Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.824
Filter
1.
Nature ; 607(7920): 762-768, 2022 07.
Article in English | MEDLINE | ID: mdl-35794484

ABSTRACT

Gastrointestinal health depends on the adaptive immune system tolerating the foreign proteins in food1,2. This tolerance is paradoxical because the immune system normally attacks foreign substances by generating inflammation. Here we addressed this conundrum by using a sensitive cell enrichment method to show that polyclonal CD4+ T cells responded to food peptides, including a natural one from gliadin, by proliferating weakly in secondary lymphoid organs of the gut-liver axis owing to the action of regulatory T cells. A few food-specific T cells then differentiated into T follicular helper cells that promoted a weak antibody response. Most cells in the expanded population, however, lacked canonical T helper lineage markers and fell into five subsets dominated by naive-like or T follicular helper-like anergic cells with limited capacity to form inflammatory T helper 1 cells. Eventually, many of the T helper lineage-negative cells became regulatory T cells themselves through an interleukin-2-dependent mechanism. Our results indicate that exposure to food antigens causes cognate CD4+ naive T cells to form a complex set of noncanonical hyporesponsive T helper cell subsets that lack the inflammatory functions needed to cause gut pathology and yet have the potential to produce regulatory T cells that may suppress it.


Subject(s)
CD4-Positive T-Lymphocytes , Food , Immune Tolerance , Allergens/immunology , Antibody Formation , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Dietary Proteins/immunology , Gastrointestinal Tract/cytology , Gastrointestinal Tract/immunology , Gliadin/immunology , Immune Tolerance/immunology , Inflammation , Interleukin-2/immunology , Liver/cytology , Liver/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Peptide Fragments/immunology , T Follicular Helper Cells/cytology , T Follicular Helper Cells/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/cytology , Th1 Cells/immunology
2.
J Paediatr Child Health ; 58(9): 1648-1652, 2022 09.
Article in English | MEDLINE | ID: mdl-35726522

ABSTRACT

AIM: Deamidated gliadin peptide-IgG (DGP-IgG) antibody serology testing is widely utilised in screening for coeliac disease in Australia; however, it is used sparingly in Europe. The aim of this study was to assess the diagnostic value of a positive DGP-IgG in the setting of a negative tissue transglutaminase-IgA (tTG-IgA) for gastrointestinal pathology among paediatric patients. METHODS: We conducted a retrospective cohort study of all children with an elevated DGP-IgG in the setting of a negative tTG-IgA who underwent gastroscopy over a 48-month period (January 2015-December 2018) at a tertiary paediatric centre. They were identified utilising the electronic pathology database and demographic and clinical data were collected from electronic medical records. Patients who had previously been diagnosed with coeliac disease were on a gluten-free diet or over the age of 18 were excluded from the study. RESULTS: Twenty-six patients with an elevated DGP-IgG in the setting of a negative tTG-IgA underwent gastroscopy. Our study yielded a positive predictive value of 1/26 (3.9% CI 95% 0.7%, 18.9%) for the diagnosis of coeliac disease. Overall, there were 25 histopathological diagnoses including 1 diagnosis of coeliac disease among the total 26 patients who were positive DGP-IgG and negative tTG-IgA and underwent gastroscopy. CONCLUSIONS: Our findings suggest that an isolated positive DGP-IgG has a very low diagnostic yield for coeliac disease in children and may be indicative of other gastrointestinal pathology.


Subject(s)
Celiac Disease , Immunoglobulin G , Autoantibodies , Celiac Disease/diagnosis , Child , Gliadin/immunology , Humans , Immunoglobulin A , Immunoglobulin G/analysis , Peptides , Retrospective Studies , Sensitivity and Specificity , Transglutaminases
3.
Rinsho Shinkeigaku ; 62(2): 112-122, 2022 Feb 19.
Article in Japanese | MEDLINE | ID: mdl-35095045

ABSTRACT

BACKGROUND: Autoimmune cerebellar ataxia (AICA) is a general term for diseases in which the cerebellum is damaged by an autoimmune mechanism. For the diagnosis of the AICA, anti-thyroid antibodies (anti-thyroid peroxidase antibody and anti-thyroglobulin antibody), anti-glutamic acid decarboxylase (GAD) antibodies, and anti-gliadin antibodies are measured. Immunotherapy is known to be effective for AICA, but some patients with effective immunotherapy lack autoantibodies associated with cerebellar ataxia. The purpose of this study was to clarify whether the effectiveness of immunotherapy in patients with suspected AICA could be predicted by anti-mouse cerebellar tissue-derived antigen antibody tests. METHODS: This study was conducted on 25 patients with idiopathic cerebellar ataxia (excluding multiple system atrophy, hereditary spinocerebellar degeneration, cancer-bearing patients, and patients taking phenytoin) who received immunotherapy from 2005 to 2016 at Tokyo Medical University Hachioji Medical Center. The patients were suspected of having AICA because they were positive for cerebellar ataxia-related autoantibodies (anti-thyroid antibody, anti-GAD antibody, anti-gliadin antibody, or anti-transglutaminase 6 antibody) or other autoantibodies. Antibodies that bind to mouse cerebellar tissue-derived antigens were defined as "anti-mouse cerebellar tissue-derived antigen antibodies" in this study, and their IgG-class antibodies were comprehensively measured using a slot blot. RESULTS: Anti-mouse cerebellar tissue-derived antigen antibody test results were correlated with immunotherapy efficacy. Furthermore, the combination of anti-mouse cerebellar tissue-derived antigen and anti-GAD antibody tests could predict the effectiveness of immunotherapy with 83% sensitivity and 100% specificity, while the combination of the anti-mouse cerebellar tissue-derived antigen, anti-GAD, and anti-gliadin (IgA class) antibody tests could predict the effectiveness of immunotherapy with 94% sensitivity and 86% specificity. CONCLUSION: Anti-mouse cerebellar tissue-derived antigen antibody tests could help to provide useful information for immunotherapy administration to patients with idiopathic cerebellar ataxia suspected to be AICA.


Subject(s)
Cerebellar Ataxia , Immunotherapy , Animals , Autoantibodies , Cerebellar Ataxia/diagnosis , Cerebellum , Gliadin/immunology , Glutamate Decarboxylase/immunology , Humans , Immunoglobulin G , Immunologic Factors
4.
Endocr Res ; 47(1): 26-31, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34374618

ABSTRACT

BACKGROUND: The general practice is to screen patients with autoimmune thyroid disease for celiac disease (CD); however, optimal timing for CD screening for patients with Graves'Disease (GD) has not been identified yet. The aim of the study was to show whether positive celiac antibodies persist after euthyroidism is achieved. MATERIALS AND METHODS: Serum samples were collected from 35 patients with GD (23 female and 12 male) who applied to the endocrine outpatient clinic. Patients and healthy controls were screened for CD with IgG and IgA antigliadin antibodies (IgG - AGA and IgA - AGA), IgA endomysial antibody (IgA-EMA) and IgA tissue transglutaminase antibody (IgA anti-tTG). These antibodies were reevaluated when patients were euthyroid under antithyroid therapy. Small intestine biopsy was offered to the patients who remained antibody positive after being euthyroid. RESULTS: Screening 35 patients with GD revealed positive results for IgA-AGA (n = 6/35, 17%), IgG-AGA (n = 9/35, 26%), IgA-EmA (n = 2/35, 6%) and IgA-tTG (n = 2/35, 6%). No patient had multiple antibodies positive. Selective IgA deficiency was not detected in patients and controls. When patients were euthyroid, baseline positive IgA-AGA, IgG-AGA, and IgA-EmA became negative, while positive anti-tTG persisted in two patients. Endoscopic duodenal biopsy showed a normal villi/crypts ratio in these patients. None of the controls had positive antibodies. CONCLUSION: Due to possibility of false seropositivity of celiac antibodies in patients with Graves' thyrotoxicosis, one should defer testing for CD until euthyroidism has been achieved.


Subject(s)
Celiac Disease , Graves Disease , Autoantibodies/blood , Celiac Disease/blood , Celiac Disease/diagnosis , Female , Gliadin/immunology , Humans , Immunoglobulin A/blood , Male , Sensitivity and Specificity , Transglutaminases/immunology
5.
Nutrients ; 13(12)2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34960101

ABSTRACT

Celiac disease (CD) is a genetically predisposed, T cell-mediated and autoimmune-like disorder caused by dietary exposure to the storage proteins of wheat and related cereals. A gluten-free diet (GFD) is the only treatment available for CD. The celiac immune response mediated by CD4+ T-cells can be assessed with a short-term oral gluten challenge. This study aimed to determine whether the consumption of bread made using flour from a low-gluten RNAi wheat line (named E82) can activate the immune response in DQ2.5-positive patients with CD after a blind crossover challenge. The experimental protocol included assessing IFN-γ production by peripheral blood mononuclear cells (PBMCs), evaluating gastrointestinal symptoms, and measuring gluten immunogenic peptides (GIP) in stool samples. The response of PBMCs was not significant to gliadin and the 33-mer peptide after E82 bread consumption. In contrast, PBMCs reacted significantly to Standard bread. This lack of immune response is correlated with the fact that, after E82 bread consumption, stool samples from patients with CD showed very low levels of GIP, and the symptoms were comparable to those of the GFD. This pilot study provides evidence that bread from RNAi E82 flour does not elicit an immune response after a short-term oral challenge and could help manage GFD in patients with CD.


Subject(s)
Bread , Celiac Disease/immunology , Diet, Gluten-Free , Gliadin/genetics , Gliadin/immunology , Glutens/immunology , RNA Interference , Triticum/genetics , Triticum/immunology , Adult , Celiac Disease/genetics , Female , Humans , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Pilot Projects , RNA Interference/immunology , Triticum/chemistry , Young Adult
6.
PLoS One ; 16(11): e0259082, 2021.
Article in English | MEDLINE | ID: mdl-34731200

ABSTRACT

A hallmark of celiac disease is the gluten-dependent production of antibodies specific for deamidated gluten peptides (DGP) and the enzyme transglutaminase 2 (TG2). Both types of antibodies are believed to result from B cells receiving help from gluten-specific CD4+ T cells and differentiating into antibody-producing plasma cells. We have here studied the collaboration between DGP- and TG2-specific B cells with gluten-specific CD4+ T cells using transgenic mice expressing celiac patient-derived T-cell and B-cell receptors, as well as between B-cell transfectants and patient-derived gluten-specific T-cell clones. We show that multivalent TG2-gluten complexes are efficient antigens for both TG2-specific and DGP-specific B cells and allow both types of B cells to receive help from gluten-specific T cells of many different specificities.


Subject(s)
Celiac Disease/genetics , Glutens/genetics , Protein Glutamine gamma Glutamyltransferase 2/genetics , Receptors, Antigen, B-Cell/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Celiac Disease/immunology , Celiac Disease/pathology , Gliadin/genetics , Gliadin/immunology , Glutens/immunology , Humans , Mice , Mice, Transgenic , Protein Glutamine gamma Glutamyltransferase 2/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
7.
J Clin Lab Anal ; 35(12): e23913, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34689365

ABSTRACT

BACKGROUND: Test utilization for the diagnosis of celiac disease may affect the prevalence and incidence of the disease in Korea. We aimed to investigate the test utilization of serological biomarkers for celiac disease in Korea. METHODS: We retrospectively investigated the test utilization of tissue transglutaminase IgA, gliadin IgA and IgG, and endomysial IgA antibody (Ab) assays between January 2011 and June 2020. RESULTS: During a nine-year-and-six-month study period, overall 307,322,606 clinical tests were requested from different clinical settings, such as local clinics, hospitals, university hospitals, and tertiary medical centers. Among them, only 58 tissue transglutaminase IgA, 22 gliadin IgA, 12 gliadin IgG, and 16 endomysial IgA Ab tests were performed on 79 Korean patients. Among them, one patient had positive transglutaminase IgA Ab result (1.3%). CONCLUSION: Low prevalence and incidence of celiac disease in Korea may be due to an underutilization of diagnostic assays.


Subject(s)
Celiac Disease/diagnosis , Serologic Tests/statistics & numerical data , Celiac Disease/epidemiology , Diagnostic Tests, Routine , Gliadin/immunology , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Protein Glutamine gamma Glutamyltransferase 2/immunology , Republic of Korea/epidemiology , Retrospective Studies
8.
Int J Mol Sci ; 22(17)2021 Aug 27.
Article in English | MEDLINE | ID: mdl-34502187

ABSTRACT

Gluten-related disorders (GRDs) are a group of diseases that involve the activation of the immune system triggered by the ingestion of gluten, with a worldwide prevalence of 5%. Among them, Celiac disease (CeD) is a T-cell-mediated autoimmune disease causing a plethora of symptoms from diarrhea and malabsorption to lymphoma. Even though GRDs have been intensively studied, the environmental triggers promoting the diverse reactions to gluten proteins in susceptible individuals remain elusive. It has been proposed that pathogens could act as disease-causing environmental triggers of CeD by molecular mimicry mechanisms. Additionally, it could also be possible that unrecognized molecular, structural, and physical parallels between gluten and pathogens have a relevant role. Herein, we report sequence, structural and physical similarities of the two most relevant gluten peptides, the 33-mer and p31-43 gliadin peptides, with bacterial pathogens using bioinformatics going beyond the molecular mimicry hypothesis. First, a stringent BLASTp search using the two gliadin peptides identified high sequence similarity regions within pathogen-derived proteins, e.g., extracellular proteins from Streptococcus pneumoniae and Granulicatella sp. Second, molecular dynamics calculations of an updated α-2-gliadin model revealed close spatial localization and solvent-exposure of the 33-mer and p31-43 peptide, which was compared with the pathogen-related proteins by homology models and localization predictors. We found putative functions of the identified pathogen-derived sequence by identifying T-cell epitopes and SH3/WW-binding domains. Finally, shape and size parallels between the pathogens and the superstructures of gliadin peptides gave rise to novel hypotheses about activation of innate immunity and dysbiosis. Based on our structural findings and the similarities with the bacterial pathogens, evidence emerges that these pathologically relevant gluten-derived peptides could behave as non-replicating pathogens opening new research questions in the interface of innate immunity, microbiome, and food research.


Subject(s)
Celiac Disease/immunology , Epitopes, T-Lymphocyte , Gliadin/metabolism , Glutens/metabolism , Molecular Mimicry , Peptide Fragments/metabolism , Carnobacteriaceae/metabolism , Computational Biology , Gliadin/chemistry , Gliadin/immunology , Glutens/chemistry , Glutens/immunology , Humans , Peptide Fragments/chemistry , Peptide Fragments/immunology , Streptococcus pneumoniae/metabolism
9.
Hum Immunol ; 82(12): 930-936, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34462157

ABSTRACT

Coeliac disease (CD) is an autoimmune gastroenteropathy triggered by gliadin and gliadin-tissue transglutaminase (tTG) complexes. CD is one of the few autoimmune diseases with an accurate, non-invasive serological test. Anti-endomysial, anti-tTG and anti-deaminated gliadin peptides (DGP) antibodies are currently used for serological tests with tTG ELISAs being the superior test. Duodenal biopsy, although invasive, is the gold standard for CD diagnosis. HLA genotyping and flow cytometry can also be used as supplementary tests. The incidence of CD is rising globally although the reasons for this remain unclear. In addition, the true incidence of coeliac disease in African populations remains unknown although recent work suggests that South African populations express the alleles associated with this disease. This review examines the pathogenesis and diagnosis of coeliac disease and considers novel and innovative biomarkers in its diagnosis specifically in an African population.


Subject(s)
Antibodies/immunology , Celiac Disease/diagnosis , Duodenum/immunology , Gliadin/immunology , HLA Antigens/immunology , Protein Glutamine gamma Glutamyltransferase 2/immunology , Biomarkers , Celiac Disease/genetics , Celiac Disease/immunology , HLA Antigens/genetics , Humans
10.
Nutrients ; 13(8)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34444682

ABSTRACT

The safety and health effects for celiac people of a novel beverage (SOFB) developed from sprouted oat flour by fermentation with Lactobacillus plantarum was explored. In vitro reactivity against anti-gliadin antibodies (AGA) and antioxidant/anti-inflammatory potential of SOFB in RAW 264.7 macrophages and Caco-2 cells were evaluated. Immunoreactivity against AGA and antioxidant activity were not detected in SOFB, but it exhibited significant anti-inflammatory activity. The tolerability and impact of SOFB consumption for 6 months on nutritional status and intestinal microbiota composition were investigated in 10 celiac adults (five treated and five control). SOFB consumption did not adversely affect duodenal mucosa nor the total IgA or anti-tissue transglutaminase antibody (IgA-tTG) levels in celiac participants, but it significantly decreased total cholesterol levels at all sampling times and folic acid levels at the end of the study compared to the placebo beverage. SOFB administration also shifted gut microbiota, leading to a higher relative abundance of some beneficial bacteria including the genera Subdoligranulum, Ruminococcus and Lactobacillus in the SOFB group. This study provides supporting evidence of the safety of health benefits of a novel functional beverage produced from sprouted oat.


Subject(s)
Avena , Celiac Disease/diet therapy , Fermented Foods , Seedlings , Animals , Anti-Inflammatory Agents , Antibodies/immunology , Antioxidants , Avena/immunology , Caco-2 Cells , Functional Food , Gastrointestinal Microbiome , Gliadin/immunology , Glutens/analysis , Humans , Immunoglobulin A/analysis , Immunoglobulin A/immunology , Lactobacillus plantarum/metabolism , Mice , Nutritional Status , RAW 264.7 Cells
11.
Int J Mol Sci ; 22(13)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34209932

ABSTRACT

Enzymatic transamidation of gliadins by microbial transglutaminase (mTG) inhibits interferon-γ (IFN-γ) secretion by intestinal T cell lines in patients with celiac disease (CD). To gain insight into the cellular mechanisms underlying the down-regulatory effects of transamidation, we tested a single recombinant α-gliadin (r-gliadin) harbouring two immunodominant peptides, p13 (aa. 120-139) and p23 (aa. 220-239), in HLA-DQ8 transgenic mice, a model of gluten sensitivity. Mice were intranasally immunised with r-gliadin or r-gliadin transamidated by mTG (K-r-gliadin) along with cholera toxin, and the response of mesenteric lymph node cells was analysed by cytokine multiplex assay. An in vitro challenge with r-gliadin was characterised by secretion of specific cytokines featuring both innate immunity and the Th1/Th2/Th17 pattern of the adaptive response. Notably, transamidation specifically down-regulated the Th1 response. Structural studies performed on K-r-gliadin confirmed that specific glutamine residues in p13 and p23, previously found to be deamidated by tissue transglutaminase, were also transamidated by mTG. In silico analysis, simulating p13 and p23 peptide binding to HLA-DQ8 showed that these glutamines, in the form of glutamate, could interact by means of salt bridges with peculiar amino acids of the alpha chain of HLA-DQ8, suggesting that their transamidation may influence the HLA-restricted recognition of these peptides. Thus, the structural findings provided a rationale to explain the down-regulation of the r-gliadin-specific Th1 response following transamidation.


Subject(s)
Celiac Disease/drug therapy , Cholera Toxin/administration & dosage , Cytokines/metabolism , Gliadin/administration & dosage , HLA-DQ Antigens/genetics , Transglutaminases/metabolism , Administration, Intranasal , Animals , Celiac Disease/genetics , Celiac Disease/immunology , Cholera Toxin/immunology , Cytokines/drug effects , Disease Models, Animal , Down-Regulation , Gene Expression Regulation , Gliadin/chemistry , Gliadin/genetics , Gliadin/immunology , HLA-DQ Antigens/metabolism , Immunization , Immunodominant Epitopes/immunology , Mice , Mice, Transgenic , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology
12.
Amino Acids ; 53(7): 1051-1063, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34059947

ABSTRACT

Celiac disease (CeD) is a T-cell-dependent enteropathy with autoimmune features where tissue transglutaminase (TG2)-mediated posttranslational modification of gliadin peptides has a decisive role in the pathomechanism. The humoral immune response is reported to target mainly TG2-deamidated γ-gliadin peptides. However, α-gliadin peptides, like p57-68, playing a crucial role in the T-cell response, and p31-43, a major trigger of innate responses, also contain B-cell gliadin epitopes and γ-gliadin like motifs. We aimed to identify if there are anti-gliadin-specific antibodies in CeD patients targeting the p31-43 and p57-68 peptides and to examine whether deamidation of these peptides could increase their antigenicity. We explored TG2-mediated deamidation of the p31-43 and p57-68 peptides, and investigated serum antibody reactivity toward the native and deamidated α and γ-gliadin peptides in children with confirmed CeD and in prospectively followed infants at increased risk for developing CeD. We affinity-purified antibody populations utilizing different single peptide gliadin antigens and tested their binding preferences for cross-reactivity in real-time interaction assays based on bio-layer interferometry. Our results demonstrate that there is serum reactivity toward p31-43 and p57-68 peptides, which is due to cross-reactive γ-gliadin specific antibodies. These γ-gliadin specific antibodies represent the first appearing antibody population in infancy and they dominate the serum reactivity of CeD patients even later on and without preference for deamidation. However, for the homologous epitope sequences in α-gliadins shorter than the core QPEQPFP heptapeptide, deamidation facilitates antibody recognition. These findings reveal the presence of cross-reactive antibodies in CeD patients recognizing the disease-relevant α-gliadins.


Subject(s)
Autoantibodies/immunology , Celiac Disease/metabolism , Gliadin/metabolism , Peptide Fragments/metabolism , Protein Glutamine gamma Glutamyltransferase 2/immunology , Adolescent , Amides/chemistry , Autoantibodies/metabolism , Celiac Disease/immunology , Child , Child, Preschool , Cross Reactions , Epitopes/immunology , Gliadin/immunology , Humans , Infant , Peptide Fragments/immunology , Protein Glutamine gamma Glutamyltransferase 2/chemistry , Protein Glutamine gamma Glutamyltransferase 2/metabolism
13.
Dermatol Online J ; 27(4)2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33999573

ABSTRACT

Non-celiac gluten sensitivity is often clinically indistinguishable from celiac disease, and patients show improvement or resolution of their symptoms with a gluten-free diet. In contrast to celiac disease, the effects of gluten on the skin and hair in the context of non-celiac gluten sensitivity are not as clear. This review aims to describe the impact of gluten on the skin and hair in patients with non-celiac gluten sensitivity and those without a definitive celiac disease diagnosis. A literature search was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) reporting guidelines for systematic reviews. Forty-two publications met inclusion criteria with five studies describing the skin manifestations of non-celiac gluten sensitivity. Trials identifying the impact of a gluten-free diet on skin disease, as well as dermatologic conditions and their associations with antigliadin antibodies were also identified. Dermatologic manifestations in patients with non-celiac gluten sensitivity vary and may be non-specific. It may be appropriate for some of these patients with skin manifestations to trial a gluten-free diet. Dermatologic conditions that may respond positively to a gluten-free diet include psoriasis, atopic dermatitis, vitiligo, and palmoplantar pustulosis, while linear IgA disease does not appear to improve with this dietary change.


Subject(s)
Diet, Gluten-Free , Glutens/adverse effects , Hair Diseases/etiology , Skin Diseases/etiology , Antibodies , Gliadin/immunology , Glutens/pharmacology , Hair/pathology , Humans , Skin/pathology
14.
Biomolecules ; 11(3)2021 03 17.
Article in English | MEDLINE | ID: mdl-33802942

ABSTRACT

Celiac disease is an autoimmune disorder triggered by toxic peptides derived from incompletely digested glutens in the stomach. Peptidases that can digest the toxic peptides may formulate an oral enzyme therapy to improve the patients' health condition. Bga1903 is a serine endopeptidase secreted by Burkholderia gladioli. The preproprotein of Bga1903 consists of an N-terminal signal peptide, a propeptide region, and an enzymatic domain that belongs to the S8 subfamily. Bga1903 could be secreted into the culture medium when it was expressed in E. coli. The purified Bga1903 is capable of hydrolyzing the gluten-derived toxic peptides, such as the 33- and 26-mer peptides, with the preference for the peptide bonds at the carbonyl site of glutamine (P1 position). The kinetic assay of Bga1903 toward the chromogenic substrate Z-HPQ-pNA at 37 °C, pH 7.0, suggests that the values of Km and kcat are 0.44 ± 0.1 mM and 17.8 ± 0.4 s-1, respectively. The addition of Bga1903 in the wort during the fermentation step of beer could help in making gluten-free beer. In summary, Bga1903 is usable to reduce the gluten content in processed foods and represents a good candidate for protein engineering/modification aimed to efficiently digest the gluten at the gastric condition.


Subject(s)
Bacterial Proteins/metabolism , Burkholderia gladioli/enzymology , Celiac Disease/metabolism , Glutens/metabolism , Peptides/metabolism , Serine Proteases/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Beer , Burkholderia gladioli/genetics , Celiac Disease/immunology , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Fermentation , Gliadin/immunology , Gliadin/metabolism , Glutens/immunology , Humans , Hydrolysis , Peptides/immunology , Recombinant Proteins/metabolism , Serine Proteases/genetics , Substrate Specificity
15.
Food Chem ; 355: 129514, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-33774225

ABSTRACT

The gluten protein found in a variety of cereal grains is a food allergen that can elicit a spectrum of immuno-inflammatory responses in people. Consumer awareness has prompted changes in food labeling requirements, expanded gluten-free food product availability and increased demand for effective gluten testing methodologies. To meet the challenges associated with gluten testing from diverse and complex foods we developed a lateral flow immunoassay (LFIA) using a pair of novel gliadin monoclonal antibodies (MAbs). Using a visual gold reporter, we show sensitive gluten detection (150 ng/mL) from complex food substrates using a fast (<5 min) and easy testing methodology. In this report we characterize the binding properties of a cohort of newly generated gliadin monoclonal antibodies suitable for gluten detection using multiple assay formats and introduce a novel plug-n-play test strip platform with integrated test components in a single-use format.


Subject(s)
Food Analysis/methods , Glutens/analysis , Immunoassay/methods , Limit of Detection , Antibodies, Monoclonal/immunology , Food Labeling , Gliadin/immunology , Glutens/immunology , Gold/chemistry , Humans , Time Factors
16.
Gastroenterology ; 161(1): 66-80.e8, 2021 07.
Article in English | MEDLINE | ID: mdl-33722583

ABSTRACT

BACKGROUND & AIMS: In celiac disease (CeD), gluten induces immune activation, leading to enteropathy. TAK-101, gluten protein (gliadin) encapsulated in negatively charged poly(dl-lactide-co-glycolic acid) nanoparticles, is designed to induce gluten-specific tolerance. METHODS: TAK-101 was evaluated in phase 1 dose escalation safety and phase 2a double-blind, randomized, placebo-controlled studies. Primary endpoints included pharmacokinetics, safety, and tolerability of TAK-101 (phase 1) and change from baseline in circulating gliadin-specific interferon-γ-producing cells at day 6 of gluten challenge, in patients with CeD (phase 2a). Secondary endpoints in the phase 2a study included changes from baseline in enteropathy (villus height to crypt depth ratio [Vh:Cd]), and frequency of intestinal intraepithelial lymphocytes and peripheral gut-homing T cells. RESULTS: In phase 2a, 33 randomized patients completed the 14-day gluten challenge. TAK-101 induced an 88% reduction in change from baseline in interferon-γ spot-forming units vs placebo (2.01 vs 17.58, P = .006). Vh:Cd deteriorated in the placebo group (-0.63, P = .002), but not in the TAK-101 group (-0.18, P = .110), although the intergroup change from baseline was not significant (P = .08). Intraepithelial lymphocyte numbers remained equal. TAK-101 reduced changes in circulating α4ß7+CD4+ (0.26 vs 1.05, P = .032), αEß7+CD8+ (0.69 vs 3.64, P = .003), and γδ (0.15 vs 1.59, P = .010) effector memory T cells. TAK-101 (up to 8 mg/kg) induced no clinically meaningful changes in vital signs or routine clinical laboratory evaluations. No serious adverse events occurred. CONCLUSIONS: TAK-101 was well tolerated and prevented gluten-induced immune activation in CeD. The findings from the present clinical trial suggest that antigen-specific tolerance was induced and represent a novel approach translatable to other immune-mediated diseases. ClinicalTrials.gov identifiers: NCT03486990 and NCT03738475.


Subject(s)
Celiac Disease/immunology , Gliadin/immunology , Immune Tolerance/immunology , Nanoparticles/administration & dosage , Celiac Disease/pathology , Double-Blind Method , Gliadin/administration & dosage , Glycolates/administration & dosage , Humans , Infusions, Intravenous
17.
Clin Exp Immunol ; 204(3): 321-334, 2021 06.
Article in English | MEDLINE | ID: mdl-33469922

ABSTRACT

Whole blood cytokine release assays (CRA) assessing cellular immunity to gluten could simplify the diagnosis and monitoring of coeliac disease (CD). We aimed to determine the effectiveness of electrochemiluminescence CRA to detect responses to immunodominant gliadin peptides. HLA-DQ2·5+ CD adults (cohort 1, n = 6; cohort 2, n = 12) and unaffected controls (cohort 3, n = 9) were enrolled. Cohort 1 had 3-day gluten challenge (GC). Blood was collected at baseline, and for cohort 1 also at 3 h, 6 h and 6 days after commencing 3-day GC. Gliadin peptide-stimulated proliferation, interferon (IFN)-γ enzyme-linked immunospot (ELISPOT) and 14- and 3-plex electrochemiluminescence CRA were performed. Poisson distribution analysis was used to estimate responding cell frequencies. In cohort 1, interleukin (IL)-2 dominated the gliadin peptide-stimulated cytokine release profile in whole blood. GC caused systemic IL-2 release acutely and increased gliadin peptide-stimulated IFN-γ ELISPOT and whole blood CRA responses. Whole blood CRA after GC was dominated by IL-2, but also included IFN-γ, C-X-C motif chemokine ligand 10/IFN-γ-induced protein 10 (CXCL10/IP-10), CXCL9/monokine induced by IFN-γ (MIG), IL-10, chemokine (C-C motif) ligand 3/macrophage inflammatory protein 1-alpha (CCL3/MIP-1α), TNF-α and IL-8/CXCL8. In cohorts 2 and 3, gliadin peptide-stimulated whole blood IL-2 release was 100% specific and 92% sensitive for CD patients on a gluten-free diet; the estimated frequency of cells in CD blood secreting IL-2 to α-gliadin peptide was 0·5 to 11 per ml. Whole blood IL-2 release successfully mapped human leucocyte antigen (HLA)-DQ2·5-restricted epitopes in an α-gliadin peptide library using CD blood before and after GC. Whole blood IL-2 release assay using electrochemiluminescence is a sensitive test for rare gliadin-specific T cells in CD, and could aid in monitoring and diagnosis. Larger studies and validation with tetramer-based assays are warranted.


Subject(s)
Celiac Disease/immunology , Glutens/immunology , Interleukin-2/immunology , T-Lymphocytes/immunology , Adult , Aged , Chemokine CXCL10/immunology , Cytokines/immunology , Epitopes, T-Lymphocyte/immunology , Female , Gliadin/immunology , HLA-DQ Antigens/immunology , Humans , Immunity, Cellular/immunology , Interferon-gamma/immunology , Interleukin-8/immunology , Male , Middle Aged , Peptide Fragments/immunology , Peptides/immunology , Young Adult
18.
Int J Mol Sci ; 22(2)2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33435615

ABSTRACT

Celiac disease is an autoimmune disorder characterized by a heightened immune response to gluten proteins in the diet, leading to gastrointestinal symptoms and mucosal damage localized to the small intestine. Despite its prevalence, the only treatment currently available for celiac disease is complete avoidance of gluten proteins in the diet. Ongoing clinical trials have focused on targeting the immune response or gluten proteins through methods such as immunosuppression, enhanced protein degradation and protein sequestration. Recent studies suggest that polyphenols may elicit protective effects within the celiac disease milieu by disrupting the enzymatic hydrolysis of gluten proteins, sequestering gluten proteins from recognition by critical receptors in pathogenesis and exerting anti-inflammatory effects on the system as a whole. This review highlights mechanisms by which polyphenols can protect against celiac disease, takes a critical look at recent works and outlines future applications for this potential treatment method.


Subject(s)
Autoimmune Diseases/immunology , Celiac Disease/immunology , Gliadin/immunology , Polyphenols/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/prevention & control , Celiac Disease/metabolism , Celiac Disease/prevention & control , Gliadin/metabolism , Glutens/immunology , Glutens/metabolism , Humans , Immunosuppression Therapy/methods , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestine, Small/drug effects , Intestine, Small/immunology , Intestine, Small/metabolism , Polyphenols/metabolism , Polyphenols/therapeutic use , Prospective Studies
20.
Anaerobe ; 68: 102237, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32721554

ABSTRACT

The human intestinal tract is colonized by a large number of diverse microorganisms that play various important physiologic functions. In inflammatory gut diseases including celiac disease (CeD), a dysbiotic state of microbiome has been observed. Interestingly, this perturbed microbiome is normalized towards eubiosis in patients showing recovery after treatment. The treatment has been observed to increase the abundance of beneficial microbes in comparison to non-treated patients. In this study, we investigated the effect of Prevotella histicola or Prevotella melaninogenica, isolated from the duodenum of a treated CeD patient, on the induction and maintenance of oral tolerance to gliadin, a CeD associated subgroup of gluten proteins, in NOD.DQ8.ABo transgenic mice. Conventionally raised mice on a gluten free diet were orally gavaged with bacteria before and after injection with pepsin trypsin digested gliadin (PTD-gliadin). P. histicola suppressed the cellular response to gliadin, whereas P. melaninogenica failed to suppress an immune response against gliadin. Interestingly, tolerance to gliadin in NOD.DQ8.ABo mice may be associated with gut microbiota as mice gavaged with P melaninogenica harbored a different microbial diversity as compared to P. histicola treated mice. This study provides experimental evidence that gut microbes like P. histicola from treated patients can suppress the immune response against gliadin epitopes.


Subject(s)
Celiac Disease/immunology , Celiac Disease/microbiology , Gastrointestinal Microbiome , Gliadin/immunology , T-Lymphocytes/immunology , Animals , Female , Humans , Immune Tolerance , Male , Mice , Mice, Inbred NOD , Prevotella/immunology , Prevotella/physiology , Prevotella melaninogenica/immunology , Prevotella melaninogenica/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...