Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
2.
J Intern Med ; 285(6): 653-669, 2019 06.
Article in English | MEDLINE | ID: mdl-30762274

ABSTRACT

BACKGROUND AND OBJECTIVES: The 52-week, randomized, double-blind, noninferiority, government-funded NOR-SWITCH trial demonstrated that switching from infliximab originator to less expensive biosimilar CT-P13 was not inferior to continued treatment with infliximab originator. The NOR-SWITCH extension trial aimed to assess efficacy, safety and immunogenicity in patients on CT-P13 throughout the 78-week study period (maintenance group) versus patients switched to CT-P13 at week 52 (switch group). The primary outcome was disease worsening during follow-up based on disease-specific composite measures. METHODS: Patients were recruited from 24 Norwegian hospitals, 380 of 438 patients who completed the main study: 197 in the maintenance group and 183 in the switch group. In the full analysis set, 127 (33%) had Crohn's disease, 80 (21%) ulcerative colitis, 67 (18%) spondyloarthritis, 55 (15%) rheumatoid arthritis, 20 (5%) psoriatic arthritis and 31 (8%) chronic plaque psoriasis. RESULTS: Baseline characteristics were similar in the two groups at the time of switching (week 52). Disease worsening occurred in 32 (16.8%) patients in the maintenance group vs. 20 (11.6%) in the switch group (per-protocol set). Adjusted risk difference was 5.9% (95% CI -1.1 to 12.9). Frequency of adverse events, anti-drug antibodies, changes in generic disease variables and disease-specific composite measures were comparable between arms. The study was inadequately powered to detect noninferiority within individual diseases. CONCLUSION: The NOR-SWITCH extension showed no difference in safety and efficacy between patients who maintained CT-P13 and patients who switched from originator infliximab to CT-P13, supporting that switching from originator infliximab to CT-P13 is safe and efficacious.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Arthritis/drug therapy , Colitis, Ulcerative/drug therapy , Infliximab/therapeutic use , Psoriasis/drug therapy , Adult , Antibodies, Monoclonal/adverse effects , Double-Blind Method , Drug Substitution , Female , Humans , Male , Middle Aged , Norway , Time Factors , Treatment Outcome
3.
Mucosal Immunol ; 11(5): 1512-1523, 2018 09.
Article in English | MEDLINE | ID: mdl-30038215

ABSTRACT

The tissue dendritic cell (DC) compartment is heterogeneous, and the ontogeny and functional specialization of human tissue conventional DC (cDC) subsets and their relationship with monocytes is unresolved. Here we identify monocyte-related CSF1R+Flt3- antigen presenting cells (APCs) that constitute about half of the cells classically defined as SIRPα+ DCs in the steady-state human small intestine. CSF1R+Flt3- APCs express calprotectin and very low levels of CD14, are transcriptionally related to monocyte-derived cells, and accumulate during inflammation. CSF1R+Flt3- APCs show typical macrophage characteristics functionally distinct from their Flt3+ cDC counterparts: under steady-state conditions they excel at antigen uptake, have a lower migratory potential, and are inefficient activators of naïve T cells. These results have important implications for the understanding of the ontogenetic and functional heterogeneity within human tissue DCs and their relation to the monocyte lineage.


Subject(s)
Dendritic Cells/physiology , Intestines/physiology , Macrophages/physiology , Monocytes/physiology , Transcription, Genetic/physiology , Transcriptome/physiology , Aged , Aged, 80 and over , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/physiology , Cell Lineage/physiology , Dendritic Cells/metabolism , Female , Humans , Inflammation/metabolism , Inflammation/physiopathology , Lipopolysaccharide Receptors/metabolism , Macrophages/metabolism , Male , Middle Aged , Monocytes/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , fms-Like Tyrosine Kinase 3/metabolism
4.
Clin Nutr ; 36(1): 275-280, 2017 02.
Article in English | MEDLINE | ID: mdl-26714791

ABSTRACT

BACKGROUND/OBJECTIVES: Adherence to gluten-free diet in self reported non-coeliac gluten sensitive subjects is scarcely researched. Objectives of the study were to compare dietary adherence in coeliac disease (CD) subjects and in non-coeliac gluten sensitive (NCGS) subjects, and to estimate gluten exposure based on weighed food records and analysis of gluten content in selected food items. SUBJECTS/METHODS: Twenty-three subjects with biopsy verified CD on a gluten-free diet and 34 HLA-DQ2+ NCGS subjects on a self-instituted gluten-free diet were enrolled. The latter group was under investigation of CD. Dietary adherence was assessed by frequency questionnaire and structured forms supplied by weighed food records. For the analyses of food samples, the sandwich R5-ELISA, Ridascreen® Gliadin competitive method was used. RESULTS: There was no difference in dietary adherence between CD and NCGS subjects (83% vs 68%, p = 0.21). NCGS subjects were mainly self-educated in gluten-free diet compared to CD subjects (91% and 39%, respectively, p < 0.001). In non-adherent subjects, there was no difference in gluten exposure between CD and NCGS (10 vs 138 mg/day, p = 0.83). There was no difference in BMR-factor between CD and NCGS subjects, or between adherent and non-adherent subjects. CONCLUSIONS: Both CD and NCGS subjects were largely adherent, and adherence did not differ between the groups. Gluten exposure varied greatly, and some CD and NCGS subjects reached gluten intake above 500 mg/day, which might have considerable health effects on the individual, especially in case of coeliac disease.


Subject(s)
Celiac Disease/diet therapy , Diet, Gluten-Free , Glutens/administration & dosage , Malabsorption Syndromes/diet therapy , Patient Compliance , Adolescent , Adult , Aged , Body Mass Index , Edible Grain/chemistry , Female , Food Hypersensitivity/diagnosis , Food Hypersensitivity/diet therapy , HLA-DQ Antigens/genetics , Humans , Male , Middle Aged , Nutrition Assessment , Prolamins/administration & dosage , Self Report , Young Adult
5.
Mucosal Immunol ; 9(6): 1455-1465, 2016 11.
Article in English | MEDLINE | ID: mdl-26982597

ABSTRACT

Common variable immunodeficiency (CVID) is the most common symptomatic primary immunodeficiency characterized by low immunoglobulin (Ig)G and IgA, and/or IgM. In addition to bacterial infections, a large subgroup has noninfectious inflammatory and autoimmune complications. We performed 16S ribosomal RNA-based profiling of stool samples in 44 CVID patients, 45 patients with inflammatory bowel disease (disease controls), and 263 healthy controls. We measured plasma lipopolysaccharide (LPS) and markers of immune cell activation (i.e., soluble (s) CD14 and sCD25) in an expanded cohort of 104 patients with CVID and in 30 healthy controls. We found a large shift in the microbiota of CVID patients characterized by a reduced within-individual bacterial diversity (alpha diversity, P<0.001) without obvious associations to antibiotics use. Plasma levels of both LPS (P=0.001) and sCD25 (P<0.0001) were elevated in CVID, correlating negatively with alpha diversity and positively with a dysbiosis index calculated from the taxonomic profile. Low alpha diversity and high dysbiosis index, LPS, and immune markers were most pronounced in the subgroup with inflammatory and autoimmune complications. Low level of IgA was associated with decreased alpha diversity, but not independently from sCD25 and LPS. Our findings suggest a link between immunodeficiency, systemic immune activation, LPS, and altered gut microbiota.


Subject(s)
Common Variable Immunodeficiency/immunology , Common Variable Immunodeficiency/microbiology , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Lipopolysaccharides/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Biodiversity , Biomarkers , Case-Control Studies , Female , Humans , Immunoglobulin A/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation/immunology , Male , Middle Aged , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Young Adult
6.
Mucosal Immunol ; 9(1): 254-64, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26153762

ABSTRACT

A hallmark of celiac disease is autoantibodies to transglutaminase 2 (TG2). By visualizing TG2-specific antibodies by antigen staining of affected gut tissue, we identified TG2-specific plasma cells in the lamina propria as well as antibodies in the subepithelial layer, inside the epithelium, and at the brush border. The frequency of TG2-specific plasma cells were found not to correlate with serum antibody titers, suggesting that antibody production at other sites may contribute to serum antibody levels. Upon commencement of a gluten-free diet, the frequency of TG2-specific plasma cells in the lesion dropped dramatically within 6 months, yet some cells remained. The frequency of TG2-specific plasma cells in the celiac lesion is thus dynamically regulated in response to gluten exposure. Laser microdissection of plasma cell patches, followed by antibody gene sequencing, demonstrated that clonal cells were seeded in distinct areas of the mucosa. This was confirmed by immunoglobulin heavy chain repertoire analysis of plasma cells isolated from individual biopsies of two untreated patients, both for TG2-specific and non-TG2-specific cells. Our results shed new light on the processes underlying the B-cell response in celiac disease, and the approach of staining for antigen-specific antibodies should be applicable to other antibody-mediated diseases.


Subject(s)
Autoantibodies/genetics , Celiac Disease/immunology , GTP-Binding Proteins/immunology , Immunoglobulin Heavy Chains/genetics , Plasma Cells/immunology , Transglutaminases/immunology , Autoantibodies/biosynthesis , Biopsy , Celiac Disease/chemically induced , Celiac Disease/diet therapy , Celiac Disease/genetics , Cell Count , Diet, Gluten-Free , Duodenum/drug effects , Duodenum/immunology , Duodenum/pathology , GTP-Binding Proteins/genetics , Gene Expression Regulation/immunology , Glutens/adverse effects , Humans , Immunoglobulin Heavy Chains/biosynthesis , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Laser Capture Microdissection , Plasma Cells/drug effects , Plasma Cells/pathology , Protein Glutamine gamma Glutamyltransferase 2 , Sequence Analysis, DNA , Transglutaminases/genetics
7.
Osteoporos Int ; 25(6): 1667-76, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24691647

ABSTRACT

Patients with celiac disease (CD) have low bone mineral density. Evidence of increased fracture risk in these patients is conflicting, and the indication for bone mineral density screening of all adult CD patients is debated. Our aim was to review current published data on fractures in CD. Cross-sectional cohort studies and one case study were identified by searching Medline and Embase. Although the identified studies are heterogeneous and difficult to compare, the overall findings indicate a positive association between CD and risk of fracture. Adult patients with CD should be considered for bone densitometry in order to estimate fracture risk.


Subject(s)
Celiac Disease/complications , Osteoporotic Fractures/etiology , Bone Density/physiology , Celiac Disease/physiopathology , Humans , Osteoporosis/etiology , Osteoporosis/physiopathology , Osteoporotic Fractures/physiopathology , Research Design , Risk Factors
8.
Mucosal Immunol ; 6(5): 985-92, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23340820

ABSTRACT

Celiac disease (CD) is a chronic small intestinal inflammation precipitated by gluten ingestion. According to case reports, interferon (IFN)-α administration may induce development of overt CD. Plasmacytoid dendritic cells (PDCs) were thought to be the source of IFN-α and promote a T helper type 1 response leading to lesion formation. Surprisingly and contradicting to earlier findings, PDCs were described as the main antigen-presenting cells (APCs) in human duodenal mucosa and particularly in CD. Here we show that when assessed by flow cytometry and in situ staining, PDCs represent < 1% of APCs in both normal duodenal mucosa and the celiac lesion. Low levels of IFN-α were detected in the celiac lesion assessed by western blot, reverse transcriptase (RT)-PCR, and immunohistochemistry. In four cell populations sorted from the celiac lesion (based on their expression of HLA-DR and CD45), we found that equally low levels of mRNA for IFN-α were distributed among these cell populations. Together, these results suggest that relatively small amount of IFN-α, produced by a variety of cell types, is present in the celiac mucosa. IFN-λ, a type III IFN important in intestinal antiviral defense, was produced mainly by APCs, but its expression was not increased in the celiac lesion.


Subject(s)
Celiac Disease/immunology , Dendritic Cells/immunology , Intestinal Mucosa/immunology , Myxovirus Resistance Proteins/metabolism , Antigen Presentation , Antigens, CD/metabolism , Cell Separation , Cells, Cultured , Flow Cytometry , Gene Expression Regulation , Glutens/immunology , HLA-DR Antigens/metabolism , Humans , Immunohistochemistry , Interferon-alpha/genetics , Interferon-alpha/metabolism , Myxovirus Resistance Proteins/genetics
9.
Scand J Immunol ; 74(2): 186-94, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21392045

ABSTRACT

Coeliac disease is a chronic inflammation of the intestinal mucosa controlled by gluten-specific T cells restricted by disease-associated HLA-DQ molecules. We have previously reported that mucosal CD11c(+) dendritic cells (DCs) are responsible for activation of gluten-reactive T cells within the coeliac lesion. In mice, intestinal CD11c(+) DCs comprise several functionally distinct subsets. Here, we report that HLA-DQ(+) antigen-presenting cells (APCs) in normal human duodenal mucosa can be divided into four subsets with striking similarities to those described in mice: CD163(+) CD11c(-) macrophages (74%), and CD11c(+) cells expressing either CD163 (7%), CD103 (11%) or CD1c (13%). CD103(+) and CD1c(+) DCs belonged to partly overlapping populations, whereas CD163(+) CD11c(+) APCs appeared to be a distinct population. In the coeliac lesion, we found increased density of CD163(+) CD11c(+) APCs, whereas the density of CD103(+) and CD1c(+) DCs was decreased, suggesting that distinct subpopulations of APCs in coeliac disease may exert different functions in the pathogenesis.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , CD11c Antigen/immunology , Celiac Disease/immunology , Dendritic Cells/immunology , HLA-DQ Antigens/immunology , Integrin alpha Chains/immunology , Receptors, Cell Surface/immunology , Adult , Aged , Celiac Disease/pathology , Cell Count , Duodenum/immunology , Duodenum/pathology , Female , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Macrophages/immunology , Male , Middle Aged , Young Adult
10.
Mucosal Immunol ; 3(6): 594-601, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20571486

ABSTRACT

We have analyzed the production of the effector cytokines interleukin (IL)-17, IL-21, and IL-22 in gluten-reactive CD4(+) T cells of celiac disease patients, either cultured from small intestinal biopsies or isolated from peripheral blood after an oral gluten challenge. Combining intracellular cytokine staining with DQ2-α-II gliadin peptide tetramer staining of intestinal polyclonal T-cell lines, we found that gluten-specific T cells produced interferon-γ (IFN-γ) and IL-21, but not IL-17 or IL-22, even if other T cells of the same lines produced these cytokines. Similarly, in DQ2-α-II-specific T cells in peripheral blood of gluten-challenged patients, very few stained for intracellular IL-17, whereas many cells stained for IFN-γ. We conclude that gluten-reactive T cells produce IL-21 and IFN-γ, but not IL-17. Their production of IL-21 suggests a role for this cytokine in the pathogenesis of celiac disease.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Celiac Disease/immunology , Gliadin/metabolism , HLA-DQ Antigens/metabolism , Interleukins/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Line , Gliadin/immunology , Glutens/immunology , HLA-DQ Antigens/immunology , Humans , Interleukin-17/metabolism , Intestines/immunology , Intestines/pathology , Peptide Fragments/immunology , Peptide Fragments/metabolism , Interleukin-22
11.
Mucosal Immunol ; 2(1): 3-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19079329

ABSTRACT

The understanding of the pathogenesis of celiac disease has made huge advances in recent years. The disease is caused by an inappropriate immune response to dietary gluten proteins. This immune response is controlled by CD4(+) T cells in the lamina propria that recognize gluten peptides in the context of disease predisposing HLA-DQ2 and HLA-DQ8 molecules.(1, 2) These T cells are specific for proline- and glutamine-rich gluten peptides that are resistant to proteolysis and that have been become deamidated by the enzyme transglutaminase 2 (TG2). Strikingly, celiac disease patients produce antibodies to this same enzyme when exposed to dietary gluten. Here we discuss how the new insight in the pathogenesis has lead to development of new diagnostics and nourished research into novel treatments.


Subject(s)
Celiac Disease/diagnosis , Celiac Disease/therapy , Antibodies/immunology , Biopsy , Celiac Disease/immunology , Celiac Disease/surgery , Glutens/immunology , Histocompatibility Antigens Class I/immunology , Humans , T-Lymphocytes/immunology
12.
Clin Exp Immunol ; 142(2): 333-40, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16232221

ABSTRACT

Various experimental models suggest that the cholesterol-lowering drugs statins may also modulate immune responses. Cellular level studies on human disorders are needed, however, to provide a rational basis for clinical testing of statins as immune therapy. Coeliac disease, a chronic small intestinal inflammation driven by HLA-DQ2 restricted mucosal T cells that are specific for ingested wheat gluten peptides, is in many ways ideal for this purpose. In addition, there is a need for alternative treatment to the gluten-free diet in this disorder. Here we have assessed the effects of atorvastatin on gluten-reactive T cells, dendritic cells and the coeliac mucosa by in vitro culture of biopsies. Atorvastatin inhibited gluten-induced proliferation and specific cytokine production of human intestinal gluten-reactive T cell clones and lines. Dendritic cells exposed to atorvastatin displayed a reduced expression of the costimulatory molecule CD83 upon maturation with lipopolysaccharide. Incubation of intestinal biopsy specimens with atorvastatin in vitro, however, did not influence gluten-induced cytokine release. In conclusion, atorvastatin has specific effects on isolated gluten-reactive T cells and dendritic cells, but does not shut down the gluten-induced production of proinflammatory cytokines in intestinal biopsies.


Subject(s)
Anticholesteremic Agents/pharmacology , Celiac Disease/immunology , Glutens/immunology , Heptanoic Acids/pharmacology , Pyrroles/pharmacology , T-Lymphocytes/drug effects , Adult , Aged , Antigens, CD/metabolism , Apoptosis/drug effects , Atorvastatin , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dose-Response Relationship, Immunologic , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Immunity, Cellular/drug effects , Immunity, Mucosal/drug effects , Immunoglobulins/metabolism , Intestinal Mucosa/immunology , Lymphocyte Activation/drug effects , Membrane Glycoproteins/metabolism , Middle Aged , Organ Culture Techniques , T-Lymphocytes/immunology , Up-Regulation/drug effects , CD83 Antigen
13.
Gut ; 52(11): 1649-52, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14570737

ABSTRACT

The current trend is to allow coeliac disease (CD) patients to introduce oats to their gluten free diet. We sought further data from the clinical setting with regards to oats consumption by coeliac patients. Several oat products were tested for wheat contamination using a commercial enzyme linked immunoassay (ELISA) kit, and six samples were examined by an ELISA using a cocktail of monoclonal antibodies, mass spectrometry, and western blot analysis. Nineteen adult CD patients on a gluten free diet were challenged with 50 g of oats per day for 12 weeks. Serological testing and gastroduodenoscopy was performed before and after the challenge. Biopsies were scored histologically and levels of mRNA specific for interferon gamma were determined by reverse transcription-polymerase chain reaction analysis. Oats were well tolerated by most patients but several reported initial abdominal discomfort and bloating. One of the patients developed partial villous atrophy and a rash during the first oats challenge. She subsequently improved on an oats free diet but developed subtotal villous atrophy and dramatic dermatitis during a second challenge. Five of the patients showed positive levels of interferon gamma mRNA after challenge. Some concerns therefore remain with respect to the safety of oats for coeliacs.


Subject(s)
Avena/adverse effects , Celiac Disease/pathology , Adult , Atrophy , Blotting, Western/methods , Celiac Disease/metabolism , Diet, Protein-Restricted/methods , Enzyme-Linked Immunosorbent Assay/methods , Female , Glutens/administration & dosage , Glutens/analysis , Humans , Interferon-gamma/analysis , Intestinal Mucosa/pathology , Intestine, Small/pathology , Male , Microvilli/pathology , Middle Aged , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods
16.
Gut ; 52(1): 7-8, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12477747

ABSTRACT

DQ8 restricted gliadin peptide is immunogenic in the intestinal mucosa of HLA-DQ8 positive patients, representing the first demonstration that a given peptide may be of pathogenic significance only for a subset of coeliacs, and strongly suggests that DQ2 and DQ8 act as immune response (Ir) genes in this disease


Subject(s)
Celiac Disease/immunology , Genes, MHC Class II , HLA-DQ Antigens/genetics , Intestinal Mucosa/immunology , Intestine, Small , Celiac Disease/genetics , Gliadin/immunology , Humans
17.
Scand J Immunol ; 56(6): 652-64, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12472679

ABSTRACT

Increased levels of interferon-gamma (IFN-gamma) transcripts have previously been found in duodenal biopsy specimens from patients with untreated coeliac disease (CD). Such samples and duodenal control mucosa were therefore studied to locate and phenotype cells spontaneously secreting IFN-gamma. Specimens were collected from consecutively recruited patients with untreated (seven), treated (four) or refractory (three) CD and from five histologically normal controls. Morphological and immunohistochemical examinations were performed, and epithelial and lamina propria cell suspensions were prepared from parallel samples. Unstimulated viable cells secreting IFN-gamma were identified and phenotyped with a new fluorescence-activated cell sorter-based assay, and IFN-gamma messenger RNA (mRNA) was analysed in snap-frozen aliquots of the same suspensions. Untreated CD cases had the highest fraction of IFN-gamma+ cells in the epithelial compartment (median 2.6%, range 1.6-6.2%) and, less strikingly, in the lamina propria compartment (1.6%, range 0.3-3.6%), followed by refractory (1.4%, 1.0-1.9%; and 0.3%, 0.0-1.2%) and treated (0.8%, 0.5-0.9%; and 0.7%, 0.2-1.1%) disease and finally the controls (0.5%, 0.3-0.9%; and 0.2%, 0.1-0.7%). IFN-gamma mRNA data supported these findings. IFN-gamma+ intraepithelial lymphocytes were mostly CD3+ and CD8+, whereas many positive lamina propria cells were CD8-. We conclude that isolated T cells spontaneously secreting IFN-gamma localize preferentially in the epithelium of patients with classical and refractory CD.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Celiac Disease/immunology , Interferon-gamma/biosynthesis , Intestinal Mucosa/immunology , Adolescent , Adult , Aged , Antigens, CD/analysis , Antigens, CD/immunology , Celiac Disease/classification , Celiac Disease/diagnosis , Cells, Cultured , Epithelium/immunology , Female , Flow Cytometry , Humans , Immunohistochemistry , Immunophenotyping , Interferon-gamma/genetics , Interferon-gamma/metabolism , Intestinal Mucosa/cytology , Male , Middle Aged , RNA, Messenger/analysis , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/immunology
18.
Dig Liver Dis ; 34(3): 238-42, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11990398

ABSTRACT

Gliadin specific T cells in the small intestines of coeliac disease patients use the disease associated human leukocyte antigen-DQ2 molecules in their antigen recognition. In an exciting interplay with tissue transglutaminase, the immune system recognises modified gliadin peptides and mounts a phlogistic response. Moreover, the role for autoimmune phenomena and the mechanism of breaking of immunological tolerance remain elusive.


Subject(s)
Celiac Disease/immunology , Animals , Epitopes , Gliadin/immunology , HLA-DQ Antigens/genetics , Humans , T-Lymphocytes/immunology , Transglutaminases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL