Elsevier

Autoimmunity Reviews

Volume 18, Issue 3, March 2019, Pages 241-246
Autoimmunity Reviews

Review
Autoimmunity in celiac disease: Extra-intestinal manifestations

https://doi.org/10.1016/j.autrev.2018.09.010Get rights and content

Abstract

Celiac disease is an autoimmune condition of the small intestine caused by prolamins in genetically susceptible individuals evoked by multiple environmental factors. The pathological luminal intricate eco-events produce multiple signals that irradiate the entire body, resulting in a plethora of extra-intestinal manifestations. Nutrients, dysbiosis, dysbiotic components and their mobilome, post-translational modification of naive proteins, inter-enterocyte's tight junction dysfunction resulting in a leaky gut, microbial lateral genetic transfer of virulent genes, the sensing network of the enteric nervous systems and the ensuing pro-inflammatory messengers are mutually orchestrating the autoimmune interplay. Genetic-environmental-luminal events-mucosal changes are driving centrifugally the remote organs autoimmunity, establishing extra-intestinal multi organ injury. Exploring the underlying intestinal eco-events, the sensing and the delivery pathways and mechanisms that induce the peripheral tissues' damages might unravel new therapeutical strategies to prevent and help the gluten affected patients.

Introduction

The target organ of celiac disease (CD) is the human proximal small bowel and in the past, symptomatology encompassed mainly small bowel presenting symptoms. Being a constantly changing disease, in the last decades and nowadays, novel features are continuously being unraveled. A lot of new observations and scientific knowledge on its genetic markers, epidemiology, clinical presentations, mode of diagnosis, therapeutic evolvement and high risk conditions were accumulated. Before embarking on its extra-intestinal manifestation (EIM), a background on CD and its shared aspects with other autoimmune diseases (ADs) is presented.

Celiac disease is a life-long autoimmune condition [1] of the enteric tract, targeting mainly the small intestine and systemically multiple extra-intestinal sites, in genetically susceptible individuals. It affects 1–2% of Western populations and its incidence is continuously increasing [2,3]. Gluten is the storage protein of wheat and its alcohol soluble gliadins are the offending toxic molecules of the disease together with structurally related components found in barley, rye and less in oat [4]. Tissue transglutaminase (tTG) is the auto antigen [5,6] and two main auto antibodies: IgA-anti endomysium and IgA-anti tTG are the most frequently used serological markers to screen and diagnose the disease [7,8]. Lately, a new serological marker immerged, targeting neoepitope complexes formed when gliadin docks the tTG enzyme [9]. It appears that compared to the traditional IgA-tTG, the neoepitope tTG out performs and is considered as a reliable diagnostic marker and reflector of the intestinal pathology [10,11]. Interestingly, the microbial transglutaminase (mTG), a heavily used industrial food additive, that imitates functionally the tTG, was recently shown to induce specific antibodies in CD patients [12]. Potentially, it might represent a new environmental player in driving CD autoimmunogenesis [[13], [14], [15]].

The genetic repertoire of CD is expanding, containing more than 50 non-HLA genes but only two of them, HLA-DQ2 and HLA-DQ8 molecules are the most important, predisposing genetic factors [16].

The pathogenesis of the disease is dominated by the intestinal innate and humoral immune systems activation, resulting in enteric inflammation and destruction. In fact, each pathogenetic step: the ingested toxic environmental gluten, the luminal enzyme resistant gluten peptides, their increased intestinal uptake, the increased IL-15 and IFNγ production, tTG gluten deamidation and transamidation (cross linking), TCD4+ cell proliferation and macrophage activation, are crucial events that form the basis for development of future potential therapeutic modalities [17,18].

Since genome-wide association studies became available and performed on various ADs, it appears that there exists an overlap of around 50% of genetic risk factors between those diseases [19]. Some of the shared genes are part of the human leukocyte antigens family that are routinely part of the diagnostic work-up in various ADs [20].

Interesting, regulatory genes, in the intronic, non-coding part of the genome, contain most of those genetic factors. Coming back to CD, the condition is associated with numerous ADs, including rheumatologic (S.L.E, rheumatoid arthritis, juvenile rheumatic/idiopathic, Sjogren's syndrome and connective tissue disease), endocrinal (Type 1 diabetes, Ashimoto thyroiditis, Addison's disease), dermatologic (dermatitis herpetiformis, psoriasis), neurologic, cardiologic, hepatic, pancreatic, gynecologic, gastrointestinal and immunological conditions [[21], [22], [23]]. In recent surveys, Hashimoto thyroiditis, type 1 diabetes, psoriasis, Sjogren's syndrome, autoimmune hepatitis, IBD and biliary ADs were the most frequently associated conditions with CD [24,25]. While the effect of gluten free diet is controversial in non-celiac ADs, the duration of the disease is considered a risk factor for polyautoimmunity [25]. It can be argued that shared genetics and environment are at the basis of the polyautoimmunity in CD patients or their family members [26]. More so, multiple screenings identified common genes shared between various ADs, supporting the autoimmune trait as part of the autoimmune tautology [[23], [24], [25], [26], [27]]. In a recent study, the following risk factors were identified for polyautoimmunity in CD patients: female gender, CD associated serology, family history of ADs, insufficient vitamin D levels, having high antinuclear antibody titer, and associate musculoskeletal conditions [28].

Conversely to the old notions upgrading genetics as the most influential, the impact of environmental exposure on autoimmune risk development, is paramount [29]. Multiple environmental factors are similarly shared between ADs. The recently coined term “autoimmune ecology” is the cross relation between us and the surrounding environment, that induce the breach of immune tolerance resulting in one or multiple ADs in an individual [29,30]. Chemicals, organic solvents, physical factors, surgery, nutrients, processed food additives, drugs and sex hormones, heavy metals, infectious agents, dysbiota, vitamins' deficiencies, smoking, alcohol, coffee, life style and stress, socioeconomic status and vaccines are some of them [13,23,29,30]. Notably, CD share some of them. Virus like enterovirus, EBV, CMV, HBV, HCV, rotavirus, reovirus, Microbes like tuberculosis, Bacteroides species, Campylobacter jejuni, pneumococcus, Helicobacter pylori, microbiotic composition and diversity, microbial transglutaminase, first year feeding pattern and antibiotic consumption, mode of delivery, amount and timing of gluten consumption, processed food additives and physical or emotional stress [4,13,14,23,31] were suggested as environmental components that influence CD induction, development or behavior. However, association and not causality was shown, except for gluten consumption in CD.

The title: “Our environment shapes us” [32], set the stage for the additional evolving topic, positioned in-between genetics and environment, meaning epigenetics. In fact, growing evidences are constantly accumulating for the pivotal role of epigenetics in driving autoimmunity. It is not only a loss of tolerance [33], since abnormal epigenetic modifications can present biomarker for ADs and be involved in multiple ADs pathogenic progression [34], CD not excluded [35,36]. Interestingly, most recently the microbiota memory was suggested to govern diseases by epigenetic regulation [37].

After mentioning the shared genes, clustering of ADs in the patients, the shared epigenetic events and environmental factors, the autoimmune trait resulting in familial coaggregation of ADs, the stage is set for extra-intestinal features of CD, shared by numerous ADs.

Section snippets

Extra-intestinal manifestations (EIM) of celiac disease

The scientific and medical literature is rich in comprehensive reviews describing EIM in CD populations. As can be expected, the list is expanding rapidly since the 90th [38] to the present [[39], [40], [41]]. Fig. 1 describes the multiple faces of the disease. The octopus intend to show that the CD arms can reach and affect any tissue/organ in the human body and many of the contemporary presenting symptoms of the disease are included. It goes without saying that patients' complains and

Nutrition

The pivotal place played by nutrients, shaping human physiology and pathology, not just only in the intestinal compartment, is increasingly appreciated. In fact, the nutritional effects are irradiated peripherally to remote organs [69,70] and even to the brain [71]. Some of their effects are channeled by the microbiome, some affect intestinal permeability and many are direct by their digested and absorbed metabolites. Two aspects, related to CD, that might drive systemic autoimmunogenesis, are

Conclusion

Multiple non-intestinal organs and tissues are affected in CD and present an end-organ disease, associated with the enteric one [21,39,41,92]. The gut originated autoimmune inflammatory network spreads peripherally and contributes to the EIM in CD population. Nutrients, bugs, chemical modifications, gut leakage, intestinal wall components like the immune, neuronal, endocrine, endo-mesodermal, hematological and vascular systems, gather together, in a very coordinated way, to initiate and

Acknowledgment

The authors would like to thank Mr. Neu Alf for the figure design and to Dr. Neidhöfer Sandra, Dr. Ramesh Ajay and Mrs. Wusterhausen Patricia for the editing and reviewing the manuscript.

References (92)

  • G. Esposito et al.

    Enteric glial-derived S100B protein stimulates nitric oxide production in celiac disease

    Gastroenterology

    (2007)
  • O. Rozenberg et al.

    A novel algorithm for the diagnosis of celiac disease and a comprehensive review of celiac disease diagnostics

    Crit Rev Allerg Immunol

    (2012)
  • A. Lerner et al.

    The world incidence of celiac disease is increasing: a review

    Int J Recent Scient Res

    (2015)
  • A. Lerner et al.

    The world incidence and prevalence of autoimmune diseases is increasing: a review

    Int J Celiac Dis

    (2015)
  • A. Lerner et al.

    Adverse effects of gluten ingestion and advantages of gluten withdrawal in nonceliac autoimmune disease

    Nutr Rev

    (2017)
  • A. Lerner et al.

    Transglutaminase 2 and anti transglutaminase 2 autoantibodies in celiac disease and beyond: part a: TG2 double-edged sword: gut and extraintestinal involvement

    Immunome Res

    (2015)
  • A. Lerner

    Serological diagnosis of celiac disease—moving beyond the tip of the iceberg

    Int J Celiac Dis

    (2014)
  • A. Lerner et al.

    Transglutaminase 2 and anti transglutaminase 2 autoantibodies in celiac disease and beyond. Part B: anti- transglutaminase 2 autoantibodies: friends or enemies

    Immunome Res

    (2015)
  • A. Lerner et al.

    Comparison of the reliability of 17 celiac disease associated bio-markers to reflect intestinal damage

    J Clin Cell Immunol

    (2017)
  • A. Lerner et al.

    Serological markers and/or intestinal biopsies in the case-finding of celiac disease

    Int. J Celiac Dis

    (2015)
  • A. Lerner et al.

    Possible association between celiac disease and bacterial transglutaminase in food processing: a hypothesis

    Nutr Rev

    (2015)
  • A. Lerner et al.

    Intestinal dysbiotic transglutaminases are potential environmental drivers of systemic autoimmunogenesis

    Front Microbiol

    (2017)
  • K.E. Lundin et al.

    Coeliac disease and autoimmune disease-genetic overlap and screening

    Nat Rev Gastroenterol Hepatol

    (2015)
  • S. Kurada et al.

    Current and novel therapeutic strategies in celiac disease

    Expert Rev Clin Pharmacol

    (2016)
  • I.H. Jonkers et al.

    Context-specific effects of genetic variants associated with autoimmune disease

    Hum Mol Genet

    (2017)
  • G. Bodis et al.

    Role of human leukocyte antigens (HLA) in autoimmune diseases

    Rheumatol Ther

    (2018 Mar 7)
  • E. Lauret et al.

    Celiac disease and autoimmune-associated conditions

    Biomed Res Int

    (2013)
  • M.U. Lodhi et al.

    Celiac disease and concomitant conditions: a case-based review

    Cureus

    (2018)
  • A. Rojas-Villarraga et al.

    Introducing polyautoimmunity: secondary autoimmune diseases no longer exist

    Autoimm Dis

    (2012)
  • S. Bibbò et al.

    Chronic autoimmune disorders are increased in coeliac disease: A case-control study

    Medicine (Baltimore)

    (2017)
  • G. Samasca et al.

    Polyautoimmunity—the missing ingredient

    Autoimm Rev

    (June 2018)
  • H.A. Deshmukh et al.

    Evaluation of 19 autoimmune disease-associated loci with rheumatoid arthritis in a Colombian population: evidence for replication and gene-gene interaction

    J Rheumatol

    (2011)
  • A. Demirezer Bolat et al.

    Risk factors for polyautoimmunity among patients with celiac disease: a cross-sectional survey

    Digestion

    (2015)
  • J.M. Anaya et al.

    The autoimmune ecology

    Front Immunol

    (2016)
  • J.M. Anaya et al.

    The autoimmune ecology: an update

    Curr Opin Rheumatol

    (2018 Feb 12)
  • A. Lerner et al.

    Microbes and viruses are bugging the gut in celiac disease. Are they friends or foes?

    Front Microbiol

    (2017)
  • M. Edwards et al.

    Our environment shapes us: the importance of environment and sex differences in regulation of autoantibody production

    Front Immunol

    (2018)
  • P. Zhang et al.

    Genetic and epigenetic influences on the loss of tolerance in autoimmunity

    Cell Mol Immunol

    (2018 Mar 5)
  • H. Wu et al.

    Epigenetics as biomarkers in autoimmune diseases

    Clin Immunol

    (Mar 21, 2018)
  • A.S. Perry et al.

    Epigenetic methodologies for the study of celiac disease

    Methods Mol Biol

    (2015)
  • K.A. Bascuñán-Gamboa et al.

    MicroRNAs: an epigenetic tool to study celiac disease

    Rev Esp Enferm Dig

    (2014)
  • Devaux CA, Raoult D. The microbiological memory, an epigenetic regulator governing the balance between good health and...
  • D. Branski et al.

    Extra intestinal manifestation and associated disorders of celiac disease

  • N. Abbasi et al.

    Extra intestinal manifestations of celiac disease and associated disorders

    Int J Celiac Dis

    (2017)
  • A. Lerner et al.

    Extraintestinal manifestations of CD: common pathways in the gut-remote organs' axes

    Int J Celiac Dis

    (2017)
  • H. Jericho et al.

    Extra-intestinal manifestation of celiac disease in children

    Nutrients

    (2018)
  • Cited by (30)

    • Food and Toxicologic Pathology

      2023, Haschek and Rousseaux's Handbook of Toxicologic Pathology, Volume 3: Environmental Toxicologic Pathology and Major Toxicant Classes
    • SARS-CoV-2 induction and COVID-19 manifestations related to autoimmune gastrointestinal diseases

      2022, Autoimmunity, COVID-19, Post-COVID19 Syndrome and COVID-19 Vaccination
    • Celiac disease in the COVID-19 pandemic

      2021, Journal of Translational Autoimmunity
      Citation Excerpt :

      CD and dermatitis herpetiformis (DH) are parts of gluten-related disorders (GRDs). Both diseases have a range of intestinal and extraintestinal clinical manifestations [2]. The GRDs incidence was at 5% of the population [3].

    • Alpha-enolase involvement in intestinal and extraintestinal manifestations of celiac disease

      2021, Journal of Translational Autoimmunity
      Citation Excerpt :

      Seldom are infantile failure to thrive, abdominal pain, bloating and diarrhea the presenting symptoms [4,5,8]. They are replaced by multiple extraintestinal manifestations [9–12], and the brain is included [13]. CD patients tend to have additional autoimmune diseases and their sera contains multiple autoimmune antibodies, including those that target the brain and the peripheral neuronal components [14].

    View all citing articles on Scopus
    View full text