells are already detectable prior to villous atrophy, and that generally their frequency increases during overt disease. By contrast, on GFD, the percentage of these cells decreases. Overall, the presence of TG2-specific plasma cells in the small bowel mucosa mirrors the presence of gluten in the diet, but the frequency is not always parallel to the level of serum or intestinal TG2 antibodies. These findings increase the knowledge about the development of the TG2 plasma cell responses especially in the early phases of coeliac disease.
Appendix
Available only for authorised users
Literature
1.
go back to reference Dieterich W, Ehnis T, Bauer M, Donner P, Volta U, Riecken E, et al. Identification of tissue transglutaminase as the autoantigen of celiac disease. Nat Med. 1997;3:797–801.CrossRefPubMed Dieterich W, Ehnis T, Bauer M, Donner P, Volta U, Riecken E, et al. Identification of tissue transglutaminase as the autoantigen of celiac disease. Nat Med. 1997;3:797–801.CrossRefPubMed
2.
go back to reference Sulkanen S, Halttunen T, Laurila K, Kolho K, Korponay-Szabó IR, Sarnesto A, et al. Tissue transglutaminase autoantibody enzyme-linked immunosorbent assay in detecting celiac disease. Gastroenterology. 1998;115:1322–8.CrossRefPubMed Sulkanen S, Halttunen T, Laurila K, Kolho K, Korponay-Szabó IR, Sarnesto A, et al. Tissue transglutaminase autoantibody enzyme-linked immunosorbent assay in detecting celiac disease. Gastroenterology. 1998;115:1322–8.CrossRefPubMed
3.
go back to reference Korponay-Szabo IR, Halttunen T, Szalai Z, Laurila K, Kiraly R, Kovacs JB, et al. In vivo targeting of intestinal and extraintestinal transglutaminase 2 by coeliac autoantibodies. Gut. 2004;53:641–8.CrossRefPubMedPubMedCentral Korponay-Szabo IR, Halttunen T, Szalai Z, Laurila K, Kiraly R, Kovacs JB, et al. In vivo targeting of intestinal and extraintestinal transglutaminase 2 by coeliac autoantibodies. Gut. 2004;53:641–8.CrossRefPubMedPubMedCentral
4.
go back to reference Kaukinen K, Peräaho M, Collin P, Partanen J, Woolley N, Kaartinen T, et al. Small-bowel mucosal transglutaminase 2-specific IgA deposits in coeliac disease without villous atrophy: a prospective and randomized clinical study. Scand J Gastroenterol. 2005;40:564–72.CrossRefPubMed Kaukinen K, Peräaho M, Collin P, Partanen J, Woolley N, Kaartinen T, et al. Small-bowel mucosal transglutaminase 2-specific IgA deposits in coeliac disease without villous atrophy: a prospective and randomized clinical study. Scand J Gastroenterol. 2005;40:564–72.CrossRefPubMed
5.
go back to reference Salmi T, Collin P, Järvinen O, Haimila K, Partanen J, Laurila K, et al. Immunoglobulin a autoantibodies against transglutaminase 2 in the small intestinal mucosa predict forthcoming coeliac disease. Aliment Pharmacol Ther. 2006;24:541–52.CrossRefPubMed Salmi T, Collin P, Järvinen O, Haimila K, Partanen J, Laurila K, et al. Immunoglobulin a autoantibodies against transglutaminase 2 in the small intestinal mucosa predict forthcoming coeliac disease. Aliment Pharmacol Ther. 2006;24:541–52.CrossRefPubMed
6.
go back to reference Salmi TT, Collin P, Korponay-Szabo IR, Laurila K, Partanen J, Huhtala H, et al. Endomysial antibody-negative coeliac disease: clinical characteristics and intestinal autoantibody deposits. Gut. 2006;55:1746–53.CrossRefPubMedPubMedCentral Salmi TT, Collin P, Korponay-Szabo IR, Laurila K, Partanen J, Huhtala H, et al. Endomysial antibody-negative coeliac disease: clinical characteristics and intestinal autoantibody deposits. Gut. 2006;55:1746–53.CrossRefPubMedPubMedCentral
7.
go back to reference Koskinen O, Collin P, Lindfors K, Laurila K, Maki M, Kaukinen K. Usefulness of small-bowel mucosal transglutaminase-2 specific autoantibody deposits in the diagnosis and follow-up of celiac disease. J Clin Gastroenterol. 2010;44:483–8.PubMed Koskinen O, Collin P, Lindfors K, Laurila K, Maki M, Kaukinen K. Usefulness of small-bowel mucosal transglutaminase-2 specific autoantibody deposits in the diagnosis and follow-up of celiac disease. J Clin Gastroenterol. 2010;44:483–8.PubMed
8.
go back to reference Ilus T, Kaukinen K, Virta L, Huhtala H, Mäki M, Kurppa K, et al. Refractory coeliac disease in a country with a high prevalence of clinically-diagnosed coeliac disease. Aliment Pharmacol Ther. 2014;39:418–25.CrossRefPubMed Ilus T, Kaukinen K, Virta L, Huhtala H, Mäki M, Kurppa K, et al. Refractory coeliac disease in a country with a high prevalence of clinically-diagnosed coeliac disease. Aliment Pharmacol Ther. 2014;39:418–25.CrossRefPubMed
9.
go back to reference Marzari R, Sblattero D, Florian F, Tongiorgi E. Not T, Tommasini a , et al. molecular dissection of the tissue transglutaminase antoantibody response in celiac disease. J Immunol. 2001;166:4170–6.CrossRefPubMed Marzari R, Sblattero D, Florian F, Tongiorgi E. Not T, Tommasini a , et al. molecular dissection of the tissue transglutaminase antoantibody response in celiac disease. J Immunol. 2001;166:4170–6.CrossRefPubMed
10.
go back to reference Di Niro R, Mesin L, Zheng N, Stamnaes J, Morrissey M, Lee J, et al. High abundance of plasma cells secreting transglutaminase 2-specific IgA autoantibodies with limited somatic hypermutation in celiac disease intestinal lesions. Nat Med. 2012;18:441–U204.CrossRefPubMedPubMedCentral Di Niro R, Mesin L, Zheng N, Stamnaes J, Morrissey M, Lee J, et al. High abundance of plasma cells secreting transglutaminase 2-specific IgA autoantibodies with limited somatic hypermutation in celiac disease intestinal lesions. Nat Med. 2012;18:441–U204.CrossRefPubMedPubMedCentral
11.
go back to reference Di Niro R, Snir O, Kaukinen K, Yaari G, Lundin K, Gupta N, et al. Responsive population dynamics and wide seeding into the duodenal lamina propria of transglutaminase-2-specific plasma cells in celiac disease. Mucosal Immunol. 2016;9:254–64. Di Niro R, Snir O, Kaukinen K, Yaari G, Lundin K, Gupta N, et al. Responsive population dynamics and wide seeding into the duodenal lamina propria of transglutaminase-2-specific plasma cells in celiac disease. Mucosal Immunol. 2016;9:254–64.
12.
go back to reference Iversen R, Snir O, Stensland M, Kroll JE, Steinsbø Ø, Korponay-Szabó IR, et al. Strong clonal relatedness between serum and gut IgA despite different plasma cell origins. Cell Rep. 2017;20:2357–67.CrossRefPubMedPubMedCentral Iversen R, Snir O, Stensland M, Kroll JE, Steinsbø Ø, Korponay-Szabó IR, et al. Strong clonal relatedness between serum and gut IgA despite different plasma cell origins. Cell Rep. 2017;20:2357–67.CrossRefPubMedPubMedCentral
13.
go back to reference Kaukinen K, Turjanmaa K, Mäki M, Partanen J, Venäläinen R, Reunala T, et al. Intolerance to cereals is not specific for coeliac disease. Scand J Gastroenterol. 2000;35:942–6.CrossRefPubMed Kaukinen K, Turjanmaa K, Mäki M, Partanen J, Venäläinen R, Reunala T, et al. Intolerance to cereals is not specific for coeliac disease. Scand J Gastroenterol. 2000;35:942–6.CrossRefPubMed
14.
go back to reference Taavela J, Koskinen O, Huhtala H, Lähdeaho M, Popp A, Laurila K, et al. Validation of morphometric analyses of small-intestinal biopsy readouts in celiac disease. PLoS One. 2013;8:e76163.CrossRefPubMedPubMedCentral Taavela J, Koskinen O, Huhtala H, Lähdeaho M, Popp A, Laurila K, et al. Validation of morphometric analyses of small-intestinal biopsy readouts in celiac disease. PLoS One. 2013;8:e76163.CrossRefPubMedPubMedCentral
15.
go back to reference Järvinen TT, Kaukinen K, Laurila K, Kyrönpalo S, Rasmussen M, Mäki M, et al. Intraepithelial lymphocytes in celiac disease. Am J Gastroenterol. 2003;98:1332–7.CrossRefPubMed Järvinen TT, Kaukinen K, Laurila K, Kyrönpalo S, Rasmussen M, Mäki M, et al. Intraepithelial lymphocytes in celiac disease. Am J Gastroenterol. 2003;98:1332–7.CrossRefPubMed
16.
go back to reference Sulkanen S, Collin P, Laurila K, Mäki M. IgA-and IgG-class antihuman umbilical cord antibody tests in adult coeliac disease. Scand J Gastroenterol. 1998;33:251–4.CrossRefPubMed Sulkanen S, Collin P, Laurila K, Mäki M. IgA-and IgG-class antihuman umbilical cord antibody tests in adult coeliac disease. Scand J Gastroenterol. 1998;33:251–4.CrossRefPubMed
17.
go back to reference Koskinen L, Romanos J, Kaukinen K, Mustalahti K, Korponay-Szabo I, Barisani D, et al. Cost-effective HLA typing with tagging SNPs predicts celiac disease risk haplotypes in the Finnish, Hungarian, and Italian populations. Immunogenetics. 2009;61:247–56.CrossRefPubMed Koskinen L, Romanos J, Kaukinen K, Mustalahti K, Korponay-Szabo I, Barisani D, et al. Cost-effective HLA typing with tagging SNPs predicts celiac disease risk haplotypes in the Finnish, Hungarian, and Italian populations. Immunogenetics. 2009;61:247–56.CrossRefPubMed
18.
19.
go back to reference Stamnaes J, Sollid LM. Celiac disease: autoimmunity in response to food antigen. Semin Immunol. 2015;27:343–52.CrossRefPubMed Stamnaes J, Sollid LM. Celiac disease: autoimmunity in response to food antigen. Semin Immunol. 2015;27:343–52.CrossRefPubMed
Metadata
Title
Small-intestinal TG2-specific plasma cells at different stages of coeliac disease
Authors
Minna Hietikko
Outi Koskinen
Kalle Kurppa
Kaija Laurila
Päivi Saavalainen
Teea Salmi
Tuire Ilus
Heini Huhtala
Katri Kaukinen
Katri Lindfors
Publication date
01-12-2018
Publisher
BioMed Central
Published in
BMC Immunology / Issue 1/2018
Electronic ISSN: 1471-2172
DOI
https://doi.org/10.1186/s12865-018-0275-7

Other articles of this Issue 1/2018

BMC Immunology 1/2018 Go to the issue