v
Search
Advanced Search

Publications > Journals > Journal of Translational Gastroenterology > Article Full Text

  • OPEN ACCESS

Gluten is a Proinflammatory Inducer of Autoimmunity

  • Aaron Lerner1,2,* ,
  • Carina Benzvi1 and
  • Aristo Vojdani3
 Author information
Journal of Translational Gastroenterology   2024;2(2):109-124

doi: 10.14218/JTG.2023.00060

Abstract

Gluten has multiple harmful effects that compromise human health, not only in gluten-dependent diseases but also in non-gluten-affected chronic inflammatory conditions. After consumption, the indigestible gluten peptides are modified by luminal microbial transglutaminase or transported through the gut epithelium to interact with the highly populated mucosal immune cells. As a disruptor of gut permeability, gluten peptides compromise tight junction integrity, allowing foreign immunogenic molecules to reach internal compartments. Gliadin peptides are distributed systemically to remote organs, where they encounter endogenous tissue transglutaminase. Following post-translational deamidation or transamidation, the peptides become immunogenic and pro-inflammatory, inducing organ dysfunction and pathology. Cross-reactivity and sequence homology between gluten/gliadin peptides and human epitopes may contribute to molecular mimicry in autoimmunity induction. A gluten-free diet can prevent these phenomena through various mechanisms. As proof of concept, gluten withdrawal alleviates disease activity in chronic inflammatory, metabolic, and autoimmune conditions, and even in neurodegeneration. We recommend combining the gluten-free and Mediterranean diets to leverage the advantages of both. Before recommending gluten withdrawal for non-gluten-dependent conditions, patients should be asked about gut symptomatology and screened for celiac-associated antibodies. The current list of gluten-induced diseases includes celiac disease, dermatitis herpetiformis, gluten ataxia, gluten allergy, and non-celiac gluten sensitivity. In view of gluten being a universal pro-inflammatory molecule, other non-celiac autoinflammatory and neurodegenerative conditions should be investigated for potential gluten avoidance.

Keywords

Pro-inflammatory nutrients, Anti-inflammatory nutrients, gluten, Gliadins, Celiac disease, Autoimmune diseases, Chronic inflammation

Introduction

Inflammation is a vital biological response that regulates interactions between humans and the environment, with nutrition playing a crucial role. Due to the surge in chronic inflammatory diseases,1 and increasing interest in anti-inflammatory dietary therapy,2,3 the exploration of pro-inflammatory nutrients has become a primary focus for clinical and scientific communities.4 In fact, the understanding of immune system-driven chronic inflammation and its associated chronic diseases are still not well-developed. The contribution of dietary constituents to inflammatory, metabolic, autoimmune, cancerous, allergic, and neurodegenerative diseases remains poorly defined. The frequently consumed Western diet is considered pro-inflammatory,5 while vegetarian, non-processed, and traditional foods are recommended as anti-inflammatory.6,7 Since it is impossible to cover all pro-inflammatory nutrients, this review will focus on the role of gluten/gliadin in celiac disease (CD)-induced inflammation, and explore their potential involvement in other non-celiac chronic inflammatory conditions. Gluten is composed of two main proteins: glutenin and gliadin. Gliadins make up about 70% of the protein in gluten and are the molecules responsible for the harmful immune response that results in intestinal injury in CD. Since the gut is the entry point for gluten and a crossroads for multiple nutrients, food additives, microbes, enzymatic digestion, and absorption, various gluten-affected luminal events irradiate peripherally, inducing remote organ, gluten-related, inflammatory damage.8,9 The luminal content impacts the enteric ecosystem. Certain dietary components, like gluten, breach tight junction integrity, resulting in increased intestinal permeability, and induce changes in the composition and diversity of the microbiome towards disease-specific dysbiosis or pathobiosis. Finally, the enhanced local enzymatic capacity for post-translational modification of proteins can turn naïve peptides to lose their tolerance and become auto-immunogenic ones. The present narrative review expands on the multiple gut-originated axes and their relationship to remote organ autoimmune diseases. Brain, joint, bone, endocrine, liver, kidney, heart, lung, and skin autoimmune diseases are connected to the deregulated events in the intestinal luminal compartment, forming the gut-systemic organ axe. Being a universal pro-inflammatory protein, affecting multiple body compartments, organs and tissue and systemically distributed, gluten peptides should be thoroughly investigated for their potential detrimental effects. If substantiated, traditional gluten dependent diseases should be ruled out and gluten-free Mediterranean diets should be recommended.

Gluten-induced inflammation in celiac disease

The incidence of celiac disease is around 1–2% of the global population, representing a chronic, autoimmune, multisystem, inflammatory, and immune-mediated condition.10 The intestine is the primary target organ; however, extra-intestinal organs are affected as well.8,9 Currently, the only accepted and proven nutrient that induces the disease is gluten, a general name for proteins found in many grains, such as wheat, barley, rye, and partially in oats.11 Interestingly, “gluten” stands for “glue” in Latin, named for its adhesive and viscoelastic properties. The autoantigen associated with CD is tissue transglutaminase (tTG),12,13 and several antibodies are used for its serological diagnosis.14–16

Celiac disease fulfills the criteria of a gluten-induced autoimmune inflammatory condition in the following aspects17–19: histologically, there is epithelial and mucosal inflammatory destruction, presented by villous atrophy and intraepithelial lymphocytosis20,21; immunologically, the adaptive22 and innate immune23 systems are activated24,25; there is a surge in pro-inflammatory cytokines,26 where gliadin peptides induce increased levels of IL-15, IFN-γ, IL-6, tumor necrosis factor (TNF)-α, IL-1β, CCL2, CCL3, and many more27; dysbiosis, a feature of many autoimmune diseases (ADs), exists in the celiac gut lumen, and gliadin can directly induce intestinal flora dysbiosis.28–30 Upon successful gluten withdrawal, all the above-mentioned inflammatory features are significantly ameliorated31,32; refractory CD and celiac crisis respond to steroids or immunosuppressive therapy33; and finally, genetic predisposition is essential for the disease’s development.25 It can be summarized that CD is a hallmark condition where genetics, inflammation, autoimmunity, dysbiosis, and environmental gluten intersect.25,34–38 It is well accepted that gluten-containing cereals contribute to chronic inflammatory conditions and various ADs, primarily by inducing gut dysbiosis, enhancing intestinal permeability, and initiating a pro-inflammatory immune response.6,39–42 Interestingly, in addition to gluten, other wheat components, like lectins and enolase, might also act as proinflammatory molecules.43–46

The place of gluten in human nutrition and as a food additive

Wheat is a major crop grown in most countries. Its annual production is 7.34 × 108 tons, cultivated on an area of 2.14 × 106 km2, which sums up to the size of Greenland.47

Wheat consumption surpasses all other crops combined, making it the world’s most favored staple food. The annual increase in gluten usage as a processed food additive over the last four to six decades is estimated at 1.8 ± 0.4%.6 The global vital wheat gluten market, valued at $2 billion in 2019, is projected to reach $2.74 billion by 2027, representing a compound annual growth rate of 4% during the forecast period.48

Discovered in the Fertile Crescent nearly 14,000 years ago,49 wheat-based foods became a staple after domestication. Wheat is a major source of protein, with gluten making up 80% of its total protein content.6,50

Gluten is a protein naturally found in certain prolamins, including wheat, barley, and rye. Although oats are considered gluten-free, they are often cross-contaminated with gluten.11,51 Besides prolamins, gluten is found in numerous non-nutritional products, such as medications, toothpaste, and cosmetics. It acts as a binder, holding substances together and adding a “stretchy” quality. When omitted in baking, the resultant dough tears easily.52

In Western countries, wheat contributes significantly to health, providing dietary fiber, B vitamins, and mineral micronutrients, notably selenium, iron, and zinc.53 There are two perspectives on wheat and gluten: public perception and reality.50,53 Recently, the roles and functions of wheat and gluten have been scrutinized in unproven or pseudoscientific publications and popular media reports, giving the impression that wheat or gluten consumption has a deleterious and addictive effect on human health.50,53 The common claim that gluten-free foods are inherently healthy is not well-supported by scientifically accepted controlled studies. Consequently, wheat/gluten withdrawal fashionistas have impacted multiple Western societies, changing their dietary habits.50,53 While gluten itself provides no essential nutrients and its consumption can be avoided without compromising human well-being.54 However, the only scientifically defined indication to withdraw gluten is in well-proven gluten-dependent conditions, namely, CD, dermatitis herpetiformis, gluten ataxia, and gluten/wheat allergy.31,32,41,53,55–57 Despite the significant interest in non-celiac gluten sensitivity, much remains to be explored about its pathogenesis, and its potential nutritional triggers are still controversial.55,58–61 A combined gluten-free and Mediterranean diet might be an attractive alternative to address the unhealthy aspects of a gluten-free diet (GFD).53,62–64

The side effects of gluten

The more gluten is explored, the more side effects are disclosed. This topic has recently been reviewed by several groups.6,8,9,41,42,65–67 There are various adverse effects of gluten that might impact health. These harmful effects are delivered through immunological and toxic pathways, leading to gut dysfunction or inadequacy.

Pro-inflammatory

As mentioned above, gluten acts as a pro-inflammatory molecule.8,9,25–27,34–39,68,69 The gliadin p31-43 peptide induces cellular stress, activates proliferative mechanisms of cryptic epithelial cells, drives enterocyte stress, induces a Ca2+ surge (thus activating the CD autoantigen tTG), triggers a local pro-inflammatory storm, activates the NFκB signaling pathway, inhibits CFTR (cystic fibrosis transmembrane conductance regulator, an ion channel protein), alters vesicular trafficking, activates the inflammasome platform, and reduces autophagy.68,70,71 Gliadin peptides are pro-oxidative, induce DNA damage, and are pro-apoptotic in in-vitro and ex-vivo studies.72

Alters the gut microbiome and increases intestinal permeability

Gluten decreases the microbiome/dysbiome ratio composition and diversity, suppressing the beneficial metabolome toward inflammation.28–30,73–77 Intestinal tight junction functional integrity is one of the most conserved protective mechanisms for human survival and is crucial for maintaining intestinal homeostasis. When disrupted, foreign molecules enter the epithelial barrier, come into contact with the subepithelial dense immune systems, and initiate chronic inflammation and autoimmunity. Increased intestinal permeability is a common feature of many of these conditions, including CD,6,35–38,41,65–68,74,75 where gluten is a major disruptor of tight junction protective function.6,78–81 Several observations strengthen the gluten-zonulin-increased permeability axis. Zonulin is a measurable blood protein that reflects tight junction functionality. Increased zonulin blood level is considered a marker of an impaired intestinal barrier.6,78–81 This active axis has been shown to operate in active and non-active CD patients and even in normal controls.82 Larazotide acetate (AT-1001), a small peptide derived from Vibrio cholerae toxin, is one of the potential non-nutritional pharmacological strategies to treat CD patients.56 Acting as a modulator of tight junction integrity with its anti-zonulin activity, it has been shown to be superior to placebo in improving gut symptoms in active CD patients.83 The drug shows promise for counteracting enhanced intestinal permeability in some other chronic systemic diseases,84–86 including severe COVID-19,87,88 although it is not yet approved.

Immunogenicity

One of the major unwanted effects of gluten is its immunological impact, closely connected to the inflammatory response. Gluten is an immunogenic protein that elicits anti-gluten/gliadin antibodies, even in non-CD patients and normal controls.89–93 In addition, microbial transglutaminase (mTG)-treated gluten peptides in patients with CD are immunogenic.94 When gliadin is cross-linked to tissue or mTG, transforming the naïve protein into an immunogenic one, CD patients mount substantial levels of neo-epitope tTG and mTG antibodies, respectively.8,9,14–16,41,66,94–99 Intriguingly, in the presence of tTG and mTG-assisted gliadin docking, gluten/gliadin loses its human body tolerance, resulting in corresponding antibody secretion, representing a classical post-translation modification of proteins.29,30,100 When tested in CD patients, the neo-epitope tTG and mTG exhibit higher immunogenic activity compared to gliadin undocked enzymes. Furthermore, the tTG neo-epitope IgA+IgG isotypes show higher optical density activity, better reflect intestinal injury, and expose higher specificity and sensitivity by targeting different autoantigens compared to the conventional tTG isotypes.98,99 The same was found for the mTG neo-epitope.14,16,95 However, the list of gluten’s immunological adverse effects is much more extensive.41

In vitro studies have shown that gluten induces macrophages to produce proinflammatory cytokines and nitric oxide (NO).101–103 Upregulated MHCII, co-stimulatory molecules, TRLs, cytokine, and chemokine production were observed in dendritic cells.104,105 Higher expression of NKG2D and CD71 on NKp46(+) cells has been shown in lymphoid organs.106 Finally, increased permeability and the production of TNFα and IL-1β were detected when gluten was applied to the Caco-2 cell line.107

In vivo studies on rats and mice have shown the following compared to controls: cytokine surge in TH1 intestinal and mesenteric lymph nodes, TH1 cytokine pattern in islet infiltrate, and increased number of intestinal pathogenic intraepithelial cells.108–110 Studies on non-obese diabetic (NOD) mice revealed increased activated intestinal CD4+ T cells, changes in TH1/TH2 intestinal cytokine ratios associated with activated dendritic and TH17 cells, increased natural killer cell cytotoxicity, and cytokine secretion of IFN-γ and IL-6.106,111–113 NKG2D is a proinflammatory, auto-immunogenic co-stimulatory molecule; it is an activating receptor mostly expressed on cells of the cytotoxic arm of the immune system. Gluten withdrawal lowered NKG2D and its ligand expression in NOD and BALB/c mice, attesting to gluten’s impact on the co-stimulatory interplay between tolerance and immune inflammation.114

When exposed to gliadin/gluten, BALB/c mice showed proportional changes in regulatory T-cell subsets, increased numbers of TH17 in peripheral lymph nodes, proinflammatory cytokine patterns in FOX3 and FOXP3+ T cells, and robust activation of innate immune and TH17 cells.115–117Ex vivo and mice studies showed gluten-induced dendritic cells’ production of IL-1β and, interestingly, enhanced neutrophil migration towards gliadin peptides.118,119

Cellular dysfunction and cytotoxicity

At the cellular level, gliadin has been found to drive cytotoxicity, decrease cell viability and differentiation, induce LDH secretion, promote apoptosis, and decrease RNA, DNA, and glycoprotein synthesis when applied to HCT116 cells.41 In 1976, Hudson et al.120 documented growth inhibition and phenotypic changes in various human cell lines induced by gliadin exposure. Several in vitro studies point to the cytotoxicity of this molecule. Gliadin induced agglutination in K562 cells, decreased F-actin content in enteric 407 cell lines, suppressed cell growth and viability, induced apoptosis, and altered redox equilibrium in Caco-2 cells and cell morphology in LoVo, two- and three-dimensional cell culture while causing rearrangement of the cytoskeleton through the zonulin molecular structure. This results in the loss of tight junction functionality in IEC-6 cells.121

Disturbance of oxidative equilibrium

Oxidative equilibrium plays an essential role in cell homeostasis, and its imbalance is involved in many chronic inflammatory diseases. Gliadin-induced oxidative stress was reported extensively on various cell lines, including Caco-2, HT29, SH-SY5Y, T84, and LoVo, and reviewed in depth.121–123 For example, the content of glutathione was reduced (−20% vs. controls), and the activity of related enzymes was inhibited.121 The dysfunctional antioxidant machinery can result in inflamed CD intestinal mucosa, making it more vulnerable to further oxidative stress and hindering mucosal recovery.123

Induce apoptosis

Intestinal homeostasis relies heavily on enterocyte viability and death to maintain the high cellular turnover necessary to cope with a hostile environment. Programmed cell death is pivotal for this equilibrium but can be detrimental in pathological conditions. The apoptotic pathway is over-activated in CD patients and plays a key role in inducing gut inflammation.73,124 Inflammatory response and enteric damage induced by gliadin p31-43 drive multiple programmed cell death pathways in the small intestine of mice.124

Gluten impacts epigenetics

The HLA-DQ2 and HLA-DQ8 haplotypes are widely associated with CD, but some people without these genes still develop the disease. Genetic predisposition can be regulated or affected by epigenetic modifications and cannot account for all reported CD cases. Environmental epigenetics adds substantial understanding to the disease’s evolution and its multi-faceted phenotypic presentations.8,125,126 The main epigenetic pathways include histone modifications, DNA methylation, non-coding RNAs, and RNA methylation, where microRNAs might be used to characterize various classes of CD patients.125 Gluten affects gene expression by changing methylation status.122,127 The impact of gliadin on epigenetics has been observed in CD and non-CD MH-SY4Y and Caco-2 cell lines.122 Wheat-derived peptide epigenetic alterations might be important during the postnatal nutritional transition from maternal breastfeeding or infant formula to complementary gluten consumption.122,127

Gluten affects cellular metabolism

Being pro-inflammatory, cytotoxic, oxidative, apoptotic, and highly immunogenic, gluten peptides can alter fundamental cellular metabolic networks. Wheat-derived peptides induce 50% inhibition in cellular proliferation, 20% suppression of colony-forming ability, and significantly lower alkaline phosphatase activity during Caco-2 cell line differentiation.128 Moreover, the peptic-tryptic digestion of wheat inhibited more than half of DNA and RNA synthesis, glycoprotein synthesis, and altered mitochondrial functions in Caco-2 cells.129,130 Wheat and gluten peptides are important in nutrigenomics and nutrigenetics, revealing various interplays between diet, specific nutritional components, and gene expression.127

Gluten affects mental health

A plethora of peripheral and central neurological manifestations affect the celiac population65,131–135 indicating that gluten consumption can also impact psychiatric behavior and mental health. Anti-neuronal antibodies such as transglutaminase 6, GAD-65, GAD-67, cerebellar peptide, and myelin-associated glycoprotein are part of the CD-associated autoantibodies.136 During intestinal digestion, resulting gluten fragments have strong opioid activity.137 These morphine-like substances, called gluten exorphins, have proven opioid effects that might affect mental health.138 Opioid receptors are scattered throughout the body, including in the gut, brain, and peripheral nervous system. Facing intestinal and blood-brain barrier disruption139,140 caused by microbes, stress, dietary components, pollutants, alcohol, or over-the-counter drugs, gluten-originated exorphins can impact mental functions.6,41,42,65,66,131,137 Cognitive impairment and “brain fog” might be associated with CD,141,142 and responsiveness to gluten withdrawal has been reported.143–145

In a more holistic view, the association between oxidative stress, gene expression, dysbiome and its mobilome, impaired gut and brain permeability146 and gut inflammation associated with gluten-derived peptides, is interrelated and interconnected during the autoimmune cascade evolution in CD.

Gluten peptides are systemically distributed

Prolamins containing gluten are main nutritional staples, and processed food gluten is heavily consumed.6,41,42,65,66,94 Consequently, gluten is widespread in the environment, in the gut lumen, and in contact with the epithelial monolayer and mucosal immune systems. CD is considered a gradually developing, mostly hypo-symptomatic or even asymptomatic chronic enteric inflammatory condition. In reality, it can abruptly erupt as an acute, symptomatic, sometimes life-threatening event involving the gut and extra-intestinal peripheral organs.8,147 Many in vivo/ex vivo or in vitro models involving CD duodenal biopsies, intestinal cell lines, or incidental gluten intake have reported acute effects within 48 h of incubation or ingestion, which were recently summarized. These acute phenotypic, cellular, and laboratory events demonstrate the potential ability of gluten peptides to impact the entire human body.8,9,147,148 A major question is whether gluten/gliadin peptides pass the protective mechanical or immunological intestinal barriers to penetrate inside the body and reach remote compartments and organs. Several observations support the systemic distribution of these peptides, and suggested mechanisms include:

Transepithelial passage of gluten peptides

The discussion on how gliadin peptides pass the gut epithelial monolayer is ongoing, but it is known that both paracellular and transcellular pathways are involved.67 There are three methods of transporting molecules through a cell: endocytosis,67,149 endoplasmic reticulum-assisted transcytosis,150 and secretory IgA-transferrin receptor-assisted translocation of intact gluten peptides below the epithelium.151 Paracellularly, following gliadin digest binding to its CXCR3 receptor, increased zonulin levels compromise tight junction function by activating the EGFR-PAR2-MyD88-mediated signaling pathways, resulting in increased intestinal permeability.152

Most recently, Stricker S. et al. visualized gliadin peptide transport into CD enterocytes using intestinal biopsies and the RACE (Rapid uptake of Antigen into the Cytosol of Enterocytes) cell line.150 The nutrition-originated peptides were transported through the endoplasmic reticulum and deposited below the enterocyte monolayer. This deposition proves that luminal gluten peptides penetrate the epithelial barrier, hence, facing the mucosal and submucosal immune networks.

In fact, gluten-dependent subepithelial deposits involving IgA-tTG are among the hallmark markers for early CD, even in seronegative patients and before histological damage occurs.153 The cohabitation of gluten peptides with these specific IgA-tTG deposits in the subepithelial space reinforces the transepithelial transport of gluten peptides.13,154 Additionally, the immunogenic CD supra-molecule, a 33-residue peptide from alpha-2 gliadin, was directly visualized in gluten-sensitive macaques,155 and gluten-stimulated CD-specific enteric T cells were shown to increase the transepithelial flux of gluten peptides.156 TTG-gluten polymeric complexes are potent antigens for tTG-specific mucosal B cells, supported by diverse subepithelial gluten-specific T cells.157 Finally, gluten peptides can be presented by subepithelial dendritic cells.158 Thus, isolated or complexed gluten/gliadin peptides located in the lamina propria are presented by local antigen-presenting cells, activating the adaptive and innate mucosal networks and inducing CD-specific autoantibodies.

Gluten metabolites are found in human body fluids

Physio-anatomical logic indicates that urinary secretion of a peptide most likely originates from the bloodstream. Recently, Upadhyay D et al. reported that gluten sensitivity expresses itself in a potential CD at the metabolic level before any intestinal damage.159 Decreased levels of histidine, tyrosine, glycine, and tryptophan, and altered levels of another six metabolites were detected in the mucosa or plasma of potential CD patients compared to active CD patients and healthy controls. Intriguingly, raising the topic of gluten addiction and mental health, gluten metabolites, namely, exorphin B4 and B5, are found in normal human blood.160,161 A hypothetical mechanism for gluten masking its own toxicity by these gluten-originated exorphins has been suggested.138 Additionally, multiple gluten-dependent circulating miRNAs that appear before IgA-tTG positivity and are responsive to gluten withdrawal have been characterized.162,163

Urinary gluten metabolites have been extensively reported. Gluten dose escalation, gluten-free diet adherence assessment, urinary gluten intake-dependent miRNAs, urine peptidomics analysis, and urinary metabolic alterations have all been documented.159,163–166

Tissue transglutaminase whole body distribution and biological functions

Before describing gluten peptide distribution in tissues and organs, it is important to remember that tTG is ubiquitous in the human body.13 It is the autoantigen in CD,12 its prime substrate is gluten,94 and the enzyme induces posttranslational modification of gluten,29 making gluten immunogenic in several gluten-dependent conditions.13,66 Cellular-wise, the enzyme spans all intracellular organelles and compartments, including transmembrane areas, and is secreted extracellularly.13,167 Tissue-wise, it is ubiquitously expressed in most human tissues.13,167 Due to its enzymatic biochemical activities, tTG is involved in multiple human biological events and diseases.13,167,168 Since gluten peptides circulate systemically, the chances of tTG encountering them are high. The missing part of the tTG-blood-gluten triangle is the localization of gluten/gliadin metabolites in remote extra-intestinal organs.

Gluten metabolites are found in human organs

Because CD is a multifaceted condition with a plethora of extra-intestinal phenotypes, patients with the disease are at risk of developing remote organ pathologies.8,9,30,41,42,55,65,66 The enzyme tTG can cross-link numerous protein substrates, and the resulting aggregates can be deposited in various organs.169 Below are the main organs where tTG and gluten might orchestrate or be involved in local pathologies:

Gluten deposits in the cerebellum

Gluten ataxia is an autoimmune ataxia and an integral part of gluten-dependent ADs. Brain IgA-tTG2 and tTG6 deposits have been reported in patients, primarily in the cerebellum, pons, and medulla.170,171 Intense perivascular deposition and inflammation might allow circulating gluten peptide and IgA-tTG2/tTG6 antibody entry through a leaky blood-brain barrier, depositing in the central nervous system.172 Indeed, when serum from gluten ataxia patients was injected into the ventricles of mice, ataxia developed within 3 h post-injection.172,173 Moreover, cross-reactive antibodies are shared between gluten peptides and Purkinje cell epitopes, suggesting a potential molecular mimicry pathway to cerebellar autoimmunogenesis.174 Despite these findings, the characterization of gluten-IgA-tTG2/tTG6 deposits in the patient’s cerebellum requires further evaluation.

Gluten peptides impact chronic inflammatory brain conditions

The gut-brain axis was recently described,131,135,175,176 but the role of nutrients in activating these pathways is not clearly defined.6,8,9,29,30,177 Recent papers strengthen the potential relationship between gluten consumption and neurodegenerative, neuroinflammatory, and central ADs in susceptible individuals.65,66,134,178 It appears that gluten peptides contribute to neurodegeneration and chronic brain inflammatory diseases.65,66,178 By specific dysbiosis, enhanced gut permeability, many cross-reactive antibodies with sequence similarity to human brain epitopes, and multiple adverse effects described above, the circulating repertoire of gluten peptides is involved in neurodegeneration.65,66,134 The combined cross-reactivity and sequence similarity suggest molecular mimicry and allude to autoimmune mechanisms resulting in gluten-related brain conditions.

Gluten deposit in the thyroid

Hashimoto’s thyroiditis and CD often overlap, sharing genetic, environmental, symptomatic, immunogenic, pathological, hormonal, and even serological aspects.179 Both entities are integral parts of polyendocrinopathy syndrome.180 There is a continuous debate concerning whether and when to screen patients for CD serology and if there is a place for a gluten-free diet in autoimmune thyroid diseases.179,181–183 The tTG enzyme resides in the thyroid follicles and extracellular matrix.184,185 In CD patients, tTG antibodies bind to these thyroid regions, and their levels correlate with thyroid peroxidase antibody activity.185 The findings suggest that CD-associated antibodies could be involved in thyroid dysfunction, thus reinforcing the gut-thyroid axis. Supporting this, Vojdani et al. applied affinity-purified antibodies made against wheat, alpha-gliadin peptide, and wheat germ agglutinin to various human tissue antigens, finding moderate to strong reactions with thyroid peroxidase and many other autoantigens.44,186

Gluten deposits in the skin

Dermatitis herpetiformis is a dermatological gluten-dependent disease characterized by cutaneous anti-tTG3 IgA deposits, where tTG3c is the autoantigen of the disease.187 Interestingly, similar aggregates can be detected in the skin of CD patients.188 tTG3 is a member of the tTG family and can cross-link with its favorable gluten/gliadin peptides. The skin anti-tTG3 IgA deposits in dermatitis herpetiformis mirror the subepithelial IgA-tTG deposits in CD. The herpetiform dermatological eruption is only one of many gluten-dependent autoimmune manifestations of the skin.189

Gluten deposits in the pancreas

Type 1 diabetes mellitus is highly associated with CD,8,136,180,190 but the role of gluten in inducing insulitis remains controversial.191 Some researchers claim that gluten-containing cereals are associated with an increased risk of pancreatic islet autoimmunity,79,192–194 while others see no such connection.195 Notably, gluten peptides have been shown to localize in the pancreatic islets, enhancing beta-cell hyperactivity, increasing the expression of beta-cell antigens, and resulting in pancreatic autoimmunity.196 Additionally, the posttranslational modifications of human islet antigens induced by local tTG increase the affinity to HLA-DQ, improving presentation to the adjacent pro-inflammatory T cells and initiating the autoimmune cascade.197 Thus, gluten intake can provoke type 1 diabetes.198

Finally, applying a GFD to diabetes-prone animals reduced tTG activity in the pancreatic islets, reduced insulitis, and delayed or reduced diabetes incidence.196 Recently, Hansen et al. substantiated these beneficial effects of gluten withdrawal in NOD mice across three generations by modulating the systemic immune system in a microbiota-independent manner, probably through epigenetic modifications.199 Additionally, a GFD was shown to modulate inflammation in the salivary glands and pancreatic islets in NOD mice.200

The role of the pancreatic gluten-tTG axis in human type 1 diabetes mellitus requires further investigation.

Gluten deposits in the Kidney

The CD is associated with several renal abnormalities,201 with IgA nephropathy, also known as Berger’s disease, being highly affected by gluten.202 It is evident that interactions between tTG and gluten peptides occur in the kidneys of IgA nephropathy patients. Indeed, tTG is overexpressed in the gut of IgA nephropathy mice, and gliadin peptides participate in the disease pathology.203 The immune mechanism of gluten-induced nephropathy involves transferrin receptors, IgA1, gliadin peptides, and soluble CD89.202 As proof of concept, several CD patients on GFDs showed resolution of their autoimmune kidney disease.42,204 The systemic circulation of gluten peptides excreted via urine further strengthens the concept of the enteric gluten-kidney axis, where gluten withdrawal might be beneficial.

Gluten deposits in the liver

The gut-liver axis operates in CD, with several liver conditions associated with hepatic disease, ranging from isolated transaminasemia to autoimmune hepatitis.8,9,30,41,42,205,206 Recently, a causal relationship was demonstrated for hepatic IgA-tTG deposits in CD patients, showing 100% sensitivity and 85% positive predictive value, establishing the association between gluten consumption, liver IgA-tTG aggregates, and liver pathology in CD.207 Not surprisingly, the resolution of liver injury and disappearance of these colocalized deposits were demonstrated with GFD in patients.207

Gluten deposits in the heart

IgG and IgA immune deposition can be detected in the pericardium of individuals with gluten-dependent dermatitis herpetiformis suffering from recurrent pericarditis.208 However, the gut-heart axis presents multiple clinical phenotypes in CD, including atrial fibrillation, dilated cardiomyopathy, pericarditis, myocarditis, angina pectoris, myocardial infarction, and even death from anoxic heart disease.209–211 Some of these manifestations arise from hypercoagulability in CD, which is caused by multifactorial mechanisms involving nutritional deficiencies and autoantibodies.212–214 Alpha-enolase, a glycolytic enzyme expressed in most tissues that plays a role in many cell functions, has been identified as an autoantigen in Hashimoto’s encephalopathy. Recently, alpha-enolase has been suggested to play a role in the cardiac manifestations of CD.46 Regarding cardiac involvement, CD IgA-tTG antibodies have shown strong fluorescence when applied to heart structures.215,216

Gluten and neurodegenerative diseases

The well-established gut-brain axis has been thoroughly reported and reviewed.131,176,217 However, the topic of gluten involvement in neurodegenerative conditions has recently generated scientific and clinical interest.8,65,66,134,217,218 The current hypothesis is that indigestible luminal immunogenic gluten peptides are transported transcellularly and enter paracellularly. After crossing the blood-brain barrier, gluten peptides, cross-linked gluten complexes, gluten-induced antibodies, or gut-originated gluten-restricted CD4 T cells initiate and maintain proinflammatory cytokines, driving neurodegenerative diseases.65,66,131,134,217,218 Most recently, cross-reactive antibodies between gluten and human brain epitopes have been described.44,45,219–222 Specifically, cross-reactive antibodies between tTG, mTG, and amyloid-beta 1-42 have been identified,223 potentially contributing to intraneuronal deposition of A-beta-P-42 in Alzheimer’s disease Similarly, cross-reactivity between tTG, mTG, wheat proteins, and alpha-synuclein has been reported,220 reinforcing the role of gluten-tTG-mTG interactions in both Alzheimer’s and Parkinson’s diseases.

Both human endogenous and microbial exogenous transglutaminases are heavily involved in CD evolution. The tTG is the autoantigen in CD,12,13 while mTG, a heavily consumed processed food additive, is described as a potential driver in CD66,94,134,224–229 and systemic autoimmunity. Gluten is a prime substrate for both enzymes.66,94 Gliadin-cross-linked complexes formed by these enzymes elicit high antibody levels in untreated CD children,13–16,66,96,98,99 and the corresponding serological markers are very reliable for diagnosing gluten-sensitive enteropathy.14–16,97,99 Finally, sequence similarity between gluten and brain epitopes was recently detected for Parkinson’s disease and other neurodegenerative conditions.65,220 Both mechanisms—cross-reactivity and sequence similarity between gluten peptides—contribute to molecular mimicry, which may result in neurodegenerative, neuroinflammatory, and neuropsychiatric conditions.

A GFD might be beneficial in many non-celiac autoimmune diseases

If gluten is a proinflammatory and auto-immunogenic nutrient and is the offending toxic inflammatory molecule in gluten-dependent ADs, a major question arises considering its beneficial curative effects when withdrawn: Might a GFD be helpful for patients affected by non-celiac ADs? This topic was recently reviewed and summarized.41,42,55,230 In a recent systematic review summarizing 83 publications, we found that 911/1,408 AD-affected patients showed improvement on a GFD. Abstaining from gluten intake was found to be efficient in 80% of the publications and clinically beneficial to 65% of the patients.42 The following ADs were screened: rheumatoid arthritis, antiphospholipid syndrome, dermatomyositis, undifferentiated connective tissue disease, Raynaud’s phenomenon, spondylarthritis, psoriasis, vitiligo, pemphigus, erythema elevatum diutinum, inflammatory bowel disease, Crohn’s disease, ulcerative colitis, autoimmune hepatitis, primary biliary cholangitis, primary sclerosing cholangitis, autoimmune pancreatitis, autoimmune enteropathy, thyroiditis, Graves’ disease, Hashimoto’s disease, type 1 diabetes, Addison’s disease, autoimmune hypopituitarism, multiple sclerosis, myasthenia gravis, autoimmune myocarditis, autoimmune pericarditis, IgA nephropathy, uveitis, idiopathic thrombocytopenic purpura, and idiopathic dilated cardiomyopathy.42 We concluded that a GFD might be beneficial for some patients affected by ADs. We suggested screening autoimmune patients for CD-associated antibodies, and only those who test positive should consider gluten withdrawal.42 Overall, there is insufficient evidence to support a GFD for all AD patients, and official guidelines for patient selection have not yet been issued.42,55,230,231 The topic is still controversial, and some studies oppose gluten withdrawal in non-celiac ADs.232–234

Logically, a GFD might counteract the harmful effects of gluten consumption. Before detailing the potential pathways and mechanisms by which gluten withdrawal might alleviate the clinical phenotype, evolution, and behavior of ADs, the following is a summary of gluten’s side effects.

A challenging puzzle is the pathophysiological pathways and mechanisms by which gluten peptides induce inflammatory pathologies in various organs. Clarifying these mechanisms will improve our understanding of the beneficial effects of a GFD. The following summary (Table 1) is based on past and recent publications.6,8,9,14–16,25–30,34–39,41,42,55,65-68,70–82,89–119,121–130,134,224-230,235 It should be stressed that most studies were done ex vivo or on animal models. Substantiation of all of them in vivo, on humans, is highly encouraged.

Table 1

Harmful effects and pathogenic mechanisms of gluten peptide-induced inflammation and cellular damage

Detrimental effects and mechanisms of gluten peptide-induced pathologyReferences
Pro-inflammatory8,9,2527,3439,42,68,235
Drive: cytotoxicity, apoptosis, LDH; Suppress: cell viability, differentiation, RNA, DNA and glycoprotein synthesis41,121,128130
Cellular stress induction42,68,70,71
Induce zonulin production78,79,121
Pro-oxidative42,72,121123
Epigenetics impact8,122,125127
Pro-apoptotic73,124
Impact nutrigenomics, nutrigenetics, gene expression127
Induce dysbiosis2830,7377
Increase macrophage’s proinflammatory cytokine101103
Increase intestinal permeability6,7882,121
Enhance NO production102
Immunogenic and induced antibodies8993
Upregulate MHCII, co-stimulatory molecules, TRLs, cytokine and chemokine production104,105
Cross-linked to mTG immunogenicity8,9,1416,29,30,41,66,94,96100
Stimulate TH1 cytokine profile108110
Enhance NO production102
Increase intraepithelial lymphocyte and intestinal damage110
Upregulate MHCII, co-stimulatory molecules, TLRs, cytokine and chemokine production104,105
Activated intestinal CD4+ T cells, dendritic and TH17 cells, natural killer cell cytotoxicity106,111113
Increase expression of NKG2D and CD71106,114
overproduction of IL-17112,115117
Induce TNFα and IL-1β107
Increase IL-1β, activate NLRP3 inflammasome118
Enhance neutrophil migration119

Potential mechanisms and pathways of GFD effectiveness in ADs

When gluten does not enter the body, all the positive and negative effects are eliminated or avoided. Several mechanisms can prevent the inflammatory or autoimmune phenomena triggered by gut-originated gluten peptides:

  • Gluten withdrawal will eliminate or attenuate the harmful inflammatory, immunogenic, oxidative, stressogenic, dysbiotic, metabolic, and cellular consequences described in Table 1.

  • Short-chain fatty acids (SCFA) are the main microbial fermentation products in the human gut. These molecules are essential for intestinal homeostasis and the proper functioning of many protective systems in our body.76,236 Untreated CD patients have disturbed microbiome diversity and composition, and their stool SCFA levels are reduced.76 After one year of abstaining from gluten, their microbiome and stool SCFA content normalized.76,237

  • Tryptophan and its metabolites regulate immune functions, are essential for enteric homeostasis, and are pivotal for serotonin-dependent human behavior. Depressed and anxious CD patients have lower free tryptophan concentrations. Applying a GFD alleviates depression and anxiety.238,239 However, a recent Iranian study did not confirm this observation.240 Notably, mood alterations, depression, phobia, and anxiety are prevalent in many ADs,241,242 and this mechanism should be studied in non-gluten-dependent ADs.

  • IgA-deficient patients are prone to ADs243 and mainly to CD.244 Mucosal IgA is a major immune protective mechanism, and its local production is encouraged by a high-fiber diet, local SCFA content, and a healthy microbiome. Increased production of IgA, a major mucosal and luminal immune barrier, is induced by a high-fiber diet, SCFAs, and the microbiome.245 A GFD sustains a physiological microbiome, increases luminal SCFA, and combined with a high-fiber diet, could enhance enteric IgA levels.

  • GFD improves macrobiotic composition and diversity compared to untreated CD patients. The resulting higher production of SCFA, acetate, and butyrate lowers the luminal and stool pH,76,246 improving colonic ecology by acting as anti-inflammatory and anti-cancer compounds.76,247,248

  • Avoiding gluten and adopting the Mediterranean diet benefit CD patients’ health.62,63,249 The recently adopted combination of the GFD-MedD pyramid avoids the harmful effects of gluten while adding the numerous benefits of the Mediterranean diet.62,250,251 MedD represents a mental and physical health-protective menu that can easily be rendered gluten-free.62,63,249251 The combined diet offers higher antioxidants, anti-inflammatory nutrients, and sufficient fiber content for the CD population.64

  • Gluten/gliadin, being primary substrates for tTG,94 are cross-linked or deamidated by post-translational modification, losing their tolerance and becoming immune- and auto-immunogenic.29,30,100 In the absence of gluten, no such reactions occur, and the body avoids gluten-dependent inflammation and tissue/organ pathologies.8,9,41,42,55,65,66

  • Cross-reactive antibodies between gluten/wheat and human tissue epitopes might induce ADs or organ pathology by molecular mimicry.44,45,220223 When no gluten peptides circulate, no cross-reactive antibodies are produced, preventing molecular mimicry.41,42,55,65,66

  • Sequence similarity between gluten and human tissue antigens has been reported recently.65,219,220 The shared homology and cross-reactivity between gluten peptides and human epitopes reinforce the molecular mimicry pathway toward inflammation and end-organ dysfunction. A GFD prevents these phenomena by curtailing shared sequences and cross-reactivity. Intriguingly, cross-reactivity and sequence similarity have recently been reported between various human antigens and a family member of tTG, namely, microbial transglutaminase.252

  • Leaky gut is reported in many, in vivo and ex vivo, metabolic, inflammatory, and ADs.235,253255 Gluten is a major disruptor of enteric tight junction functional integrity.41,42,65,66,235,254,255 Gluten avoidance might protect the body from this abnormality. However, the enigma of “Gluten: yes, no, maybe” is far from being resolved.256

  • The pathogenic mechanisms by which gluten\/gliadin peptides induce inflammation in remote organs, and the extended potential harmful effects in non-celiac ADs are described in Figure 1

Pathogenic mechanisms by which gluten/gliadin peptides induce inflammation in remote organs, and the extended potential harmful effects in non-celiac ADs.
Fig. 1  Pathogenic mechanisms by which gluten/gliadin peptides induce inflammation in remote organs, and the extended potential harmful effects in non-celiac ADs.

(a) Gluten is ingested and digested, reaching the gut lumen as gliadin peptides; (b) Gliadins are a prime substrate for deamidation and cross-linking by luminal transglutaminases. These post-translationally modified proteins (PTMP) increase their immunogenicity. Luminal digestive enzymes cannot further break down these protein complexes, leading to an inflammatory cascade that results in mucus disruption, dysbiosis, intestinal epithelial damage, and leaky gut; (c) Gliadin peptides and transglutaminases can potentially infiltrate through open junctions or trans-enterocytically into the lamina propria, exposing the highly immunoreactive sub-epithelium to foreign antigens or complexes; (d) In the lamina propria, dendritic cells (DCs) encounter gliadin-transglutaminase cross-linked complexes and migrate to lymph nodes as antigen-presenting cells to activate T cells. Secretion of IFNγ, IL-17, and IL-22 by Th1 and Th17 cells activates B cells, which secrete anti-tTG, anti-neo-tTG, and anti-endomysial autoantibodies (EMA); (e) Mucosal immune cells, immunogenic modified peptides, proinflammatory cytokines, autoantibodies, and small particles that escape the immune system enter the blood vessels. They can eventually reach remote organs and trigger an autoimmune response; (f) Some examples of inflammatory conditions that can be affected by the presence of gliadin peptides and transglutaminases or cross-linked complexes in peripheral organs. AD, Alzheimer’s Disease; AIH, Autoimmune Hepatitis; AIM, Autoimmune Myocarditis; AIP, Autoimmune Pericarditis; AT, Autoimmune Thyroiditis; CD, Celiac disease; DM, Dermatomyositis; GA, Gluten Ataxia; IBD, Inflammatory Bowel Diseases; IEL, intraepithelial lymphocytes.; IgA-Neph, IgA nephropathy (Berger’s disease); MS, Multiple Sclerosis; mTG, microbial transglutaminase; PA, Psoriatic Arthritis; PD, Parkinson’s Disease; RA, Rheumatoid Arthritis; T1D, Type 1 Diabetes; tTG, tissue transglutaminase.

Conclusion

Gluten has many side effects that compromise human health, not only in gluten-dependent conditions but also in non-gluten-related chronic diseases. After entering the gut lumen, undigestible gluten peptides are modified by luminal mTG or transported through the enteric epithelium to meet mucosal immune cells or distributed systemically to remote organs where they encounter tTG. The modified peptides become immunogenic and pro-inflammatory, inducing organ dysfunction and pathology. A GFD can prevent these phenomena by multiple mechanisms: suppressing gluten-associated detrimental effects, improving the microbiome/dysbiome ratio, avoiding post-translational modification of gluten peptides, preventing cross-reactivity and sequence similarity between gluten and human epitopes, and reducing gut leakage.

As proof of concept, gluten withdrawal alleviates disease activity in multiple chronic inflammatory, metabolic, autoimmune conditions, and even neurodegeneration. However, caution is needed. GFD consumers should be aware of the disadvantages of a gluten-restricted diet. It is advised to combine a GFD with the Mediterranean diet to harness the advantages of both. Before recommending a GFD for non-gluten-dependent conditions, patients should be assessed for gut symptomatology and screened for celiac-associated antibodies. Notably, this topic is still under discussion and is not included in the guidelines of professional decision-making societies.

It is hoped that this narrative review will encourage the scientific, nutritional, and medical communities to further explore the mechanisms by which gluten peptides induce inflammation and end-organ damage. Understanding these pathways will clarify gluten’s role in the induction of human chronic inflammatory diseases.

Declarations

Acknowledgement

None.

Funding

None.

Conflict of interest

AV is the CEO of Immunosciences Lab., Inc. The authors have no other conflict of interests to declare.

Authors’ contributions

Screening the literature (AL, CB), designing and writing the manuscript (AL), editing and revising the manuscript, designing the figure with BioRender.com permission (CB), writing part of the manuscript, revising the manuscript, and summarizing the cross-reactivity results (AV). The three authors agreed to the published version of the manuscript.

References

  1. Furman D, Campisi J, Verdin E, Carrera-Bastos P, Targ S, Franceschi C, et al. Chronic inflammation in the etiology of disease across the life span. Nat Med 2019;25(12):1822-1832 View Article PubMed/NCBI
  2. Sears B. Anti-inflammatory Diets. J Am Coll Nutr 2015;34(Suppl 1):14-21 View Article PubMed/NCBI
  3. Haß U, Herpich C, Norman K. Anti-Inflammatory Diets and Fatigue. Nutrients 2019;11(10):2315 View Article PubMed/NCBI
  4. Gill PA, Inniss S, Kumagai T, Rahman FZ, Smith AM. The Role of Diet and Gut Microbiota in Regulating Gastrointestinal and Inflammatory Disease. Front Immunol 2022;13:866059 View Article PubMed/NCBI
  5. Malesza IJ, Malesza M, Walkowiak J, Mussin N, Walkowiak D, Aringazina R, et al. High-Fat, Western-Style Diet, Systemic Inflammation, and Gut Microbiota: A Narrative Review. Cells 2021;10(11):3164 View Article PubMed/NCBI
  6. Lerner A, Matthias T. Changes in intestinal tight junction permeability associated with industrial food additives explain the rising incidence of autoimmune disease. Autoimmun Rev 2015;14(6):479-489 View Article PubMed/NCBI
  7. Liu W, Chen X, Li H, Zhang J, An J, Liu X. Anti-Inflammatory Function of Plant-Derived Bioactive Peptides: A Review. Foods 2022;11(15):2361 View Article PubMed/NCBI
  8. Aaron L, Torsten M, Patricia W. Autoimmunity in celiac disease: Extra-intestinal manifestations. Autoimmun Rev 2019;18(3):241-246 View Article PubMed/NCBI
  9. Lerner A, Matthias T. GUT-the Trojan Horse in remote organs’ Autoimmunity. J Clin Cell Immunol 2016;7:1-10 View Article
  10. Catassi C, Verdu EF, Bai JC, Lionetti E. Coeliac disease. Lancet 2022;399(10344):2413-2426 View Article PubMed/NCBI
  11. Lerner A. The Enigma of Oats in Nutritional Therapy for Celiac Disease. Int J Celiac Dis 2014;2:110-114 View Article
  12. Reif S, Lerner A. Tissue transglutaminase—the key player in celiac disease: a review. Autoimmun Rev 2004;3(1):40-45 View Article PubMed/NCBI
  13. Lerner A, Neidhöfer S, Matthias T. Transglutaminase 2 and anti transglutaminase 2 autoantibodies in celiac disease and beyond: TG2 double-edged sword: Gut and extraintestinal involvement. Immunome Res 2015;11:1-4 View Article
  14. Lerner A, Ramesh A, Matthias T. Serologic Diagnosis of Celiac Disease: New Biomarkers. Gastroenterol Clin North Am 2019;48(2):307-317 View Article PubMed/NCBI
  15. Lerner A, Jeremias P, Neidhofer S, Matthias T. Comparison of the Reliability of 17 Celiac Disease Associated Bio-Markers to Reflect Intestinal Damage. J Clin Cell Immunol 2017;8:486 View Article
  16. Agardh D, Matthias T, Wusterhausen P, Neidhöfer S, Heller A, Lerner A. Antibodies against neo-epitope of microbial and human transglutaminase complexes as biomarkers of childhood celiac disease. Clin Exp Immunol 2020;199(3):294-302 View Article PubMed/NCBI
  17. Lerner A, Blank M, Shoenfeld Y. Celiac disease and autoimmunity. Isr J Med Sci 1996;32:33-36
  18. Sollid LM, Jabri B. Is celiac disease an autoimmune disorder?. Curr Opin Immunol 2005;17(6):595-600 View Article PubMed/NCBI
  19. Troncone R, Discepolo V. Celiac disease and autoimmunity. J Pediatr Gastroenterol Nutr 2014;59(Suppl 1):S9-S11 View Article PubMed/NCBI
  20. Villanacci V, Vanoli A, Leoncini G, Arpa G, Salviato T, Bonetti LR, et al. Celiac disease: histology-differential diagnosis-complications. A practical approach. Pathologica 2020;112(3):186-196 View Article PubMed/NCBI
  21. Lerner A, Matthias T. Intraepithelial lymphocyte normal cut-off level in celiac disease: The debate continues. Int J Celiac Dis 2016;4:4-6 View Article
  22. Lindeman I, Sollid LM. Single-cell approaches to dissect adaptive immune responses involved in autoimmunity: the case of celiac disease. Mucosal Immunol 2022;15(1):51-63 View Article PubMed/NCBI
  23. Kim SM, Mayassi T, Jabri B. Innate immunity: actuating the gears of celiac disease pathogenesis. Best Pract Res Clin Gastroenterol 2015;29(3):425-435 View Article PubMed/NCBI
  24. Anderson RP. Innate and adaptive immunity in celiac disease. Curr Opin Gastroenterol 2020;36(6):470-478 View Article PubMed/NCBI
  25. Voisine J, Abadie V. Interplay Between Gluten, HLA, Innate and Adaptive Immunity Orchestrates the Development of Coeliac Disease. Front Immunol 2021;12:674313 View Article PubMed/NCBI
  26. Vincentini O, Maialetti F, Gonnelli E, Silano M. Gliadin-dependent cytokine production in a bidimensional cellular model of celiac intestinal mucosa. Clin Exp Med 2015;15(4):447-454 View Article PubMed/NCBI
  27. Garrote JA, Gómez-González E, Bernardo D, Arranz E, Chirdo F. Celiac disease pathogenesis: the proinflammatory cytokine network. J Pediatr Gastroenterol Nutr 2008;47(Suppl 1):S27-S32 View Article PubMed/NCBI
  28. Wu X, Qian L, Liu K, Wu J, Shan Z. Gastrointestinal microbiome and gluten in celiac disease. Ann Med 2021;53(1):1797-1805 View Article PubMed/NCBI
  29. Lerner A, Aminov R, Matthias T. Dysbiosis May Trigger Autoimmune Diseases via Inappropriate Post-Translational Modification of Host Proteins. Front Microbiol 2016;7:84 View Article PubMed/NCBI
  30. Lerner A, Aminov R, Matthias T. Transglutaminases in Dysbiosis As Potential Environmental Drivers of Autoimmunity. Front Microbiol 2017;8:66 View Article PubMed/NCBI
  31. Lerner A, Matthias T. The Yin and Yang of dietary gluten transgressions in real-life scenarios of celiac patients. BMC Med 2020;18(1):70 View Article PubMed/NCBI
  32. Samasca G, Lerner A, Girbovan A, Sur G, Lupan I, Makovicky P, et al. Challenges in gluten-free diet in coeliac disease: Prague consensus. Eur J Clin Invest 2017;47(5):394-397 View Article PubMed/NCBI
  33. Hujoel IA, Murray JA. Refractory Celiac Disease. Curr Gastroenterol Rep 2020;22(4):18 View Article PubMed/NCBI
  34. Patel N, Robert ME. Frontiers in Celiac Disease: Where Autoimmunity and Environment Meet. Am J Surg Pathol 2022;46(1):e43-e54 View Article PubMed/NCBI
  35. Mayassi T, Ladell K, Gudjonson H, McLaren JE, Shaw DG, Tran MT, et al. Chronic Inflammation Permanently Reshapes Tissue-Resident Immunity in Celiac Disease. Cell 2019;176(5):967-981.e19 View Article PubMed/NCBI
  36. Auricchio R, Calabrese I, Galatola M, Cielo D, Carbone F, Mancuso M, et al. Gluten consumption and inflammation affect the development of celiac disease in at-risk children. Sci Rep 2022;12(1):5396 View Article PubMed/NCBI
  37. Levescot A, Malamut G, Cerf-Bensussan N. Immunopathogenesis and environmental triggers in coeliac disease. Gut 2022;71(11):2337-2349 View Article PubMed/NCBI
  38. Porpora M, Conte M, Lania G, Bellomo C, Rapacciuolo L, Chirdo FG, et al. Inflammation Is Present, Persistent and More Sensitive to Proinflammatory Triggers in Celiac Disease Enterocytes. Int J Mol Sci 2022;23(4):1973 View Article PubMed/NCBI
  39. de Punder K, Pruimboom L. The dietary intake of wheat and other cereal grains and their role in inflammation. Nutrients 2013;5(3):771-787 View Article PubMed/NCBI
  40. Zingone F. Grain Intake and Human Health. Nutrients 2020;12(12):3733 View Article PubMed/NCBI
  41. Lerner A, Shoenfeld Y, Matthias T. Adverse effects of gluten ingestion and advantages of gluten withdrawal in nonceliac autoimmune disease. Nutr Rev 2017;75(12):1046-1058 View Article PubMed/NCBI
  42. Lerner A, Freire de Carvalho J, Kotrova A, Shoenfeld Y. Gluten-free diet can ameliorate the symptoms of non-celiac autoimmune diseases. Nutr Rev 2022;80(3):525-543 View Article PubMed/NCBI
  43. Garutti M, Nevola G, Mazzeo R, Cucciniello L, Totaro F, Bertuzzi CA, et al. The Impact of Cereal Grain Composition on the Health and Disease Outcomes. Front Nutr 2022;9:888974 View Article PubMed/NCBI
  44. Vojdani A. Reaction of food-specific antibodies with different tissue antigens. Int J Food Sci Technol 2020;55:1800-1815 View Article
  45. Vojdani A, Gushgari LR, Vojdani E. Interaction between food antigens and the immune system: Association with autoimmune disorders. Autoimmun Rev 2020;19(3):102459 View Article PubMed/NCBI
  46. Lerner A, Sobolevskaia P, Churilov L, Shoenfeld Y. Alpha-enolase involvement in intestinal and extraintestinal manifestations of celiac disease. J Transl Autoimmun 2021;4:100109 View Article PubMed/NCBI
  47. FAOSTAT Food and Agriculture Organization of the United Nations. Available from: http://www.fao.org/faostat/en/#data. Accessed October 13, 2022. 2020
  48. https://www.globenewswire.com/news-release/2020/06/30/2055191/0/en/Global-Vital-Wheat-Gluten-Industry-Assessment-2018-2020-2024-2027.html
  49. Harari YN. Sapiens: A Brief History of Humankind. Oxford: Signal; 2017
  50. Wieser H, Koehler P, Scherf KA. The Two Faces of Wheat. Front Nutr 2020;7:517313 View Article PubMed/NCBI
  51. Vojdani A, Tarash I. Cross-Reaction between Gliadin and Different Food and Tissue Antigens. Food Nutr Sci 2013;04:20-32 View Article
  52. Shewry P. What Is Gluten-Why Is It Special?. Front Nutr 2019;6:101 View Article PubMed/NCBI
  53. Lerner A, O’Bryan T, Matthias T. Navigating the Gluten-Free Boom: The Dark Side of Gluten Free Diet. Front Pediatr 2019;7:414 View Article PubMed/NCBI
  54. Shmerling RH. Harvard Health Publishing. Harvard Medical School. Ditch the Gluten, Improve Your Health? April 14 ,2022. https://www.health.harvard.edu/staying-healthy/ditch-the-gluten-improve-your-health (accessed October 13, 2022)
  55. Lerner A, Ramesh A, Matthias T. Going gluten free in non-celiac autoimmune diseases: the missing ingredient. Expert Rev Clin Immunol 2018;14(11):873-875 View Article PubMed/NCBI
  56. Lerner A. New therapeutic strategies for celiac disease. Autoimmun Rev 2010;9(3):144-147 View Article PubMed/NCBI
  57. Lerner A, Matthias T. Gluten-free diet tough alley in torrid time. Int J Celiac Dis 2017;5:50-5 View Article
  58. Sergi C, Villanacci V, Carroccio A. Non-celiac wheat sensitivity: rationality and irrationality of a gluten-free diet in individuals affected with non-celiac disease: a review. BMC Gastroenterol 2021;21(1):5 View Article PubMed/NCBI
  59. Siddiqui UN, Pervaiz A, Khan ZB, Sultana T. Diagnostic Dilemma, Possible Non-celiac Gluten Sensitivity: Consideration in Approach and Management. Cureus 2022;14(5):e25302 View Article PubMed/NCBI
  60. Reese I, Schäfer C, Kleine-Tebbe J, Ahrens B, Bachmann O, Ballmer-Weber B, et al. Non-celiac gluten/wheat sensitivity (NCGS)-a currently undefined disorder without validated diagnostic criteria and of unknown prevalence: Position statement of the task force on food allergy of the German Society of Allergology and Clinical Immunology (DGAKI). Allergo J Int 2018;27(5):147-151 View Article PubMed/NCBI
  61. Mumolo MG, Rettura F, Melissari S, Costa F, Ricchiuti A, Ceccarelli L, et al. Is Gluten the Only Culprit for Non-Celiac Gluten/Wheat Sensitivity?. Nutrients 2020;12(12):3785 View Article PubMed/NCBI
  62. Bascuñán KA, Elli L, Vecchi M, Scricciolo A, Mascaretti F, Parisi M, et al. Mediterranean Gluten-Free Diet: Is It a Fair Bet for the Treatment of Gluten-Related Disorders?. Front Nutr 2020;7:583981 View Article PubMed/NCBI
  63. Nestares T, Martín-Masot R, de Teresa C, Bonillo R, Maldonado J, Flor-Alemany M, et al. Influence of Mediterranean Diet Adherence and Physical Activity on Bone Health in Celiac Children on a Gluten-Free Diet. Nutrients 2021;13(5):1636 View Article PubMed/NCBI
  64. Cenni S, Sesenna V, Boiardi G, Casertano M, Di Nardo G, Esposito S, et al. The Mediterranean Diet in Paediatric Gastrointestinal Disorders. Nutrients 2022;15(1):79 View Article PubMed/NCBI
  65. Lerner A, Benzvi C. “Let Food Be Thy Medicine”: Gluten and Potential Role in Neurodegeneration. Cells 2021;10(4):756 View Article PubMed/NCBI
  66. Lerner A, Matthias T. Gluten and Autoimmunogenesis. In Mosaic of Autoimmunity: The Novel Factors of Autoimmune Diseases Revisited. 2nd ed. Academic Press; 2019 View Article
  67. Lammers KM, Herrera MG, Dodero VI. Translational Chemistry Meets Gluten-Related Disorders. ChemistryOpen 2018;7(3):217-232 View Article PubMed/NCBI
  68. Chirdo FG, Auricchio S, Troncone R, Barone MV. The gliadin p31-43 peptide: Inducer of multiple proinflammatory effects. Int Rev Cell Mol Biol 2021;358:165-205 View Article PubMed/NCBI
  69. Barone MV, Auricchio R, Nanayakkara M, Greco L, Troncone R, Auricchio S. Pivotal Role of Inflammation in Celiac Disease. Int J Mol Sci 2022;23(13):7177 View Article PubMed/NCBI
  70. Goel G, Tye-Din JA, Qiao SW, Russell AK, Mayassi T, Ciszewski C, et al. Cytokine release and gastrointestinal symptoms after gluten challenge in celiac disease. Sci Adv 2019;5(8):eaaw7756 View Article PubMed/NCBI
  71. Conte M, Nigro F, Porpora M, Bellomo C, Furone F, Budelli AL, et al. Gliadin Peptide P31-43 Induces mTOR/NFkβ Activation and Reduces Autophagy: The Role of Lactobacillus paracasei CBA L74 Postbiotc. Int J Mol Sci 2022;23(7):3655 View Article PubMed/NCBI
  72. Monguzzi E, Marabini L, Elli L, Vaira V, Ferrero S, Ferretti F, et al. Gliadin effect on the oxidative balance and DNA damage: An in-vitro, ex-vivo study. Dig Liver Dis 2019;51(1):47-54 View Article PubMed/NCBI
  73. Perez F, Ruera CN, Miculan E, Carasi P, Chirdo FG. Programmed Cell Death in the Small Intestine: Implications for the Pathogenesis of Celiac Disease. Int J Mol Sci 2021;22(14):7426 View Article PubMed/NCBI
  74. Chander AM, Yadav H, Jain S, Bhadada SK, Dhawan DK. Cross-Talk Between Gluten, Intestinal Microbiota and Intestinal Mucosa in Celiac Disease: Recent Advances and Basis of Autoimmunity. Front Microbiol 2018;9:2597 View Article PubMed/NCBI
  75. Ren Z, Pan LL, Huang Y, Chen H, Liu Y, Liu H, et al. Gut microbiota-CRAMP axis shapes intestinal barrier function and immune responses in dietary gluten-induced enteropathy. EMBO Mol Med 2021;13(8):e14059 View Article PubMed/NCBI
  76. Lerner A, Patricia J, Matthias T. Nutrients, Bugs and Us: The Short-chain Fatty Acids Story in Celiac Disease. Int J Celiac Dis 2016;4:92-4 View Article
  77. Lerner A, Arleevskaya M, Schmiedl A, Matthias T. Microbes and Viruses Are Bugging the Gut in Celiac Disease. Are They Friends or Foes?. Front Microbiol 2017;8:1392 View Article PubMed/NCBI
  78. Guerreiro CS, Calado Â, Sousa J, Fonseca JE. Diet, Microbiota, and Gut Permeability-The Unknown Triad in Rheumatoid Arthritis. Front Med (Lausanne) 2018;5:349 View Article PubMed/NCBI
  79. Wood Heickman LK, DeBoer MD, Fasano A. Zonulin as a potential putative biomarker of risk for shared type 1 diabetes and celiac disease autoimmunity. Diabetes Metab Res Rev 2020;36(5):e3309 View Article PubMed/NCBI
  80. An J, Liu Y, Wang Y, Fan R, Hu X, Zhang F, et al. The Role of Intestinal Mucosal Barrier in Autoimmune Disease: A Potential Target. Front Immunol 2022;13:871713 View Article PubMed/NCBI
  81. Jauregi-Miguel A. The tight junction and the epithelial barrier in coeliac disease. Int Rev Cell Mol Biol 2021;358:105-132 View Article PubMed/NCBI
  82. Drago S, El Asmar R, Di Pierro M, Grazia Clemente M, Tripathi A, Sapone A, et al. Gliadin, zonulin and gut permeability: Effects on celiac and non-celiac intestinal mucosa and intestinal cell lines. Scand J Gastroenterol 2006;41(4):408-419 View Article PubMed/NCBI
  83. Hoilat GJ, Altowairqi AK, Ayas MF, Alhaddab NT, Alnujaidi RA, Alharbi HA, et al. Larazotide acetate for treatment of celiac disease: A systematic review and meta-analysis of randomized controlled trials. Clin Res Hepatol Gastroenterol 2022;46(1):101782 View Article PubMed/NCBI
  84. Matei DE, Menon M, Alber DG, Smith AM, Nedjat-Shokouhi B, Fasano A, et al. Intestinal barrier dysfunction plays an integral role in arthritis pathology and can be targeted to ameliorate disease. Med 2021;2(7):864-883.e9 View Article PubMed/NCBI
  85. Yonker LM, Gilboa T, Ogata AF, Senussi Y, Lazarovits R, Boribong BP, et al. Multisystem inflammatory syndrome in children is driven by zonulin-dependent loss of gut mucosal barrier. J Clin Invest 2021;131(14):149633 View Article PubMed/NCBI
  86. Slifer ZM, Krishnan BR, Madan J, Blikslager AT. Larazotide acetate: a pharmacological peptide approach to tight junction regulation. Am J Physiol Gastrointest Liver Physiol 2021;320(6):G983-G989 View Article PubMed/NCBI
  87. Di Micco S, Musella S, Sala M, Scala MC, Andrei G, Snoeck R, et al. Peptide Derivatives of the Zonulin Inhibitor Larazotide (AT1001) as Potential Anti SARS-CoV-2: Molecular Modelling, Synthesis and Bioactivity Evaluation. Int J Mol Sci 2021;22(17):9427 View Article PubMed/NCBI
  88. Ailioaie LM, Ailioaie C, Litscher G, Chiran DA. Celiac Disease and Targeting the Molecular Mechanisms of Autoimmunity in COVID Pandemic. Int J Mol Sci 2022;23(14):7719 View Article PubMed/NCBI
  89. Gillett HR, Freeman HJ. Serological testing in screening for adult celiac disease. Can J Gastroenterol 1999;13(3):265-269 View Article PubMed/NCBI
  90. Tucker NT, Barghuthy FS, Prihoda TJ, Kumar V, Lerner A, Lebenthal E. Antigliadin antibodies detected by enzyme-linked immunosorbent assay as a marker of childhood celiac disease. J Pediatr 1988;113(2):286-289 View Article PubMed/NCBI
  91. Lerner A, Kumar V, Iancu TC. Immunological diagnosis of childhood coeliac disease: comparison between antigliadin, antireticulin and antiendomysial antibodies. Clin Exp Immunol 1994;95(1):78-82 View Article PubMed/NCBI
  92. Lerner A, Lebenthal E. The controversy of the use of anti-gluten antibody (AGA) as a diagnostic tool in celiac disease. J Pediatr Gastroenterol Nutr 1991;12(4):407-409 View Article PubMed/NCBI
  93. Vojdani A. Detection of IgE, IgG, IgA and IgM antibodies against raw and processed food antigens. Nutr Metab (Lond) 2009;6:22 View Article PubMed/NCBI
  94. Lerner A, Matthias T. Possible association between celiac disease and bacterial transglutaminase in food processing: a hypothesis. Nutr Rev 2015;73(8):544-552 View Article PubMed/NCBI
  95. Matthias T, Jeremias P, Neidhöfer S, Lerner A. The industrial food additive, microbial transglutaminase, mimics tissue transglutaminase and is immunogenic in celiac disease patients. Autoimmun Rev 2016;15(12):1111-1119 View Article PubMed/NCBI
  96. Lerner A. More novel diagnostic antibodies for celiac disease. Expert Rev Gastroenterol Hepatol 2016;10(7):767-768 View Article PubMed/NCBI
  97. Lerner A, Matthias T. Food Industrial Microbial Transglutaminase in Celiac Disease: Treat or Trick. Int J Celiac Dis 2015;3:1-6 View Article
  98. Matthias T, Neidhöfer S, Pfeiffer S, Prager K, Reuter S, Gershwin ME. Novel trends in celiac disease. Cell Mol Immunol 2011;8(2):121-125 View Article PubMed/NCBI
  99. Lerner A, Jeremias P, Neidhöfer S, Matthias T. Antibodies against neo-epitope tTg complexed to gliadin are different and more reliable then anti-tTg for the diagnosis of pediatric celiac disease. J Immunol Methods 2016;429:15-20 View Article PubMed/NCBI
  100. Lerner A, Matthias T. Rheumatoid arthritis-celiac disease relationship: joints get that gut feeling. Autoimmun Rev 2015;14(11):1038-1047 View Article PubMed/NCBI
  101. Thomas KE, Sapone A, Fasano A, Vogel SN. Gliadin stimulation of murine macrophage inflammatory gene expression and intestinal permeability are MyD88-dependent: role of the innate immune response in Celiac disease. J Immunol 2006;176(4):2512-2521 View Article PubMed/NCBI
  102. Tucková L, Flegelová Z, Tlaskalová-Hogenová H, Zídek Z. Activation of macrophages by food antigens: enhancing effect of gluten on nitric oxide and cytokine production. J Leukoc Biol 2000;67(3):312-318 View Article PubMed/NCBI
  103. Tucková L, Novotná J, Novák P, Flegelová Z, Kveton T, Jelínková L, et al. Activation of macrophages by gliadin fragments: isolation and characterization of active peptide. J Leukoc Biol 2002;71(4):625-631 PubMed/NCBI
  104. Palová-Jelínková L, Rozková D, Pecharová B, Bártová J, Sedivá A, Tlaskalová-Hogenová H, et al. Gliadin fragments induce phenotypic and functional maturation of human dendritic cells. J Immunol 2005;175(10):7038-7045 View Article PubMed/NCBI
  105. Ciccocioppo R, Rossi M, Pesce I, Ricci G, Millimaggi D, Maurano F, et al. Effects of gliadin stimulation on bone marrow-derived dendritic cells from HLA-DQ8 transgenic MICE. Dig Liver Dis 2008;40(12):927-935 View Article PubMed/NCBI
  106. Larsen J, Dall M, Antvorskov JC, Weile C, Engkilde K, Josefsen K, et al. Dietary gluten increases natural killer cell cytotoxicity and cytokine secretion. Eur J Immunol 2014;44(10):3056-3067 View Article PubMed/NCBI
  107. Gujral N, Suh JW, Sunwoo HH. Effect of anti-gliadin IgY antibody on epithelial intestinal integrity and inflammatory response induced by gliadin. BMC Immunol 2015;16:41 View Article PubMed/NCBI
  108. Scott FW, Rowsell P, Wang GS, Burghardt K, Kolb H, Flohé S. Oral exposure to diabetes-promoting food or immunomodulators in neonates alters gut cytokines and diabetes. Diabetes 2002;51(1):73-78 View Article PubMed/NCBI
  109. Scott FW, Cloutier HE, Kleemann R, Wöerz-Pagenstert U, Rowsell P, Modler HW, et al. Potential mechanisms by which certain foods promote or inhibit the development of spontaneous diabetes in BB rats: dose, timing, early effect on islet area, and switch in infiltrate from Th1 to Th2 cells. Diabetes 1997;46(4):589-598 View Article PubMed/NCBI
  110. Stĕpánková R, Tlaskalová-Hogenová H, Sinkora J, Jodl J, Fric P. Changes in jejunal mucosa after long-term feeding of germfree rats with gluten. Scand J Gastroenterol 1996;31(6):551-557 View Article PubMed/NCBI
  111. Flohé SB, Wasmuth HE, Kerad JB, Beales PE, Pozzilli P, Elliott RB, et al. A wheat-based, diabetes-promoting diet induces a Th1-type cytokine bias in the gut of NOD mice. Cytokine 2003;21(3):149-154 View Article PubMed/NCBI
  112. Alam C, Valkonen S, Palagani V, Jalava J, Eerola E, Hänninen A. Inflammatory tendencies and overproduction of IL-17 in the colon of young NOD mice are counteracted with diet change. Diabetes 2010;59(9):2237-2246 View Article PubMed/NCBI
  113. Larsen J, Weile C, Antvorskov JC, Engkilde K, Nielsen SM, Josefsen K, et al. Effect of dietary gluten on dendritic cells and innate immune subsets in BALB/c and NOD mice. PLoS One 2015;10(3):e0118618 View Article PubMed/NCBI
  114. Adlercreutz EH, Weile C, Larsen J, Engkilde K, Agardh D, Buschard K, et al. A gluten-free diet lowers NKG2D and ligand expression in BALB/c and non-obese diabetic (NOD) mice. Clin Exp Immunol 2014;177(2):391-403 View Article PubMed/NCBI
  115. Antvorskov JC, Fundova P, Buschard K, Funda DP. Impact of dietary gluten on regulatory T cells and Th17 cells in BALB/c mice. PLoS One 2012;7(3):e33315 View Article PubMed/NCBI
  116. Antvorskov JC, Fundova P, Buschard K, Funda DP. Dietary gluten alters the balance of pro-inflammatory and anti-inflammatory cytokines in T cells of BALB/c mice. Immunology 2013;138(1):23-33 View Article PubMed/NCBI
  117. Bernardo D, Garrote JA, Fernández-Salazar L, Riestra S, Arranz E. Is gliadin really safe for non-coeliac individuals? Production of interleukin 15 in biopsy culture from non-coeliac individuals challenged with gliadin peptides. Gut 2007;56(6):889-890 View Article PubMed/NCBI
  118. Palová-Jelínková L, Dáňová K, Drašarová H, Dvořák M, Funda DP, Fundová P, et al. Pepsin digest of wheat gliadin fraction increases production of IL-1β via TLR4/MyD88/TRIF/MAPK/NF-κB signaling pathway and an NLRP3 inflammasome activation. PLoS One 2013;8(4):e62426 View Article PubMed/NCBI
  119. Lammers KM, Chieppa M, Liu L, Liu S, Omatsu T, Janka-Junttila M, et al. Gliadin Induces Neutrophil Migration via Engagement of the Formyl Peptide Receptor, FPR1. PLoS One 2015;10(9):e0138338 View Article PubMed/NCBI
  120. Hudson DA, Cornell HJ, Purdham DR, Rolles CJ. Non-specific cytotoxicity of wheat gliadin components towards cultured human cells. Lancet 1976;1(7955):339-341 View Article PubMed/NCBI
  121. Dolfini E, Elli L, Roncoroni L, Costa B, Colleoni MP, Lorusso V, et al. Damaging effects of gliadin on three-dimensional cell culture model. World J Gastroenterol 2005;11(38):5973-5977 View Article PubMed/NCBI
  122. Trivedi MS, Shah JS, Al-Mughairy S, Hodgson NW, Simms B, Trooskens GA, et al. Food-derived opioid peptides inhibit cysteine uptake with redox and epigenetic consequences. J Nutr Biochem 2014;25(10):1011-1018 View Article PubMed/NCBI
  123. Kaplan M, Ates I, Yüksel M, Ozin YO, Akpinar MY, Topcuoglu C, et al. The Role of Oxidative Stress in the Etiopathogenesis of Gluten-sensitive Enteropathy Disease. J Med Biochem 2017;36(3):243-250 View Article PubMed/NCBI
  124. Ruera CN, Miculán E, Pérez F, Ducca G, Carasi P, Chirdo FG. Sterile inflammation drives multiple programmed cell death pathways in the gut. J Leukoc Biol 2021;109(1):211-221 View Article PubMed/NCBI
  125. Gnodi E, Meneveri R, Barisani D. Celiac disease: From genetics to epigenetics. World J Gastroenterol 2022;28(4):449-463 View Article PubMed/NCBI
  126. Olazagoitia-Garmendia A, Sebastian-delaCruz M, Castellanos-Rubio A. Involvement of lncRNAs in celiac disease pathogenesis. Int Rev Cell Mol Biol 2021;358:241-264 View Article PubMed/NCBI
  127. Ferretti G, Bacchetti T, Masciangelo S, Saturni L. Celiac disease, inflammation and oxidative damage: a nutrigenetic approach. Nutrients 2012;4(4):243-257 View Article PubMed/NCBI
  128. Giovannini C, Maiuri L, De Vincenzi M. Cytotoxic effect of prolamin-derived peptides on in vitro cultures of cell line Caco-2: Implications for coeliac disease. Toxicol In Vitro 1995;9(3):251-255 View Article PubMed/NCBI
  129. Giovannini C, Mancini E, De Vincenzi M. Inhibition of the cellular metabolism of Caco-2 cells by prolamin peptides from cereals toxic for coeliacs. Toxicol In Vitro 1996;10(5):533-538 View Article PubMed/NCBI
  130. Orlando A, Chimienti G, Pesce V, Fracasso F, Lezza AMS, Russo F. An In Vitro Study on Mitochondrial Compensatory Response Induced by Gliadin Peptides in Caco-2 Cells. Int J Mol Sci 2019;20(8):1862 View Article PubMed/NCBI
  131. Lerner A, Neidhöfer S, Matthias T. The Gut Microbiome Feelings of the Brain: A Perspective for Non-Microbiologists. Microorganisms 2017;5(4):66 View Article PubMed/NCBI
  132. Zelnik N, Pacht A, Obeid R, Lerner A. Range of neurologic disorders in patients with celiac disease. Pediatrics 2004;113(6):1672-1676 View Article PubMed/NCBI
  133. Lerner A, Makhoul BF, Eliakim R. Neurological Manifestations of Celiac Disease in Children and Adults Affiliations Celiac disease and environment View project Neurological Manifestations of Celiac Disease in Children and Adults. Eur Neurol J 2012;4:15-20
  134. Lerner A, Matthias T. Don’t forget the exogenous microbial transglutaminases: it is immunogenic and potentially pathogenic. AIMS Biophys 2016;3:546-552 View Article
  135. Patel SC, Shreya D, Zamora DI, Patel GS, Grossmann I, Rodriguez K, et al. Celiac Disease, Beyond the Bowel: A Review of Its Neurological Manifestations. Cureus 2021;13(12):e20112 View Article
  136. Shaoul R, Lerner A. Associated autoantibodies in celiac disease. Autoimmun Rev 2007;6(8):559-565 View Article PubMed/NCBI
  137. Bressan P, Kramer P. Bread and Other Edible Agents of Mental Disease. Front Hum Neurosci 2016;10:130 View Article PubMed/NCBI
  138. Pruimboom L, de Punder K. The opioid effects of gluten exorphins: asymptomatic celiac disease. J Health Popul Nutr 2015;33:24 View Article PubMed/NCBI
  139. Singh S, Sharma P, Pal N, Kumawat M, Shubham S, Sarma DK, et al. Impact of Environmental Pollutants on Gut Microbiome and Mental Health via the Gut-Brain Axis. Microorganisms 2022;10(7):1457 View Article PubMed/NCBI
  140. Tran VTA, Lee LP, Cho H. Neuroinflammation in neurodegeneration via microbial infections. Front Immunol 2022;13:907804 View Article PubMed/NCBI
  141. Yelland GW. Gluten-induced cognitive impairment (“brain fog”) in coeliac disease. J Gastroenterol Hepatol 2017;32(Suppl 1):90-93 View Article PubMed/NCBI
  142. Edwards George JB, Aideyan B, Yates K, Voorhees KN, O’Flynn J, Sweet K, et al. Gluten-induced Neurocognitive Impairment: Results of a Nationwide Study. J Clin Gastroenterol 2022;56(7):584-591 View Article PubMed/NCBI
  143. Makhlouf S, Messelmani M, Zaouali J, Mrissa R. Cognitive impairment in celiac disease and non-celiac gluten sensitivity: review of literature on the main cognitive impairments, the imaging and the effect of gluten free diet. Acta Neurol Belg 2018;118(1):21-27 View Article PubMed/NCBI
  144. Lichtwark IT, Newnham ED, Robinson SR, Shepherd SJ, Hosking P, Gibson PR, et al. Cognitive impairment in coeliac disease improves on a gluten-free diet and correlates with histological and serological indices of disease severity. Aliment Pharmacol Ther 2014;40(2):160-170 View Article PubMed/NCBI
  145. Kristensen VA, Valeur J, Brackmann S, Jahnsen J, Brunborg C, Tveito K. Attention deficit and hyperactivity disorder symptoms respond to gluten-free diet in patients with coeliac disease. Scand J Gastroenterol 2019;54(5):571-576 View Article PubMed/NCBI
  146. Obrenovich MEM. Leaky Gut, Leaky Brain?. Microorganisms 2018;6(4):107 View Article PubMed/NCBI
  147. Lerner A, Matthias T. A Silent or Hypo-symptomatic Disease Can Erupt: Acute Presentations of Celiac Disease. Int J Celiac Dis 2017;5:129-132 View Article
  148. Guarino M, Gambuti E, Alfano F, Strada A, Ciccocioppo R, Lungaro L, et al. Life-threatening onset of coeliac disease: a case report and literature review. BMJ Open Gastroenterol 2020;7(1):e000406 View Article PubMed/NCBI
  149. Reinke Y, Zimmer KP, Naim HY. Toxic peptides in Frazer’s fraction interact with the actin cytoskeleton and affect the targeting and function of intestinal proteins. Exp Cell Res 2009;315(19):3442-3452 View Article PubMed/NCBI
  150. Stricker S, de Laffolie J, Rudloff S, Komorowski L, Zimmer KP. Intracellular Localization of Microbial Transglutaminase and Its Influence on the Transport of Gliadin in Enterocytes. J Pediatr Gastroenterol Nutr 2019;68(3):e43-e50 View Article PubMed/NCBI
  151. Heyman M, Abed J, Lebreton C, Cerf-Bensussan N. Intestinal permeability in coeliac disease: insight into mechanisms and relevance to pathogenesis. Gut 2012;61(9):1355-1364 View Article PubMed/NCBI
  152. Fasano A. All disease begins in the (leaky) gut: role of zonulin-mediated gut permeability in the pathogenesis of some chronic inflammatory diseases. F1000Res 2020;9(F1000 Faculty Rev):69 View Article PubMed/NCBI
  153. Salmi TT, Collin P, Korponay-Szabó IR, Laurila K, Partanen J, Huhtala H, et al. Endomysial antibody-negative coeliac disease: clinical characteristics and intestinal autoantibody deposits. Gut 2006;55(12):1746-1753 View Article PubMed/NCBI
  154. Lindfors K, Mäki M, Kaukinen K. Transglutaminase 2-targeted autoantibodies in celiac disease: Pathogenetic players in addition to diagnostic tools?. Autoimmun Rev 2010;9(11):744-749 View Article PubMed/NCBI
  155. Mazumdar K, Alvarez X, Borda JT, Dufour J, Martin E, Bethune MT, et al. Visualization of transepithelial passage of the immunogenic 33-residue peptide from alpha-2 gliadin in gluten-sensitive macaques. PLoS One 2010;5(4):e10228 View Article PubMed/NCBI
  156. Bethune MT, Siegel M, Howles-Banerji S, Khosla C. Interferon-gamma released by gluten-stimulated celiac disease-specific intestinal T cells enhances the transepithelial flux of gluten peptides. J Pharmacol Exp Ther 2009;329(2):657-668 View Article PubMed/NCBI
  157. Lindstad CB, Dewan AE, Stamnaes J, Sollid LM, du Pré MF. TG2-gluten complexes as antigens for gluten-specific and transglutaminase-2 specific B cells in celiac disease. PLoS One 2021;16(11):e0259082 View Article PubMed/NCBI
  158. Discepolo V, Lania G, Ten Eikelder MLG, Nanayakkara M, Sepe L, Tufano R, et al. Pediatric Celiac Disease Patients Show Alterations of Dendritic Cell Shape and Actin Rearrangement. Int J Mol Sci 2021;22(5):2708 View Article PubMed/NCBI
  159. Upadhyay D, Das P, Dattagupta S, Makharia GK, Jagannathan NR, Sharma U. NMR based metabolic profiling of patients with potential celiac disease elucidating early biochemical changes of gluten-sensitivity: A pilot study. Clin Chim Acta 2022;531:291-301 View Article PubMed/NCBI
  160. Pennington CL, Dufresne CP, Fanciulli G, Wood TD. Detection of Gluten Exorphin B4 and B5 in Human Blood by Liquid Chromatography-Mass Spectrometry/Mass Spectrometry. Open Spectrosc J 2007;1:9-16 View Article
  161. Fanciulli G, Pennington CL, Dufresne CP, Wood TD. Gluten exorphins in human blood. Pharmacol Res 2020;160:105084 View Article PubMed/NCBI
  162. Tan IL, Coutinho de Almeida R, Modderman R, Stachurska A, Dekens J, Barisani D, et al. Circulating miRNAs as Potential Biomarkers for Celiac Disease Development. Front Immunol 2021;12:734763 View Article PubMed/NCBI
  163. Paolini A, Sarshar M, Felli C, Bruno SP, Rostami-Nejad M, Ferretti F, et al. Biomarkers to Monitor Adherence to Gluten-Free Diet by Celiac Disease Patients: Gluten Immunogenic Peptides and Urinary miRNAs. Foods 2022;11(10):1380 View Article PubMed/NCBI
  164. Burger JPW, van Lochem EG, Roovers EA, Drenth JPH, Wahab PJ. Dose-Escalating (50-500 mg) Gluten Administration Leads to Detectable Gluten-Immunogenic-Peptides in Urine of Patients with Coeliac Disease Which Is Unrelated to Symptoms, a Placebo Controlled Trial. Nutrients 2022;14(9):1771 View Article PubMed/NCBI
  165. Monachesi C, Verma AK, Catassi GN, Franceschini E, Gatti S, Gesuita R, et al. Determination of Urinary Gluten Immunogenic Peptides to Assess Adherence to the Gluten-Free Diet: A Randomized, Double-Blind, Controlled Study. Clin Transl Gastroenterol 2021;12(10):e00411 View Article PubMed/NCBI
  166. Palanski BA, Weng N, Zhang L, Hilmer AJ, Fall LA, Swaminathan K, et al. An efficient urine peptidomics workflow identifies chemically defined dietary gluten peptides from patients with celiac disease. Nat Commun 2022;13(1):888 View Article PubMed/NCBI
  167. Piacentini M, D’Eletto M, Farrace MG, Rodolfo C, Del Nonno F, Ippolito G, et al. Characterization of distinct sub-cellular location of transglutaminase type II: changes in intracellular distribution in physiological and pathological states. Cell Tissue Res 2014;358(3):793-805 View Article PubMed/NCBI
  168. Tatsukawa H, Hitomi K. Role of Transglutaminase 2 in Cell Death, Survival, and Fibrosis. Cells 2021;10(7):1842 View Article PubMed/NCBI
  169. Esposito C, Paparo F, Caputo I, Rossi M, Maglio M, Sblattero D, et al. Anti-tissue transglutaminase antibodies from coeliac patients inhibit transglutaminase activity both in vitro and in situ. Gut 2002;51(2):177-181 View Article PubMed/NCBI
  170. Hadjivassiliou M, Mäki M, Sanders DS, Williamson CA, Grünewald RA, Woodroofe NM, et al. Autoantibody targeting of brain and intestinal transglutaminase in gluten ataxia. Neurology 2006;66(3):373-377 View Article PubMed/NCBI
  171. Hadjivassiliou M, Aeschlimann P, Sanders DS, Mäki M, Kaukinen K, Grünewald RA, et al. Transglutaminase 6 antibodies in the diagnosis of gluten ataxia. Neurology 2013;80(19):1740-1745 View Article PubMed/NCBI
  172. Mitoma H, Adhikari K, Aeschlimann D, Chattopadhyay P, Hadjivassiliou M, Hampe CS, et al. Consensus Paper: Neuroimmune Mechanisms of Cerebellar Ataxias. Cerebellum 2016;15(2):213-232 View Article PubMed/NCBI
  173. Boscolo S, Lorenzon A, Sblattero D, Florian F, Stebel M, Marzari R, et al. Anti transglutaminase antibodies cause ataxia in mice. PLoS One 2010;5(3):e9698 View Article PubMed/NCBI
  174. Hadjivassiliou M, Williamson CA, Woodroofe N. The immunology of gluten sensitivity: beyond the gut. Trends Immunol 2004;25(11):578-582 View Article PubMed/NCBI
  175. Giuffrè M, Gazzin S, Zoratti C, Llido JP, Lanza G, Tiribelli C, et al. Celiac Disease and Neurological Manifestations: From Gluten to Neuroinflammation. Int J Mol Sci 2022;23(24):15564 View Article PubMed/NCBI
  176. Bashir Y, Khan AU. The interplay between the gut-brain axis and the microbiome: A perspective on psychiatric and neurodegenerative disorders. Front Neurosci 2022;16:1030694 View Article PubMed/NCBI
  177. Lerner A, Matthias T, Aminov R. Potential Effects of Horizontal Gene Exchange in the Human Gut. Front Immunol 2017;8:1630 View Article PubMed/NCBI
  178. Philip A, White ND. Gluten, Inflammation, and Neurodegeneration. Am J Lifestyle Med 2022;16(1):32-35 View Article PubMed/NCBI
  179. Lerner A, Jeremias P, Matthias T. Gut-thyroid axis and celiac disease. Endocr Connect 2017;6(4):R52-R58 View Article PubMed/NCBI
  180. Samasca G, Ajay R, Sur D, Aldea C, Sur L, Floca E, et al. Polyautoimmunity - The missing ingredient. Autoimmun Rev 2018;17(8):840-841 View Article PubMed/NCBI
  181. Lerner A, Matthias T. Autoimmune Thyroid Diseases in Celiac Disease: If and When to Screen?. Int J Celiac Dis 2016;4(4):124-126 View Article
  182. Liontiris MI, Mazokopakis EE. A concise review of Hashimoto thyroiditis (HT) and the importance of iodine, selenium, vitamin D and gluten on the autoimmunity and dietary management of HT patients.Points that need more investigation. Hell J Nucl Med 2017;20(1):51-56 View Article PubMed/NCBI
  183. Malandrini S, Trimboli P, Guzzaloni G, Virili C, Lucchini B. What about TSH and Anti-Thyroid Antibodies in Patients with Autoimmune Thyroiditis and Celiac Disease Using a Gluten-Free Diet? A Systematic Review. Nutrients 2022;14(8):1681 View Article PubMed/NCBI
  184. Duntas LH. Does celiac disease trigger autoimmune thyroiditis?. Nat Rev Endocrinol 2009;5(4):190-191 View Article PubMed/NCBI
  185. Naiyer AJ, Shah J, Hernandez L, Kim SY, Ciaccio EJ, Cheng J, et al. Tissue transglutaminase antibodies in individuals with celiac disease bind to thyroid follicles and extracellular matrix and may contribute to thyroid dysfunction. Thyroid 2008;18(11):1171-1178 View Article PubMed/NCBI
  186. Vojdani A, Afar D, Vojdani E. Reaction of Lectin-Specific Antibody with Human Tissue: Possible Contributions to Autoimmunity. J Immunol Res 2020;2020:1438957 View Article PubMed/NCBI
  187. Kemppainen E, Salmi T, Lindfors K. Missing Insight Into T and B Cell Responses in Dermatitis Herpetiformis. Front Immunol 2021;12:657280 View Article PubMed/NCBI
  188. Cannistraci C, Lesnoni La Parola I, Cardinali G, Bolasco G, Aspite N, Stigliano V, et al. Co-localization of IgA and TG3 on healthy skin of coeliac patients. J Eur Acad Dermatol Venereol 2007;21(4):509-514 View Article PubMed/NCBI
  189. Malkovics T, Koszorú K, Kárpáti S, Arató A, Görög A, Sárdy M. The many-faced gluten sensitivity: Gluten-induced autoimmunity from dermatological point of view. Orv Hetil 2021;162(28):1107-1118 View Article PubMed/NCBI
  190. Goodwin G. Type 1 Diabetes Mellitus and Celiac Disease: Distinct Autoimmune Disorders That Share Common Pathogenic Mechanisms. Horm Res Paediatr 2019;92(5):285-292 View Article PubMed/NCBI
  191. Hamilton-Williams EE, Lorca GL, Norris JM, Dunne JL. A Triple Threat? The Role of Diet, Nutrition, and the Microbiota in T1D Pathogenesis. Front Nutr 2021;8:600756 View Article PubMed/NCBI
  192. Hakola L, Miettinen ME, Syrjälä E, Åkerlund M, Takkinen HM, Korhonen TE, et al. Association of Cereal, Gluten, and Dietary Fiber Intake With Islet Autoimmunity and Type 1 Diabetes. JAMA Pediatr 2019;173(10):953-960 View Article PubMed/NCBI
  193. Al Theyab A, Almutairi T, Al-Suwaidi AM, Bendriss G, McVeigh C, Chaari A. Epigenetic Effects of Gut Metabolites: Exploring the Path of Dietary Prevention of Type 1 Diabetes. Front Nutr 2020;7:563605 View Article PubMed/NCBI
  194. Lund-Blix NA, Tapia G, Mårild K, Brantsaeter AL, Njølstad PR, Joner G, et al. Maternal and child gluten intake and association with type 1 diabetes: The Norwegian Mother and Child Cohort Study. PLoS Med 2020;17(3):e1003032 View Article PubMed/NCBI
  195. Meijer CR, Discepolo V, Troncone R, Mearin ML. Does infant feeding modulate the manifestation of celiac disease and type 1 diabetes?. Curr Opin Clin Nutr Metab Care 2017;20(3):222-226 View Article PubMed/NCBI
  196. Haupt-Jorgensen M, Holm LJ, Josefsen K, Buschard K. Possible Prevention of Diabetes with a Gluten-Free Diet. Nutrients 2018;10(11):1746 View Article PubMed/NCBI
  197. van Lummel M, Duinkerken G, van Veelen PA, de Ru A, Cordfunke R, Zaldumbide A, et al. Posttranslational modification of HLA-DQ binding islet autoantigens in type 1 diabetes. Diabetes 2014;63(1):237-247 View Article PubMed/NCBI
  198. Füchtenbusch M, Ziegler AG, Hummel M. Elimination of dietary gluten and development of type 1 diabetes in high risk subjects. Rev Diabet Stud 2004;1(1):39-41 View Article PubMed/NCBI
  199. Hansen CHF, Larsen CS, Zachariassen LF, Mentzel CMJ, Laigaard A, Krych L, et al. Gluten-free diet reduces autoimmune diabetes mellitus in mice across multiple generations in a microbiota-independent manner. J Autoimmun 2022;127:102795 View Article PubMed/NCBI
  200. Haupt-Jorgensen M, Groule V, Reibel J, Buschard K, Pedersen AML. Gluten-free diet modulates inflammation in salivary glands and pancreatic islets. Oral Dis 2022;28(3):639-647 View Article PubMed/NCBI
  201. Boonpheng B, Cheungpasitporn W, Wijarnpreecha K. Renal disease in patients with celiac disease. Minerva Med 2018;109(2):126-140 View Article PubMed/NCBI
  202. Lerner A, Berthelot L, Jeremias P, Matthias T, Abbad L, Monteiro RC. Gluten, Transglutaminase, Celiac Disease and IgA Nephropathy. J Clin Cell Immunol 2017;8:2 View Article
  203. Abbad L, Monteiro RC, Berthelot L. Food antigens and Transglutaminase 2 in IgA nephropathy: Molecular links between gut and kidney. Mol Immunol 2020;121:1-6 View Article PubMed/NCBI
  204. Habura I, Fiedorowicz K, Woźniak A, Idasiak-Piechocka I, Kosikowski P, Oko A. IgA nephropathy associated with coeliac disease. Cent Eur J Immunol 2019;44(1):106-108 View Article PubMed/NCBI
  205. Aggarwal M, Garg R, Kumar P, Lindenmeyer CC, Wakim-Fleming J, Jansson-Knodell C, et al. Bi-directional Relationship Between Celiac Disease and Liver Chemistries: A Systematic Review and Meta-Analysis. Dig Dis Sci 2023;68(4):1369-1380 View Article PubMed/NCBI
  206. Rubio-Tapia A, Murray JA. The Liver and Celiac Disease. Clin Liver Dis 2019;23(2):167-176 View Article PubMed/NCBI
  207. Dutta R, Iqbal A, Das P, Palanichamy JK, Singh A, Mehtab W, et al. Liver involvement in patients with coeliac disease: proof of causality using IgA/anti-TG2 colocalisation techniques. J Clin Pathol 2021;74(12):766-773 View Article PubMed/NCBI
  208. Afrasiabi R, Sirop PA, Albini SM, Rosenbaum HM, Piscatelli RL. Recurrent pericarditis and dermatitis herpetiformis. Evidence for immune complex deposition in the pericardium. Chest 1990;97(4):1006-1007 View Article PubMed/NCBI
  209. Santoro L, De Matteis G, Fuorlo M, Giupponi B, Martone AM, Landi F, et al. Atherosclerosis and cardiovascular involvement in celiac disease: the role of autoimmunity and inflammation. Eur Rev Med Pharmacol Sci 2017;21:5437-5444 View Article
  210. Lerner A, Matthias T. Celiac Disease: Intestinal, Heart and Skin Interconnections. Undefined 2015;3:28-30 View Article
  211. Lerner A. The Gut Feeling of the Heart: Pathophysiological Pathways in the Gut-heart Axis in Celiac Disease. Int J Celiac Dis 2020;8(4):120-123 View Article
  212. Fousekis FS, Beka ET, Mitselos IV, Milionis H, Christodoulou DK. Thromboembolic complications and cardiovascular events associated with celiac disease. Ir J Med Sci 2021;190(1):133-141 View Article PubMed/NCBI
  213. Lerner A, Blank M. Hypercoagulability in celiac disease—an update. Autoimmun Rev 2014;13(11):1138-1141 View Article PubMed/NCBI
  214. Lerner A, Agmon-Levin N, Shapira Y, Gilburd B, Reuter S, Lavi I, et al. The thrombophilic network of autoantibodies in celiac disease. BMC Med 2013;11:89 View Article PubMed/NCBI
  215. Sategna-Guidetti C, Franco E, Martini S, Bobbio M. Binding by serum IgA antibodies from patients with coeliac disease to monkey heart tissue. Scand J Gastroenterol 2004;39(6):540-543 View Article PubMed/NCBI
  216. Volta U, Ballardini G, Molinaro N, De Franceschi L, Groff P, Bianchi FB. Specific reactivity of fluorescein isothiocyanate-conjugated separated IgG and IgA from celiac disease sera on human tissues. Int J Clin Lab Res 1995;25(2):110-115 View Article PubMed/NCBI
  217. Mayer EA, Nance K, Chen S. The Gut-Brain Axis. Annu Rev Med 2022;73:439-453 View Article PubMed/NCBI
  218. Mohan M, Okeoma CM, Sestak K. Dietary Gluten and Neurodegeneration: A Case for Preclinical Studies. Int J Mol Sci 2020;21(15):5407 View Article PubMed/NCBI
  219. Vojdani A, Vojdani E. The Role of Exposomes in the Pathophysiology of Autoimmune Diseases I: Toxic Chemicals and Food. Pathophysiology 2021;28(4):513-543 View Article PubMed/NCBI
  220. Vojdani A, Lerner A, Vojdani E. Cross-Reactivity and Sequence Homology Between Alpha-Synuclein and Food Products: A Step Further for Parkinson’s Disease Synucleinopathy. Cells 2021;10(5):1111 View Article PubMed/NCBI
  221. Vojdani A, Kharrazian D, Mukherjee PS. The prevalence of antibodies against wheat and milk proteins in blood donors and their contribution to neuroimmune reactivities. Nutrients 2013;6(1):15-36 View Article PubMed/NCBI
  222. Vojdani A, O’Bryan T, Green JA, Mccandless J, Woeller KN, Vojdani E, et al. Immune response to dietary proteins, gliadin and cerebellar peptides in children with autism. Nutr Neurosci 2004;7(3):151-161 View Article PubMed/NCBI
  223. Vojdani A, Vojdani E. Amyloid-Beta 1-42 Cross-Reactive Antibody Prevalent in Human Sera May Contribute to Intraneuronal Deposition of A-Beta-P-42. Int J Alzheimers Dis 2018;2018:1672568 View Article PubMed/NCBI
  224. Torsten M, Aaron L. Microbial Transglutaminase Is Immunogenic and Potentially Pathogenic in Pediatric Celiac Disease. Front Pediatr 2018;6:389 View Article PubMed/NCBI
  225. Aaron L, Torsten M. Microbial transglutaminase: A new potential player in celiac disease. Clin Immunol 2019;199:37-43 View Article PubMed/NCBI
  226. Lerner A, Matthias T. Microbial Transglutaminase is Beneficial to Food Industries but a Caveat to Public Health. Med One 2019;4:e190001 View Article
  227. Lerner A, Matthias T. Processed Food Additive Microbial Transglutaminase and Its Cross-Linked Gliadin Complexes Are Potential Public Health Concerns in Celiac Disease. Int J Mol Sci 2020;21(3):1127 View Article PubMed/NCBI
  228. Lerner A, Matthias T. Microbial transglutaminase should be considered as an environmental inducer of celiac disease. World J Clin Cases 2019;7(22):3912-3914 View Article PubMed/NCBI
  229. Lerner A, Benzvi C. Microbial Transglutaminase Is a Very Frequently Used Food Additive and Is a Potential Inducer of Autoimmune/Neurodegenerative Diseases. Toxics 2021;9(10):233 View Article PubMed/NCBI
  230. Lerner A, Ramesh A, Matthias T. Are Non-Celiac Autoimmune Diseases Responsive to Gluten-Free Diet?. Int J Celiac Dis 2017;5:164-167 View Article
  231. Mikulska AA, Karaźniewicz-Łada M, Filipowicz D, Ruchała M, Główka FK. Metabolic Characteristics of Hashimoto’s Thyroiditis Patients and the Role of Microelements and Diet in the Disease Management-An Overview. Int J Mol Sci 2022;23(12):6580 View Article PubMed/NCBI
  232. Passali M, Josefsen K, Frederiksen JL, Antvorskov JC. Current Evidence on the Efficacy of Gluten-Free Diets in Multiple Sclerosis, Psoriasis, Type 1 Diabetes and Autoimmune Thyroid Diseases. Nutrients 2020;12(8):2316 View Article PubMed/NCBI
  233. Thomsen HL, Jessen EB, Passali M, Frederiksen JL. The role of gluten in multiple sclerosis: A systematic review. Mult Scler Relat Disord 2019;27:156-163 View Article PubMed/NCBI
  234. Lidón AC, Patricia ML, Vinesh D, Marta MS. Evaluation of Gluten Exclusion for the Improvement of Rheumatoid Arthritis in Adults. Nutrients 2022;14(24):5396 View Article PubMed/NCBI
  235. Truzzi F, Tibaldi C, Whittaker A, Dilloo S, Spisni E, Dinelli G. Pro-Inflammatory Effect of Gliadins and Glutenins Extracted from Different Wheat Cultivars on an In Vitro 3D Intestinal Epithelium Model. Int J Mol Sci 2020;22(1):172 View Article PubMed/NCBI
  236. Cai Y, Folkerts J, Folkerts G, Maurer M, Braber S. Microbiota-dependent and -independent effects of dietary fibre on human health. Br J Pharmacol 2020;177(6):1363-1381 View Article PubMed/NCBI
  237. Tjellström B, Högberg L, Stenhammar L, Fälth-Magnusson K, Magnusson KE, Norin E, et al. Faecal short-chain fatty acid pattern in childhood coeliac disease is normalised after more than one year’s gluten-free diet. Microb Ecol Health Dis 2013;24(1):20905 View Article PubMed/NCBI
  238. Pynnönen PA, Isometsä ET, Verkasalo MA, Kähkönen SA, Sipilä I, Savilahti E, et al. Gluten-free diet may alleviate depressive and behavioural symptoms in adolescents with coeliac disease: a prospective follow-up case-series study. BMC Psychiatry 2005;5:14 View Article PubMed/NCBI
  239. Addolorato G, Capristo E, Ghittoni G, Valeri C, Mascianà R, Ancona C, et al. Anxiety but not depression decreases in coeliac patients after one-year gluten-free diet: a longitudinal study. Scand J Gastroenterol 2001;36(5):502-506 View Article PubMed/NCBI
  240. Rostami-Nejad M, Taraghikhah N, Ciacci C, Pourhoseingholi MA, Barzegar F, Rezaei-Tavirani M, et al. Anxiety Symptoms in Adult Celiac Patients and the Effect of a Gluten-Free Diet: An Iranian Nationwide Study. Inflamm Intest Dis 2020;5(1):42-47 View Article PubMed/NCBI
  241. Reinhorn IM, Bernstein CN, Graff LA, Patten SB, Sareen J, Fisk JD, et al. Social phobia in immune-mediated inflammatory diseases. J Psychosom Res 2020;128:109890 View Article PubMed/NCBI
  242. Euesden J, Danese A, Lewis CM, Maughan B. A bidirectional relationship between depression and the autoimmune disorders - New perspectives from the National Child Development Study. PLoS One 2017;12(3):e0173015 View Article PubMed/NCBI
  243. Odineal DD, Gershwin ME. The Epidemiology and Clinical Manifestations of Autoimmunity in Selective IgA Deficiency. Clin Rev Allergy Immunol 2020;58(1):107-133 View Article PubMed/NCBI
  244. Lerner A, Neidhöfer S, Matthias T. The gut-gut axis: Cohabitation of celiac, Crohn’s disease and IgA deficiency. Int J Celiac Dis 2016;4:68-70 View Article
  245. Tan J, McKenzie C, Vuillermin PJ, Goverse G, Vinuesa CG, Mebius RE, et al. Dietary Fiber and Bacterial SCFA Enhance Oral Tolerance and Protect against Food Allergy through Diverse Cellular Pathways. Cell Rep 2016;15(12):2809-2824 View Article PubMed/NCBI
  246. Drabińska N, Jarocka-Cyrta E, Markiewicz LH, Krupa-Kozak U. The Effect of Oligofructose-Enriched Inulin on Faecal Bacterial Counts and Microbiota-Associated Characteristics in Celiac Disease Children Following a Gluten-Free Diet: Results of a Randomized, Placebo-Controlled Trial. Nutrients 2018;10(2):201 View Article PubMed/NCBI
  247. Dicks LMT, Vermeulen W. Do Bacteria Provide an Alternative to Cancer Treatment and What Role Does Lactic Acid Bacteria Play?. Microorganisms 2022;10(9):1733 View Article PubMed/NCBI
  248. Liu P, Wang Y, Yang G, Zhang Q, Meng L, Xin Y, et al. The role of short-chain fatty acids in intestinal barrier function, inflammation, oxidative stress, and colonic carcinogenesis. Pharmacol Res 2021;165:105420 View Article PubMed/NCBI
  249. Casas R, Sacanella E, Estruch R. The immune protective effect of the Mediterranean diet against chronic low-grade inflammatory diseases. Endocr Metab Immune Disord Drug Targets 2014;14(4):245-254 View Article PubMed/NCBI
  250. Spyridaki A, Psylinakis E, Chatzivasili D, Thalassinos N, Kounelaki V, Charonitaki A, et al. Adherence to the Mediterranean diet is linked to reduced psychopathology in female celiac disease patients. Psychol Health Med 2023;28(6):1634-1639 View Article PubMed/NCBI
  251. Morreale F, Agnoli C, Roncoroni L, Sieri S, Lombardo V, Mazzeo T, et al. Are the dietary habits of treated individuals with celiac disease adherent to a Mediterranean diet?. Nutr Metab Cardiovasc Dis 2018;28(11):1148-1154 View Article PubMed/NCBI
  252. Lerner A, Benzvi C, Vojdani A. Cross-reactivity and sequence similarity between microbial transglutaminase and human tissue antigens. Sci Rep 2023;13(1):17526 View Article PubMed/NCBI
  253. Bischoff SC, Barbara G, Buurman W, Ockhuizen T, Schulzke JD, Serino M, et al. Intestinal permeability—a new target for disease prevention and therapy. BMC Gastroenterol 2014;14:189 View Article PubMed/NCBI
  254. Barone MV, Salvatore A. Pro-Inflammatory Nutrient: Focus on Gliadin and Celiac Disease. Int J Mol Sci 2022;23(10):5577 View Article PubMed/NCBI
  255. English J, Connolly L, Stewart LD. Increased Intestinal Permeability: An Avenue for the Development of Autoimmune Disease?. Springer Netherlands 2024;16:575-605 View Article
  256. Guandalini S. Editorial: Gluten: yes, no, maybe. Front Med (Lausanne) 2023;10:1225139 View Article PubMed/NCBI
  • Journal of Translational Gastroenterology
  • eISSN 2994-8754
Back to Top

Gluten is a Proinflammatory Inducer of Autoimmunity

Aaron Lerner, Carina Benzvi, Aristo Vojdani
  • Reset Zoom
  • Download TIFF