Next Article in Journal
Antioxidant and Inflammation-Attenuating Ability of Human Milk, Infant Formulas and Their Oligosaccharides
Next Article in Special Issue
Enhancing the Functional and Emulsifying Properties of Potato Protein via Enzymatic Hydrolysis with Papain and Bromelain for Gluten-Free Cake Emulsifiers
Previous Article in Journal
Raman Spectroscopy in the Characterization of Food Carotenoids: Challenges and Prospects
Previous Article in Special Issue
Determination and Comparison of Fat and Fibre Contents in Gluten-Free and Gluten-Containing Flours and Breads: Nutritional Implications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Celiac Disease—Narrative Review on Progress in Celiac Disease

by
Marek K. Kowalski
,
Danuta Domżał-Magrowska
and
Ewa Małecka-Wojciesko
*
Department of Digestive Tract Diseases, Norbert Barlicki University Hospital, 90-153 Lodz, Poland
*
Author to whom correspondence should be addressed.
Foods 2025, 14(6), 959; https://doi.org/10.3390/foods14060959
Submission received: 3 February 2025 / Revised: 28 February 2025 / Accepted: 9 March 2025 / Published: 11 March 2025
(This article belongs to the Special Issue Gluten-Free Food and Celiac Disease: 2nd Edition)

Abstract

:
Celiac disease is defined as a systemic immunological disorder caused by gluten (gliadin and other prolamin) in genetically predisposed individuals, who present with a variety of gluten-dependent symptoms, specific antibodies, the presence of the HLA DQ2 and DQ8 histocompatibility antigen, and enteropathy. Its prevalence, depending on the studied population and methodology, is estimated at 0.75–1.6% of the general population. During the complex immune reaction it induces, most cells involved in inflammatory processes are activated, which leads to the gradual atrophy of intestinal villi and the proliferation of enterocytes within intestinal crypts. The pathogenesis of celiac disease is extremely complicated and is still the subject of research. According to the current diagnostic guidelines, the following criteria should be taken into account: clinical symptoms (intestinal and extraintestinal), the presence of antibodies against tissue transglutaminase in the IgA class, the level of total IgA, and the presence of typical histological changes in duodenal biopsies. Diet-resistant celiac disease is one of the most important clinical challenges, causing serious complications. Currently, the basic method for treating celiac disease is an elimination diet (i.e., the exclusion of products that may contain gluten from the diet), however, new therapeutic strategies are still being sought, mainly based on supplementation with exogenous endopeptidases, modification of the immune response, and the use of zonulin inhibitors and transglutaminase 2 inhibitors. Clinical trials of new drugs are ongoing. The gradually expanding knowledge about the pathogenesis of celiac disease may allow for the development of new therapeutic strategies for both patients with a mild disease course, as well as those that are diet-resistant.

1. Introduction

Celiac disease (CD) is defined as a systemic autoimmune disorder induced by cereal prolamins in genetically predisposed individuals, who experience a variety of gluten-dependent symptoms, specific antibodies, the presence of HLA-DQ2 and DQ8 histocompatibility antigens, and enteropathy [1,2]. Characteristic intestinal symptoms of celiac disease may include diarrhea (13–96%), abdominal pain (8–90%), vomiting (26–33%), flatulence (5–10%), and fatty stools [3,4,5,6]. There are also extraintestinal symptoms that are related to gastrointestinal dysfunction, mainly in the course of malabsorption, ultimately leading to numerous disorders affecting most body systems.
Celiac disease, untreated for years, can also lead to the development of malignant tumors, such as esophageal cancer, small intestine cancer, and lymphoma, especially T-cell lymphoma, which occurs mainly in the small intestine. These cancers are rare, but they occur significantly more often in patients with celiac disease than in the general population [7].
Patients with celiac disease also suffer from other comorbidities—the most prevalent are autoimmune diseases such as type 1 diabetes (7%), Duhring’s disease (3%), and thyroid diseases (5–21%) [8,9], but also neuropsychiatric disorders (3.9–35.9%) [10].
In this publication, we would like to present the progress in knowledge on the pathogenesis of celiac disease, the latest diagnostics novel management strategies, and treatment monitoring.

1.1. Classification

Chronic inflammation in celiac disease leads to gradual changes in the duodenal mucosa. The histopathological picture of the small intestinal mucosa was described and systematized initially by Marsh et al. and then modified by Oberhuber et al. [11,12,13,14,15] According to current classification, the histopathological changes in the small intestinal mucosa are divided into five types. In type 0 (preinfiltrative), the mucosal picture is normal and the ratio of intraepithelial lymphocytes (IELs) to enterocytes is less than 30. In type 1 (infiltrative stage), an increase in the number of intraepithelial lymphocytes (IELs) above 30 per 100 enterocytes is observed, without other changes. In type 2 (infiltrative–hyperplastic), in addition to an increased number of IELs, intestinal crypt hyperplasia and an elevated mitotic index are observed, with a normal villi structure. Within type 3 (flat destructive), three subtypes are distinguished. In subtype 3a (progressive villous atrophy), in addition to the changes characteristic of type 2, there also comes mild villous atrophy. In subtype 3b (almost complete shortening of the villi), villous atrophy is clearly marked, with crypt hyperplasia and a number of IELs > 30/100 enterocytes, and in 3c (complete villous atrophy), the villi are flat, with crypt hyperplasia and a number of IELs > 30/100 enterocytes. Type 4, hypoplastic–atrophic, involves complete villous atrophy with a normal crypt structure and normal number of IELs [11,12,13,14,15].
According to the current Oslo definitions for celiac disease, depending on the clinical picture and additional tests, several forms of celiac disease have been identified, as follows: typical, atypical, asymptomatic, potential, and refractory [16,17]. Typical celiac disease is diagnosed based on intestinal symptoms (such as weight loss, chronic diarrhea, abdominal pain, vomiting, flatulence, and steatorrhea), Marsh 3 villus atrophy, the presence of characteristic antibodies, and the presence of HLA-DQ 2 or DQ8. Atypical celiac disease differs from overt celiac disease by the presence of extraintestinal symptoms and the absence of classical symptoms. The most frequent extraintestinal symptoms are anemia (3–30%), iron deficiency anemia (40%), folic acid deficiency (20%), and vitamin B12 deficiency (17%). Among celiac disease patients, deficiencies in the fat-soluble vitamins A, D, K, and E and elevated levels of aminotransferases are observed more frequently than in the general population [18]. Osteopenia (54%) and osteoporosis (12%), mainly due to a vitamin D deficiency (34%), and hypocalcemia leading to tetany are among the most common complications, which may present as the first symptoms of the disease [19,20,21,22,23,24]. A meta-analysis of 26 studies showed lower levels of vitamin D among celiac disease patients who did not adhere to a gluten-free diet than in the general population. At the same time, it was found that, after the introduction of a gluten-free diet, vitamin D levels were elevated [25]. It should be remembered that vitamin D deficiency is also a common phenomenon [26] among residents of countries with a high sun exposure [27]. Zanchetta et al. reported that bone loss is observed in about 50% of patients with CD [23,28] (Table 1). In another study by Bai et al., osteopenia incidence in CD was even higher—84% [21]. Galli et al., Ganji et al., Walker et al., and Sayar et al. estimated osteopenia frequency to be between 32 and 67% in all CD patients. Osteoporosis is less frequent and, among CD patients, yields from about 11 to 38% [29,30,31,32,33] (Table 1). In the newest study, Skoracka et al. revealed that women with CD have a decreased bone mass density (BMD) and anthropometric parameters such as body mass, BMI, fat tissue, muscle mass, and fat-free mass [34] (Table 1). There are also many neurological diseases associated with celiac disease, like gluten-dependent cerebellar ataxia, progressive cerebellar ataxia, spinocerebellar degeneration, increased epileptic activity on EEG recording and epilepsy, cerebral calcifications with convulsions, restless legs syndrome, myopathy, and peripheral neuropathy in the course of vitamin B1 and B12 deficiency, as well as dementia [35,36,37,38]. Patients often suffer from mental illnesses, especially schizophrenia and depression, as well as autism [39,40,41,42,43].
The clinical manifestation of pediatric patients with celiac disease has changed over the years. Nowadays, children present many different symptoms in the gastrointestinal tract, such as diarrhea, bloating, and abdominal pain, but also constipation, gastroesophageal reflux disease, vomiting, and dyspepsia. They also present extraintestinal symptoms like malabsorption syndrome, failure to thrive, short stature, dental enamel defects, delayed puberty, and joint pain or anemia due to isolated iron deficiency [44,45]. In children, there are more frequent neuropsychiatric symptoms, like mood changes, learning disabilities, confusion, fatigue, memory loss, depression, persecutory delusions, and psychosis [46,47].
In asymptomatic celiac disease, only the presence of serum anti-tTG, -EMA, and -DPG antibodies and villous atrophy detected in duodenal bioptates are observed. Potential celiac disease is characterized by the presence of characteristic HLA and elevated levels of anti-tTG, -EMA, and -DGP antibodies, but not enteropathy. It concerns individuals who have shown symptoms of gluten-sensitive enteropathy in the past. It should be noted that patients with potential celiac disease are at risk for villus atrophy [2,48]. Refractory celiac disease (RCD) occurs when a patient, despite adherence to a strict gluten-free diet for 12 months, does not achieve villous regeneration [49,50,51]. The two following forms of RCD are distinguished: type I, in which in a histopathological examination, activated T cells constitute up to 20% of all those visible in the preparation, and type II, when their presence is higher [52,53,54]. RCD accounts for 0.04–1.5% of all celiac disease cases and is mainly observed in patients diagnosed over the age of 50 [55,56]. In spite of research progress, RCD represents the important clinical challenge and its management is difficult.
Once a correct diagnosis has been made, in accordance with the guidelines for patients with celiac disease, serologic surveillance is recommended every 3–6 months for the first year after diagnosis and then every 1–2 years. It has been considered that a lack of normalization of antibody levels within a period of 12 months indicates gluten contamination of consumed food or RCD [2,49,51,57,58,59,60,61,62,63].

1.2. Epidemiology

Celiac disease is considered one of the most common autoimmune diseases. Its prevalence, depending on the investigated population and methodology, is estimated at 0.28–5.6% of all screened people [16,64,65,66]. When analyzing the distribution of celiac disease incidence, there is no clear geographical latitude or continent where celiac disease occurs more often (Figure 1) [65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89]. A detailed analysis conducted among the inhabitants of the United States of America showed that celiac disease occurs significantly more frequently in Caucasian people (1.01%) than in African American (0.2%) or Latino (0.3%) people [67].
Celiac disease develops in genetically predisposed subjects. It has been shown to occur more frequently in relatives of celiac patients than in the general population. The risk of developing celiac disease was 4.5–7.5% among first-degree relatives and 2.3–2.6% among second-degree relatives. It was diagnosed more often in women than in men who were first-degree relatives of celiac patients (8.4% vs. 5.2%) [68,69,70].
Studies conducted around the world have observed considerable geographic variations in the prevalence of celiac disease. It occurs mainly in Europe (1.5–2.4% in Finland, 0.7% in Italy, and 0.3% in Germany [71]), North Africa (0.28–5.6%), the Middle East (1.9–2%), North America (0.75%), and India. It is much less frequent in Australia (0.3%). It is equally rare in Asia, excluding India, as well as in Central and South Africa (0.4–0.6%) (Table 2) [72,73,74,75,76,77].
Most population studies are based on an analysis of the frequency of increased serum anti-tTG antibodies, which does not fully correspond to the frequency of celiac disease [78]. Many studies assessing the incidence of celiac disease are conducted among patients with type 1 diabetes [74].

1.3. Etiopathogenesis

Celiac disease develops due to an abnormal immune response to ingested prolamin contained in cereals such as rye, wheat, barley, and oats, which are, respectively, gliadin, secalin, hordein, or avenin, in genetically predisposed individuals with specific histocompatibility antigens (HLA-DQ2 or HLA-DQ8) [90]. Gluten proteins are a heterogeneous group consisting of high-molecular-weight (HMW) and low-molecular-weight (LMW) subunits. Although oats contain prolamins that are toxic to some patients with CD, pure oats are not contraindicated in patients with celiac disease in some high-income countries. However, because of this controversy, their use should be associated with more careful monitoring of the patient [91,92,93]. A meta-analysis of 433 studies examining the immunogenicity of oats did not show any effect of their consumption on the health of patients with celiac disease [94]. Also, a double-blind study on a group of 177 patients with confirmed celiac disease, lasting 15 months, did not reveal an increase in antibody levels and did not cause symptoms or an increased intestinal permeability [95]. Perhaps this is the reason that ESPGHAN, WGO, ECD, NICE, BCG, and ACG guidelines allow the use of oats in CD [2,49,57,59,60,61,63]. On the other hand, the presence of t-cell lymphocytes with specific DQ 2.5-ave-1a-c and DQ 2.5-ave-2 receptors has been demonstrated [96]. In addition, Russian guidelines categorically exclude the possibility of using refined oats in a gluten-free diet [58].
After ingesting gluten-rich cereals, gluten is partially broken down in the stomach by pepsin. The long-chain protein fragments obtained in this way, after contact with the gastrointestinal mucosa, show significant immunogenicity, causing an increase in the concentration of inflammatory interleukins in the blood [97].
Prolamin degradation products are further digested in the duodenum and small intestine with prolyl oligopeptidase [98]. Gluten treated by endopeptidases is broken down into short glutamine and proline-rich peptide chains. The level of degradation resulting from the digestive process depends on the type of the contained protein. Protein digestion results in the formation of oligopeptides which are then deamidated by tissue transglutaminase and stimulate the activity of T lymphocytes [99,100,101].
The intestinal microbiota play an active role in the process of gluten protein digestion. In celiac patients, especially those not complying with a gluten-free diet, the presence of a greater number of fermenting bacteria in the gut has been demonstrated [102]. It should also be noted that the introduction of a gluten-free diet leads to an increase in the number of Enterobacteriaceae and Escherichia coli with a simultaneous reduction in the number of Bifidobacter sp., both in celiac patients and in healthy individuals. In Sanz and De Palma’s study, ten healthy subjects followed a gluten-free diet for a month. After this period, fecal samples revealed significantly reduced counts of Bifidobacterium and Lactobacillus and increased counts of Prevotella, Clostridium, Enterobacteriaceae, and E. coli [103,104]. The same microflora changes were observed in patients both with celiac disease strictly adhering to the diet and in patients with untreated celiac disease. A reduction in the number of probiotic strains and the promotion of non-beneficial strains can lead to numerous gastrointestinal complaints such as flatulence, abdominal pain, empty belching, and even fever and diarrhea [105,106,107]. Many different oral and intestinal bacteria produce their own endopeptidases and hydrolases that contribute to the digestion of protein chains resulting from the degradation of gliadin. Therefore, excluding gluten from the diet may promote other bacterial strains, leading to adverse changes in the intestinal flora described above [108,109,110,111,112,113].
Fragments of undigested proline-rich proteins in contact with the intestinal mucosa increase in IL-15, which, in turn, stimulates activated T and NK cells to produce chemokines and proinflammatory cytokines [114,115,116]. Moreover, IL-15 promotes NK cell apoptosis [117,118]. This process is additionally intensified by a direct contact reaction between gliadin fraction (p31–43) and the intestinal mucosa, leading to enterocyte cytoskeleton reconstruction and, subsequently, to apoptosis [119,120,121] Furthermore, fragments of partially digested gliadin have been shown to bind to CD95/FAS receptors belonging to the group of TNF-alpha (Tumor Necrosis Factor alpha) family receptors, leading to apoptosis by activating caspase 8 and 10 [122].
Moreover, an increased intestinal mucosal permeability is caused by the loosening of the tight junctions between enterocytes [123] The pathomechanism of this phenomenon was described by Fasano et al. They showed that gliadin fragments (111–130 and 151–170) in the small intestine bind to the CXCR3 receptor [124]. This leads to the release of zonulin (a protein similar to the toxin produced by cholera bacillus) from enterocytes of the small intestinal mucosa [125,126,127]. This protein binds to the protease-activating receptor (PAR2). As a result, the tight junctions between enterocytes are disrupted and transepithelial electrical resistance is reduced, leading to increased intestinal wall permeability [128,129,130,131]. Both described processes are responsible for the increased penetration of undigested short-chain proline-rich proteins into the submucosa. At this stage, the intestinal bacterial flora may also play an important role. In untreated celiac patients, B. lactis and L. fermentum have been shown to increase intestinal mucosal permeability by reducing transepithelial resistance and increasing zonulin expression [132].
In vitro studies have demonstrated that gliadin fragments (mainly p62–75 and p57–68) undergo tissue transglutaminase-mediated deamidation. The deamination process has been shown to be necessary to trigger further immune responses [133,134,135]. In the next step, deamidated gliadin peptides bind with DQ2 or DQ8 receptors expressed on antigen-presenting cells (APCs) [136,137,138]. Two heterodimers, DQ2 (DQ2.2 and DQ 2.5) and DQ8, participate in the antigen presentation process [139,140,141,142]. As a result of the reaction, HLA-DQ2 and DQ8 are complexed with deamidated α-gliadin fragments [143]. The protein fragments are then presented to B lymphocytes by APCs, initiating further humoral response.
The existence of molecular mimicry resulting from a similar three-dimensional appearance of proteins, mainly tissue transglutaminase and specific gliadin fragments, has been proven [144]. As a result, B cells produce antibodies against both tissue transglutaminase and gliadin and deamidated gliadin peptides [145,146,147,148]. It should be noted that, under the influence of a number of inflammatory cytokines produced by monocytes and T cells, such as INFα, IL-15, IL-18, and IL-21, the inflammatory response in the course of celiac disease is directed towards the Th1-dependent inflammatory pathway [149,150,151,152,153]. At the same time, INF γ and TGF α stimulate monocytes and myofibroblasts, damaging the enterocyte stroma by producing matrix metalloproteinases (MMP-1, MMP-3, and MMP-12). This leads directly to the intensification of the apoptosis process and to increased mucosal permeability [154,155,156].
The above-mentioned CXCR3 receptor present on enterocytes also interacts with fragments of gliadin (p261–277 and p270–286), leading to an increase in the concentration of inflammatory interleukins, especially IL-8, a neutrophil chemokine. This reaction was observed only in celiac patients and persisted despite a gluten-free diet [157].
On the other hand, the contact of gliadin fragments with enterocytes leads to an increased production of EGF (epidermal growth factor), which stimulates crypt enterocyte proliferation, leading to their hyperplasia [158,159].
As it results from the above description, during a complex immune reaction, most cells involved in inflammatory processes are activated, both in terms of cellular and humoral responses (Figure 2). The above-mentioned prolamins, adequately to the HLA epitopes present, contribute to CD development in a complex mechanism led to direct the apoptosis of enterocytes, which leads to gradual villous atrophy. On the other hand, gliadin induces an increased production of EGF, which leads to cell hyperproliferation within intestinal crypts. The pathogenesis of celiac disease is extremely complex and still not fully elucidated yet.

1.4. Diagnosis

Knowledge about the pathogenesis of celiac disease has deepened over recent decades. As a result, the criteria for diagnosing celiac disease have changed over the years (Figure 3). According to the 2012 ESPGHAN (European Society for Pediatric Gastroenterology, Hepatology and Nutrition) criteria, diagnosis is based on the clinical picture (both typical and atypical symptoms) and intestinal villus atrophy type 2 or 3 according to the Marsh scale. Biopsy is recommended at upper G.I. endoscopy according to the following scheme—two samples from the duodenal bulb and four from the descending duodenum. In addition, the detection of specific anti-endomysial antibodies, anti-tissue transglutaminase 2 (anti-tTG), or anti-deamidated gliadin peptides (anti-DGP) at a concentration exceeding the norm by at least three times should be noted, as well as an elevated level of endomysial antibodies (EMAs) and the presence of the specific histocompatibility antigens HLA-DQ2 or DQ8. The sensitivity and specificity of serum antibody tests for celiac disease according to the different sources are presented in Table 3 [160,161,162].
Avoiding biopsy in pediatric patients who have levels of IgA anti-tTG antibodies ten times higher than the upper limit of normal, a high level of EMA, and fulfil the genetic criteria has been considered [163]. However, the 2020 modification of the above guidelines places the main emphasis on the determination of IgA anti-tTG antibodies and on the determination of the total IgA level. The determination of total IgA is necessary due to its frequent deficiency in CD patients. As a result, low levels of IgA anti-tTG antibodies may result from total IgA deficiency and produce a false-negative result. In the case of children and adolescents, the simultaneous finding of a ten times increase in the upper limit of IgA anti-tTG antibodies with a normal level of total IgA and a typical clinical picture allows for the diagnosis of celiac disease without the need for duodenal histology. It should be noted that IgA anti-tTG antibodies are also useful in the detection of gluten free-diet non-adherence in patients with established CD.
In adults, the presence of typical histological changes is necessary for CD diagnosis. Genetic tests assessing the presence of HLA-DQ2 and DQ8 antigens are used to exclude celiac disease in subjects observing a gluten-free diet. Nevertheless, HLA-DQ2/DQ8 has a limited role in the diagnosis of CD. This role is based on a negative predictive value in order to rule out CD in patients who are seronegative with typical histologic changes, in patients seronegative at the time of diagnosis, and in those patients with previously diagnosed CD before the introduction of celiac-specific serology [2].
These diagnostic criteria are also recommended by the American College of Gastroenterology (ACG). The ACG devotes part of its guidelines to intestinal permeability tests. Permeability tests are not recommended for the diagnosis of CD. They might be useful for the detection of gross changes in intestinal permeability associated with intestinal inflammation, but their sensitivity and specificity are too low for the diagnosis of CD [1,61].

1.5. New Diagnostic Techniques for Celiac Disease

New diagnostic techniques are still being sought to enable diagnosis with a greater precision. Recent studies indicate a higher reliability of the determination of antibodies against neo-epitope tTG complexed to gliadin (98–100% sensitivity and 93–96% specificity) in comparison with the assessment of anti-tissue transglutaminase antibodies (sensitivity: 74–100%, specificity: 78–100%) [160,161,162,164,165,166,167,168].
An additional test confirming celiac disease in the future may be the determination of the presence of T cells’ response to HLA-DQ2–α-gliadin complexes. A positive correlation was demonstrated between the number of gluten-reactive T cells in duodenal biopsy and histological damage in the course of celiac disease, as well as the concentration of anti-tissue transglutaminase antibodies [169]. It has been shown that three days of ingestion of gluten-containing food renders the memory T lymphocytes to be reactive against gliadin from gut-associated lymphoid tissue (GALT) and be detected in the peripheral blood of CD patients. These antigen-specific T-cells can be detected with enzyme-linked immunospot (ELISPOT) assays or by flow cytometry tetramer technology. Moreover, studies have been conducted on T cells collected from the peripheral blood of patients for the presence of the histocompatibility antigen HLA-DQ2 [170,171,172]. In the future, this test may become not only a new diagnostic method for celiac disease detection, but also a test to confirm the diagnosis of celiac disease in patients already observing a gluten-free diet, without exposing them to a long-term gluten challenge. The recent popularity of self-administered gluten-free diets without a clear indication represents a frequent challenge for clinicians when comes to the CD confirmation or exclusion. Analysis of the presence of gluten-reactive T cells in peripheral blood can also be used to assess adherence to a gluten-free diet [170,171,172,173]. Such a test could be helpful in diagnosing celiac disease, especially since current studies in healthy individuals with HLA-DQ 2.5+ have not shown any reactivity of memory T cells specific for immunodominant gluten epitopes [174]. Although Özgenel et al. and Cecilio et al. showed an increased frequency of HLA-DQ2/DQ8 in first-degree relatives of celiac patients [175,176], their use in the primary diagnosis of celiac disease is not confirmed [177].
New genetic determinants of celiac disease are still being sought due to the significant genotypic–phenotypic divergence among individuals with HLA DQ2/DQ8 antigens. A GWAS (genome-wide association study) study conducted in 336 celiac patients from Poland demonstrated a significant association between the development of celiac disease and the presence of the MSH5 gene [178]. In a study evaluating single-nucleotide polymorphisms (SNPs), 57 non-HLA variants predisposing to the development of CD were identified. In turn, within HLA, a significant predictive value was demonstrated for the presence of HLA-DQ 2.5 rs2187668, HLA-DQ7 rs4639334, and DQ8 rs7454108 [179,180,181,182]. So far, few studies have been published examining the association of non-HLA genes with the risk of developing and subsequent severity of CD.
In some patients with celiac disease, despite gluten-free diet adherence, the intestinal villi do not recover and chronic symptoms do not subside. In order to monitor and detect a group of patients who may require more careful surveillance and the introduction of additional management, it may be useful to detect patients’ whole-blood IL-2 release [183,184]. This relationship was confirmed by Tye-Din et al. in a study of 295 patients on a gluten-free diet who were challenged with gluten [185]. Gliadin-specific T cells found both in the G.I. tract [134,186,187] and selected from the peripheral blood [137,188] can also be used to assess unconscious exposure to ingested gluten. Zühlke et al. demonstrated an increased expression of CD38 on gluten-specific CD4+ T cells in patients after gliadin exposure. The study was conducted using blood samples from patients who underwent a gluten challenge test. Samples were incubated with an equal mixture of HLA-DQ2.5:gluten tetramers and were stained with the following antibody mixture: CD38-FITC, CD45RA-PE-Cy7. The activation status of tetramer + β7 + TEM cells was assessed by the percentage of cell surface expression of CD38 on tetramer  +  β7  +  TEM cells [189]. These tests can be used in the future to monitor gluten free diet adherence.

1.6. Treatment

According to the current guidelines, the basic method for treating celiac disease is an elimination diet (i.e., excluding products that may contain gluten) [1,2], i.e., foods produced using substrates derived from wheat, rye, and barley [61]. There is a lot of controversy about the use of oats in CD. On the one hand, they can enrich the diet with whole grains and nutritious dishes [190], but on the other hand, some studies show that they may contain traces of gluten [191]. It should be noted that the avenin present in oats does not have epitopes considered to be the source of the immunological reaction to gluten proteins in wheat, barley, and rye [192]. Current studies indicate that the consumption of 10 mg of gluten daily in patients with celiac disease should not cause an exacerbation of the disease, although in some cases, the daily dose may be several times higher [193,194,195,196]. The applicable certification standards allow for a gluten content of 20 ppm (20 mg per kilogram of product) in gluten-free products and 100 ppm in low-gluten products [197]. In the European Union, gluten-free products are marked with the crossed-out ear of wheat symbol and in accordance with “Commission Regulation (EU) 41/2009 on the composition and labelling of foodstuffs suitable for people intolerant to gluten”. The gluten content in food products marked with this logo may not exceed 20 mg per kilogram of product. A similar legal regulation was also introduced in the United States [198]. It should be noted that even trace amounts of gluten can lead to chronic inflammation in the intestinal mucosa [199,200].
In current times, considering that we encounter highly processed food on a daily basis, a gluten-free diet has become an extremely demanding form of treatment for patients. Patients must be aware of how to properly read food labels and follow the current product tests for gluten content. At the same time, a proper balance of the diet in terms of vitamins, minerals, and fiber should be taken into account [201]. This constant effort can affect quality of life [202]. In addition, gluten-free products, compared to those containing gluten, have a lower protein and fiber content, as well as nutritional value (i.e., lower intake of folate, iron, magnesium, selenium, calcium, and vitamins D, E, and some of group B [nestares, cardo]) compared to their counterparts containing gluten. Due to the consumption of a large amount of meat, a gluten-free diet is also a diet rich in saturated fats [203,204,205,206]. As a result, a poorly balanced gluten-free diet can lead to amino acid and protein deficiency, vitamin deficiency (especially vitamin A, B1, B6, B12, E, and D), electrolyte deficiency (primarily iron, zinc, calcium, and magnesium), and folic acid deficiency [207,208,209,210]. Moreover, gluten-free products are much more expensive than their gluten-containing counterparts, and some patients will be forced to limit their food intake or choose products of a lower quality, which, in many cases, may be an additional factor increasing such deficiencies [211,212,213].
However, in most cases, constant adherence to a gluten-free diet, especially among young patients, leads to complete recovery of the villi and resolution of the inflammatory infiltrate, despite the presence of trace amounts of gluten contamination in food [214]. In adults, especially over the age of 60, histological changes may not undergo complete remission despite strict adherence to a gluten-free diet [215,216].
It should be underlined that a gluten-free diet is connected to numerous economic, social, and psychological problems [202]. This is very expensive, and patients have to search for special stores with the adequate products, which are not uniformly localized. According to the studies conducted, trace amounts of gluten are currently detected even in naturally gluten-free products due to contamination resulting from production processes [217,218,219,220,221,222,223]. Even in products marked with the crossed-out ear of grain symbol, the safe gluten concentration is often exceeded [217,224,225,226]. The high quality of these products is not always warranted [211,212,213]. Gluten-free diet adherence poses social problems when visiting restaurants with friends, where gluten free food is less available [227,228,229]. In addition, this is connected with stigmatization, as being a person with special requirements can be traumatic, in particular for young people.

2. New Potential Treatment Strategies

2.1. The Use of Bacteria in the Treatment of Celiac Disease

The use of endopeptidases naturally produced by bacterial strains and fungi is one of the suggested treatment methods. Flavobacterium meningosepticum were the first strains in which the presence of endopeptidases capable of digesting prolamine-rich protein fragments was detected. As a result of gliadin degradation with endopeptidases, fragments are formed that are non-immunogenic for celiac patients [113]. Further studies have also shown the presence of similar endopeptidases in other bacteria and fungi [111,112]. Proteases have also been purified from the probiotic bacteria of Lactococcus and Lactobacillus [230,231,232]. Alpha-gliadins were reduced by more than 50% by peptidases produced by Lactobacillus spp. [233]. The endopeptidases described above have a very limited effect on gluten digestion. To achieve effective treatment, a fermentation process is necessary, which limits their use as an enzymatic supplement. Endopeptidases derived from Lactobacillus sp. are used in the fermentation process of flour, which can be used in further stages for bakery products [234,235,236,237]. Enzymes produced by Lactococcus, Lactobacillus, and Flovabacterium are not suitable for the oral supplementation of gluten-digesting enzymes. Further research in this area led to the discovery of endopeptidases that effectively break down gluten in the upper gastrointestinal tract, as described below.

2.2. Oral Supplementation of Endopeptidases

Clinical trials are currently underway on the oral administration of endopeptidase derived from Aspergillus Niger (AN-PEP). Although first reports indicate an effective reduction in the frequency of immune responses, the authors indicate that the dose of enzymes necessary to digest the gluten contained in food strictly depends on the type of meal and the method of its preparation, therefore, effective supplementation may prove difficult [238,239].
Oral preparations of endopeptidase mixture (ALV003) obtained from Sphingomonas capsulata (SC-PEP) and endopeptidase from barley seeds (EP-B2) have also been used in clinical trials. Studies conducted so far indicate that subjecting gluten-containing products to enzymatic treatment with ALV003 before consumption significantly reduces the immune response to prolamins contained in the meal [240,241,242]. Phase 2 clinical trials have shown that MGX003 reduced gluten-induced intestinal mucosal damage and symptom severity [243].
After a successful trial using 1.2 g of gluten per day for 6 weeks, which showed a significant decrease in the inflammatory response and a reduction in symptoms [243], further clinical trials with Latiglutenase were initiated in a large group of patients. Research has also been initiated on a computer-modified enzyme, Kuma 030 obtained from Alicyclobacillus sendaiensis, which is an endopeptidase that effectively degrades the linkage between proline and glutamine (TAK-062) [244]. In preliminary studies, it has shown a high efficiency in digesting a significant amount of gluten [245].

2.3. Modification of Immune Response

Apart from the application of bacterial and fungal endopeptidases, research is still ongoing on probiotic strains that reduce the inflammatory process. Strains of the genus Bifidobacterium spp. are mainly used in such studies. They were shown to reduce the level of TNF α, the number of IELs, and the level of antibodies in patients with celiac disease compared to a group of patients with celiac disease who did not receive probiotics [246,247,248,249].
Systemic steroid therapy has been used for years to modify the inflammatory response. Its possible use in celiac disease, especially if resistant to diet, has been considered for many years [250]. Studies using prednisolone at a dose of 1 mg/kg bw did not demonstrate any significant effect on villous regeneration [251]. However, the administration of 9 mg of budesonide in patients with refractory celiac disease for 3 months resulted in improvement and led to villous regeneration [252]. Numerous studies have indicated the effectiveness of budesonide in patients with celiac disease not responding to a gluten-free diet (NRCD), as well as refractory celiac disease (RCD) [253,254,255]. Azathioprine is also used in RCD. Remissions have been demonstrated in small groups of patients with this type of CD [256]. In subsequent studies, the use of azathioprine was found to be effective in type I RCD, but in some cases of type II RCD, its effect seems to be unsatisfactory [257,258]
There are very few case reports of the successful use of anti-TNFα antibodies in RCD. However, so far, there has been no broader analysis of such treatment. Therefore, such management should be considered as non-standard and limited to selected cases [259,260,261].
The monoclonal anti-IL-15 antibody (AMG 714; currently PRV-015) has been evaluated in CD [262,263,264]. In one study, the authors did not demonstrate any significant difference between the use of the preparation and a placebo in terms of pathological changes in the intestinal mucosa in patients exposed to a gluten challenge, but they did demonstrate a significant reduction in symptoms [265]. In another phase 2a study in a group of 28 patients, no effect on the course of the inflammatory process was demonstrated, but a reduction in symptoms was confirmed compared to the group receiving placebo [266]. A phase 2b clinical study (NCT04424927) is currently underway in adult patients with refractory celiac disease.
In vitro studies have shown that tofacitinib, a Janus kinase inhibitor, has the potential to regulate the activity of abnormal IEL cell populations. In a phase 2 open-label clinical study [(EudraCT): 2018-001678-10] in patients with RCD type 2, 12-week treatment with tofacitinib led to the resolution of diarrhea/loose stools and disappearance of abdominal pain and weight gain, however, the primary immunologic end point of an absolute decrease in total IELs was not met and mucosal improvement as a secondary end point was observed in four of six patients. In all patients, a rapid recurrence of symptoms, including weight loss, was observed after treatment discontinuation, while the reintroduction of therapy led to a rapid and complete improvement [267].
According to current knowledge, celiac disease is a Th1-mediated autoimmune process. Attempts are being made to modulate this response by redirecting patients’ immune response to a Th2-mediated pathway. For this purpose, CD patients received hookworm larvae (Necator americanus) transcutaneously. In the studies conducted so far, in small groups, patients undergoing the procedure developed gluten tolerance without other clinical implications [268,269,270]. Moreover, N. americanus infection in gluten-challenged patients leads to an increased microbial richness by improving homeostasis, which may normalize inflammatory parameters and increase gluten tolerance [271,272]. Although a study conducted in 54 patients with celiac disease showed that hookworm infection reduces symptoms after gluten ingestion in patients with celiac disease, it does not restore tolerance to long-term moderate gluten consumption (2 g/day) [273].
A polymer conjugated to a deamidated gliadin peptide (KAN-101) has also been developed, which, when administered intravenously, liver-targeted, is expected to induce immune tolerance to gluten. The first-in-human study of KAN-101 demonstrated an acceptable safety profile in patients with celiac disease. Furthermore, KAN-101 showed the potential to induce gluten tolerance by blunting the inflammatory response of gliadin-specific CD4+ cells and intestinal CD8+ cells after gluten challenge (NCT04248855) [274]. KAN-101 is currently being evaluated in phase Ib/II and phase II studies (NCT05574010, NCT06001177). Another similar strategy is the use of nanoparticles as a copolymer of gluten particles and PLGA (TAK-101). Currently, the second phase of clinical studies has been completed, confirming the safety of this preparation and demonstrating the lack of an immune reaction to 14-day gluten challenge—TAK-101 was well-tolerated in celiac patients and no evidence of systemic immunosuppression was observed (NCT03486990 and NCT03738475) [275]. A phase II study is currently underway to investigate the efficacy and safety of TAK-101 in preventing gluten-specific T cell activation in celiac patients on a gluten-free diet (NCT04530123).
There has also been an attempt to create a vaccine (Nexvax2) designed to induce gluten tolerance by modifying the T cell response. Clinical trials have been initiated in this aspect. After vaccination, the immune response to gliadin was significantly lower than in unvaccinated patients. Studies have confirmed lower concentrations of IL-2 and INF-γ, as well as significantly lower CD4+ T cells proliferation [276,277,278,279]. Although the initial phase 2 study by Hardy et al. showed a reduction in the immune response to gluten in the group receiving nexvax2 [279], a further study in 178 patients with celiac disease, including an analysis of symptoms following gluten ingestion, did not show a reduction in symptoms in the group receiving the vaccine compared to the group receiving a placebo [280].

2.4. Zonulin Inhibitors

In celiac disease, according to the pathophysiology described above, the contact of gluten with the intestinal mucosa results in an increase in zonulin release. This leads to enterocyte tight junction dysfunction and increased mucosal permeability [127,281,282,283]. Knowledge of this pathomechanism was used to develop a protein substance (larazotide acetate) with properties that regulate the tight junctions between enterocytes, modulate intercellular tension (TEER), and inhibit the zonulin effect. This leads to a reduction in the permeability of partially digested gliadin fragments and, thus, a reduction in the immune response. Additionally, larazotide promotes the repair of enterocyte structural defects resulting from direct reaction with gliadin [284,285,286,287,288]. Developed by 9 Meters Biopharma, it was investigated as an adjunctive treatment for celiac disease patients who continued to have symptoms despite adherence to a gluten-free diet. The trial was discontinued in 2022 after an interim analysis explaining that the additional number of patients needed to determine a significant clinical outcome between the placebo and larazotide was too large to support trial continuation [289]. The therapeutic potential of larazotide acetate was assessed to be lower than expected due to the presence of both paracellular and transcellular gliadin transport pathways, whereas larazotide acetate is intended to block only the paracellular pathway. On the other hand, the study conducted studies on the use of AT-1001, which is an inhibitor of paracellular permeability derived from a protein secreted by Vibrio cholerae, and found that it ameliorated the impaired intestinal permeability [290].

2.5. Tissue Transglutaminase 2 Inhibitors

A number of substances have been developed as tissue transglutaminase 2 (tTG2) inhibitors [291,292,293]. However, studies in mice have shown that a complete congenital deficiency of tissue transglutaminase 2 leads to numerous complications, such as glomerulonephritis, splenomegaly, and impaired phagocytosis [294,295]. For this reason, it is impossible to introduce complete transglutaminase 2 inhibition in clinical practice. There are ongoing studies investigating the use of partial tTG2 inhibitors in patients with celiac disease [296,297]. A phase II clinical trial of a selective oral inhibitor of activated tissue transglutaminase 2, ZED 1227, has been completed. In the initial phases of the study, ZED 1227 was shown to be effective in preventing gliadin deamidation. The application of the preparation in a group of CD patients undergoing gluten challenge also demonstrated good results, including a reduction in mucosal damage compared to the group receiving placebo [298,299]. A phase IIb trial is currently ongoing in CD patients experiencing symptoms despite following a gluten-free diet (EudraCT 2020-004612-97).

3. Conclusions

Celiac disease results from a complex immune reaction to gluten. The gradually expanding knowledge about its pathogenesis enables the development of new therapeutic strategies both in patients with a mild course of the disease and in those who do not observe clinical improvement after following a gluten-free diet. However, currently the only recognized treatment for celiac disease remains a gluten-free diet. The main clinical challenges are diet-refractory disease and the increased risk of small intestine neoplasia, which is particularly difficult to detect. Early small intestine cancers symptoms are not characteristic and diagnostic methods such as MR enterography and enteroscopy are not widely available and highly operator-dependent, although it is possible that, in the coming years, new diagnostic and treatment methods will also find their application in clinical practice.

Funding

This study was supported by the Scientific grant No. 503/1-002-01/503-11-001 from the Medical University of Lodz, Poland to Ewa Malecka Wojciesko and from the Polish Society for Digestive Tract Neoplasms Prevention.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rubio-Tapia, A.; Hill, I.D.; Semrad, C.; Kelly, C.P.; Greer, K.B.; Limketkai, B.N.; Lebwohl, B. American College of Gastroenterology Guidelines Update: Diagnosis and Management of Celiac Disease. Am. J. Gastroenterol. 2023, 118, 59–76. [Google Scholar] [CrossRef]
  2. Husby, S.; Koletzko, S.; Korponay-Szabó, I.; Kurppa, K.; Mearin, M.L.; Ribes-Koninckx, C.; Shamir, R.; Troncone, R.; Auricchio, R.; Castillejo, G.; et al. European Society Paediatric Gastroenterology, Hepatology and Nutrition Guidelines for Diagnosing Coeliac Disease 2020. J. Pediatr. Gastroenterol. Nutr. 2020, 70, 141–156. [Google Scholar] [CrossRef] [PubMed]
  3. Garampazzi, A.; Rapa, A.; Mura, S.; Capelli, A.; Valori, A.; Boldorini, R.; Oderda, G. Clinical pattern of celiac disease is still changing. J. Pediatr. Gastroenterol. Nutr. 2007, 45, 611–614. [Google Scholar] [CrossRef] [PubMed]
  4. Dinler, G.; Atalay, E.; Kalayci, A.G. Celiac disease in 87 children with typical and atypical symptoms in Black Sea region of Turkey. World J. Pediatr. 2009, 5, 282–286. [Google Scholar] [CrossRef] [PubMed]
  5. Bottaro, G.; Failla, P.; Rotolo, N.; Sanfilippo, G.; Azzaro, F.; Spina, M.; Patane, R. Changes in coeliac disease behaviour over the years. Acta Paediatr. 1993, 82, 566–568. [Google Scholar] [CrossRef]
  6. Faulkner-Hogg, K.B.; Selby, W.S.; Loblay, R.H. Dietary analysis in symptomatic patients with coeliac disease on a gluten-free diet: The role of trace amounts of gluten and non-gluten food intolerances. Scand. J. Gastroenterol. 1999, 34, 784–789. [Google Scholar] [CrossRef]
  7. Marek, K.; Kowalski, A.G. Czy choroba trzewna predysponuje do rozwoju chorób nowotworowych? Onkol. Prakt. Klin. 2015, 2, 140–148. [Google Scholar]
  8. Schosler, L.; Christensen, L.A.; Hvas, C.L. Symptoms and findings in adult-onset celiac disease in a historical Danish patient cohort. Scand. J. Gastroenterol. 2016, 51, 288–294. [Google Scholar] [CrossRef]
  9. Zingone, F.; Bai, J.C.; Cellier, C.; Ludvigsson, J.F. Celiac Disease-Related Conditions: Who to Test? Gastroenterology 2024, 167, 64–78. [Google Scholar] [CrossRef]
  10. Hansen, S.; Osler, M.; Thysen, S.M.; Rumessen, J.J.; Linneberg, A.; Kårhus, L.L. Celiac disease and risk of neuropsychiatric disorders: A nationwide cohort study. Acta Psychiatr. Scand. 2023, 148, 60–70. [Google Scholar] [CrossRef]
  11. Marsh, M.N. Grains of truth: Evolutionary changes in small intestinal mucosa in response to environmental antigen challenge. Gut 1990, 31, 111–114. [Google Scholar] [CrossRef] [PubMed]
  12. Marsh, M.N. Studies of intestinal lymphoid tissue. III. Quantitative analyses of epithelial lymphocytes in the small intestine of human control subjects and of patients with celiac sprue. Gastroenterology 1980, 79, 481–492. [Google Scholar] [CrossRef] [PubMed]
  13. Oberhuber, G.; Granditsch, G.; Vogelsang, H. The histopathology of coeliac disease: Time for a standardized report scheme for pathologists. Eur. J. Gastroenterol. Hepatol. 1999, 11, 1185–1194. [Google Scholar] [CrossRef] [PubMed]
  14. Oberhuber, G.; Caspary, W.F.; Kirchner, T.; Borchard, F.; Stolte, M. Study Group of Gastroenterological Pathology of the German Society of Pathology. Recommendations for celiac disease/sprue diagnosis. Z. Gastroenterol. 2001, 39, 157–166. [Google Scholar] [CrossRef]
  15. Oberhuber, G. Histopathology of celiac disease. Biomed. Pharmacother. 2000, 54, 368–372. [Google Scholar] [CrossRef]
  16. Bingley, P.J.; Williams, A.J.; Norcross, A.J.; Unsworth, D.J.; Lock, R.J.; Ness, A.R.; Jones, R.W. Undiagnosed coeliac disease at age seven: Population based prospective birth cohort study. BMJ 2004, 328, 322–323. [Google Scholar] [CrossRef]
  17. Ludvigsson, J.F.; Leffler, D.A.; Bai, J.C.; Biagi, F.; Fasano, A.; Green, P.H.; Hadjivassiliou, M.; Kaukinen, K.; Kelly, C.P.; Leonard, J.N.; et al. The Oslo definitions for coeliac disease and related terms. Gut 2013, 62, 43–52. [Google Scholar] [CrossRef]
  18. Montón Rodríguez, C.; Sánchez Serrano, J.; Poyatos García, P.; Abril García, C.; Gómez Medina, C.; Capilla-Lozano, M.; Lluch Garcia, P.; Pascual Moreno, I. Liver disorders and celiac disease. Rev. Esp. Enferm. Dig. 2024, 116, 41–42. [Google Scholar] [CrossRef]
  19. Kalayci, A.G.; Kansu, A.; Girgin, N.; Kucuk, O.; Aras, G. Bone mineral density and importance of a gluten-free diet in patients with celiac disease in childhood. Pediatrics 2001, 108, E89. [Google Scholar] [CrossRef]
  20. Mautalen, C.; Gonzalez, D.; Mazure, R.; Vazquez, H.; Lorenzetti, M.P.; Maurino, E.; Niveloni, S.; Pedreira, S.; Smecuol, E.; Boerr, L.A.; et al. Effect of treatment on bone mass, mineral metabolism, and body composition in untreated celiac disease patients. Am. J. Gastroenterol. 1997, 92, 313–318. [Google Scholar]
  21. Bai, J.C.; Gonzalez, D.; Mautalen, C.; Mazure, R.; Pedreira, S.; Vazquez, H.; Smecuol, E.; Siccardi, A.; Cataldi, M.; Niveloni, S.; et al. Long-term effect of gluten restriction on bone mineral density of patients with coeliac disease. Aliment. Pharmacol. Ther. 1997, 11, 157–164. [Google Scholar] [CrossRef] [PubMed]
  22. Ciacci, C.; Maurelli, L.; Klain, M.; Savino, G.; Salvatore, M.; Mazzacca, G.; Cirillo, M. Effects of dietary treatment on bone mineral density in adults with celiac disease: Factors predicting response. Am. J. Gastroenterol. 1997, 92, 992–996. [Google Scholar] [PubMed]
  23. Zanchetta, M.B.; Longobardi, V.; Costa, F.; Longarini, G.; Mazure, R.M.; Moreno, M.L.; Vazquez, H.; Silveira, F.; Niveloni, S.; Smecuol, E.; et al. Impaired Bone Microarchitecture Improves After One Year On Gluten-Free Diet: A Prospective Longitudinal HRpQCT Study in Women with Celiac Disease. J. Bone Miner. Res. 2016, 32, 135–142. [Google Scholar] [CrossRef] [PubMed]
  24. Santonicola, A.; Wieser, H.; Gizzi, C.; Soldaini, C.; Ciacci, C. Associations between Celiac Disease, Extra-Gastrointestinal Manifestations, and Gluten-Free Diet: A Narrative Overview. Nutrients 2024, 16, 1814. [Google Scholar] [CrossRef]
  25. Sun, Y.; Zhou, Q.; Tian, D.; Zhou, J.; Dong, S. Relationship between vitamin D levels and pediatric celiac disease: A systematic review and meta-analysis. BMC Pediatr. 2024, 24, 185. [Google Scholar] [CrossRef]
  26. Bolland, M.J.; Hofman, P.; Grey, A. Prevalence of Vitamin D Deficiency With Biochemical Abnormalities in Children Undergoing Vitamin D Testing. Clin. Endocrinol. 2024, 102, 255–263. [Google Scholar] [CrossRef]
  27. Asseri, A.A. Serum Vitamin D Profiles of Children with Asthma in Southwest Saudi: A Comparative Cross-Sectional Study. Int. J. Gen. Med. 2024, 17, 6323–6333. [Google Scholar] [CrossRef]
  28. Zanchetta, M.B.; Costa, A.F.; Longobardi, V.; Mazure, R.; Silveira, F.; Temprano, M.P.; Vázquez, H.; Bogado, C.; Niveloni, S.I.; Smecuol, E.; et al. Improved Bone Microarchitecture in Patients With Celiac Disease After 3 Years on a Gluten-Free Diet. Clin. Gastroenterol. Hepatol. 2018, 16, 774–775. [Google Scholar] [CrossRef]
  29. Galli, G.; Carabotti, M.; Conti, L.; Scalamonti, S.; Annibale, B.; Lahner, E. Comparison of Clinical, Biochemical and Histological Features between Adult Celiac Patients with High and Low Anti-Transglutaminase IgA Titer at Diagnosis and Follow-Up. Nutrients 2023, 15, 2151. [Google Scholar] [CrossRef]
  30. Ganji, R.; Moghbeli, M.; Sadeghi, R.; Bayat, G.; Ganji, A. Prevalence of osteoporosis and osteopenia in men and premenopausal women with celiac disease: A systematic review. Nutr. J. 2019, 18, 9. [Google Scholar] [CrossRef]
  31. Ganji, A.; Moghbeli, M.; Moradi, Y.; Babaei, N.; Baniasad, A. Bone Loss Correlated with Parathyroid Hormone Levels in Adult Celiac Patients. Middle East J. Dig. Dis. 2022, 14, 103–109. [Google Scholar] [CrossRef] [PubMed]
  32. Walker, M.D.; Williams, J.; Lewis, S.K.; Bai, J.C.; Lebwohl, B.; Green, P.H.R. Measurement of Forearm Bone Density by Dual Energy X-Ray Absorptiometry Increases the Prevalence of Osteoporosis in Men With Celiac Disease. Clin. Gastroenterol. Hepatol. 2020, 18, 99–106. [Google Scholar] [CrossRef] [PubMed]
  33. Sayar, S.; Aykut, H.; Kaya, Ö.; Kürbüz, K.; Ak, Ç.; Gökçen, P.; Mutlu Bilgiç, N.; Adalı, G.; Kahraman, R.; Doganay, L.; et al. Bone Mineral Density Screening and the Frequency of Osteopenia/Osteoporosis in Turkish Adult Patients With Celiac Disease. Turk. J. Gastroenterol. 2021, 32, 600–607. [Google Scholar] [CrossRef] [PubMed]
  34. Kinga, S.; Marciniak, M.D.; Michalak, M.; Zawada, A.; Ratajczak-Pawłowska, A.E.; Dobrowolska, A.; Krela-Kaźmierczak, I. The other side of celiac disease—Assessment of bone mineral density and body composition in patients with celiac disease. Gastroenterol. Rev. Przegląd Gastroenterol. 2024, 19, 434–438. [Google Scholar] [CrossRef]
  35. Weinstock, L.B.; Walters, A.S.; Mullin, G.E.; Duntley, S.P. Celiac disease is associated with restless legs syndrome. Dig. Dis. Sci. 2010, 55, 1667–1673. [Google Scholar] [CrossRef]
  36. Manchanda, S.; Davies, C.R.; Picchietti, D. Celiac disease as a possible cause for low serum ferritin in patients with restless legs syndrome. Sleep. Med. 2009, 10, 763–765. [Google Scholar] [CrossRef]
  37. Rodrigo, L.; Hernandez-Lahoz, C.; Lauret, E.; Rodriguez-Pelaez, M.; Soucek, M.; Ciccocioppo, R.; Kruzliak, P. Gluten ataxia is better classified as non-celiac gluten sensitivity than as celiac disease: A comparative clinical study. Immunol. Res. 2016, 64, 558–564. [Google Scholar] [CrossRef]
  38. Isikay, S.; Hizli, S.; Coskun, S.; Yilmaz, K. Increased tissue transglutaminase levels are associated with increased epileptiform activity in electroencephalography among patients with celiac disease. Arq. Gastroenterol. 2015, 52, 272–277. [Google Scholar] [CrossRef]
  39. Smith, D.F.; Gerdes, L.U. Meta-analysis on anxiety and depression in adult celiac disease. Acta Psychiatr. Scand. 2012, 125, 189–193. [Google Scholar] [CrossRef]
  40. Chen, S.J.; Chao, Y.L.; Chen, C.Y.; Chang, C.M.; Wu, E.C.; Wu, C.S.; Yeh, H.H.; Chen, C.H.; Tsai, H.J. Prevalence of autoimmune diseases in in-patients with schizophrenia: Nationwide population-based study. Br. J. Psychiatry 2012, 200, 374–380. [Google Scholar] [CrossRef]
  41. Ludvigsson, J.F.; Osby, U.; Ekbom, A.; Montgomery, S.M. Coeliac disease and risk of schizophrenia and other psychosis: A general population cohort study. Scand. J. Gastroenterol. 2007, 42, 179–185. [Google Scholar] [CrossRef] [PubMed]
  42. Garud, S.; Leffler, D.; Dennis, M.; Edwards-George, J.; Saryan, D.; Sheth, S.; Schuppan, D.; Jamma, S.; Kelly, C.P. Interaction between psychiatric and autoimmune disorders in coeliac disease patients in the Northeastern United States. Aliment. Pharmacol. Ther. 2009, 29, 898–905. [Google Scholar] [CrossRef] [PubMed]
  43. Porcelli, B.; Verdino, V.; Bossini, L.; Terzuoli, L.; Fagiolini, A. Celiac and non-celiac gluten sensitivity: A review on the association with schizophrenia and mood disorders. Auto. Immun. Highlights 2014, 5, 55–61. [Google Scholar] [CrossRef] [PubMed]
  44. Pedretti, M.; Sbravati, F.; Allegri, D.; Labriola, F.; Lombardo, V.; Spisni, E.; Zarbo, C.; Alvisi, P. Is the clinical pattern of pediatric celiac disease changing? A thirty-years real-life experience of an Italian center. Ital. J. Pediatr. 2021, 47, 235. [Google Scholar] [CrossRef]
  45. Popp, A.; Mäki, M. Changing Pattern of Childhood Celiac Disease Epidemiology: Contributing Factors. Front. Pediatr. 2019, 7, 357. [Google Scholar] [CrossRef]
  46. Bianchi, P.I.; Lenti, M.V.; Petrucci, C.; Gambini, G.; Aronico, N.; Varallo, M.; Rossi, C.M.; Pozzi, E.; Groppali, E.; Siccardo, F.; et al. Diagnostic Delay of Celiac Disease in Childhood. JAMA Netw. Open 2024, 7, e245671. [Google Scholar] [CrossRef]
  47. Smith, L.B.; Lynch, K.F.; Kurppa, K.; Koletzko, S.; Krischer, J.; Liu, E.; Johnson, S.B.; Agardh, D. Psychological Manifestations of Celiac Disease Autoimmunity in Young Children. Pediatrics 2017, 139, e20162848. [Google Scholar] [CrossRef]
  48. Iwańczak, F.; Iwańczak, B. New guidelines for diagnosis and treatment of coeliac disease in children and adolescents. Gastroenterol. Rev. Przegląd Gastroenterol. 2012, 7, 185–191. [Google Scholar] [CrossRef]
  49. Al-Toma, A.; Volta, U.; Auricchio, R.; Castillejo, G.; Sanders, D.S.; Cellier, C.; Mulder, C.J.; Lundin, K.E.A. European Society for the Study of Coeliac Disease (ESsCD) guideline for coeliac disease and other gluten-related disorders. United Eur. Gastroenterol. J. 2019, 7, 583–613. [Google Scholar] [CrossRef]
  50. Villanacci, V.; Ciacci, C.; Salviato, T.; Leoncini, G.; Bonetti, L.R.; Ragazzini, T.; Limarzi, F.; Saragoni, L. Histopathology of Celiac Disease. Position Statements of the Italian Group of Gastrointestinal Pathologists (GIPAD-SIAPEC). Transl. Med. UniSa 2020, 23, 28–36. [Google Scholar] [CrossRef]
  51. Green, P.H.R.; Paski, S.; Ko, C.W.; Rubio-Tapia, A. AGA Clinical Practice Update on Management of Refractory Celiac Disease: Expert Review. Gastroenterology 2022, 163, 1461–1469. [Google Scholar] [CrossRef] [PubMed]
  52. Ondrejka, S.; Jagadeesh, D. Enteropathy-Associated T-Cell Lymphoma. Curr. Hematol. Malig. Rep. 2016, 11, 504–513. [Google Scholar] [CrossRef] [PubMed]
  53. Olaussen, R.W.; Løvik, A.; Tollefsen, S.; Andresen, P.A.; Vatn, M.H.; De Lange, T.; Bratlie, J.; Brandtzaeg, P.; Farstad, I.N.; Lundin, K.E. Effect of elemental diet on mucosal immunopathology and clinical symptoms in type 1 refractory celiac disease. Clin. Gastroenterol. Hepatol. 2005, 3, 875–885. [Google Scholar] [CrossRef]
  54. Demiroren, K. Possible relationship between refractory celiac disease and malignancies. World J. Clin. Oncol. 2022, 13, 200–208. [Google Scholar] [CrossRef]
  55. Daum, S.; Cellier, C.; Mulder, C.J. Refractory coeliac disease. Best. Pract. Res. Clin. Gastroenterol. 2005, 19, 413–424. [Google Scholar] [CrossRef]
  56. van Wanrooij, R.L.; Bouma, G.; Bontkes, H.J.; Neefjes-Borst, A.; van Grieken, N.C.; von Blomberg, B.M.; Mulder, C.J. Outcome of Referrals for Non-Responsive Celiac Disease in a Tertiary Center: Low Incidence of Refractory Celiac Disease in the Netherlands. Clin. Transl. Gastroenterol. 2017, 8, e218. [Google Scholar] [CrossRef]
  57. Bai, J.C.; Ciacci, C. World Gastroenterology Organisation Global Guidelines: Celiac Disease February 2017. J. Clin. Gastroenterol. 2017, 51, 755–768. [Google Scholar] [CrossRef]
  58. Parfenov, A.I.; Bykova, S.V.; Sabel’nikova, E.A.; Maev, I.V.; Baranov, A.A.; Bakulin, I.G.; Krums, L.M.; Bel’mer, S.V.; Borovik, T.E.; Zakharova, I.N.; et al. All-Russian Consensus on Diagnosis and Treatment of Celiac Disease in Children and Adults. Ter. Arkh. 2017, 89, 94–107. [Google Scholar] [CrossRef]
  59. Downey, L.; Houten, R.; Murch, S.; Longson, D. Recognition, assessment, and management of coeliac disease: Summary of updated NICE guidance. BMJ 2015, 351, h4513. [Google Scholar] [CrossRef]
  60. Ludvigsson, J.F.; Bai, J.C.; Biagi, F.; Card, T.R.; Ciacci, C.; Ciclitira, P.J.; Green, P.H.; Hadjivassiliou, M.; Holdoway, A.; van Heel, D.A.; et al. Diagnosis and management of adult coeliac disease: Guidelines from the British Society of Gastroenterology. Gut 2014, 63, 1210–1228. [Google Scholar] [CrossRef]
  61. Rubio-Tapia, A.; Hill, I.D.; Kelly, C.P.; Calderwood, A.H.; Murray, J.A. ACG clinical guidelines: Diagnosis and management of celiac disease. Am. J. Gastroenterol. 2013, 108, 656–676, quiz 677. [Google Scholar] [CrossRef] [PubMed]
  62. Remes-Troche, J.M.; Uscanga-Domínguez, L.F.; Aceves-Tavares, R.G.; Calderón de la Barca, A.M.; Carmona-Sánchez, R.I.; Cerda-Contreras, E.; Coss-Adame, E.; Icaza-Chávez, M.E.; Lopéz-Colombo, A.; Milke-García, M.P.; et al. Clinical guidelines on the diagnosis and treatment of celiac disease in Mexico. Rev. Gastroenterol. México 2018, 83, 434–450. [Google Scholar] [CrossRef] [PubMed]
  63. Raiteri, A.; Granito, A.; Giamperoli, A.; Catenaro, T.; Negrini, G.; Tovoli, F. Current guidelines for the management of celiac disease: A systematic review with comparative analysis. World J. Gastroenterol. 2022, 28, 154–175. [Google Scholar] [CrossRef] [PubMed]
  64. Maki, M.; Mustalahti, K.; Kokkonen, J.; Kulmala, P.; Haapalahti, M.; Karttunen, T.; Ilonen, J.; Laurila, K.; Dahlbom, I.; Hansson, T.; et al. Prevalence of Celiac disease among children in Finland. N. Engl. J. Med. 2003, 348, 2517–2524. [Google Scholar] [CrossRef]
  65. Laass, M.W.; Schmitz, R.; Uhlig, H.H.; Zimmer, K.P.; Thamm, M.; Koletzko, S. The prevalence of celiac disease in children and adolescents in Germany. Dtsch. Arztebl. Int. 2015, 112, 553–560. [Google Scholar] [CrossRef]
  66. Singh, P.; Arora, S.; Singh, A.; Strand, T.A.; Makharia, G.K. Prevalence of Celiac disease in Asia: A systematic review and meta-analysis. J. Gastroenterol. Hepatol. 2015, 31, 1095–1101. [Google Scholar] [CrossRef]
  67. Choung, R.S.; Ditah, I.C.; Nadeau, A.M.; Rubio-Tapia, A.; Marietta, E.V.; Brantner, T.L.; Camilleri, M.J.; Rajkumar, S.V.; Landgren, O.; Everhart, J.E.; et al. Trends and racial/ethnic disparities in gluten-sensitive problems in the United States: Findings from the National Health and Nutrition Examination Surveys from 1988 to 2012. Am. J. Gastroenterol. 2015, 110, 455–461. [Google Scholar] [CrossRef]
  68. Fasano, A.; Berti, I.; Gerarduzzi, T.; Not, T.; Colletti, R.B.; Drago, S.; Elitsur, Y.; Green, P.H.; Guandalini, S.; Hill, I.D.; et al. Prevalence of celiac disease in at-risk and not-at-risk groups in the United States: A large multicenter study. Arch. Intern. Med. 2003, 163, 286–292. [Google Scholar] [CrossRef]
  69. Singh, P.; Arora, S.; Lal, S.; Strand, T.A.; Makharia, G.K. Risk of Celiac Disease in the First- and Second-Degree Relatives of Patients with Celiac Disease: A Systematic Review and Meta-Analysis. Am. J. Gastroenterol. 2015, 110, 1539–1548. [Google Scholar] [CrossRef]
  70. Sahin, Y.; Mermer, S. Frequency of celiac disease and distribution of HLA-DQ2/DQ8 haplotypes among siblings of children with celiac disease. World J. Clin. Pediatr. 2022, 11, 351–359. [Google Scholar] [CrossRef]
  71. Mustalahti, K.; Catassi, C.; Reunanen, A.; Fabiani, E.; Heier, M.; McMillan, S.; Murray, L.; Metzger, M.H.; Gasparin, M.; Bravi, E.; et al. The prevalence of celiac disease in Europe: Results of a centralized, international mass screening project. Ann. Med. 2010, 42, 587–595. [Google Scholar] [CrossRef] [PubMed]
  72. Lionetti, E.; Gatti, S.; Pulvirenti, A.; Catassi, C. Celiac disease from a global perspective. Best. Pract. Res. Clin. Gastroenterol. 2015, 29, 365–379. [Google Scholar] [CrossRef] [PubMed]
  73. Singh, P.; Arora, A.; Strand, T.A.; Leffler, D.A.; Catassi, C.; Green, P.H.; Kelly, C.P.; Ahuja, V.; Makharia, G.K. Global Prevalence of Celiac Disease: Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. 2018, 16, 823–836.e2. [Google Scholar] [CrossRef] [PubMed]
  74. El-Metwally, A.; Toivola, P.; AlAhmary, K.; Bahkali, S.; AlKhathaami, A.; AlSaqabi, M.K.; Al Ammar, S.A.; Jawed, M.; Alosaimi, S.M. The Epidemiology of Celiac Disease in the General Population and High-Risk Groups in Arab Countries: A Systematic Review. Biomed. Res. Int. 2020, 2020, 6865917. [Google Scholar] [CrossRef]
  75. Tamai, T.; Ihara, K. Celiac Disease Genetics, Pathogenesis, and Standard Therapy for Japanese Patients. Int. J. Mol. Sci. 2023, 24, 2075. [Google Scholar] [CrossRef]
  76. Poddighe, D.; Abdukhakimova, D. Celiac Disease in Asia beyond the Middle East and Indian subcontinent: Epidemiological burden and diagnostic barriers. World J. Gastroenterol. 2021, 27, 2251–2256. [Google Scholar] [CrossRef]
  77. Ashtari, S.; Najafimehr, H.; Pourhoseingholi, M.A.; Rostami, K.; Asadzadeh-Aghdaei, H.; Rostami-Nejad, M.; Tavirani, M.R.; Olfatifar, M.; Makharia, G.K.; Zali, M.R. Prevalence of celiac disease in low and high risk population in Asia-Pacific region: A systematic review and meta-analysis. Sci. Rep. 2021, 11, 2383. [Google Scholar] [CrossRef]
  78. Ludvigsson, J.F.; Card, T.R.; Kaukinen, K.; Bai, J.; Zingone, F.; Sanders, D.S.; Murray, J.A. Screening for celiac disease in the general population and in high-risk groups. United Eur. Gastroenterol. J. 2015, 3, 106–120. [Google Scholar] [CrossRef]
  79. Parra-Medina, R.; Molano-Gonzalez, N.; Rojas-Villarraga, A.; Agmon-Levin, N.; Arango, M.T.; Shoenfeld, Y.; Anaya, J.M. Prevalence of celiac disease in latin america: A systematic review and meta-regression. PLoS ONE 2015, 10, e0124040. [Google Scholar] [CrossRef]
  80. Savvateeva, L.V.; Erdes, S.I.; Antishin, A.S.; Zamyatnin, A.A., Jr. Overview of Celiac Disease in Russia: Regional Data and Estimated Prevalence. J. Immunol. Res. 2017, 2017, 2314813. [Google Scholar] [CrossRef]
  81. Zhou, W.Y.; Liu, X.Y.; Wang, M.M.; Liang, L.P.; Liu, L.; Zheng, K.; Silvester, J.A.; Ma, W.J.; Wu, W.; Ji, G.Y.; et al. Prevalence of celiac disease in China: Meta-analysis and serological survey in high-risk populations. J. Dig. Dis. 2021, 22, 645–655. [Google Scholar] [CrossRef] [PubMed]
  82. Makharia, G.K.; Verma, A.K.; Amarchand, R.; Bhatnagar, S.; Das, P.; Goswami, A.; Bhatia, V.; Ahuja, V.; Datta Gupta, S.; Anand, K. Prevalence of celiac disease in the northern part of India: A community based study. J. Gastroenterol. Hepatol. 2011, 26, 894–900. [Google Scholar] [CrossRef]
  83. Chin, M.W.; Mallon, D.F.; Cullen, D.J.; Olynyk, J.K.; Mollison, L.C.; Pearce, C.B. Screening for coeliac disease using anti-tissue transglutaminase antibody assays, and prevalence of the disease in an Australian community. Med. J. Aust. 2009, 190, 429–432. [Google Scholar] [CrossRef] [PubMed]
  84. Dehghani, S.M.; Haghighat, M.; Mobayen, A.; Rezaianzadeh, A.; Geramizadeh, B. Prevalence of celiac disease in healthy Iranian school children. Ann. Saudi Med. 2013, 33, 159–161. [Google Scholar] [CrossRef]
  85. Al-Ajlan, A.S. Screening of coeliac disease in undetected adults and patients diagnosed with irritable bowel syndrome in Riyadh, Saudi Arabia. Saudi J. Biol. Sci. 2016, 23, 462–466. [Google Scholar] [CrossRef]
  86. Lechtman, N.; Shamir, R.; Cohen, S.; Chodick, G.; Kariv, R.; Supino-Rosin, L.; Weintraub, Y.; Yerushalmy-Feler, A.; Ben Tov, A. Increased incidence of coeliac disease autoimmunity rate in Israel: A 9-year analysis of population-based data. Aliment. Pharmacol. Ther. 2021, 53, 696–703. [Google Scholar] [CrossRef]
  87. Mankaï, A.; Landolsi, H.; Chahed, A.; Gueddah, L.; Limem, M.; Ben Abdessalem, M.; Yacoub-Jemni, S.; Ghannem, H.; Jeddi, M.; Ghedira, I. Celiac disease in Tunisia: Serological screening in healthy blood donors. Pathol. Biol. 2006, 54, 10–13. [Google Scholar] [CrossRef]
  88. Abu-Zekry, M.; Kryszak, D.; Diab, M.; Catassi, C.; Fasano, A. Prevalence of celiac disease in Egyptian children disputes the east-west agriculture-dependent spread of the disease. J. Pediatr. Gastroenterol. Nutr. 2008, 47, 136–140. [Google Scholar] [CrossRef]
  89. Teresi, S.; Crapisi, M.; Vallejo, M.D.; Castellaneta, S.P.; Francavilla, R.; Iacono, G.; Ravelli, A.; Menegazzi, P.; Louali, M.; Catassi, C. Celiac disease seropositivity in Saharawi children: A follow-up and family study. J. Pediatr. Gastroenterol. Nutr. 2010, 50, 506–509. [Google Scholar] [CrossRef]
  90. Aboulaghras, S.; Piancatelli, D.; Taghzouti, K.; Balahbib, A.; Alshahrani, M.M.; Al Awadh, A.A.; Goh, K.W.; Ming, L.C.; Bouyahya, A.; Oumhani, K. Meta-Analysis and Systematic Review of HLA DQ2/DQ8 in Adults with Celiac Disease. Int. J. Mol. Sci. 2023, 24, 1188. [Google Scholar] [CrossRef]
  91. Comino, I.; Moreno Mde, L.; Sousa, C. Role of oats in celiac disease. World J. Gastroenterol. 2015, 21, 11825–11831. [Google Scholar] [CrossRef] [PubMed]
  92. Pulido, O.M.; Gillespie, Z.; Zarkadas, M.; Dubois, S.; Vavasour, E.; Rashid, M.; Switzer, C.; Godefroy, S.B. Introduction of oats in the diet of individuals with celiac disease: A systematic review. Adv. Food Nutr. Res. 2009, 57, 235–285. [Google Scholar] [CrossRef] [PubMed]
  93. La Vieille, S.; Pulido, O.M.; Abbott, M.; Koerner, T.B.; Godefroy, S. Celiac Disease and Gluten-Free Oats: A Canadian Position Based on a Literature Review. Can. J. Gastroenterol. Hepatol. 2016, 2016, 1870305. [Google Scholar] [CrossRef] [PubMed]
  94. Aboulaghras, S.; Piancatelli, D.; Taghzouti, K.; Balahbib, A.; Alshahrani, M.M.; Al Awadh, A.A.; Goh, K.W.; Ming, L.C.; Bouyahya, A.; Oumhani, K. Safety of Adding Oats to a Gluten-Free Diet for Patients with Celiac Disease: Systematic Review and Meta-analysis of Clinical and Observational Studies. Gastroenterology 2017, 153, 395–409. [Google Scholar] [CrossRef]
  95. Lionetti, E.; Gatti, S.; Galeazzi, T.; Caporelli, N.; Francavilla, R.; Cucchiara, S.; Roggero, P.; Malamisura, B.; Iacono, G.; Tomarchio, S.; et al. Safety of Oats in Children with Celiac Disease: A Double-Blind, Randomized, Placebo-Controlled Trial. J. Pediatr. 2018, 194, 116–122.e2. [Google Scholar] [CrossRef]
  96. Tanner, G.; Juhász, A.; Florides, C.G.; Nye-Wood, M.; Békés, F.; Colgrave, M.L.; Russell, A.K.; Hardy, M.Y.; Tye-Din, J.A. Preparation and Characterization of Avenin-Enriched Oat Protein by Chill Precipitation for Feeding Trials in Celiac Disease. Front. Nutr. 2019, 6, 162. [Google Scholar] [CrossRef]
  97. Palova-Jelinkova, L.; Danova, K.; Drasarova, H.; Dvorak, M.; Funda, D.P.; Fundova, P.; Kotrbova-Kozak, A.; Cerna, M.; Kamanova, J.; Martin, S.F.; et al. Pepsin digest of wheat gliadin fraction increases production of IL-1beta via TLR4/MyD88/TRIF/MAPK/NF-kappaB signaling pathway and an NLRP3 inflammasome activation. PLoS ONE 2013, 8, e62426. [Google Scholar] [CrossRef]
  98. Garcia-Horsman, J.A.; Venalainen, J.I.; Lohi, O.; Auriola, I.S.; Korponay-Szabo, I.R.; Kaukinen, K.; Maki, M.; Mannisto, P.T. Deficient activity of mammalian prolyl oligopeptidase on the immunoactive peptide digestion in coeliac disease. Scand. J. Gastroenterol. 2007, 42, 562–571. [Google Scholar] [CrossRef]
  99. Matysiak-Budnik, T.; Candalh, C.; Cellier, C.; Dugave, C.; Namane, A.; Vidal-Martinez, T.; Cerf-Bensussan, N.; Heyman, M. Limited efficiency of prolyl-endopeptidase in the detoxification of gliadin peptides in celiac disease. Gastroenterology 2005, 129, 786–796. [Google Scholar] [CrossRef]
  100. Wei, G.; Helmerhorst, E.J.; Darwish, G.; Blumenkranz, G.; Schuppan, D. Gluten Degrading Enzymes for Treatment of Celiac Disease. Nutrients 2020, 12, 2095. [Google Scholar] [CrossRef]
  101. Dunaevsky, Y.E.; Tereshchenkova, V.F.; Belozersky, M.A.; Filippova, I.Y.; Oppert, B.; Elpidina, E.N. Effective Degradation of Gluten and Its Fragments by Gluten-Specific Peptidases: A Review on Application for the Treatment of Patients with Gluten Sensitivity. Pharmaceutics 2021, 13, 1603. [Google Scholar] [CrossRef] [PubMed]
  102. Nistal, E.; Caminero, A.; Vivas, S.; Ruiz de Morales, J.M.; Saenz de Miera, L.E.; Rodriguez-Aparicio, L.B.; Casqueiro, J. Differences in faecal bacteria populations and faecal bacteria metabolism in healthy adults and celiac disease patients. Biochimie 2012, 94, 1724–1729. [Google Scholar] [CrossRef] [PubMed]
  103. Sanz, Y. Effects of a gluten-free diet on gut microbiota and immune function in healthy adult humans. Gut Microbes 2010, 1, 135–137. [Google Scholar] [CrossRef]
  104. De Palma, G.; Nadal, I.; Collado, M.C.; Sanz, Y. Effects of a gluten-free diet on gut microbiota and immune function in healthy adult human subjects. Br. J. Nutr. 2009, 102, 1154–1160. [Google Scholar] [CrossRef] [PubMed]
  105. Di Cagno, R.; Rizzello, C.G.; Gagliardi, F.; Ricciuti, P.; Ndagijimana, M.; Francavilla, R.; Guerzoni, M.E.; Crecchio, C.; Gobbetti, M.; De Angelis, M. Different fecal microbiotas and volatile organic compounds in treated and untreated children with celiac disease. Appl. Environ. Microbiol. 2009, 75, 3963–3971. [Google Scholar] [CrossRef]
  106. Bonder, M.J.; Tigchelaar, E.F.; Cai, X.; Trynka, G.; Cenit, M.C.; Hrdlickova, B.; Zhong, H.; Vatanen, T.; Gevers, D.; Wijmenga, C.; et al. The influence of a short-term gluten-free diet on the human gut microbiome. Genome Med. 2016, 8, 45. [Google Scholar] [CrossRef]
  107. Szczuciński, W.; Salamon, D.; Sopel, A.; Gosiewski, T. Celiac disease and human gut microbiota—How can we study the composition of microorganisms? Gastroenterol. Rev. Przegląd Gastroenterol. 2024, 19, 139574. [Google Scholar] [CrossRef]
  108. Caminero, A.; Nistal, E.; Herran, A.R.; Perez-Andres, J.; Ferrero, M.A.; Vaquero Ayala, L.; Vivas, S.; Ruiz de Morales, J.M.; Albillos, S.M.; Casqueiro, F.J. Differences in gluten metabolism among healthy volunteers, coeliac disease patients and first-degree relatives. Br. J. Nutr. 2015, 114, 1157–1167. [Google Scholar] [CrossRef]
  109. Zamakhchari, M.; Wei, G.; Dewhirst, F.; Lee, J.; Schuppan, D.; Oppenheim, F.G.; Helmerhorst, E.J. Identification of Rothia bacteria as gluten-degrading natural colonizers of the upper gastro-intestinal tract. PLoS ONE 2011, 6, e24455. [Google Scholar] [CrossRef]
  110. Helmerhorst, E.J.; Zamakhchari, M.; Schuppan, D.; Oppenheim, F.G. Discovery of a novel and rich source of gluten-degrading microbial enzymes in the oral cavity. PLoS ONE 2010, 5, e13264. [Google Scholar] [CrossRef]
  111. Stepniak, D.; Spaenij-Dekking, L.; Mitea, C.; Moester, M.; de Ru, A.; Baak-Pablo, R.; van Veelen, P.; Edens, L.; Koning, F. Highly efficient gluten degradation with a newly identified prolyl endoprotease: Implications for celiac disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2006, 291, G621–G629. [Google Scholar] [CrossRef] [PubMed]
  112. Shan, L.; Marti, T.; Sollid, L.M.; Gray, G.M.; Khosla, C. Comparative biochemical analysis of three bacterial prolyl endopeptidases: Implications for coeliac sprue. Biochem. J. 2004, 383 Pt 2, 311–318. [Google Scholar] [CrossRef] [PubMed]
  113. Yoshimoto, T.; Walter, R.; Tsuru, D. Proline-specific endopeptidase from Flavobacterium. Purification and properties. J. Biol. Chem. 1980, 255, 4786–4792. [Google Scholar] [CrossRef] [PubMed]
  114. Meresse, B.; Chen, Z.; Ciszewski, C.; Tretiakova, M.; Bhagat, G.; Krausz, T.N.; Raulet, D.H.; Lanier, L.L.; Groh, V.; Spies, T.; et al. Coordinated induction by IL15 of a TCR-independent NKG2D signaling pathway converts CTL into lymphokine-activated killer cells in celiac disease. Immunity 2004, 21, 357–366. [Google Scholar] [CrossRef]
  115. Londei, M.; Ciacci, C.; Ricciardelli, I.; Vacca, L.; Quaratino, S.; Maiuri, L. Gliadin as a stimulator of innate responses in celiac disease. Mol. Immunol. 2005, 42, 913–918. [Google Scholar] [CrossRef]
  116. Maiuri, L.; Ciacci, C.; Ricciardelli, I.; Vacca, L.; Raia, V.; Auricchio, S.; Picard, J.; Osman, M.; Quaratino, S.; Londei, M. Association between innate response to gliadin and activation of pathogenic T cells in coeliac disease. Lancet 2003, 362, 30–37. [Google Scholar] [CrossRef]
  117. Andre, P.; Castriconi, R.; Espeli, M.; Anfossi, N.; Juarez, T.; Hue, S.; Conway, H.; Romagne, F.; Dondero, A.; Nanni, M.; et al. Comparative analysis of human NK cell activation induced by NKG2D and natural cytotoxicity receptors. Eur. J. Immunol. 2004, 34, 961–971. [Google Scholar] [CrossRef]
  118. Hue, S.; Mention, J.J.; Monteiro, R.C.; Zhang, S.; Cellier, C.; Schmitz, J.; Verkarre, V.; Fodil, N.; Bahram, S.; Cerf-Bensussan, N.; et al. A direct role for NKG2D/MICA interaction in villous atrophy during celiac disease. Immunity 2004, 21, 367–377. [Google Scholar] [CrossRef]
  119. Vilasi, S.; Sirangelo, I.; Irace, G.; Caputo, I.; Barone, M.V.; Esposito, C.; Ragone, R. Interaction of ‘toxic’ and ‘immunogenic’ A-gliadin peptides with a membrane-mimetic environment. J. Mol. Recognit. 2010, 23, 322–328. [Google Scholar] [CrossRef]
  120. Nanayakkara, M.; Kosova, R.; Lania, G.; Sarno, M.; Gaito, A.; Galatola, M.; Greco, L.; Cuomo, M.; Troncone, R.; Auricchio, S.; et al. A celiac cellular phenotype, with altered LPP sub-cellular distribution, is inducible in controls by the toxic gliadin peptide P31-43. PLoS ONE 2013, 8, e79763. [Google Scholar] [CrossRef]
  121. Barone, M.V.; Gimigliano, A.; Castoria, G.; Paolella, G.; Maurano, F.; Paparo, F.; Maglio, M.; Mineo, A.; Miele, E.; Nanayakkara, M.; et al. Growth factor-like activity of gliadin, an alimentary protein: Implications for coeliac disease. Gut 2007, 56, 480–488. [Google Scholar] [CrossRef] [PubMed]
  122. Giovannini, C.; Matarrese, P.; Scazzocchio, B.; Vari, R.; D’Archivio, M.; Straface, E.; Masella, R.; Malorni, W.; De Vincenzi, M. Wheat gliadin induces apoptosis of intestinal cells via an autocrine mechanism involving Fas-Fas ligand pathway. FEBS Lett. 2003, 540, 117–124. [Google Scholar] [CrossRef] [PubMed]
  123. Menard, S.; Lebreton, C.; Schumann, M.; Matysiak-Budnik, T.; Dugave, C.; Bouhnik, Y.; Malamut, G.; Cellier, C.; Allez, M.; Crenn, P.; et al. Paracellular versus transcellular intestinal permeability to gliadin peptides in active celiac disease. Am. J. Pathol. 2012, 180, 608–615. [Google Scholar] [CrossRef] [PubMed]
  124. Lammers, K.M.; Lu, R.; Brownley, J.; Lu, B.; Gerard, C.; Thomas, K.; Rallabhandi, P.; Shea-Donohue, T.; Tamiz, A.; Alkan, S.; et al. Gliadin induces an increase in intestinal permeability and zonulin release by binding to the chemokine receptor CXCR3. Gastroenterology 2008, 135, 194–204.e3. [Google Scholar] [CrossRef]
  125. Clemente, M.G.; De Virgiliis, S.; Kang, J.S.; Macatagney, R.; Musu, M.P.; Di Pierro, M.R.; Drago, S.; Congia, M.; Fasano, A. Early effects of gliadin on enterocyte intracellular signalling involved in intestinal barrier function. Gut 2003, 52, 218–223. [Google Scholar] [CrossRef]
  126. Drago, S.; El Asmar, R.; Di Pierro, M.; Grazia Clemente, M.; Tripathi, A.; Sapone, A.; Thakar, M.; Iacono, G.; Carroccio, A.; D’Agate, C.; et al. Gliadin, zonulin and gut permeability: Effects on celiac and non-celiac intestinal mucosa and intestinal cell lines. Scand. J. Gastroenterol. 2006, 41, 408–419. [Google Scholar] [CrossRef]
  127. Fasano, A.; Not, T.; Wang, W.; Uzzau, S.; Berti, I.; Tommasini, A.; Goldblum, S.E. Zonulin, a newly discovered modulator of intestinal permeability, and its expression in coeliac disease. Lancet 2000, 355, 1518–1519. [Google Scholar] [CrossRef]
  128. Tripathi, A.; Lammers, K.M.; Goldblum, S.; Shea-Donohue, T.; Netzel-Arnett, S.; Buzza, M.S.; Antalis, T.M.; Vogel, S.N.; Zhao, A.; Yang, S.; et al. Identification of human zonulin, a physiological modulator of tight junctions, as prehaptoglobin-2. Proc. Natl. Acad. Sci. USA 2009, 106, 16799–16804. [Google Scholar] [CrossRef]
  129. Sapone, A.; de Magistris, L.; Pietzak, M.; Clemente, M.G.; Tripathi, A.; Cucca, F.; Lampis, R.; Kryszak, D.; Carteni, M.; Generoso, M.; et al. Zonulin upregulation is associated with increased gut permeability in subjects with type 1 diabetes and their relatives. Diabetes 2006, 55, 1443–1449. [Google Scholar] [CrossRef]
  130. El Asmar, R.; Panigrahi, P.; Bamford, P.; Berti, I.; Not, T.; Coppa, G.V.; Catassi, C.; Fasano, A. Host-dependent zonulin secretion causes the impairment of the small intestine barrier function after bacterial exposure. Gastroenterology 2002, 123, 1607–1615. [Google Scholar] [CrossRef]
  131. Wang, W.; Uzzau, S.; Goldblum, S.E.; Fasano, A. Human zonulin, a potential modulator of intestinal tight junctions. J. Cell Sci. 2000, 113 Pt 24, 4435–4440. [Google Scholar] [CrossRef] [PubMed]
  132. Lindfors, K.; Blomqvist, T.; Juuti-Uusitalo, K.; Stenman, S.; Venalainen, J.; Maki, M.; Kaukinen, K. Live probiotic Bifidobacterium lactis bacteria inhibit the toxic effects induced by wheat gliadin in epithelial cell culture. Clin. Exp. Immunol. 2008, 152, 552–558. [Google Scholar] [CrossRef] [PubMed]
  133. Arentz-Hansen, H.; Korner, R.; Molberg, O.; Quarsten, H.; Vader, W.; Kooy, Y.M.; Lundin, K.E.; Koning, F.; Roepstorff, P.; Sollid, L.M.; et al. The intestinal T cell response to alpha-gliadin in adult celiac disease is focused on a single deamidated glutamine targeted by tissue transglutaminase. J. Exp. Med. 2000, 191, 603–612. [Google Scholar] [CrossRef]
  134. Molberg, O.; McAdam, S.; Lundin, K.E.; Kristiansen, C.; Arentz-Hansen, H.; Kett, K.; Sollid, L.M. T cells from celiac disease lesions recognize gliadin epitopes deamidated in situ by endogenous tissue transglutaminase. Eur. J. Immunol. 2001, 31, 1317–1323. [Google Scholar] [CrossRef]
  135. Quarsten, H.; Molberg, O.; Fugger, L.; McAdam, S.N.; Sollid, L.M. HLA binding and T cell recognition of a tissue transglutaminase-modified gliadin epitope. Eur. J. Immunol. 1999, 29, 2506–2514. [Google Scholar] [CrossRef]
  136. Camarca, A.; Anderson, R.P.; Mamone, G.; Fierro, O.; Facchiano, A.; Costantini, S.; Zanzi, D.; Sidney, J.; Auricchio, S.; Sette, A.; et al. Intestinal T cell responses to gluten peptides are largely heterogeneous: Implications for a peptide-based therapy in celiac disease. J. Immunol. 2009, 182, 4158–4166. [Google Scholar] [CrossRef]
  137. Anderson, R.P.; van Heel, D.A.; Tye-Din, J.A.; Barnardo, M.; Salio, M.; Jewell, D.P.; Hill, A.V. T cells in peripheral blood after gluten challenge in coeliac disease. Gut 2005, 54, 1217–1223. [Google Scholar] [CrossRef]
  138. van de Wal, Y.; Kooy, Y.; van Veelen, P.; Pena, S.; Mearin, L.; Papadopoulos, G.; Koning, F. Selective deamidation by tissue transglutaminase strongly enhances gliadin-specific T cell reactivity. J. Immunol. 1998, 161, 1585–1588. [Google Scholar] [CrossRef]
  139. Dorum, S.; Qiao, S.W.; Sollid, L.M.; Fleckenstein, B. A quantitative analysis of transglutaminase 2-mediated deamidation of gluten peptides: Implications for the T-cell response in celiac disease. J. Proteome Res. 2009, 8, 1748–1755. [Google Scholar] [CrossRef]
  140. Qiao, S.W.; Bergseng, E.; Molberg, O.; Jung, G.; Fleckenstein, B.; Sollid, L.M. Refining the rules of gliadin T cell epitope binding to the disease-associated DQ2 molecule in celiac disease: Importance of proline spacing and glutamine deamidation. J. Immunol. 2005, 175, 254–261. [Google Scholar] [CrossRef]
  141. Tollefsen, S.; Arentz-Hansen, H.; Fleckenstein, B.; Molberg, O.; Raki, M.; Kwok, W.W.; Jung, G.; Lundin, K.E.; Sollid, L.M. HLA-DQ2 and -DQ8 signatures of gluten T cell epitopes in celiac disease. J. Clin. Investig. 2006, 116, 2226–2236. [Google Scholar] [CrossRef]
  142. Vader, W.; Stepniak, D.; Kooy, Y.; Mearin, L.; Thompson, A.; van Rood, J.J.; Spaenij, L.; Koning, F. The HLA-DQ2 gene dose effect in celiac disease is directly related to the magnitude and breadth of gluten-specific T cell responses. Proc. Natl. Acad. Sci. USA 2003, 100, 12390–12395. [Google Scholar] [CrossRef] [PubMed]
  143. Henderson, K.N.; Reid, H.H.; Borg, N.A.; Broughton, S.E.; Huyton, T.; Anderson, R.P.; McCluskey, J.; Rossjohn, J. The production and crystallization of the human leukocyte antigen class II molecules HLA-DQ2 and HLA-DQ8 complexed with deamidated gliadin peptides implicated in coeliac disease. Acta Crystallogr. Sect. F Struct. Biol. Cryst. Commun. 2007, 63 Pt 12, 1021–1025. [Google Scholar] [CrossRef] [PubMed]
  144. Korponay-Szabo, I.R.; Vecsei, Z.; Kiraly, R.; Dahlbom, I.; Chirdo, F.; Nemes, E.; Fesus, L.; Maki, M. Deamidated gliadin peptides form epitopes that transglutaminase antibodies recognize. J. Pediatr. Gastroenterol. Nutr. 2008, 46, 253–261. [Google Scholar] [CrossRef] [PubMed]
  145. Vitoria, J.C.; Arrieta, A.; Arranz, C.; Ayesta, A.; Sojo, A.; Maruri, N.; Garcia-Masdevall, M.D. Antibodies to gliadin, endomysium, and tissue transglutaminase for the diagnosis of celiac disease. J. Pediatr. Gastroenterol. Nutr. 1999, 29, 571–574. [Google Scholar]
  146. Aleanzi, M.; Demonte, A.M.; Esper, C.; Garcilazo, S.; Waggener, M. Celiac disease: Antibody recognition against native and selectively deamidated gliadin peptides. Clin. Chem. 2001, 47, 2023–2028. [Google Scholar] [CrossRef]
  147. Agardh, D.; Dahlbom, I.; Daniels, T.; Lorinc, E.; Ivarsson, S.A.; Lernmark, A.; Hansson, T. Autoantibodies against soluble and immobilized human recombinant tissue transglutaminase in children with celiac disease. J. Pediatr. Gastroenterol. Nutr. 2005, 41, 322–327. [Google Scholar] [CrossRef]
  148. Sulkanen, S.; Halttunen, T.; Laurila, K.; Kolho, K.L.; Korponay-Szabo, I.R.; Sarnesto, A.; Savilahti, E.; Collin, P.; Maki, M. Tissue transglutaminase autoantibody enzyme-linked immunosorbent assay in detecting celiac disease. Gastroenterology 1998, 115, 1322–1328. [Google Scholar] [CrossRef]
  149. Bodd, M.; Raki, M.; Tollefsen, S.; Fallang, L.E.; Bergseng, E.; Lundin, K.E.; Sollid, L.M. HLA-DQ2-restricted gluten-reactive T cells produce IL-21 but not IL-17 or IL-22. Mucosal Immunol. 2010, 3, 594–601. [Google Scholar] [CrossRef]
  150. Harris, K.M.; Fasano, A.; Mann, D.L. Monocytes differentiated with IL-15 support Th17 and Th1 responses to wheat gliadin: Implications for celiac disease. Clin. Immunol. 2010, 135, 430–439. [Google Scholar] [CrossRef]
  151. Salvati, V.M.; MacDonald, T.T.; Bajaj-Elliott, M.; Borrelli, M.; Staiano, A.; Auricchio, S.; Troncone, R.; Monteleone, G. Interleukin 18 and associated markers of T helper cell type 1 activity in coeliac disease. Gut 2002, 50, 186–190. [Google Scholar] [CrossRef] [PubMed]
  152. Fina, D.; Sarra, M.; Caruso, R.; Del Vecchio Blanco, G.; Pallone, F.; MacDonald, T.T.; Monteleone, G. Interleukin 21 contributes to the mucosal T helper cell type 1 response in coeliac disease. Gut 2008, 57, 887–892. [Google Scholar] [CrossRef] [PubMed]
  153. Di Sabatino, A.; Pickard, K.M.; Gordon, J.N.; Salvati, V.; Mazzarella, G.; Beattie, R.M.; Vossenkaemper, A.; Rovedatti, L.; Leakey, N.A.; Croft, N.M.; et al. Evidence for the role of interferon-alfa production by dendritic cells in the Th1 response in celiac disease. Gastroenterology 2007, 133, 1175–1187. [Google Scholar] [CrossRef]
  154. Mora, B.; Bonamico, M.; Ferri, M.; Megiorni, F.; Osborn, J.; Pizzuti, A.; Mazzilli, M.C. Association of the matrix metalloproteinase-3 (MMP-3) promoter polymorphism with celiac disease in male subjects. Hum. Immunol. 2005, 66, 716–720. [Google Scholar] [CrossRef]
  155. Ciccocioppo, R.; Di Sabatino, A.; Bauer, M.; Della Riccia, D.N.; Bizzini, F.; Biagi, F.; Cifone, M.G.; Corazza, G.R.; Schuppan, D. Matrix metalloproteinase pattern in celiac duodenal mucosa. Lab. Investig. 2005, 85, 397–407. [Google Scholar] [CrossRef]
  156. Bister, V.; Kolho, K.L.; Karikoski, R.; Westerholm-Ormio, M.; Savilahti, E.; Saarialho-Kere, U. Metalloelastase (MMP-12) is upregulated in the gut of pediatric patients with potential celiac disease and in type 1 diabetes. Scand. J. Gastroenterol. 2005, 40, 1413–1422. [Google Scholar] [CrossRef]
  157. Lammers, K.M.; Khandelwal, S.; Chaudhry, F.; Kryszak, D.; Puppa, E.L.; Casolaro, V.; Fasano, A. Identification of a novel immunomodulatory gliadin peptide that causes interleukin-8 release in a chemokine receptor CXCR3-dependent manner only in patients with coeliac disease. Immunology 2011, 132, 432–440. [Google Scholar] [CrossRef]
  158. Nanayakkara, M.; Lania, G.; Maglio, M.; Discepolo, V.; Sarno, M.; Gaito, A.; Troncone, R.; Auricchio, S.; Auricchio, R.; Barone, M.V. An undigested gliadin peptide activates innate immunity and proliferative signaling in enterocytes: The role in celiac disease. Am. J. Clin. Nutr. 2013, 98, 1123–1135. [Google Scholar] [CrossRef]
  159. Barone, M.V.; Troncone, R.; Auricchio, S. Gliadin peptides as triggers of the proliferative and stress/innate immune response of the celiac small intestinal mucosa. Int. J. Mol. Sci. 2014, 15, 20518–20537. [Google Scholar] [CrossRef]
  160. Volta, U.; Granito, A.; Fiorini, E.; Parisi, C.; Piscaglia, M.; Pappas, G.; Muratori, P.; Bianchi, F.B. Usefulness of antibodies to deamidated gliadin peptides in celiac disease diagnosis and follow-up. Dig. Dis. Sci. 2008, 53, 1582–1588. [Google Scholar] [CrossRef]
  161. Majsiak, E.; Cukrowska, B.; Choina, M.; Bielawski, K.; Cielecka-Kuszyk, J.; Konopka, E.; Wysokiński, M.; Bierła, J.B. Evaluation of the Usefulness of a Serological Test for Diagnosis of Celiac Disease Simultaneously Detecting Specific Antibodies and Total IgA. Nutrients 2022, 15, 202. [Google Scholar] [CrossRef] [PubMed]
  162. Rostom, A.; Dubé, C.; Cranney, A.; Saloojee, N.; Sy, R.; Garritty, C.; Sampson, M.; Zhang, L.; Yazdi, F.; Mamaladze, V.; et al. The diagnostic accuracy of serologic tests for celiac disease: A systematic review. Gastroenterology 2005, 128 (Suppl. S1), S38–S46. [Google Scholar] [CrossRef] [PubMed]
  163. Husby, S.; Koletzko, S.; Korponay-Szabo, I.R.; Mearin, M.L.; Phillips, A.; Shamir, R.; Troncone, R.; Giersiepen, K.; Branski, D.; Catassi, C.; et al. European Society for Pediatric Gastroenterology, Hepatology, and Nutrition guidelines for the diagnosis of coeliac disease. J. Pediatr. Gastroenterol. Nutr. 2012, 54, 136–160. [Google Scholar] [CrossRef] [PubMed]
  164. Konopka, E.; Grzywnowicz, M.; Oralewska, B.; Cielecka-Kuszyk, J.; Trojanowska, I.; Cukrowska, B. Clinical utility of quantitative multi-antibody Polycheck immunoassays in the diagnosis of coeliac disease. World J. Gastrointest. Pharmacol. Ther. 2016, 7, 254–260. [Google Scholar] [CrossRef]
  165. Agardh, D. Antibodies against synthetic deamidated gliadin peptides and tissue transglutaminase for the identification of childhood celiac disease. Clin. Gastroenterol. Hepatol. 2007, 5, 1276–1281. [Google Scholar] [CrossRef]
  166. Giersiepen, K.; Lelgemann, M.; Stuhldreher, N.; Ronfani, L.; Husby, S.; Koletzko, S.; Korponay-Szabo, I.R. Accuracy of diagnostic antibody tests for coeliac disease in children: Summary of an evidence report. J. Pediatr. Gastroenterol. Nutr. 2012, 54, 229–241. [Google Scholar] [CrossRef]
  167. Lytton, S.D.; Antiga, E.; Pfeiffer, S.; Matthias, T.; Szaflarska-Poplawska, A.; Ulaganathan, V.K.; Placek, W.; Fabbri, P.; Hall, R.; Caproni, M. Neo-epitope tissue transglutaminase autoantibodies as a biomarker of the gluten sensitive skin disease—Dermatitis herpetiformis. Clin. Chim. Acta 2013, 415, 346–349. [Google Scholar] [CrossRef]
  168. Lerner, A.; Jeremias, P.; Neidhofer, 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. [Google Scholar] [CrossRef]
  169. Bodd, M.; Raki, M.; Bergseng, E.; Jahnsen, J.; Lundin, K.E.; Sollid, L.M. Direct cloning and tetramer staining to measure the frequency of intestinal gluten-reactive T cells in celiac disease. Eur. J. Immunol. 2013, 43, 2605–2612. [Google Scholar] [CrossRef]
  170. Picascia, S.; Mandile, R.; Auricchio, R.; Troncone, R.; Gianfrani, C. Gliadin-Specific T-Cells Mobilized in the Peripheral Blood of Coeliac Patients by Short Oral Gluten Challenge: Clinical Applications. Nutrients 2015, 7, 10020–10031. [Google Scholar] [CrossRef]
  171. Raki, M.; Fallang, L.E.; Brottveit, M.; Bergseng, E.; Quarsten, H.; Lundin, K.E.; Sollid, L.M. Tetramer visualization of gut-homing gluten-specific T cells in the peripheral blood of celiac disease patients. Proc. Natl. Acad. Sci. USA 2007, 104, 2831–2836. [Google Scholar] [CrossRef] [PubMed]
  172. Brottveit, M.; Raki, M.; Bergseng, E.; Fallang, L.E.; Simonsen, B.; Lovik, A.; Larsen, S.; Loberg, E.M.; Jahnsen, F.L.; Sollid, L.M.; et al. Assessing possible celiac disease by an HLA-DQ2-gliadin Tetramer Test. Am. J. Gastroenterol. 2011, 106, 1318–1324. [Google Scholar] [CrossRef] [PubMed]
  173. Christophersen, A.; Raki, M.; Bergseng, E.; Lundin, K.E.; Jahnsen, J.; Sollid, L.M.; Qiao, S.W. Tetramer-visualized gluten-specific CD4+ T cells in blood as a potential diagnostic marker for coeliac disease without oral gluten challenge. United Eur. Gastroenterol. J. 2014, 2, 268–278. [Google Scholar] [CrossRef] [PubMed]
  174. Christophersen, A.; Risnes, L.F.; Bergseng, E.; Lundin, K.E.; Sollid, L.M.; Qiao, S.W. Healthy HLA-DQ2.5+ Subjects Lack Regulatory and Memory T Cells Specific for Immunodominant Gluten Epitopes of Celiac Disease. J. Immunol. 2016, 196, 2819–2826. [Google Scholar] [CrossRef]
  175. Özgenel, Ş.M.; Temel, T.; Üsküdar Teke, H.; Yıldız, P.; Korkmaz, H.; Özakyol, A. HLA-DQ2/DQ8 frequency in adult patients with celiac disease, their first-degree relatives, and normal population in Turkey. Turk. J. Gastroenterol. 2019, 30, 321–325. [Google Scholar] [CrossRef]
  176. Cecilio, L.A.; Bonatto, M.W. The prevalence of HLA DQ2 and DQ8 in patients with celiac disease, in family and in general population. Arq. Bras. Cir. Dig. 2015, 28, 183–185. [Google Scholar] [CrossRef]
  177. Mansouri, M.; Dadfar, M.; Rostami-Nejad, M.; Ekhlasi, G.; Shahbazkhani, A.; Shahbazkhani, B. The frequency of HLA-DQ2/DQ8 haplotypes and celiac disease among the first-degree relatives of patients with celiac disease. Gastroenterol. Hepatol. Bed Bench 2021, 14, 36–43. [Google Scholar]
  178. Paziewska, A.; Cukrowska, B.; Dabrowska, M.; Goryca, K.; Piatkowska, M.; Kluska, A.; Mikula, M.; Karczmarski, J.; Oralewska, B.; Rybak, A.; et al. Combination Testing Using a Single MSH5 Variant alongside HLA Haplotypes Improves the Sensitivity of Predicting Coeliac Disease Risk in the Polish Population. PLoS ONE 2015, 10, e0139197. [Google Scholar] [CrossRef]
  179. Romanos, J.; Rosen, A.; Kumar, V.; Trynka, G.; Franke, L.; Szperl, A.; Gutierrez-Achury, J.; van Diemen, C.C.; Kanninga, R.; Jankipersadsing, S.A.; et al. Improving coeliac disease risk prediction by testing non-HLA variants additional to HLA variants. Gut 2014, 63, 415–422. [Google Scholar] [CrossRef]
  180. Koskinen, L.; Romanos, J.; Kaukinen, K.; Mustalahti, K.; Korponay-Szabo, I.; Barisani, D.; Bardella, M.T.; Ziberna, F.; Vatta, S.; Szeles, G.; et al. Cost-effective HLA typing with tagging SNPs predicts celiac disease risk haplotypes in the Finnish, Hungarian, and Italian populations. Immunogenetics 2009, 61, 247–256. [Google Scholar] [CrossRef]
  181. Monsuur, A.J.; de Bakker, P.I.; Zhernakova, A.; Pinto, D.; Verduijn, W.; Romanos, J.; Auricchio, R.; Lopez, A.; van Heel, D.A.; Crusius, J.B.; et al. Effective detection of human leukocyte antigen risk alleles in celiac disease using tag single nucleotide polymorphisms. PLoS ONE 2008, 3, e2270. [Google Scholar] [CrossRef] [PubMed]
  182. Huang, S.Q.; Zhang, N.; Zhou, Z.X.; Huang, C.C.; Zeng, C.L.; Xiao, D.; Guo, C.C.; Han, Y.J.; Ye, X.H.; Ye, X.G.; et al. Association of LPP and TAGAP Polymorphisms with Celiac Disease Risk: A Meta-Analysis. Int. J. Environ. Res. Public. Health 2017, 14, 171. [Google Scholar] [CrossRef] [PubMed]
  183. Anderson, R.P.; Goel, G.; Hardy, M.Y.; Russell, A.K.; Wang, S.; Szymczak, E.; Zhang, R.; Goldstein, K.E.; Neff, K.; Truitt, K.E.; et al. Whole blood interleukin-2 release test to detect and characterize rare circulating gluten-specific T cell responses in coeliac disease. Clin. Exp. Immunol. 2021, 204, 321–334. [Google Scholar] [CrossRef] [PubMed]
  184. Goel, G.; Tye-Din, J.A.; Qiao, S.W.; Russell, A.K.; Mayassi, T.; Ciszewski, C.; Sarna, V.K.; Wang, S.; Goldstein, K.E.; Dzuris, J.L.; et al. Cytokine release and gastrointestinal symptoms after gluten challenge in celiac disease. Sci. Adv. 2019, 5, eaaw7756. [Google Scholar] [CrossRef]
  185. Tye-Din, J.A.; Daveson, A.J.M.; Goldstein, K.E.; Hand, H.L.; Neff, K.M.; Goel, G.; Williams, L.J.; Truitt, K.E.; Anderson, R.P. Patient factors influencing acute gluten reactions and cytokine release in treated coeliac disease. BMC Med. 2020, 18, 362. [Google Scholar] [CrossRef]
  186. Molberg, O.; Lundin, K.E.; Nilsen, E.M.; Scott, H.; Kett, K.; Brandtzaeg, P.; Thorsby, E.; Sollid, L.M. HLA restriction patterns of gliadin- and astrovirus-specific CD4+ T cells isolated in parallel from the small intestine of celiac disease patients. Tissue Antigens 1998, 52, 407–415. [Google Scholar] [CrossRef]
  187. Anderson, R.P.; Degano, P.; Godkin, A.J.; Jewell, D.P.; Hill, A.V. In vivo antigen challenge in celiac disease identifies a single transglutaminase-modified peptide as the dominant A-gliadin T-cell epitope. Nat. Med. 2000, 6, 337–342. [Google Scholar] [CrossRef]
  188. Gjertsen, H.A.; Sollid, L.M.; Ek, J.; Thorsby, E.; Lundin, K.E. T cells from the peripheral blood of coeliac disease patients recognize gluten antigens when presented by HLA-DR, -DQ, or -DP molecules. Scand. J. Immunol. 1994, 39, 567–574. [Google Scholar] [CrossRef]
  189. Zühlke, S.; Risnes, L.F.; Dahal-Koirala, S.; Christophersen, A.; Sollid, L.M.; Lundin, K.E. CD38 expression on gluten-specific T cells is a robust marker of gluten re-exposure in coeliac disease. United Eur. Gastroenterol. J. 2019, 7, 1337–1344. [Google Scholar] [CrossRef]
  190. Fritz, R.D.; Chen, Y. Oat safety for celiac disease patients: Theoretical analysis correlates adverse symptoms in clinical studies to contaminated study oats. Nutr. Res. 2018, 60, 54–67. [Google Scholar] [CrossRef]
  191. Thompson, T.; Keller, A. Gluten cross contact in oats: Retrospective database analysis 2011 to 2023. Front. Nutr. 2023, 10, 1284636. [Google Scholar] [CrossRef] [PubMed]
  192. Smulders, M.J.M.; van de Wiel, C.C.M.; van den Broeck, H.C.; van der Meer, I.M.; Israel-Hoevelaken, T.P.M.; Timmer, R.D.; van Dinter, B.J.; Braun, S.; Gilissen, L. Oats in healthy gluten-free and regular diets: A perspective. Food Res. Int. 2018, 110, 3–10. [Google Scholar] [CrossRef] [PubMed]
  193. Lahdeaho, M.L.; Maki, M.; Laurila, K.; Huhtala, H.; Kaukinen, K. Small- bowel mucosal changes and antibody responses after low- and moderate-dose gluten challenge in celiac disease. BMC Gastroenterol. 2011, 11, 129. [Google Scholar] [CrossRef] [PubMed]
  194. Catassi, C.; Fabiani, E.; Iacono, G.; D’Agate, C.; Francavilla, R.; Biagi, F.; Volta, U.; Accomando, S.; Picarelli, A.; De Vitis, I.; et al. A prospective, double-blind, placebo-controlled trial to establish a safe gluten threshold for patients with celiac disease. Am. J. Clin. Nutr. 2007, 85, 160–166. [Google Scholar] [CrossRef]
  195. Bruins, M.J. The clinical response to gluten challenge: A review of the literature. Nutrients 2013, 5, 4614–4641. [Google Scholar] [CrossRef]
  196. Akobeng, A.K.; Thomas, A.G. Systematic review: Tolerable amount of gluten for people with coeliac disease. Aliment. Pharmacol. Ther. 2008, 27, 1044–1052. [Google Scholar] [CrossRef]
  197. Gibert, A.; Espadaler, M.; Angel Canela, M.; Sanchez, A.; Vaque, C.; Rafecas, M. Consumption of gluten-free products: Should the threshold value for trace amounts of gluten be at 20, 100 or 200 p.p.m.? Eur. J. Gastroenterol. Hepatol. 2006, 18, 1187–1195. [Google Scholar] [CrossRef]
  198. Food and Drug Administration. Food labeling: Gluten-free labeling of foods. Final rule. Fed. Regist. 2013, 78, 47154–47179. [Google Scholar]
  199. Tuire, I.; Marja-Leena, L.; Teea, S.; Katri, H.; Jukka, P.; Paivi, S.; Heini, H.; Markku, M.; Pekka, C.; Katri, K. Persistent duodenal intraepithelial lymphocytosis despite a long-term strict gluten-free diet in celiac disease. Am. J. Gastroenterol. 2012, 107, 1563–1569. [Google Scholar] [CrossRef]
  200. Hollon, J.R.; Cureton, P.A.; Martin, M.L.; Puppa, E.L.; Fasano, A. Trace gluten contamination may play a role in mucosal and clinical recovery in a subgroup of diet-adherent non-responsive celiac disease patients. BMC Gastroenterol. 2013, 13, 40. [Google Scholar] [CrossRef]
  201. Bascunan, K.A.; Vespa, M.C.; Araya, M. Celiac disease: Understanding the gluten-free diet. Eur. J. Nutr. 2016, 56, 449–459. [Google Scholar] [CrossRef] [PubMed]
  202. Samasca, G.; Sur, G.; Lupan, I.; Deleanu, D. Gluten-free diet and quality of life in celiac disease. Gastroenterol. Hepatol. Bed Bench 2014, 7, 139–143. [Google Scholar] [PubMed]
  203. Balamtekin, N.; Aksoy, C.; Baysoy, G.; Uslu, N.; Demir, H.; Koksal, G.; Saltik-Temizel, I.N.; Ozen, H.; Gurakan, F.; Yuce, A. Is compliance with gluten-free diet sufficient? Diet composition of celiac patients. Turk. J. Pediatr. 2015, 57, 374–379. [Google Scholar]
  204. Churruca, I.; Miranda, J.; Lasa, A.; Bustamante, M.A.; Larretxi, I.; Simon, E. Analysis of Body Composition and Food Habits of Spanish Celiac Women. Nutrients 2015, 7, 5515–5531. [Google Scholar] [CrossRef]
  205. Kautto, E.; Ivarsson, A.; Norstrom, F.; Hogberg, L.; Carlsson, A.; Hornell, A. Nutrient intake in adolescent girls and boys diagnosed with coeliac disease at an early age is mostly comparable to their non-coeliac contemporaries. J. Hum. Nutr. Diet. 2014, 27, 41–53. [Google Scholar] [CrossRef]
  206. Wu, J.H.; Neal, B.; Trevena, H.; Crino, M.; Stuart-Smith, W.; Faulkner-Hogg, K.; Yu Louie, J.C.; Dunford, E. Are gluten-free foods healthier than non-gluten-free foods? An evaluation of supermarket products in Australia. Br. J. Nutr. 2015, 114, 448–454. [Google Scholar] [CrossRef]
  207. Shepherd, S.J.; Gibson, P.R. Nutritional inadequacies of the gluten-free diet in both recently-diagnosed and long-term patients with coeliac disease. J. Hum. Nutr. Diet. 2013, 26, 349–358. [Google Scholar] [CrossRef]
  208. Miranda, J.; Lasa, A.; Bustamante, M.A.; Churruca, I.; Simon, E. Nutritional differences between a gluten-free diet and a diet containing equivalent products with gluten. Plant Foods Hum. Nutr. 2014, 69, 182–187. [Google Scholar] [CrossRef]
  209. Vici, G.; Belli, L.; Biondi, M.; Polzonetti, V. Gluten free diet and nutrient deficiencies: A review. Clin. Nutr. 2016, 35, 1236–1241. [Google Scholar] [CrossRef]
  210. Martin, J.; Geisel, T.; Maresch, C.; Krieger, K.; Stein, J. Inadequate nutrient intake in patients with celiac disease: Results from a German dietary survey. Digestion 2013, 87, 240–246. [Google Scholar] [CrossRef]
  211. MacCulloch, K.; Rashid, M. Factors affecting adherence to a gluten-free diet in children with celiac disease. Paediatr. Child. Health 2014, 19, 305–309. [Google Scholar] [CrossRef] [PubMed]
  212. Burden, M.; Mooney, P.D.; Blanshard, R.J.; White, W.L.; Cambray-Deakin, D.R.; Sanders, D.S. Cost and availability of gluten-free food in the UK: In store and online. Postgrad. Med. J. 2015, 91, 622–626. [Google Scholar] [CrossRef] [PubMed]
  213. Missbach, B.; Schwingshackl, L.; Billmann, A.; Mystek, A.; Hickelsberger, M.; Bauer, G.; Konig, J. Gluten-free food database: The nutritional quality and cost of packaged gluten-free foods. PeerJ 2015, 3, e1337. [Google Scholar] [CrossRef] [PubMed]
  214. Zanini, B.; Marullo, M.; Villanacci, V.; Salemme, M.; Lanzarotto, F.; Ricci, C.; Lanzini, A. Persistent Intraepithelial Lymphocytosis in Celiac Patients Adhering to Gluten-Free Diet Is Not Abolished Despite a Gluten Contamination Elimination Diet. Nutrients 2016, 8, 525. [Google Scholar] [CrossRef]
  215. Lanzini, A.; Lanzarotto, F.; Villanacci, V.; Mora, A.; Bertolazzi, S.; Turini, D.; Carella, G.; Malagoli, A.; Ferrante, G.; Cesana, B.M.; et al. Complete recovery of intestinal mucosa occurs very rarely in adult coeliac patients despite adherence to gluten-free diet. Aliment. Pharmacol. Ther. 2009, 29, 1299–1308. [Google Scholar] [CrossRef]
  216. Tursi, A.; Brandimarte, G.; Giorgetti, G.M.; Elisei, W.; Inchingolo, C.D.; Monardo, E.; Aiello, F. Endoscopic and histological findings in the duodenum of adults with celiac disease before and after changing to a gluten-free diet: A 2-year prospective study. Endoscopy 2006, 38, 702–707. [Google Scholar] [CrossRef]
  217. Daniewski, W.; Wojtasik, A.; Kunachowicz, H. Gluten content in special dietary use gluten-free products and other food products. Rocz. Panstw. Zakl. Hig. 2010, 61, 51–55. [Google Scholar]
  218. Cichańska, B.A. Problemy z Rozróżnianiem Żywności Bezglutenowej. Pediatria Współczesna. Gastroenterologia, Hepatologia i Żywienie Dziecka. 2009, pp. 117–122. Available online: https://katalogi.bn.org.pl/discovery/fulldisplay?docid=alma9910307223805606&vid=48OMNIS_NLOP:48OMNIS_NLOP (accessed on 8 March 2025).
  219. Wojtasik, W.D.A.; Kunachowicz, H. Zawartość glutenu (gliadyny) w wybranych produktach spożywczych. Bromat. Chem. Toksykol. 2010, XLIII, 362–371. [Google Scholar]
  220. Koerner, T.B.; Cleroux, C.; Poirier, C.; Cantin, I.; La Vieille, S.; Hayward, S.; Dubois, S. Gluten contamination of naturally gluten-free flours and starches used by Canadians with celiac disease. Food Addit. Contam. Part. A Chem. Anal. Control Expo. Risk Assess. 2013, 30, 2017–2021. [Google Scholar] [CrossRef]
  221. Thompson, T.; Lee, A.R.; Grace, T. Gluten contamination of grains, seeds, and flours in the United States: A pilot study. J. Am. Diet. Assoc. 2010, 110, 937–940. [Google Scholar] [CrossRef]
  222. Miller, K.; McGough, N.; Urwin, H. Catering Gluten-Free When Simultaneously Using Wheat Flour. J. Food Prot. 2016, 79, 282–287. [Google Scholar] [CrossRef] [PubMed]
  223. Colgrave, M.L.; Goswami, H.; Byrne, K.; Blundell, M.; Howitt, C.A.; Tanner, G.J. Proteomic profiling of 16 cereal grains and the application of targeted proteomics to detect wheat contamination. J. Proteome Res. 2015, 14, 2659–2668. [Google Scholar] [CrossRef] [PubMed]
  224. Gibert, A.; Kruizinga, A.G.; Neuhold, S.; Houben, G.F.; Canela, M.A.; Fasano, A.; Catassi, C. Might gluten traces in wheat substitutes pose a risk in patients with celiac disease? A population-based probabilistic approach to risk estimation. Am. J. Clin. Nutr. 2013, 97, 109–116. [Google Scholar] [CrossRef] [PubMed]
  225. La Vieille, S.; Dubois, S.; Hayward, S.; Koerner, T.B. Estimated levels of gluten incidentally present in a Canadian gluten-free diet. Nutrients 2014, 6, 881–896. [Google Scholar] [CrossRef]
  226. Lee, H.J.; Anderson, Z.; Ryu, D. Gluten contamination in foods labeled as “gluten free” in the United States. J. Food Prot. 2014, 77, 1830–1833. [Google Scholar] [CrossRef]
  227. Silvester, J.A.; Graff, L.A.; Rigaux, L.; Walker, J.R.; Duerksen, D.R. Symptomatic suspected gluten exposure is common among patients with coeliac disease on a gluten-free diet. Aliment. Pharmacol. Ther. 2016, 44, 612–619. [Google Scholar] [CrossRef]
  228. Roma, E.; Roubani, A.; Kolia, E.; Panayiotou, J.; Zellos, A.; Syriopoulou, V.P. Dietary compliance and life style of children with coeliac disease. J. Hum. Nutr. Diet. 2010, 23, 176–182. [Google Scholar] [CrossRef]
  229. Black, J.L.; Orfila, C. Impact of coeliac disease on dietary habits and quality of life. J. Hum. Nutr. Diet. 2011, 24, 582–587. [Google Scholar] [CrossRef]
  230. Sasaki, M.; Bosman, B.W.; Tan, P.S. A new, broad-substrate-specificity aminopeptidase from the dairy organism Lactobacillus helveticus SBT 2171. Microbiology 1996, 142 Pt 4, 799–808. [Google Scholar] [CrossRef]
  231. Tan, P.S.; Pos, K.M.; Konings, W.N. Purification and characterization of an endopeptidase from Lactococcus lactis subsp. cremoris Wg2. Appl. Environ. Microbiol. 1991, 57, 3593–3599. [Google Scholar] [CrossRef]
  232. Laloi, P.; Atlan, D.; Blanc, B.; Gilbert, C.; Portalier, R. Cell-wall-associated proteinase of Lactobacillus delbrueckii subsp. bulgaricus CNRZ 397: Differential extraction, purification and properties of the enzyme. Appl. Microbiol. Biotechnol. 1991, 36, 196–204. [Google Scholar] [CrossRef] [PubMed]
  233. Di Cagno, R.; De Angelis, M.; Lavermicocca, P.; De Vincenzi, M.; Giovannini, C.; Faccia, M.; Gobbetti, M. Proteolysis by sourdough lactic acid bacteria: Effects on wheat flour protein fractions and gliadin peptides involved in human cereal intolerance. Appl. Environ. Microbiol. 2002, 68, 623–633. [Google Scholar] [CrossRef] [PubMed]
  234. Di Cagno, R.; De Angelis, M.; Auricchio, S.; Greco, L.; Clarke, C.; De Vincenzi, M.; Giovannini, C.; D’Archivio, M.; Landolfo, F.; Parrilli, G.; et al. Sourdough bread made from wheat and nontoxic flours and started with selected lactobacilli is tolerated in celiac sprue patients. Appl. Environ. Microbiol. 2004, 70, 1088–1096. [Google Scholar] [CrossRef]
  235. di Cagno, R.; de Angelis, M.; Alfonsi, G.; de Vincenzi, M.; Silano, M.; Vincentini, O.; Gobbetti, M. Pasta made from durum wheat semolina fermented with selected lactobacilli as a tool for a potential decrease of the gluten intolerance. J. Agric. Food Chem. 2005, 53, 4393–4402. [Google Scholar] [CrossRef]
  236. Gerez, C.L.; Font de Valdez, G.; Rollan, G.C. Functionality of lactic acid bacteria peptidase activities in the hydrolysis of gliadin-like fragments. Lett. Appl. Microbiol. 2008, 47, 427–432. [Google Scholar] [CrossRef]
  237. Rollan, G.; De Angelis, M.; Gobbetti, M.; de Valdez, G.F. Proteolytic activity and reduction of gliadin-like fractions by sourdough lactobacilli. J. Appl. Microbiol. 2005, 99, 1495–1502. [Google Scholar] [CrossRef]
  238. Montserrat, V.; Bruins, M.J.; Edens, L.; Koning, F. Influence of dietary components on Aspergillus niger prolyl endoprotease mediated gluten degradation. Food Chem. 2015, 174, 440–445. [Google Scholar] [CrossRef]
  239. Tack, G.J.; van de Water, J.M.; Bruins, M.J.; Kooy-Winkelaar, E.M.; van Bergen, J.; Bonnet, P.; Vreugdenhil, A.C.; Korponay-Szabo, I.; Edens, L.; von Blomberg, B.M.; et al. Consumption of gluten with gluten-degrading enzyme by celiac patients: A pilot-study. World J. Gastroenterol. 2013, 19, 5837–5847. [Google Scholar] [CrossRef]
  240. Tye-Din, J.A.; Anderson, R.P.; Ffrench, R.A.; Brown, G.J.; Hodsman, P.; Siegel, M.; Botwick, W.; Shreeniwas, R. The effects of ALV003 pre-digestion of gluten on immune response and symptoms in celiac disease in vivo. Clin. Immunol. 2010, 134, 289–295. [Google Scholar] [CrossRef]
  241. Siegel, M.; Garber, M.E.; Spencer, A.G.; Botwick, W.; Kumar, P.; Williams, R.N.; Kozuka, K.; Shreeniwas, R.; Pratha, V.; Adelman, D.C. Safety, tolerability, and activity of ALV003: Results from two phase 1 single, escalating-dose clinical trials. Dig. Dis. Sci. 2012, 57, 440–450. [Google Scholar] [CrossRef]
  242. Lahdeaho, M.L.; Kaukinen, K.; Laurila, K.; Vuotikka, P.; Koivurova, O.P.; Karja-Lahdensuu, T.; Marcantonio, A.; Adelman, D.C.; Maki, M. Glutenase ALV003 attenuates gluten-induced mucosal injury in patients with celiac disease. Gastroenterology 2014, 146, 1649–1658. [Google Scholar] [CrossRef] [PubMed]
  243. Murray, J.A.; Syage, J.A.; Wu, T.-T.; Dickason, M.A.; Ramos, A.G.; Van Dyke, C.; Horwath, I.; Lavin, P.T.; Mäki, M.; Hujoel, I.; et al. Latiglutenase Protects the Mucosa and Attenuates Symptom Severity in Patients with Celiac Disease Exposed to a Gluten Challenge. Gastroenterology 2022, 163, 1510–1521.e6. [Google Scholar] [CrossRef] [PubMed]
  244. Wolf, C.; Siegel, J.B.; Tinberg, C.; Camarca, A.; Gianfrani, C.; Paski, S.; Guan, R.; Montelione, G.; Baker, D.; Pultz, I.S. Engineering of Kuma030: A Gliadin Peptidase That Rapidly Degrades Immunogenic Gliadin Peptides in Gastric Conditions. J. Am. Chem. Soc. 2015, 137, 13106–13113. [Google Scholar] [CrossRef] [PubMed]
  245. Pultz, I.S.; Hill, M.; Vitanza, J.M.; Wolf, C.; Saaby, L.; Liu, T.; Winkle, P.; Leffler, D.A. Gluten Degradation, Pharmacokinetics, Safety, and Tolerability of TAK-062, an Engineered Enzyme to Treat Celiac Disease. Gastroenterology 2021, 161, 81–93.e3. [Google Scholar] [CrossRef]
  246. Klemenak, M.; Dolinsek, J.; Langerholc, T.; Di Gioia, D.; Micetic-Turk, D. Administration of Bifidobacterium breve Decreases the Production of TNF-alpha in Children with Celiac Disease. Dig. Dis. Sci. 2015, 60, 3386–3392. [Google Scholar] [CrossRef]
  247. Olivares, M.; Laparra, M.; Sanz, Y.; Cinova, J.; De Palma, G.; Stepankova, R.; Kofronova, O.; Kverka, M.; Tuckova, L. Influence of Bifidobacterium longum CECT 7347 and gliadin peptides on intestinal epithelial cell proteome. J. Agric. Food Chem. 2011, 59, 7666–7671. [Google Scholar] [CrossRef]
  248. Olivares, M.; Castillejo, G.; Varea, V.; Sanz, Y. Double-blind, randomised, placebo-controlled intervention trial to evaluate the effects of Bifidobacterium longum CECT 7347 in children with newly diagnosed coeliac disease. Br. J. Nutr. 2014, 112, 30–40. [Google Scholar] [CrossRef]
  249. Smecuol, E.; Hwang, H.J.; Sugai, E.; Corso, L.; Chernavsky, A.C.; Bellavite, F.P.; Gonzalez, A.; Vodanovich, F.; Moreno, M.L.; Vazquez, H.; et al. Exploratory, randomized, double-blind, placebo-controlled study on the effects of Bifidobacterium infantis natren life start strain super strain in active celiac disease. J. Clin. Gastroenterol. 2013, 47, 139–147. [Google Scholar] [CrossRef]
  250. Lloyd-Still, J.D.; Grand, R.J.; Khaw, K.T.; Shwachman, H. The use of corticosteroids in celiac crisis. J. Pediatr. 1972, 81, 1074–1081. [Google Scholar] [CrossRef]
  251. Abbas, A.; Shahab, T.; Sherwani, R.K.; Alam, S. Addition of a Short Course of Prednisolone to a Gluten-Free Diet vs. Gluten-Free Diet Alone in Recovery of Celiac Disease: A Pilot Randomized Controlled Trial. Cureus 2018, 10, e2118. [Google Scholar] [CrossRef]
  252. Ali Ibrahim, A.; Kenyon, V.; Fasano, A.; Leonard, M.M. Budesonide and the Gluten Containing Elimination Diet as Treatments for Non-responsive Celiac Disease in Children. J. Pediatr. Gastroenterol. Nutr. 2022, 75, 616–622. [Google Scholar] [CrossRef] [PubMed]
  253. Rubio-Tapia, A.; Murray, J.A. Classification and management of refractory coeliac disease. Gut 2010, 59, 547–557. [Google Scholar] [CrossRef] [PubMed]
  254. Mukewar, S.S.; Sharma, A.; Rubio-Tapia, A.; Wu, T.T.; Jabri, B.; Murray, J.A. Open-Capsule Budesonide for Refractory Celiac Disease. Am. J. Gastroenterol. 2017, 112, 959–967. [Google Scholar] [CrossRef] [PubMed]
  255. Therrien, A.; Silvester, J.A.; Leonard, M.M.; Leffler, D.A.; Fasano, A.; Kelly, C.P. Enteric-Release Budesonide May Be Useful in the Management of Non-Responsive Celiac Disease. Dig. Dis. Sci. 2021, 66, 1989–1997. [Google Scholar] [CrossRef]
  256. Mauriño, E.; Niveloni, S.; Cherñavsky, A.; Pedreira, S.; Mazure, R.; Vazquez, H.; Reyes, H.; Fiorini, A.; Smecuol, E.; Cabanne, A.; et al. Azathioprine in refractory sprue: Results from a prospective, open-label study. Am. J. Gastroenterol. 2002, 97, 2595–2602. [Google Scholar] [CrossRef]
  257. Goerres, M.S.; Meijer, J.W.; Wahab, P.J.; Kerckhaert, J.A.; Groenen, P.J.; Van Krieken, J.H.; Mulder, C.J. Azathioprine and prednisone combination therapy in refractory coeliac disease. Aliment. Pharmacol. Ther. 2003, 18, 487–494. [Google Scholar] [CrossRef]
  258. Iqbal, U.; Chaudhary, A.; Karim, M.A.; Anwar, H.; Merrell, N. Refractory Celiac Disease Successfully Treated with Azathioprine. Gastroenterol. Res. 2017, 10, 199–201. [Google Scholar] [CrossRef]
  259. Rawal, N.; Twaddell, W.; Fasano, A.; Blanchard, S.; Safta, A. Remission of Refractory Celiac Disease With Infliximab in a Pediatric Patient. ACG Case Rep. J. 2015, 2, 121–123. [Google Scholar] [CrossRef]
  260. Valitutti, F.; Barbato, M.; Aloi, M.; Marcheggiano, A.; Di Nardo, G.; Leoni, S.; Iorfida, D.; Corazza, G.R.; Cucchiara, S. Autoimmune enteropathy in a 13-year-old celiac girl successfully treated with infliximab. J. Clin. Gastroenterol. 2014, 48, 264–266. [Google Scholar] [CrossRef]
  261. Costantino, G.; della Torre, A.; Lo Presti, M.A.; Caruso, R.; Mazzon, E.; Fries, W. Treatment of life-threatening type I refractory coeliac disease with long-term infliximab. Dig. Liver Dis. 2008, 40, 74–77. [Google Scholar] [CrossRef]
  262. Senolt, L.; Vencovsky, J.; Pavelka, K.; Ospelt, C.; Gay, S. Prospective new biological therapies for rheumatoid arthritis. Autoimmun. Rev. 2009, 9, 102–107. [Google Scholar] [CrossRef] [PubMed]
  263. Waldmann, T.A.; Conlon, K.C.; Stewart, D.M.; Worthy, T.A.; Janik, J.E.; Fleisher, T.A.; Albert, P.S.; Figg, W.D.; Spencer, S.D.; Raffeld, M.; et al. Phase 1 trial of IL-15 trans presentation blockade using humanized Mikbeta1 mAb in patients with T-cell large granular lymphocytic leukemia. Blood 2013, 121, 476–484. [Google Scholar] [CrossRef] [PubMed]
  264. Morris, J.C.; Janik, J.E.; White, J.D.; Fleisher, T.A.; Brown, M.; Tsudo, M.; Goldman, C.K.; Bryant, B.; Petrus, M.; Top, L.; et al. Preclinical and phase I clinical trial of blockade of IL-15 using Mikbeta1 monoclonal antibody in T cell large granular lymphocyte leukemia. Proc. Natl. Acad. Sci. USA 2006, 103, 401–406. [Google Scholar] [CrossRef] [PubMed]
  265. Lähdeaho, M.L.; Scheinin, M.; Vuotikka, P.; Taavela, J.; Popp, A.; Laukkarinen, J.; Koffert, J.; Koivurova, O.P.; Pesu, M.; Kivelä, L.; et al. Safety and efficacy of AMG 714 in adults with coeliac disease exposed to gluten challenge: A phase 2a, randomised, double-blind, placebo-controlled study. Lancet Gastroenterol. Hepatol. 2019, 4, 948–959. [Google Scholar] [CrossRef]
  266. Cellier, C.; Bouma, G.; Van Gils, T.; Khater, S.; Malamut, G.; Crespo, L.; Collin, P.; Green, P.H.; Crowe, S.E.; Tsuji, W.; et al. Safety and efficacy of AMG 714 in patients with type 2 refractory coeliac disease: A phase 2a, randomised, double-blind, placebo-controlled, parallel-group study. Lancet Gastroenterol. Hepatol. 2019, 4, 960–970. [Google Scholar] [CrossRef]
  267. Dieckman, T.; Schumann, M.; Beaumont, H.; Bontkes, H.J.; Koning, F.; Bouma, G. Enduring Clinical Remission in Refractory Celiac Disease Type II with Tofacitinib: An Open-Label Clinical Study. Clin. Gastroenterol. Hepatol. 2024, 22, 2334–2336. [Google Scholar] [CrossRef]
  268. Croese, J.; Giacomin, P.; Navarro, S.; Clouston, A.; McCann, L.; Dougall, A.; Ferreira, I.; Susianto, A.; O’Rourke, P.; Howlett, M.; et al. Experimental hookworm infection and gluten microchallenge promote tolerance in celiac disease. J. Allergy Clin. Immunol. 2015, 135, 508–516. [Google Scholar] [CrossRef]
  269. McSorley, H.J.; Gaze, S.; Daveson, J.; Jones, D.; Anderson, R.P.; Clouston, A.; Ruyssers, N.E.; Speare, R.; McCarthy, J.S.; Engwerda, C.R.; et al. Suppression of inflammatory immune responses in celiac disease by experimental hookworm infection. PLoS ONE 2011, 6, e24092. [Google Scholar] [CrossRef]
  270. Daveson, A.J.; Jones, D.M.; Gaze, S.; McSorley, H.; Clouston, A.; Pascoe, A.; Cooke, S.; Speare, R.; Macdonald, G.A.; Anderson, R.; et al. Effect of hookworm infection on wheat challenge in celiac disease--a randomised double-blinded placebo controlled trial. PLoS ONE 2011, 6, e17366. [Google Scholar] [CrossRef]
  271. Giacomin, P.; Zakrzewski, M.; Croese, J.; Su, X.; Sotillo, J.; McCann, L.; Navarro, S.; Mitreva, M.; Krause, L.; Loukas, A.; et al. Experimental hookworm infection and escalating gluten challenges are associated with increased microbial richness in celiac subjects. Sci. Rep. 2015, 5, 13797. [Google Scholar] [CrossRef]
  272. Cantacessi, C.; Giacomin, P.; Croese, J.; Zakrzewski, M.; Sotillo, J.; McCann, L.; Nolan, M.J.; Mitreva, M.; Krause, L.; Loukas, A. Impact of experimental hookworm infection on the human gut microbiota. J. Infect. Dis. 2014, 210, 1431–1434. [Google Scholar] [CrossRef] [PubMed]
  273. Croese, J.; Miller, G.C.; Marquart, L.; Llewellyn, S.; Gupta, R.; Becker, L.; Clouston, A.D.; Welch, C.; Sidorenko, J.; Wallace, L.; et al. Randomized, Placebo Controlled Trial of Experimental Hookworm Infection for Improving Gluten Tolerance in Celiac Disease. Clin. Transl. Gastroenterol. 2020, 11, e00274. [Google Scholar] [CrossRef] [PubMed]
  274. Murray, J.A.; Wassaf, D.; Dunn, K.; Arora, S.; Winkle, P.; Stacey, H.; Cooper, S.; Goldstein, K.E.; Manchanda, R.; Kontos, S.; et al. Safety and tolerability of KAN-101, a liver-targeted immune tolerance therapy, in patients with coeliac disease (ACeD): A phase 1 trial. Lancet Gastroenterol. Hepatol. 2023, 8, 735–747. [Google Scholar] [CrossRef]
  275. Kelly, C.P.; Murray, J.A.; Leffler, D.A.; Getts, D.R.; Bledsoe, A.C.; Smithson, G.; First, M.R.; Morris, A.; Boyne, M.; Elhofy, A.; et al. TAK-101 Nanoparticles Induce Gluten-Specific Tolerance in Celiac Disease: A Randomized, Double-Blind, Placebo-Controlled Study. Gastroenterology 2021, 161, 66–80.e8. [Google Scholar] [CrossRef]
  276. Riedmann, E.M. Human vaccines: News. Hum. Vaccin. Immunother. 2012, 8, 1550–1553. [Google Scholar] [CrossRef]
  277. Daveson, A.J.M.; Ee, H.C.; Andrews, J.M.; King, T.; Goldstein, K.E.; Dzuris, J.L.; MacDougall, J.A.; Williams, L.J.; Treohan, A.; Cooreman, M.P.; et al. Epitope-Specific Immunotherapy Targeting CD4-Positive T Cells in Celiac Disease: Safety, Pharmacokinetics, and Effects on Intestinal Histology and Plasma Cytokines with Escalating Dose Regimens of Nexvax2 in a Randomized, Double-Blind, Placebo-Controlled Phase 1 Study. EBioMedicine 2017, 26, 78–90. [Google Scholar] [CrossRef]
  278. Truitt, K.E.; Daveson, A.J.M.; Ee, H.C.; Goel, G.; MacDougall, J.; Neff, K.; Anderson, R.P. Randomised clinical trial: A placebo-controlled study of subcutaneous or intradermal NEXVAX2, an investigational immunomodulatory peptide therapy for coeliac disease. Aliment. Pharmacol. Ther. 2019, 50, 547–555. [Google Scholar] [CrossRef]
  279. Hardy, M.Y.; Goel, G.; Russell, A.K.; Chen Yi Mei, S.L.G.; Brown, G.J.E.; Wang, S.; Szymczak, E.; Zhang, R.; Goldstein, K.E.; Neff, K.M.; et al. A Sensitive Whole Blood Assay Detects Antigen-Stimulated Cytokine Release From CD4+ T Cells and Facilitates Immunomonitoring in a Phase 2 Clinical Trial of Nexvax2 in Coeliac Disease. Front. Immunol. 2021, 12, 661622. [Google Scholar] [CrossRef]
  280. Tye-Din, J.A.; Daveson, A.J.; Goel, G.; Goldstein, K.E.; Hand, H.L.; Neff, K.M.; Popp, A.; Taavela, J.; Maki, M.; Isola, J.; et al. Efficacy and safety of gluten peptide-based antigen-specific immunotherapy (Nexvax2) in adults with coeliac disease after bolus exposure to gluten (RESET CeD): An interim analysis of a terminated randomised, double-blind, placebo-controlled phase 2 study. Lancet Gastroenterol. Hepatol. 2023, 8, 446–457. [Google Scholar] [CrossRef]
  281. Fasano, A. Zonulin and its regulation of intestinal barrier function: The biological door to inflammation, autoimmunity, and cancer. Physiol. Rev. 2011, 91, 151–175. [Google Scholar] [CrossRef]
  282. Fasano, A. Intestinal permeability and its regulation by zonulin: Diagnostic and therapeutic implications. Clin. Gastroenterol. Hepatol. 2012, 10, 1096–1100. [Google Scholar] [CrossRef] [PubMed]
  283. Fasano, A. Zonulin, regulation of tight junctions, and autoimmune diseases. Ann. N. Y. Acad. Sci. 2012, 1258, 25–33. [Google Scholar] [CrossRef] [PubMed]
  284. Leffler, D.A.; Kelly, C.P.; Abdallah, H.Z.; Colatrella, A.M.; Harris, L.A.; Leon, F.; Arterburn, L.A.; Paterson, B.M.; Lan, Z.H.; Murray, J.A. A randomized, double-blind study of larazotide acetate to prevent the activation of celiac disease during gluten challenge. Am. J. Gastroenterol. 2012, 107, 1554–1562. [Google Scholar] [CrossRef] [PubMed]
  285. Kelly, C.P.; Green, P.H.; Murray, J.A.; Dimarino, A.; Colatrella, A.; Leffler, D.A.; Alexander, T.; Arsenescu, R.; Leon, F.; Jiang, J.G.; et al. Larazotide acetate in patients with coeliac disease undergoing a gluten challenge: A randomised placebo-controlled study. Aliment. Pharmacol. Ther. 2013, 37, 252–262. [Google Scholar] [CrossRef]
  286. Leffler, D.A.; Kelly, C.P.; Green, P.H.; Fedorak, R.N.; DiMarino, A.; Perrow, W.; Rasmussen, H.; Wang, C.; Bercik, P.; Bachir, N.M.; et al. Larazotide acetate for persistent symptoms of celiac disease despite a gluten-free diet: A randomized controlled trial. Gastroenterology 2015, 148, 1311–1319.e6. [Google Scholar] [CrossRef]
  287. Paterson, B.M.; Lammers, K.M.; Arrieta, M.C.; Fasano, A.; Meddings, J.B. The safety, tolerance, pharmacokinetic and pharmacodynamic effects of single doses of AT-1001 in coeliac disease subjects: A proof of concept study. Aliment. Pharmacol. Ther. 2007, 26, 757–766. [Google Scholar] [CrossRef]
  288. Gopalakrishnan, S.; Durai, M.; Kitchens, K.; Tamiz, A.P.; Somerville, R.; Ginski, M.; Paterson, B.M.; Murray, J.A.; Verdu, E.F.; Alkan, S.S.; et al. Larazotide acetate regulates epithelial tight junctions in vitro and in vivo. Peptides 2012, 35, 86–94. [Google Scholar] [CrossRef]
  289. Biopharma. 9 Meters Biopharma Announces Interim Analysis of Phase 3 Study of Larazotide for Celiac Disease Does Not Support Trial Continuation (Press Release 21 June 2022). Available online: https://www.biospace.com/poor-results-end-9-meters-phase-iii-trial-for-larazotide (accessed on 8 March 2025).
  290. Hoilat, G.J.; Altowairqi, A.K.; Ayas, M.F.; Alhaddab, N.T.; Alnujaidi, R.A.; Alharbi, H.A.; Alyahyawi, N.; Kamal, A.; Alhabeeb, H.; Albazee, E.; 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, 101782. [Google Scholar] [CrossRef]
  291. Pardin, C.; Pelletier, J.N.; Lubell, W.D.; Keillor, J.W. Cinnamoyl inhibitors of tissue transglutaminase. J. Org. Chem. 2008, 73, 5766–5775. [Google Scholar] [CrossRef]
  292. Klock, C.; Jin, X.; Choi, K.; Khosla, C.; Madrid, P.B.; Spencer, A.; Raimundo, B.C.; Boardman, P.; Lanza, G.; Griffin, J.H. Acylideneoxoindoles: A new class of reversible inhibitors of human transglutaminase 2. Bioorg Med. Chem. Lett. 2011, 21, 2692–2696. [Google Scholar] [CrossRef]
  293. Hausch, F.; Halttunen, T.; Maki, M.; Khosla, C. Design, synthesis, and evaluation of gluten peptide analogs as selective inhibitors of human tissue transglutaminase. Chem. Biol. 2003, 10, 225–231. [Google Scholar] [CrossRef] [PubMed]
  294. Toth, B.; Garabuczi, E.; Sarang, Z.; Vereb, G.; Vamosi, G.; Aeschlimann, D.; Blasko, B.; Becsi, B.; Erdodi, F.; Lacy-Hulbert, A.; et al. Transglutaminase 2 is needed for the formation of an efficient phagocyte portal in macrophages engulfing apoptotic cells. J. Immunol. 2009, 182, 2084–2092. [Google Scholar] [CrossRef] [PubMed]
  295. Szondy, Z.; Sarang, Z.; Molnar, P.; Nemeth, T.; Piacentini, M.; Mastroberardino, P.G.; Falasca, L.; Aeschlimann, D.; Kovacs, J.; Kiss, I.; et al. Transglutaminase 2-/- mice reveal a phagocytosis-associated crosstalk between macrophages and apoptotic cells. Proc. Natl. Acad. Sci. USA 2003, 100, 7812–7817. [Google Scholar] [CrossRef]
  296. Rauhavirta, T.; Oittinen, M.; Kivisto, R.; Mannisto, P.T.; Garcia-Horsman, J.A.; Wang, Z.; Griffin, M.; Maki, M.; Kaukinen, K.; Lindfors, K. Are transglutaminase 2 inhibitors able to reduce gliadin-induced toxicity related to celiac disease? A proof-of-concept study. J. Clin. Immunol. 2013, 33, 134–142. [Google Scholar] [CrossRef]
  297. Sulic, A.M.; Kurppa, K.; Rauhavirta, T.; Kaukinen, K.; Lindfors, K. Transglutaminase as a therapeutic target for celiac disease. Expert Opin. Ther. Targets 2015, 19, 335–348. [Google Scholar] [CrossRef]
  298. Schuppan, D.; Mäki, M.; Lundin, K.E.A.; Isola, J.; Friesing-Sosnik, T.; Taavela, J.; Popp, A.; Koskenpato, J.; Langhorst, J.; Hovde, Ø.; et al. A Randomized Trial of a Transglutaminase 2 Inhibitor for Celiac Disease. N. Engl. J. Med. 2021, 385, 35–45. [Google Scholar] [CrossRef]
  299. Dotsenko, V.; Tewes, B.; Hils, M.; Pasternack, R.; Isola, J.; Taavela, J.; Popp, A.; Sarin, J.; Huhtala, H.; Hiltunen, P.; et al. Transcriptomic analysis of intestine following administration of a transglutaminase 2 inhibitor to prevent gluten-induced intestinal damage in celiac disease. Nat. Immunol. 2024, 25, 1218–1230. [Google Scholar] [CrossRef]
Figure 1. World map of celiac disease prevalence.
Figure 1. World map of celiac disease prevalence.
Foods 14 00959 g001
Figure 2. Schematic diagram of the basic immune reactions leading to the development of celiac disease. APC—antigen-presenting cell. TGF-α—transforming growth factor, α IFN-γ—interferon γ, TEER—transepithelial electrical resistance, PPARγ—Peroxisome proliferator-activated receptors, CXCR3—chemokine receptor, 3MMP—matrix metalloproteinases, and EGFR—epidermal growth factor receptor.
Figure 2. Schematic diagram of the basic immune reactions leading to the development of celiac disease. APC—antigen-presenting cell. TGF-α—transforming growth factor, α IFN-γ—interferon γ, TEER—transepithelial electrical resistance, PPARγ—Peroxisome proliferator-activated receptors, CXCR3—chemokine receptor, 3MMP—matrix metalloproteinases, and EGFR—epidermal growth factor receptor.
Foods 14 00959 g002
Figure 3. Changes in the diagnostic criteria for celiac disease over the years.
Figure 3. Changes in the diagnostic criteria for celiac disease over the years.
Foods 14 00959 g003
Table 1. Analysis of studies evaluating osteopenia and osteoporosis among patients with celiac disease. n/c—no data at that point.
Table 1. Analysis of studies evaluating osteopenia and osteoporosis among patients with celiac disease. n/c—no data at that point.
AuthorYears of StudyCountryAgeNo. of
Participation
SexOsteoporosis
Frequency
Zanchetta et al. [23,25]2011–2015ArgentinaAdults30Womenn/c
Bai et al. [21]n/cArgentinaAdults25Womenn/c
Galli et al. [29]2010–2021ItaliAdults291Women–men 3:1Osteopenia + osteoporosis 63.5%
Ganji et al. [30]Review of 56 article tile 2018UK, Brazil, India, Hungary, PolandAdults Women–men 3:114.4%
Ganji et al. [31]2014–2019IranAdults under 55387Women–men 3:116.4%
Walker et al. [32] USAAdults721Women–men 3:119.6%
Sayar et al. [33]2010–2019TurkeyAdults under 5084Women and men15.2%
Soracka et al. [34] PolandAdults under 5030Women13.3%
Table 2. Prevalence of celiac disease in population studies across the world.
Table 2. Prevalence of celiac disease in population studies across the world.
Country/RegionYears of StudyStudied
Population
Test Used for
Screening
Studied
Population
Number
Prevalence of CD
North America
-USA [68]
Anty-EMA, anty-tTG, HLA DQ2/DQ8
1996–2001At all ages13,1450.75%
South America [79]2000–2013Metanalyses of 72 seroprevalence studies, at all agesAnty-tTG, anty-EMA 0.46–0.64%
Europe [73]1991–2012Metanalyses of 49 seroprevalence studies, at all ageAnty-tTG 1.3%
-Finland [64,71]1994SchoolchildrenAnty-EMA, anty-tTG36451.5%
2000–2001Adults (30–93 years old)Anty-EMA, anty-tTG, biopsies64032.4%
-German [71]1990, 2001Adults (25–74 years old)Anty-EMA, anty-tTG, biopsies92010.3%
-Italy [71]2000–2002At all agesAnty-EMA, anty-tTG, biopsies74270.7%
-Russian [80]1997–2001At all agesAnty-EMA, anty-tTG, biopsies11,0700.2–0.57%
Asia [66] Metanalyses of 19 seroprevalence studies, at all agesAnty-tTG, anty-EMA 1.6%
-China [81] Metanalyses of 18 seroprevalence studies, at all ageAnty-tTG, anty-DGP 0.56%
-India [82]1994–1995Seroprevalence at all agesAnty-tTG10,4881.44%
-Australia [83]2007–2015Seroprevalence at all agesAnty-tTG30111.56%
-Iran [84] Seroprevalence, schoolchildrenAnty-tTG15002%
-Saudi Arabia [85] Healthy adults aged 20–60 yearsAnty-EMA, anty-tTG, biopsies9801.9%
-Israel [86]2012–2013Seroprevalence at all agesAnty-tTG403,2831.56%
Africa:
-Tunisia [87]Adults (blood donors)Anty-EMA25000.28%
-Egypt [88]ChildrenAnty-tTG15000.53%
-Saharawi population [89]At all agesAnty-tTG, anty-EMA9755.6%
Table 3. Sensitivity and specificity of serum antibodies tests for celiac disease [160,161,162].
Table 3. Sensitivity and specificity of serum antibodies tests for celiac disease [160,161,162].
AntibodySensitivity (%)Specificity (%)
Anti-tTG IgA63–96.891.0–100
EMA IgA80–93.795–100
DGP IgG18–84.498.5
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kowalski, M.K.; Domżał-Magrowska, D.; Małecka-Wojciesko, E. Celiac Disease—Narrative Review on Progress in Celiac Disease. Foods 2025, 14, 959. https://doi.org/10.3390/foods14060959

AMA Style

Kowalski MK, Domżał-Magrowska D, Małecka-Wojciesko E. Celiac Disease—Narrative Review on Progress in Celiac Disease. Foods. 2025; 14(6):959. https://doi.org/10.3390/foods14060959

Chicago/Turabian Style

Kowalski, Marek K., Danuta Domżał-Magrowska, and Ewa Małecka-Wojciesko. 2025. "Celiac Disease—Narrative Review on Progress in Celiac Disease" Foods 14, no. 6: 959. https://doi.org/10.3390/foods14060959

APA Style

Kowalski, M. K., Domżał-Magrowska, D., & Małecka-Wojciesko, E. (2025). Celiac Disease—Narrative Review on Progress in Celiac Disease. Foods, 14(6), 959. https://doi.org/10.3390/foods14060959

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop