eISSN: 1644-4124
ISSN: 1426-3912
Central European Journal of Immunology
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Abstracting and indexing Subscription Contact Instructions for authors
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
3/2014
vol. 39
 
Share:
Share:

Review paper
Celiac disease as an autoimmune condition

Gabriel Samasca
,
Genel Sur
,
Iulia Lupan
,
Mariana Tilinca
,
Diana Deleanu

(Centr Eur J Immunol 2014; 39 (3): 396-399)
Online publish date: 2014/10/14
Article file
- Celiac disease.pdf  [0.12 MB]
Get citation
 
PlumX metrics:
 

Introduction

In the United States, the increase in prevalence of celiac disease (CD) during a 15-year period was attributable to the increasing number of the subjects who lost the immunological tolerance to gluten in their adulthood [1]. In Europe, CD is widespread in the Western countries [2]. A clear environmental trigger has been identified in CD [3], namely gluten from wheat, barley, and rye [4]. Patients with CD are continuously exposed to the exogenous dietary antigen gluten [5]. Therefore, CD is a self-perpetuating autoimmune condition [6], influenced by disorders of the gut microbiota [7]. Why is it necessary to study separate autoimmune phenomena from CD? Innate gluten sensitivity, adaptive gluten sensitivity and autoimmunity are independent phenomena and are all essential in the development of CD [8].

Material and methods

In a recent study we have analyzed the immune response and new immunological challenges in CD [9]. The aim of our study was to follow the changes in the clinical autoimmunity picture of CD from recent years. We conducted the study on the PubMed database. Eligibility criteria consisted of data found in articles indexed in the PubMed database about autoimmunity in CD.

Results and discussion

Clinical presentation

Gluten exposure in infants genetically susceptible to CD leads to an immune response against gluten. These are more common than at-risk infants, in which gluten exposure occurs late until 12 months of age [10]. The single life-long treatment is a gluten-free diet (GFD) [11]. Some study showed an increased incidence and prevalence of CD in developed countries [12]. However, autoimmune disease prevalence including CD was 1.4% in older women (95% CI: 1.3%, 1.5%), which is the evidence that GFD is beneficial [13]. But gender differences were also found in CD [14].
The prevalence of type 1 diabetes mellitus (T1DM) among CD families was 0.9% [15]. Within the framework of polyglandular disorders, T1DM can coexist with CD. But in CD organ-specific autoantibodies can occur [16]. Children diagnosed with T1DM before the age of 6 had a major risk of developing CD, compared with those diagnosed after this age [17]. After long-term monitoring of diabetes [18], CD appearance in the T1DM was identified as one of the autoimmune morbidities [19]. Researchers analysed three functional 5’ un-translated region -defensin 1 (DEFB1) single nucleotide polymorphisms (SNPs) in a group of 170 T1DM patients. They found no association of DEFB1 SNPs with the onset of thyroid autoimmunity disease (TA), CD, and both TA and CD in T1DM patients [20]. But the prevalence of CD is 5.4% in patients with TA [21]. Thyroid autoimmunity disease appeared to be as common in paediatric and adolescent patients with CD on a GFD as in controls [22]. Another recent study highlighted the fact that there is a prevalence of 1.2% of CD for Graves’ and 1.2% for Hashimoto’s diseases [23].
Integrin, alpha M (complement component 3 receptor 3 subunit) and its related ‘predisposing’ variant (rs1143679, Arg77His) are predicted to alter the tertiary structures of the ligand-binding domain and can play a key role in systemic lupus erythematosus (SLE) pathogenesis. A group of researchers evaluated case-control associations between rs1143679 and autoimmune diseases (n = 18,457) including CD. The conclusions were that integrin, alpha M (complement component 3 receptor 3 subunit) could not possibly be a general autoimmunity gene but this variant could eventually be associated with SLE and systemic sclerosis [24].
In CD, bone metabolic changes are more frequently associated with inflammatory joint disorders. Several disorders have been revealed as associated diseases/extraintestinal manifestations, including rheumatological diseases [25]. According to Iqbal et al., a high rate of spondyloarthritis could not be observed in CD patients but increased rates of T1DM, TA, SLE, and psoriasis could [26]. Another possible association with CD, namely the risk of psoriasis was found in CD patients with vitamin D deficiency (7% vs. 3%, p = 0.04) [27].
An association between active CD and hyperprolactinemia was found suggestively higher in patients with active CD than in patients with an inactive one [28]. A 2.7% prevalence of CD in Turner syndrome (TS) [29] was found. The most common clinical features of TS are short stature and gonadal dysgenesis. Grossi et al. presented a unique case of mosaic TS with a complex rearrangement involving a partial deletion of chromosome 2q and duplication of chromosome 10p. Thyroid autoimmunity disease was associated with a group of multiorgan autoimmunity-related manifestations including Hashimoto’s thyroiditis, CD, T1DM. Their conclusion was that timely genetic analysis in TS patients with complex associations of multiorgan autoimmune manifestations would enable a detailed diagnostic classification [30].
Another rare, chronic endocrine disorder in which the adrenal glands do not produce enough steroid hormones is autoimmune Addison’s disease (AAD). In AAD, a 3.5% prevalence of CD [31] was identified. Celiac disease was occasionally associated with a neurologic disease (4 patients among 70 CD patients), and with antibody reactivity to neuronal antigens [32]. But a recent study described CD as one of the autoimmune diseases most often associated with sensory neuronopathies [33].

Diagnosis

Tissue transglutaminase (tTG) is a deamidating enzyme that can increase the immunostimulatory effect of gluten, and a target autoantigen in the immune response [34, 35]. Autoantibodies to tTG showed high CD specificity and sensitivity [36]. Immune reactivity to other autoantigens, including transglutaminase 3, actin, ganglioside, collagen, calreticulin or zonulin has also been reported in CD [37]. The small-bowel mucosa and other tissues deposited CD specific tTG-targeted autoantibodies. Therefore, extraintestinal manifestations of the CD [38] have been reported. Marwaha et al. found high positivity of anti-glutamic acid decarboxylase and anti-tTG antibodies, i.e. 6.9 and 12.5 per cent in subjects among cases compared to 3.5 per cent (p = 0.015) and 4.3 per cent (p = 0.001) in controls, among subjects with TA [39]. The researchers observed a slight and probably irrelevant increase in IgG tTG antibody in patients with rheumatoid arthritis treated with adalimumab [40]. Tissue transglutaminase has also been associated with angiogenesis disturbance in CD [41].
One or more CD HLA-DQ heterodimers increased the risk of developing CD (p < 0.001) [42]. The HLA-DR3-DQ2 haplotype in children with T1DM was associated with CD in the extended family (p < 0.001) [43]. By stratifying the HLA-DQ, Östensson et al. identified a new genome-wide significant risk locus covering the DUSP10 gene [44].

New immunological research directions

Interleukin-1 has a systemic influence on the loss of immunological tolerance and NALP3 inflammasome was directly involved in the pro-inflammatory cytokine production. Pontillo et al. highlighted an association of NLRP3 rs358294199 SNP to CD (p = 5exp-4). They hypothesized that variations in NLRP3 could belong to a predisposing genetic background that triggers autoimmune diseases development [45]. The increase in Th1 serum cytokines could possibly be associated with CD in offspring [46]. The associations for interleukin-2 gene polymorphisms and its - and -chain receptor (IL2RA and IL2RB) variants of different autoimmune diseases including CD were also suggested [47]. Protein tyrosine phosphatase non-receptor type 22 (PTPN22) is a strong susceptibility gene spread among many autoimmune diseases. PTPN22 showed a negligible association with CD [48]. Hmida et al. isolated intraepithelial lymphocytes (IELs) and lamina propria lymphocytes (LPLs) from duodenal biopsy specimens of CD patients and controls. CD4+CD25+ T lymphocytes (Tregs) were purified from blood. They tested responses of IELs, LPLs and peripheral lymphocytes (PBLs) to suppression by Tregs. Their results indicated that effector T lymphocytes from active CD become resistant to suppression by Tregs. Their conclusion was that this resistance might cause loss of tolerance to gluten, but also to self-antigens [49]. The level of soluble CTLA-4 (Cytotoxic T-Lymphocyte Antigen 4), also known as CD152 (cluster of differentiation 152) was evaluated in the sera of CD patients with overlapping autoimmune diseases (obliterative airway disease (OAD), T1DM, TA, inflammatory bowel diseases, and autoimmune polyendocrine syndromes). The results showed a statistically significant association for serum sCTLA-4 levels with associated autoimmune disease (i.e. OAD) in CD patients vs. patients with CD only [50].
The infection with Necator americanus remains a challenge, which elicited parasite-specific immunity and modified the host’s immune response to gluten: mucosal IL-1 and IL-22 responses were enhanced, but IFN- and IL-17A levels and circulating regulatory T cells following gluten challenges were suppressed [51]. The biggest challenge remains the definitive identification of the pathogenic T cell epitopes, which would outline the antigen-specific immunotherapy in CD [52].

Authors declare no conflict of interest.
This paper was published under the framework of the European Social Fund, Human Resources Development Operational Programme 2007-2013, project no. POSDRU/159/1.5/S/138776.

References

1. Catassi C, Kryszak D, Bhatti B, et al. (2010): Natural history of celiac disease autoimmunity in a USA cohort followed since 1974. Ann Med 42: 530-538.
2. Kaukinen K, Mäki M (2014): Coeliac disease in 2013: new insights in dietary-gluten-induced autoimmunity. Nat Rev Gastroenterol Hepatol 11: 80-82.
3. Selmi C (2012): Cutting-edge issues in autoimmunity and allergy of the digestive system. Clin Rev Allergy Immunol 42: 265-268.
4. Denham JM, Hill ID (2013): Celiac disease and autoimmunity: review and controversies. Curr Allergy Asthma Rep 13: 347-353.
5. Sollid LM, Jabri B (2013): Triggers and drivers of autoimmunity: lessons from coeliac disease. Nat Rev Immunol 13: 294-302.
6. Rossi M, Bot A (2011): Celiac disease: progress towards diagnosis and definition of pathogenic mechanisms. Int Rev Immunol 30: 183-184.
7. Lupan I, Sur G, Deleanu D, et al. (2013): Celiac disease microbiota and its applications. Ann Microbiol 64: 899-903.
8. Marietta EV, David CS, Murray JA (2011): Important lessons derived from animal models of celiac disease. Int Rev Immunol 30: 197-206.
9. Samasca G, Lupan I, Deleanu D, et al. (2014): Immunological approach of the challenges of the XXI century in celiac disease. Int Rev Immunol 33: 3-8.
10. Sellitto M, Bai G, Serena G, et al. (2012): Proof of concept of microbiome-metabolome analysis and delayed gluten exposure on celiac disease autoimmunity in genetically at-risk infants. PLoS One 7: e33387.
11. Lerner A (2010): New therapeutic strategies for celiac disease. Autoimmun Rev 9: 144-147.
12. Logan I, Bowlus CL (2010): The geoepidemiology of autoimmune intestinal diseases. Autoimmun Rev 9: A372-378.
13. Rogers MA, Levine DA, Blumberg N, et al. (2012): Antigenic challenge in the etiology of autoimmune disease in women. J Autoimmun 38: J97-J102.
14. Nussinovitch U, Shoenfeld Y (2012): The role of gender and organ specific autoimmunity. Autoimmun Rev 11: A377-385.
15. Neuhausen SL, Steele L, Ryan S, et al. (2008): Co-occurrence of celiac disease and other autoimmune diseases in celiacs and their first-degree relatives. J Autoimmun 31: 160-165.
16. Tsirogianni A, Pipi E, Soufleros K (2009): Specificity of islet cell autoantibodies and coexistence with other organ specific autoantibodies in type 1 diabetes mellitus. Autoimmun Rev 8: 687-691.
17. Levy-Shraga Y, Lerner-Geva L, Boyko V, et al. (2012): Type 1 diabetes in pre-school children – long-term metabolic control, associated autoimmunity and complications. Diabet Med 29: 1291-1296.
18. Rewers M, Eisenbarth GS (2011): Autoimmunity: celiac disease in T1DM-the need to look long term. Nat Rev Endocrinol 8: 7-8.
19. H Nagy K, Rózsai B, Kürti K, et al. (2012): Autoimmune comorbidity and microvascular complications in childhood-onset type 1 diabetes after 20 years of diabetes duration: a questionnaire study. Orv Hetil 153: 222-226.
20. Guimarães RL, Segat L, Rocha CR, et al. (2009): Functional polymorphisms of DEFB1 gene in type 1 diabetes Brazilian children. Autoimmunity 42: 406-413.
21. Spadaccino AC, Basso D, Chiarelli S, et al. (2008): Celiac disease in North Italian patients with autoimmune thyroid diseases. Autoimmunity 41: 116-121.
22. Diamanti A, Ferretti F, Guglielmi R, et al. (2011): Thyroid autoimmunity in children with coeliac disease: a prospective survey. Arch Dis Child 96: 1038-1041.
23. Wiebolt J, Achterbergh R, den Boer A, et al. (2011): Clustering of additional autoimmunity behaves differently in Hashimoto’s patients compared with Graves’ patients. Eur J Endocrinol 164: 789-794.
24. Anaya JM, Kim-Howard X, Prahalad S, et al. (2012): Evaluation of genetic association between an ITGAM non-synonymous SNP (rs1143679) and multiple autoimmune diseases. Autoimmun Rev 11: 276-280.
25. Vereckei E, Szodoray P, Poor G, Kiss E (2011): Genetic and immunological processes in the pathomechanism of gluten-sensitive enteropathy and associated metabolic bone disorders. Autoimmun Rev 10: 336-340.
26. Iqbal T, Zaidi MA, Wells GA, Karsh J (2013): Celiac disease arthropathy and autoimmunity study. J Gastroenterol Hepatol 28: 99-105.
27. Tavakkoli A, DiGiacomo D, Green PH, Lebwohl B (2013): Vitamin D status and concomitant autoimmunity in celiac disease. J Clin Gastroenterol 47: 515-519.
28. Shelly S, Boaz M, Orbach H (2012): Prolactin and autoimmunity. Autoimmun Rev 11: A465-470.
29. Bakalov VK, Gutin L, Cheng CM, et al. (2012): Autoimmune disorders in women with turner syndrome and women with karyotypically normal primary ovarian insufficiency. J Autoimmun 38: 315-321.
30. Grossi A, Palma A, Zanni G, et al. (2013): Multiorgan autoimmunity in a Turner syndrome patient with partial monosomy 2q and trisomy 10p. Gene 515: 439-443.
31. Fichna M, Fichna P, Gryczyńska M, et al. (2010): Screening for associated autoimmune disorders in Polish patients with Addison’s disease. Endocrine 37: 349-360.
32. Briani C, Doria A, Ruggero S, et al. (2018): Antibodies to muscle and ganglionic acetylcholine receptors (AchR) in celiac disease. Autoimmunity 41: 100-104.
33. Martinez AR, Nunes MB, Nucci A, França MC Jr. (2012): Sensory neuronopathy and autoimmune diseases. Autoimmune Dis 2012: 873587.
34. Di Sabatino A, Vanoli A, Giuffrida P, et al. (2012): The function of tissue transglutaminase in celiac disease. Autoimmun Rev 11: 746-753.
35. Gornowicz-Porowska J, Seraszek-Jaros A, Kaczmarek E, et al. (2012): IgA autoantibodies to gliadin nonapeptides, tissue transglutaminase and epidermal transglutaminase are associated, but unrelated to neutrophil elastase expression in lesional skin in human dermatitis herpetiformis. Postep Derm Alergol 29: 233-239.
36. Sollid LM, Jabri B (2011): Celiac disease and transglutaminase 2: a model for posttranslational modification of antigens and HLA association in the pathogenesis of autoimmune disorders. Curr Opin Immunol 23: 732-738.
37. Alaedini A, Green PH (2008): Autoantibodies in celiac disease. Autoimmunity 41: 19-26.
38. Lindfors K, Mäki M, Kaukinen K (2010): Transglutaminase 2-targeted autoantibodies in celiac disease: Pathogenetic players in addition to diagnostic tools? Autoimmun Rev 9: 744-749.
39. Marwaha RK, Garg MK, Tandon N, et al. (2013): Glutamic acid decarboxylase (anti-GAD) & tissue transglutaminase (anti-TTG) antibodies in patients with thyroid autoimmunity. Indian J Med Res 137: 82-86.
40. Atzeni F, Doria A, Ghirardello A, et al. (2008): Organ-specific autoantibodies in patients with rheumatoid arthritis treated with adalimumab: a prospective long-term follow-up. Autoimmunity 41: 87-91.
41. Castellanos-Rubio A, Caja S, Irastorza I, et al. (2012): Angiogenesis-related gene expression analysis in celiac disease. Autoimmunity 45: 264-270.
42. Larizza D, Calcaterra V, Klersy C, et al. (2012): Common immunogenetic profile in children with multiple autoimmune diseases: the signature of HLA-DQ pleiotropic genes. Autoimmunity 45: 470-475.
43. Parkkola A, Härkönen T, Ryhänen SJ, et al. (2013): Finnish Pediatric Diabetes Register. Extended family history of autoimmune diseases and phenotype and genotype of children with newly diagnosed type 1 diabetes. Eur J Endocrinol 169: 171-178.
44. Östensson M, Montén C, Bacelis J, et al. (2013): A possible mechanism behind autoimmune disorders discovered by genome-wide linkage and association analysis in celiac disease. PLoS One 8: e70174.
45. Pontillo A, Brandao L, Guimaraes R, et al. (2010): Two SNPs in NLRP3 gene are involved in the predisposition to type-1 diabetes and celiac disease in a pediatric population from northeast Brazil. Autoimmunity 43: 583-589.
46. Lindehammer SR, Björck S, Lynch K, et al. (2011): Early human pregnancy serum cytokine levels predict autoimmunity in offspring. Autoimmunity 44: 445-452.
47. Espino-Paisán L, De La Calle H, Fernández-Arquero M, et al. (2011): Study of polymorphisms in 4q27, 10p15, and 22q13 regions in autoantibodies stratified type 1 diabetes patients. Autoimmunity 44: 624-630.
48. Zheng J, Ibrahim S, Petersen F, Yu X (2012): Meta-analysis reveals an association of PTPN22 C1858T with autoimmune diseases, which depends on the localization of the affected tissue. Genes Immun 13: 641-652.
49. Hmida NB, Ben Ahmed M, Moussa A, et al. (2012): Impaired control of effector T cells by regulatory T cells: a clue to loss of oral tolerance and autoimmunity in celiac disease? Am J Gastroenterol 107: 604-611.
50. Pesce G, Auricchio R, Bagnasco M, Saverino D (2014): Oversecretion of soluble CTLA-4 in various autoimmune diseases overlapping celiac disease. Genet Test Mol Biomarkers 18: 8-11.
51. Croese J, Gaze ST, Loukas A (2013): Changed gluten immunity in celiac disease by Necator americanus provides new insights into autoimmunity. Int J Parasitol 43: 275-282.
52. Anderson RP, Jabri B (2013): Vaccine against autoimmune disease: antigen-specific immunotherapy. Curr Opin Immunol 25: 410-417.
Copyright: © 2014 Polish Society of Experimental and Clinical Immunology This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.

Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.