Figure 1 - uploaded by Ilma Korponay-Szabo
Content may be subject to copyright.
IgA (green) and transglutaminase 2 (TG2, red) in the normal jejunum (A, D, G), in the early developing stage of coeliac disease (B, E, H) and in the coeliac flat lesion (C, F, I). (B) Subepithelial IgA deposits in the villi and around crypts in the mucosa with a normal architecture and (C) in the flat mucosa developed four months later in the same patient. IgA deposits colocalised with TG2 (E, F, H. I), as indicated by the yellow merging of the labels. Both deposited IgA and TG2 were closely related to vessels shown in blue (I, J, arrows). (K) Similar jejunal IgA deposits in a patient with dermatitis herpetiformis and negative serum endomysial antibodies. (L) Crypt basement membrane laminin (red band) with coeliac IgA (green) deposits. There was only partial overlap (yellow) on the extracellular surface of the laminin. Bar = 50 m m. 

IgA (green) and transglutaminase 2 (TG2, red) in the normal jejunum (A, D, G), in the early developing stage of coeliac disease (B, E, H) and in the coeliac flat lesion (C, F, I). (B) Subepithelial IgA deposits in the villi and around crypts in the mucosa with a normal architecture and (C) in the flat mucosa developed four months later in the same patient. IgA deposits colocalised with TG2 (E, F, H. I), as indicated by the yellow merging of the labels. Both deposited IgA and TG2 were closely related to vessels shown in blue (I, J, arrows). (K) Similar jejunal IgA deposits in a patient with dermatitis herpetiformis and negative serum endomysial antibodies. (L) Crypt basement membrane laminin (red band) with coeliac IgA (green) deposits. There was only partial overlap (yellow) on the extracellular surface of the laminin. Bar = 50 m m. 

Source publication
Article
Full-text available
IgA class serum autoantibodies against type 2 (tissue) transglutaminase (TG2) bind to both intestinal and extraintestinal normal tissue sections in vitro, eliciting endomysial, reticulin, and jejunal antibody reactions. It is not known whether similar binding also occurs in coeliac patients in vivo, and may thereby contribute to disease manifestati...

Context in source publication

Context 1
... frozen tissue samples from six additional patients who underwent biopsies from organs other than the small bowel while they had active coeliac disease. All of these biopsies had been performed based on clinical indications independent of the evaluation of coeliac disease (table 2). Samples were traced retrospectively and included two lymph nodes, one skeletal muscle, two liver biopsy samples, and a non-inflamed appendix. As controls, one lymph node, three liver, and six appendix samples from non-coeliac patients were included. These had been removed in unrelated surgical interventions and were histologically normal. All biopsies were performed for diagnostic purposes and informed consent was obtained. Unfixed 5 m m thick sections of patient tissues were investigated for deposited immunoglobulins by direct immunofluorescence using fluorescein isothiocyanate labelled rabbit antibodies against human IgA, IgG, and IgM (Dako AS, Glostrup, Denmark) at a dilution of 1:40 in phosphate buffered saline (PBS), pH 7.4. These stainings were also performed as multicolour procedures where patient immunoglobulins were shown in green and extracellular matrix proteins in red using rhodamine conjugated antimouse or antirabbit secondary antibodies, as described elsewhere. 18 19 The primary antibodies used were monoclonal mouse antibodies against TG2 (CUB7402; NeoMarkers, Fremont, California, USA), and rabbit antibodies against fibronectin (Dako) and laminin (Dako), respectively, diluted 1:200 in PBS. For investigation of the relation of patient immunoglobulins to blood vessels, green was used for detection of patient IgA, red for TG2, and blue for an endothelial marker (rabbit antibodies against von Willebrand factor (Dako) followed by AMCA conjugated secondary antirabbit antibodies (Vector Laboratories, Burlingame, California, USA)). Unfixed cryosections were washed twice in PBS to remove blood and related antibodies. A solution of 0.25% chloroacetic acid (Fluka Chemie AG, Buchs, Switzerland) with 0.2 M/l NaCl, pH 2.7, was then used as the eluent 28 and applied to the sections for 20 minutes. The eluates with chloroacetic acid were neutralised with 1 M/l imidazole, washed in PBS, concentrated by centrifugation on 10K membranes (Microsep, Pall Corporation, Ann Harbor, Michigan, USA), and lyophilised. Treated sections were washed twice in PBS and examined by immunofluorescence for the remaining IgA and TG2 in tissues, as described above. Sections from extraintestinal samples, from six coeliac and three dermatitis herpetiformis jejunum samples, were investigated by immunofluorescence after choroacetic acid treat- ment. Lyophilised tissue eluates were prepared from a coeliac liver, lymph node, and a non-coeliac liver. Endomysial antibodies were investigated in serum samples and tissue eluates by an indirect immunofluorescent method on monkey oesophagus, normal human umbilical cord, jejunum, and appendix sections, as described elsewhere. 18 The initial dilution was 1:2.5 in PBS, and samples non-reactive at this dilution were considered negative. Eluates were also tested on TG2 knockout mouse (TG2 2 / 2 ) jejunum as a substrate, and on TG2 2 / 2 jejunum coated with human recombinant TG2 expressed in Escherichia coli , 29 as previously described. 19 Tissue eluates were tested for the presence TG2 and IgA by western blot 30 and for TG2 specific IgA by ELISA using microtitre plates coated with fibronectin (Sigma, St Louis, Missouri, USA) and recombinant TG2, as described by Achyuthan and colleagues. 31 Also, plates coated with rabbit antibodies against mouse IgG1 (ICN, Aurora; dissolved in 0.03 M bicarbonate buffer, pH 9.6) and CUB7402 monoclonal TG2 specific capture antibodies were applied to further verify the presence of both TG2 specific IgA and TG2 in the eluates. Bound IgA was detected with peroxidase conjugated rabbit antibodies against human IgA, as described previously. 30 In non-coeliac jejunal biopsy samples, IgA was detected only inside plasma cells and epithelial cells (fig1A). In contrast, clear subepithelial IgA positivity was found along the villous and crypt basement membranes in jejunal samples from all 10 coeliac patients, both in the developing stage of the disease when the villous architecture was still normal and also later when the mucosa had deteriorated to the typical flat lesion (fig 1B, C, same patient). This IgA positivity pattern was identical to the normal tissue localisation of TG2 (fig 1D, G; red) which appeared merged into yellow in the coeliac mucosa due to colocalisation with IgA deposits labelled in green (fig 1E, F, H, I). This colocalisation was also seen on reticulin fibres of lymphoid follicles (fig 1H, asterisk) and on trabecular structures which appeared fragmented in the flat lesion (fig 1F, arrow). IgA in plasma cells or inside crypt epithelial cells did not merge into yellow in either normal or coeliac mucosa. Both coeliac IgA and TG2 were arranged around mucosal vessels, shown in blue (fig 1 I, J, arrows). Coeliac IgA was located on the extracellular surface of laminin (fig 1L), and also showed a close relation to fibronectin (not shown). Similar IgA deposits were present in seven of 11 patients with dermatitis herpetiformis who had a normal jejunal villous structure at diagnosis. Two dermatitis herpetiformis patients who had clear jejunal IgA deposits were negative for circulating IgA class serum endomysial antibodies (fig 1K). Jejunal IgM and IgG positivity did not differ between coeliac and control samples. To establish whether coeliac IgA can reach TG2 in extraintestinal tissues, we examined by double immunofluorescence two coeliac lymph nodes, two liver samples, one appendix, and one skeletal muscle specimen from coeliac patients (table 2, figs 2, 3). Specimens were normal (cases 1, 2, 3, 5, and 6 in table 2) or showed only non-specific changes (case 4, liver steatosis; table 2) by conventional microscopy. All of these tissues contained abundant TG2 in endomysial localisations or around reticulin fibres both in non-coeliac controls (fig 2A, 3B) and in coeliacs (fig 2G, J; fig 3D, F, H, red). Tissue IgA was not related to endomysial or reticulin structures in non-coeliac lymph node (n = 1), appendix (n = 6), or liver (n = 3) samples and appeared only inside epithelial cells or in the centre of lymphoid follicles (fig 2A, D; fig 3A, B, green). However, in coeliac samples there were heavy IgA deposits on reticulin fibres also (lymph nodes, appendix, liver (fig 2B, E, H; fig 3C)) and on endomysial structures (muscular layer of the appendix, skeletal muscle (fig 2F, K)) which were colocalised with tissue TG2 (fig 2C, I, L; fig 3D). In the case of IgA deficiency (cases 5 and 6 in table 2), IgA deposits were missing (fig 3E, F) but TG2 related IgG deposits were seen in the liver (fig 3G, H) and also on archive electron microscopic kidney pictures of case 6 (not shown). Colocalisation of in vivo deposited antibodies with TG2, both in the jejunum and also in extraintestinal tissues, strongly suggested that the specific target for coeliac antibody binding is TG2. To obtain direct evidence, we eluted deposited IgA from extraintestinal tissues for further studies. We used chloroacetic acid to disrupt TG2 binding to fibronectin 28 and to release the in vivo deposited IgA together with released TG2. Treatment of the sections resulted in disappearance of extracellular IgA deposits in sections from both the extrajejunal and jejunal coeliac samples, while IgA in plasma cells and epithelial cells remained visible on immunofluorescence (fig 4A, B). Likewise, in tissues, TG2 became undetectable by monoclonal TG2 specific autoantibodies after elutions with choroacetic acid. To ascertain that both TG2 and IgA are found in eluates, western blot and ELISA studies were conducted with concentrated eluates prepared from a coeliac and a non- coeliac control liver, and from a coeliac lymph node. A clear band at 80 kDa, corresponding to the molecular weight of TG2 was, as expected, equally detectable with monoclonal TG2 antibodies both in coeliac and non-coeliac eluates (fig 4C). Human IgA was also detected both in coeliac and non-coeliac eluates (data not shown). However, when the eluates were tested by ELISA for TG2 specific IgA class antibodies, high optical density values were obtained only with IgA eluted from the coeliac liver and lymph node (0.578 and 2.551, respectively) but not with IgA eluted from the control liver (0.052, representative values from two independent elution experiments). This finding indicates that IgA originating from non-coeliac tissues was not directed against TG2. The result was confirmed in an additional experiment where the eluates were added to ELISA plates coated with TG2 specific monoclonal antibodies and IgA was measured from the bound TG2 human IgA complexes; coeliac liver eluate gave an optical density of 0.493 versus 0.107 obtained with the control liver eluate (fig 4D). Similarly, IgA eluted from coeliac tissues bound to human recombinant TG2 applied on TG2 2 / 2 mouse tissues (fig 4E, F), and also to the endomysial and reticulin structures of human umbilical cord (fig 4I), monkey oesophagus (fig 4J), and normal appendix (fig 4K) sections. In contrast, IgA eluted from the non-coeliac liver did not bind to any of these structures (fig 4G, H). Furthermore, there was no IgA binding to the TG2 2 / 2 mouse tissues with either the coeliac or non-coeliac eluates. The experiments above collectively demonstrate that deposited IgA seen in coeliac tissues is targeted against TG2. In the present study, we showed for the first time that in vivo targeting of TG2 in coeliac disease occurs in the form of in situ endomysial, reticulin, and jejunal subepithelial autoantibody binding. Patient IgA found deposited on extracellular TG2 in the liver, lymph nodes, and muscles indicates that the coeliac disease autoantigen is widely accessible to the intestinally produced 21 circulating ...

Citations

... 2% of the patients we studied presented dental enamel damage. Dental enamel damage is related to malnutrition and hypocalcemia in particular, but also to autoimmunity [5]. Alopecia was encountered in 2% of cases in our study, from the literature it is reported that it was observed in 1% of celiac children, ranging from Alopecia Areata to totalis. ...
Article
Full-text available
Celiac disease, also known as gluten-sensitive enteropathy, is a chronic disorder of digestive tract mediated by immune mechanisms, resulting in an inability to tolerate gluten and its related proteins in genetically predisposed individuals. Gluten is one protein commonly found in wheat, rye and barley. This disease has a wide spectrum clinical presentations and affects a wide age range on which it has a significant impact in terms of development and morbidity. Therefore, we found it important to study investigation of the variety of extraintestinal manifestations in pediatric celiac patients and identification of possible strategies that can be adapted to diagnose and treat them as soon as possible.
... Припускають, що підвищена кишкова проникність може спричинити надходження в портальну циркуляцію (а потім у печінку) токсинів, мікробних та інших антигенів, цитокінів та/або інших медіаторів пошкодження печінки (рис. 2) [27,37,53,68].Однак ураження печінки зазвичай не спостерігається за іншої кишкової патології, пов'язаної з підвищеною кишковою проникністю [30]. ...
Article
Full-text available
Celiac hepatitis is a gluten‑dependent liver disease that usually eliminates after 12 months of a strict gluten‑free diet. What is important is that liver histology also improves after a gluten‑free diet. Hypertransaminasemia (in 13—60% of cases) is common at the untreated celiac disease (CD). Conversely, CD presents in 9% of patients with unexplained hypertransaminasemia. Patients with CD have a higher risk of both subsequent liver disease and death caused by cirrhosis compared to the general population. Pathogenetic mechanisms underlying CD, have not been sufficiently studied. The permeability of the intestinal mucosa appears to be considerably higher in patients with CD and hypertransaminasemia than in patients with CD and normal liver tests. The normalization of intestinal permeability and transaminase levels after following a gluten‑free diet demonstrates that this phenomenon is gluten‑dependent. It has been suggested that increased intestinal permeability promotes the admission of toxins, microbial and other antigens, cytokines, and/or other mediators of liver damage into the portal circulation (and hence to the liver). However, liver damage is not usually observed in other intestinal pathologies associated with increased intestinal permeability. A liver biopsy is rarely performed in celiac hepatitis. Mild and/or non‑specific histological changes are observed. Severe liver fibrosis and cirrhosis occur rarely. Primary biliary cholangitis and autoimmune hepatitis may be associated with CD. The incidence of CD in patients with primary biliary cholangitis is 1—7%, while the incidence of primary biliary cholangitis in patients with CD is 0.1—3%. CD is revealed in 4—6% of patients with autoimmune hepatitis, both type 1 and type 2. Cases of primary sclerosing cholangitis combined with CD have been reported, too. No correlation has been found between CD and chronic hepatitis C. Treatment of hepatitis C with interferon‑a and/or ribavirin may activate occult or latent CD. The incidence of CD in patients with non‑alcoholic fatty liver disease ranges from 3% to 7%. The prevalence of CD in patients after liver transplantation for a variety of reasons ranges from 3% to 4.3%. Liver‑caused mortality is increased in patients with CD, although the absolute risk of mortality from liver failure is low.
... The enzyme transglutaminase 2 (TG2) plays a key role in celiac disease (CeD) pathogenesis, both as a CeD autoantigen and through the generation of immunogenic, deamidated gliadin peptide epitopes recognized by the T-cells [1]. TG2 is thought to localize mainly extracellularly in the lamina propria of the small bowel mucosa [2], where it is a target for humoral immunity [3]. In celiac disease, TG2 forms Int. ...
... A hematoxylineosin-stained biopsy from a CeD patient is shown for reference in Supplementary Figure S3. [2], where it is a target for humoral immunity [3]. In celiac disease, TG2 forms complexes with immunoglobulin A on the subepithelial basement membrane, forming the IgA deposits detectable by immunofluorescence microscopy [3]. ...
... Here, our novel and unexpected finding was that the oral TG2 inhibitor ZED1227 concentrated mainly in the villous enterocytes and not in the lamina propria, where the majority of TG2 protein resides in the duodenal mucosa. The localization of the TG2 protein in the lamina propria, especially in the basement membrane, is extensively demonstrated in the literature [2,3]. Its localization on the luminal surface of the enterocytes has also been documented [6,7,13], but the role of the luminal TG2 in celiac disease pathogenesis has remained unclear. ...
Article
Full-text available
The enzyme transglutaminase 2 (TG2) plays a key role in celiac disease (CeD) pathogenesis. Active TG2 is located mainly extracellularly in the lamina propria but also in the villous enterocytes of the duodenum. The TG2 inhibitor ZED1227 is a promising drug candidate for treating CeD and is designed to block the TG2-catalyzed deamidation and crosslinking of gliadin peptides. Our aim was to study the accumulation of ZED1227 after oral administration of the drug. We studied duodenal biopsies derived from a phase 2a clinical drug trial using an antibody that detects ZED1227 when bound to the catalytic center of TG2. Human epithelial organoids were studied in vitro for the effect of ZED1227 on the activity of TG2 using the 5-biotin-pentylamine assay. The ZED1227-TG2 complex was found mainly in the villous enterocytes in post-treatment biopsies. The signal of ZED1227-TG2 was strongest in the luminal epithelial brush border, while the intensity of the signal in the lamina propria was only ~20% of that in the villous enterocytes. No signal specific to ZED1227 could be detected in pretreatment biopsies or in biopsies from patients randomized to the placebo treatment arm. ZED1227-TG2 staining co-localized with total TG2 and native and deamidated gliadin peptides on the enterocyte luminal surface. Inhibition of TG2 activity by ZED1227 was demonstrated in epithelial organoids. Our findings suggest that active TG2 is present at the luminal side of the villous epithelium and that inhibition of TG2 activity by ZED1227 occurs already there before gliadin peptides enter the lamina propria.
... Finally, studies have shown that tissue transglutaminase 2, a ubiquitous enzyme active in tissue repair, fibrogenesis, apoptosis, and inflammation plays a role in celiac-associated liver damage. It is an important target of the anti-tissue transglutaminase (anti-TTG) autoantibody, often found in patients with CeD, which inhibits enzyme activity leading to liver injury [16,19]. ...
... In our study, the higher odds of having AIH, PBC and PSC when anti-TTG is positive is thought-provoking, and in the case of AIH and PBC, the odds remain significant after adjusting for the presence of CeD. This corroborates the notion that subjects with positive anti-TTG are at higher risk of developing AIH, PBC and PSC, as suggested in prior studies [16,19]. ...
Article
Background: While there is higher prevalence of autoimmune, cholestatic and fatty liver disease in celiac disease (CeD), most data is from small-scale studies. We evaluated the prevalence and risk factors of the same using large cohort data. Methods: A population-based cross-sectional study was conducted using Explorys, a multi-institutional database. Prevalence and risk factors of autoimmune hepatitis (AIH), primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC) and nonalcoholic fatty liver disease (NAFLD) in CeD were assessed. Results: Out of 70 352 325 subjects, 136 735 had CeD (0.19%). The prevalence of AIH (0.32%), PBC (0.15%), PSC (0.004%) and NAFLD (0.7%) were high in CeD. After adjusting for age, gender, Caucasian race and anti-tissue transglutaminase antibody (anti-TTG), CeD subjects had higher odds of AIH [adjusted odds ratio (aOR) 7.06, 95% confidence interval (CI) 6.32-7.89] and PBC (aOR 4.16, 95% CI 3.46-5.0). Even after adjusting for CeD, anti-TTG positivity concurred with higher odds of AIH (aOR 4.79, 95% CI 3.88-5.92) and PBC (aOR 9.22, 95% CI 7.03-12.1). After adjusting for age, gender, Caucasian race, diabetes mellitus (DM), obesity, hypothyroidism and metabolic syndrome, there was higher prevalence of NAFLD in CeD, with the aOR in the presence of DM type 1 being 2.1 (95% CI 1.96-2.25), and in the presence of DM type 2 being 2.92 (95% CI 2.72-3.14). Conclusion: Subjects with CeD are more likely to have AIH, PBC, PSC and NAFLD. AIH and PBC have higher odds in the presence of anti-TTG. The odds of NAFLD in CeD are high regardless of type of DM.
... A B-sejt aktiváció végeredménye mind coeliakiában, mind DH-ban TG2-ellenes IgA antitestek termelése lesz (1,70), melyek egyrészt kimutathatók a keringésből TG2 ELISA-val és EMA-vizsgálattal, másrészt in vivo is kötődnek a beteg különböző kötőszöveteihez, pl. a tápcsatorna részeihez, nyirokcsomókhoz, májhoz, vázizom endomysiumhoz (71). A fenti immun-és autoimmun folyamatok hatására enteropathiához, ill. ...
Article
In this review article, after a short introduction, the authors present the pathogenesis and pathomechanism of dermatitis herpetiformis from the perspective of the transglutaminase enzyme family. According to our current knowledge, these isoenzymes consist of nine members in humans, and four of them play a role in the development of the disease. The main autoantigen is probably epidermal transglutaminase, but the role of other isozymes cannot be neglected either. The mechanism of the development of gluten-sensitive enteropathy is presented, followed by the theory of formation of IgA-epidermal transglutaminase immune complexes. The article also briefly covers the associated disorders of the blood coagulation system and neurological conditions.
... Extent of involvement usually correlates with overall disease severity (histopathological and serological). Duodenal damage may expose the liver to hepatotoxins because of the increased permeability, and autoimmune factors may also play a role suggested by the deposition of CD antibodies in the liver [43,44]. Liver involvement is usually mild and reversible and rarely causes liver failure [45]. ...
Chapter
Full-text available
Celiac disease can involve any organ system, leading to various non-classical or atypical manifestations. These atypical signs and symptoms have been seen increasingly in the last few decades, both in children and adults, which may or may not involve the gastrointestinal system. This transition from a malabsorptive disorder causing GI symptoms and malnutrition to a more subtle condition causing a variety of extraintestinal manifestations led to newer nomenclature of gastrointestinal and extraintestinal signs and symptoms. Infancy and early childhood onset celiac disease may have a predominance of gastrointestinal manifestations leading to protein energy malnutrition and failure to thrive. The late presentation may have subtle manifestations, and extraintestinal signs and symptoms may be commoner. Short stature, delayed puberty, osteopenia, neuropsychiatric manifestations, iron-deficiency anemia, and elevated liver enzymes are common extraintestinal symptoms. The pathogenesis of extraintestinal manifestations may be due to malabsorption or associated with a systemic autoimmune response. These atypical presentations, especially in the absence of gastrointestinal symptoms and family history, may be missed, leading to a delay in diagnosis and management. A suitable case-finding strategy and liberal use of serological tests may improve the detection rate of CD.
... I-anti-TG2 can be detected by double immunofluorescence staining on frozen duodenal sections (deposit technique) (8) or using an endomysial antibody assay in the supernatants of duodenal biopsies after incubation with gliadin fragments for 72 hours (EMA biopsy) (9). The diagnostic accuracy of these techniques is comparable because they both have high sensitivity and specificity. ...
Article
Full-text available
Introduction: Intestinal antitransglutaminase antibodies (I-anti-TG2) are a specific marker of celiac disease (CeD). The aim of this study was to evaluate the diagnostic accuracy of a novel application of an immunochromatographic assay referred to as Rapid_AntiTG2 to detect I-anti-TG2 on intestinal biopsy lysate. Methods: Consecutive pediatric patients referred to a single center for elective upper endoscopy were enrolled. Biopsies were taken from duodenal bulb and distal duodenum. For each sampling site, 2 biopsies were analyzed for standard histology, 1 biopsy was cultured to perform the reference standard assay for I-anti-TG2 detection (endomysium [EMA] biopsy), and 1 biopsy was mechanically lysed to perform Rapid_AntiTG2. The primary outcome was the diagnostic accuracy of Rapid_AntiTG2 on biopsy lysate compared with that of the gold standard (serology + histopathology) for CeD diagnosis. The secondary outcome was the agreement of Rapid_AntiTG2 with EMA biopsy. Results: One hundred forty-eight patients were included. Of them, 79 were those with CeD (64 classical CeD, 2 seronegative CeD, and 13 potential CeD) and 69 were controls. Rapid_AntiTG2 on biopsy lysate had very high diagnostic accuracy (sensitivity 100%, specificity 97%, LR+ 34.1, LR- 0.01) in separating patients with CeD from controls. Diagnostic accuracy was unchanged in patients with potential and seronegative CeD. Rapid_AntiTG2 on biopsy lysate had almost perfect agreement with the EMA biopsy reference test (99% agreement, Cohen K 0.97). Discussion: I-anti-TG2 can be detected with an immunochromatographic assay after simple mechanical lysis of fresh intestinal biopsy with very high diagnostic accuracy. The test is quick and easy to perform and can be widely available in any endoscopy unit. Its implementation would allow a better understanding of the prognostic value of I-anti-TG2 and help clinicians in cases of suspected CeD that are difficult to classify.
... TG2 contributes to ataxia and CD development in at least two ways: by deamidating gluten peptides, increasing their affinity for HLA-DQ2/DQ8, and potentiating the eventual T cell-mediated response [81]. ATG2A are deposited in the small bowel mucosa of CD patients, even in the absence of enteropathy, and in extra-intestinal locations, such as muscle and liver [82]. The broad deposition of ATGA has also been detected in the proximity of brain blood vessels in patients with gluten ataxia [25,63,71]. ...
Article
Full-text available
Celiac disease (CD) is a complex multi-organ disease with a high prevalence of extra-intestinal involvement, including neurological and psychiatric manifestations, such as cerebellar ataxia, peripheral neuropathy, epilepsy, headache, cognitive impairment, and depression. However, the mechanisms behind the neurological involvement in CD remain controversial. Recent evidence shows these can be related to gluten-mediated pathogenesis, including antibody cross-reaction, deposition of immune-complex, direct neurotoxicity, and in severe cases, vitamins or nutrients deficiency. Here, we have summarized new evidence related to gut microbiota and the so-called “gut-liver-brain axis” involved in CD-related neurological manifestations. Additionally, there has yet to be an agreement on whether serological or neurophysiological findings can effectively early diagnose and properly monitor CD-associated neurological involvement; notably, most of them can revert to normal with a rigorous gluten-free diet. Moving from a molecular level to a symptom-based approach, clinical, serological, and neurophysiology data might help to disentangle the many-faceted interactions between the gut and brain in CD. Eventually, the identification of multimodal biomarkers might help diagnose, monitor, and improve the quality of life of patients with “neuroCD”.
... The same group also reported 68% positivity for deposits in non-diabetic PCD, although 90% of overt CD patients had these deposits. 17 In our study, 93.8% of T1D patients were dsIF positive, compared to 85.7% of nondiabetics. Similar to a report by Kaukinen et al., all our controls were dsIF negative. ...
Article
Full-text available
Background Celiac disease (CD) is frequently associated with type I diabetes mellitus (T1D), where its diagnosis may be a challenging task. This study aims to test the usefulness of the double staining immunofluorescence (dsIF) technique for the detection of intestinal anti-tissue transglutaminase specific IgA antibody (tTG-IgA) deposits in CD and T1D children with coexisting CD. Methods A total of 46 patients (30 cases of CD and 16 cases of T1D with CD) and 16 non-diabetic, non-celiac children were recruited. Endoscopic biopsies were taken and analyzed by light microscopy, quantitative histology (QH), and a dsIF technique. Results Histologically, villous atrophy was most severe in CD, followed by T1D with CD, while all control biopsies except 1 were normal. QH showed a statistically significant difference in villous height (Vh), crypt depth (CrD), and Vh:CrD ratio between diabetic and non-diabetic patients with CD. dsIF technique could detect tTG-IgA deposits in 85.7% of cases of CD alone and 93.8% of biopsies from diabetic children. Surprisingly, deposits were more extensive in biopsies with minimal villous shortening. Also, all 5 biopsies from T1D patients with normal histology were dsIF positive. Conclusion In-situ analysis of tTG-IgA immune deposits facilitates the detection of positive serology early-onset CD. Quantitative analysis may be used as an ancillary tool to increase the reliability of histological findings in these patients.
... Продукция антител у людей с целиакией происходит в слизистой оболочке кишечника, о чем свидетельствует наличие иммунных комплексов, обнаруживаемых с помощью иммунофлуоресценции. Аутоантитела последовательно проникают в кровеносные сосуды через слизистую оболочку [20]. Однако при СНЦ антитела не могут проходить через собственную пластинку и не попадают в кровоток. ...
Article
Full-text available
It is about seronegative celiac disease