Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.455
Filter
1.
Life Sci Alliance ; 7(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38719750

ABSTRACT

Celiac disease (CD) is an autoimmune enteropathy resulting from an interaction between diet, genome, and immunity. Although many patients respond to a gluten-free diet, in a substantive number of individuals, the intestinal injury persists. Thus, other factors might amplify the ongoing inflammation. Candida albicans is a commensal fungus that is well adapted to the intestinal life. However, specific conditions increase Candida pathogenicity. The hypothesis that Candida may be a trigger in CD has been proposed after the observation of similarity between a fungal wall component and two CD-related gliadin T-cell epitopes. However, despite being implicated in intestinal disorders, Candida may also protect against immune pathologies highlighting a more intriguing role in the gut. Herein, we postulated that a state of chronic inflammation associated with microbial dysbiosis and leaky gut are favorable conditions that promote C. albicans pathogenicity eventually contributing to CD pathology via a mast cells (MC)-IL-9 axis. However, the restoration of immune and microbial homeostasis promotes a beneficial C. albicans-MC cross-talk favoring the attenuation of CD pathology to alleviate CD pathology and symptoms.


Subject(s)
Candida albicans , Celiac Disease , Homeostasis , Mast Cells , Celiac Disease/immunology , Celiac Disease/microbiology , Celiac Disease/metabolism , Humans , Candida albicans/pathogenicity , Candida albicans/immunology , Mast Cells/immunology , Mast Cells/metabolism , Gastrointestinal Microbiome/immunology , Dysbiosis/immunology , Candidiasis/immunology , Candidiasis/microbiology , Animals , Candida/pathogenicity , Candida/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism
2.
Front Immunol ; 15: 1390755, 2024.
Article in English | MEDLINE | ID: mdl-38715620

ABSTRACT

Being defined as an autoimmune, chronic pathology, frequently encountered in any age group, but especially in pediatrics, celiac disease (also called gluten enteropathy), is gaining more and more ground in terms of diagnosis, but also interest in research. The data from the literature of the last decades attest the chameleonic way of its presentation, there may be both classic onset symptoms and atypical symptoms. Given the impact played by celiac disease, especially in the optimal growth and development of children, the current narrative review aims to highlight the atypical presentation methods, intended to guide the clinician towards the inclusion of the pathology in the differential diagnosis scheme. To these we add the summary presentation of the general data and therapeutic lines regarding the underlying condition and the existing comorbidities. In order to place the related information up to date, we performed a literature review of the recent articles published in international databases. We bring forward the current theories and approaches regarding both classic celiac disease and its atypical manifestations. Among these we note mainly constitutional, skin or mucous, bone, neuro-psychic, renal, reproductive injuries, but also disorders of biological constants and association with multiple autoimmunities. Knowing and correlating them with celiac disease is the key to optimal management of patients, thus reducing the subsequent burden of the disease.


Subject(s)
Celiac Disease , Celiac Disease/diagnosis , Celiac Disease/immunology , Humans , Child , Diagnosis, Differential
3.
Int Immunopharmacol ; 133: 112020, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38608449

ABSTRACT

Celiac disease (CD) is a chronic autoimmune enteropathy and multifactorial disease caused by inappropriate immune responses to gluten in the small intestine. Weight loss, anemia, osteoporosis, arthritis, and hepatitis are among the extraintestinal manifestations of active CD. Currently, a strict lifelong gluten-free diet (GFD) is the only safe, effective, and available treatment. Despite the social burden, high expenses, and challenges of following a GFD, 2 to 5 percent of patients do not demonstrate clinical or pathophysiological improvement. Therefore, we need novel and alternative therapeutic approaches for patients. Innovative approaches encompass a broad spectrum of strategies, including enzymatic degradation of gluten, inhibition of intestinal permeability, modulation of the immune response, inhibition of the transglutaminase 2 (TG2) enzyme, blocking antigen presentation by HLA-DQ2/8, and induction of tolerance. Hence, this review is focused on comprehensive therapeutic strategies ranging from dietary approaches to novel methods such as antigen-based immunotherapy, cell and gene therapy, and the usage of nanoparticles for CD treatment.


Subject(s)
Celiac Disease , Diet, Gluten-Free , Humans , Celiac Disease/diet therapy , Celiac Disease/therapy , Celiac Disease/immunology , Animals , Cell- and Tissue-Based Therapy/methods , Protein Glutamine gamma Glutamyltransferase 2 , Immunotherapy/methods , Glutens/immunology , Transglutaminases/immunology , Transglutaminases/metabolism
5.
Cell Rep ; 43(4): 114045, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38578826

ABSTRACT

Autoantibodies against the enzyme transglutaminase 2 (TG2) are characteristic of celiac disease (CeD), and TG2-specific immunoglobulin (Ig) A plasma cells are abundant in gut biopsies of patients. Here, we describe the corresponding population of autoreactive B cells in blood. Circulating TG2-specific IgA cells are present in untreated patients on a gluten-containing diet but not in controls. They are clonally related to TG2-specific small intestinal plasma cells, and they express gut-homing molecules, indicating that they are plasma cell precursors. Unlike other IgA-switched cells, the TG2-specific cells are negative for CD27, placing them in the double-negative (IgD-CD27-) category. They have a plasmablast or activated memory B cell phenotype, and they harbor fewer variable region mutations than other IgA cells. Based on their similarity to naive B cells, we propose that autoreactive IgA cells in CeD are generated mainly through chronic recruitment of naive B cells via an extrafollicular response involving gluten-specific CD4+ T cells.


Subject(s)
B-Lymphocytes , Celiac Disease , GTP-Binding Proteins , Immunoglobulin A , Plasma Cells , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases , Celiac Disease/immunology , Celiac Disease/pathology , Humans , Transglutaminases/immunology , Transglutaminases/metabolism , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Immunoglobulin A/blood , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , GTP-Binding Proteins/immunology , GTP-Binding Proteins/metabolism , Autoantibodies/immunology , Autoantibodies/blood , Adult , Male , Female , Middle Aged , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Glutens/immunology
6.
Int Immunopharmacol ; 133: 112059, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38615385

ABSTRACT

Many immune-mediated diseases have the common genetic basis, as an autoimmune disorder, celiac disease (CeD) primarily affects the small intestine, and is caused by the ingestion of gluten in genetically susceptible individuals. As for ulcerative colitis (UC), which most likely involves a complex interplay between some components of the commensal microbiota and other environmental factors in its origin. These two autoimmune diseases share a specific target organ, the bowel. The etiology and immunopathogenesis of both conditions characterized by chronic intestinal inflammation, ulcerative colitis and celiac disease, are not completely understood. Both are complex diseases with genetics and the environmental factors contributing to dysregulation of innate and adaptive immune responses, leading to chronic inflammation and disease. This study is designed to further clarify the relationship between UC and CeD. The GEO database was used to download gene expression profiles for CeD (GSE112102) and UC (GSE75214). The GSEA KEGG pathway analysis revealed that immune-related pathways were significantly associated with both diseases. Further, we screened 187 shared differentially expressed genes (DEGs) of the two diseases. Gene Ontology (GO) and WikiPathways were carried out to perform the biological process and pathway enrichment analysis. Subsequently, based on the DEGs, the least absolute shrinkage and selection operator (LASSO) analysis was performed to screen for the diagnostic biomarkers of the diseases. Moreover, single-cell RNA-sequencing (RNA-seq) data from five colonic propria with UC showed that REG4 expression was present in Goblet cell, Enteroendocrine cell, and Epithelial. Finally, our work identified REG4 is the shared gene of UC and CeD via external data validation, cellular experiments, and immunohistochemistry. In conclusion, our study elucidated that abnormal immune response could be the common pathogenesis of UC and CeD, and REG4 might be a key potential biomarker and therapeutic target for the comorbidity of these two diseases.


Subject(s)
Celiac Disease , Colitis, Ulcerative , Single-Cell Analysis , Celiac Disease/genetics , Celiac Disease/immunology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Humans , Transcriptome , Gene Expression Profiling , Sequence Analysis, RNA
7.
Eur J Immunol ; 54(5): e2350839, 2024 May.
Article in English | MEDLINE | ID: mdl-38430190

ABSTRACT

The active vitamin A metabolite, all-trans-retinoic acid (RA), primes precursor dendritic cells (DCs) into a mucosal phenotype with tolerogenic properties characterized by the expression of integrin CD103. CD103+ DCs can counteract pathogenic Th1 and Th17 in inflammatory bowel disease (IBD) or celiac disease (CD). Tolerogenic manipulation of DCs using nanoparticles carrying tolerogenic adjuvants and disease-specific antigens is a valuable treatment strategy to induce antigen-specific mucosal tolerance in vivo. Here, we investigated the effects of RA-loaded liposomes on human DC phenotype and function, including DC-driven T-cell development, both during the generation of monocyte-derived DCs (moDCs) as well as by priming immature moDCs. RA liposomes drove CD103+ DC differentiation as well as ALDH1A2 expression in DCs. Neutrophil-dependent Th17 cell development was reduced by RA-liposome-differentiated and RA-liposome-primed DCs. Moreover, RA liposome treatment shifted T-cell development toward a Th2 cell profile. Importantly, RA liposomes induced the development of IL-10-producing and FoxP3+ regulatory T cells (Tregs) of various Treg subsets, including ICOS+ Tregs, that were potent inhibitors of bystander memory T-cell proliferation. Taken together, RA-loaded liposomes could be a novel treatment avenue for IBD or CD patients.


Subject(s)
Aldehyde Dehydrogenase 1 Family , Antigens, CD , Cell Differentiation , Dendritic Cells , Integrin alpha Chains , Liposomes , Retinal Dehydrogenase , T-Lymphocytes, Regulatory , Th17 Cells , Tretinoin , Humans , Tretinoin/pharmacology , Integrin alpha Chains/metabolism , Th17 Cells/immunology , Dendritic Cells/immunology , Dendritic Cells/drug effects , Antigens, CD/immunology , Antigens, CD/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Cell Differentiation/drug effects , Cell Differentiation/immunology , Retinal Dehydrogenase/metabolism , Immune Tolerance/drug effects , Cells, Cultured , Interleukin-10/metabolism , Interleukin-10/immunology , Forkhead Transcription Factors/metabolism , Inflammatory Bowel Diseases/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Celiac Disease/immunology
8.
J Pediatr Gastroenterol Nutr ; 78(5): 1143-1148, 2024 May.
Article in English | MEDLINE | ID: mdl-38477348

ABSTRACT

OBJECTIVES: Autoantibodies against tissue transglutaminase (tTG) are serological markers of celiac disease. The aim was to study the applicability of human leukocyte antigen (HLA)-genotyping and tTG autoantibodies in the screening of celiac disease in a longitudinal birth cohort followed to age 15 years. METHODS: Included were 13,860 HLA-DQ-genotyped children at birth and previously invited to a screening at age 3 and 9 years, respectively. HLA-DQB1*02 and/or DQB1*03:02 (HLA-risk) children were compared with non-HLA-DQB1*02 and non-DQB1*03:02 (HLA-nonrisk) children. The present study reinvited 12,948/13,860 (93.4%) children at age 15 years of whom 1056/2374 (44.5%) participated in screening at both age 3 and 9 years. Both immunoglobulin A (IgA) and G (IgG) autoantibodies against tTG were analyzed separately in radiobinding assays. Persistently tTG autoantibody-positive children were examined with intestinal biopsy to confirm the diagnosis of celiac disease. RESULTS: At age 3 years, celiac disease was diagnosed in 56/1635 (3.4%) HLA-risk children compared with 0/1824 HLA-nonrisk children (p < 0.001). By age 9 years, celiac disease was diagnosed in 72/1910 (3.8%) HLA-risk children compared with 0/2167 HLA-nonrisk children (p < 0.001). Screening at age 15 years detected 14/1071 (1.3%) HLA-risk children positive for IgA-tTG and/or IgG-tTG of whom 12/1071 (1.1%) remained persistently positive. Among those, 10/1071 (0.9%, 95% confidence interval: 0.4%-1.7%) HLA-risk children were diagnosed with celiac disease compared with 0/1303 HLA-nonrisk children (p < 0.001) and 5/491 (1.0%) were negative in screenings at both 3 and 9 years of age. CONCLUSIONS: Screening for celiac disease needs to be performed at multiple timepoints to detect all cases but can be restricted to children at HLA-risk.


Subject(s)
Autoantibodies , Celiac Disease , GTP-Binding Proteins , Immunoglobulin A , Transglutaminases , Humans , Celiac Disease/diagnosis , Celiac Disease/immunology , Celiac Disease/genetics , Child , Child, Preschool , Transglutaminases/immunology , Longitudinal Studies , Autoantibodies/blood , Adolescent , Female , Male , Immunoglobulin A/blood , GTP-Binding Proteins/immunology , Immunoglobulin G/blood , Protein Glutamine gamma Glutamyltransferase 2 , HLA-DQ Antigens/genetics , Mass Screening/methods , Genotype , HLA-DQ beta-Chains/genetics , Risk Factors , Genetic Predisposition to Disease
9.
Nat Rev Gastroenterol Hepatol ; 21(5): 335-347, 2024 May.
Article in English | MEDLINE | ID: mdl-38336920

ABSTRACT

Coeliac disease is an autoinflammatory condition caused by immune reactions to cereal gluten proteins. Currently, the only available treatment for the condition is a lifelong avoidance of gluten proteins in the diet. There is an unmet need for alternative therapies. Coeliac disease has a strong association with certain HLA-DQ allotypes (DQ2.5, DQ2.2 and DQ8), and these disease-associated HLA-DQ molecules present deamidated gluten peptides to gluten-specific CD4+ T cells. The gluten-specific CD4+ T cells are the drivers of the immune reactions leading to coeliac disease. Once established, the clonotypes of gluten-specific CD4+ T cells persist for decades, explaining why patients must adhere to a gluten-free diet for life. Given the key pathogenic role of gluten-specific CD4+ T cells, tolerance-inducing therapies that target these T cells are attractive for treatment of the disorder. Lessons learned from coeliac disease might provide clues for treatment of other HLA-associated diseases for which the disease-driving antigens are unknown. Thus, intensive efforts have been and are currently implemented to bring an effective tolerance-inducing therapy for coeliac disease. This Review discusses mechanisms of the various approaches taken, summarizing the progress made, and highlights future directions in this field.


Subject(s)
Celiac Disease , Celiac Disease/immunology , Celiac Disease/therapy , Humans , Immune Tolerance/immunology , Glutens/immunology , Glutens/adverse effects , Diet, Gluten-Free , HLA-DQ Antigens/immunology , CD4-Positive T-Lymphocytes/immunology
10.
J Neurol ; 271(5): 2596-2604, 2024 May.
Article in English | MEDLINE | ID: mdl-38337123

ABSTRACT

BACKGROUND: A prior small-scale single center study suggested an association between celiac disease (CD)-type immunity and refractory temporal lobe epilepsy (TLE) with hippocampal sclerosis (HS). The present study addresses this putative association in a large, well-characterized group of drug-resistant epilepsy (DRE) patients. These patients were grouped based on the spectrum of CD and gluten sensitivity-associated antibodies. METHODS: In this cross-sectional study, 253 consecutive adult epilepsy patients (135 females, 118 males; age 16-76 years) were categorized into three groups: (i) CD-positive group with either prior diagnosis of CD or CD-specific TG2/EmA antibodies, (ii) AGA-positive group with antigliadin antibodies (AGA) but without CD, and (iii) CD/AGA-negative group without any gluten sensitivity-associated antibodies or CD. Clinical and immunological findings were then compared among the groups. RESULTS: TLE with HS was more common in the CD-positive group compared to CD/AGA-negative group (31.8% versus 11.9%, P = 0.019). Autoimmune disorders were more common in the AGA-positive group than in the CD/AGA-negative group (P = 0.025). Considering HS lateralization; left lateralization was more common in CD-positive group compared to CD/AGA-negative group (71.4% versus 25%, P = 0.030). TG6 seropositivity did not differ among the groups (P > 0.05). CONCLUSIONS: This study provides further evidence linking TLE with HS and CD-type autoimmunity suggesting that CD-type immune response to gluten can be one potential mechanism as a disease modifier leading to DRE and HS. Understanding these immunological factors is imperative for developing immunomodulatory or dietary treatments for DRE potentially preventing HS progression.


Subject(s)
Celiac Disease , Drug Resistant Epilepsy , Epilepsy, Temporal Lobe , Hippocampus , Sclerosis , Humans , Female , Male , Adult , Middle Aged , Celiac Disease/complications , Celiac Disease/immunology , Epilepsy, Temporal Lobe/immunology , Epilepsy, Temporal Lobe/complications , Drug Resistant Epilepsy/immunology , Drug Resistant Epilepsy/etiology , Sclerosis/immunology , Young Adult , Adolescent , Cross-Sectional Studies , Aged , Hippocampus/pathology , Hippocampus/immunology , Autoantibodies/blood , Gliadin/immunology , Transglutaminases/immunology , GTP-Binding Proteins/immunology , Protein Glutamine gamma Glutamyltransferase 2 , Hippocampal Sclerosis
11.
Immunol Cell Biol ; 102(5): 331-335, 2024.
Article in English | MEDLINE | ID: mdl-38111272

ABSTRACT

As part of the Monash Sensory Science Exhibition, our team guided participants through a multisensory journey unraveling coeliac disease development and pathology. Through tactile and sensory exhibits, we showed how benign dietary gluten can be transformed into a harmful entity for the 1 in 70 Australians with this illness. In contrast to the common misconception of coeliac disease as a food allergy, our exhibits revealed its closer association with autoimmune diseases such as type 1 diabetes, involving genetic susceptibility linked to specific human leukocyte antigens, crucial antigen-specific T- and B-cell responses and autoantibody production. Tactile models underscored the severe consequences of the proinflammatory immune response to gluten on patient health and quality of life. This educational event affirmed to us the value and importance of fostering inclusivity in science education.


Subject(s)
Celiac Disease , Glutens , Celiac Disease/immunology , Celiac Disease/etiology , Humans , Glutens/immunology , Touch , Australia , Diabetes Mellitus, Type 1/immunology , Autoantibodies/immunology
12.
Nature ; 624(7992): 653-662, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37993717

ABSTRACT

Ameloblasts are specialized epithelial cells in the jaw that have an indispensable role in tooth enamel formation-amelogenesis1. Amelogenesis depends on multiple ameloblast-derived proteins that function as a scaffold for hydroxyapatite crystals. The loss of function of ameloblast-derived proteins results in a group of rare congenital disorders called amelogenesis imperfecta2. Defects in enamel formation are also found in patients with autoimmune polyglandular syndrome type-1 (APS-1), caused by AIRE deficiency3,4, and in patients diagnosed with coeliac disease5-7. However, the underlying mechanisms remain unclear. Here we show that the vast majority of patients with APS-1 and coeliac disease develop autoantibodies (mostly of the IgA isotype) against ameloblast-specific proteins, the expression of which is induced by AIRE in the thymus. This in turn results in a breakdown of central tolerance, and subsequent generation of corresponding autoantibodies that interfere with enamel formation. However, in coeliac disease, the generation of such autoantibodies seems to be driven by a breakdown of peripheral tolerance to intestinal antigens that are also expressed in enamel tissue. Both conditions are examples of a previously unidentified type of IgA-dependent autoimmune disorder that we collectively name autoimmune amelogenesis imperfecta.


Subject(s)
Amelogenesis Imperfecta , Autoantibodies , Celiac Disease , Polyendocrinopathies, Autoimmune , Humans , Amelogenesis Imperfecta/complications , Amelogenesis Imperfecta/immunology , Autoantibodies/immunology , Celiac Disease/complications , Celiac Disease/immunology , Immunoglobulin A/immunology , Polyendocrinopathies, Autoimmune/complications , Polyendocrinopathies, Autoimmune/immunology , Proteins/immunology , Proteins/metabolism , Ameloblasts/metabolism , Dental Enamel/immunology , Dental Enamel/metabolism , AIRE Protein/deficiency , Antigens/immunology , Antigens/metabolism , Intestines/immunology , Intestines/metabolism
13.
Immunol Res ; 71(6): 839-848, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37221348

ABSTRACT

Celiac disease (CD) is an autoimmune disease with inflammatory characteristics, having a condition of chronic malabsorption, affecting approximately 1% of the population at any age. In recent years, a concrete correlation between eating disorders and CD has emerged. Hypothalamus plays a central role in determining eating behaviour, regulating appetite and, consequently, food intake. One hundred and ten sera from celiac patients (40 active and 70 following a gluten-free diet) were tested for the presence of autoantibodies against primate hypothalamic periventricular neurons by immunofluorescence and by a home-made ELISA assay. In addition, ghrelin was measured by ELISA. As control, 45 blood serums from healthy age matched were analysed. Among active CD, all patients resulted positive for anti-hypothalamus autoantibodies and sera showed significantly higher levels of ghrelin. All of the free-gluten CD were negative for anti-hypothalamus autoantibodies and had low levels of ghrelin, as well as healthy controls. Of interest, anti-hypothalamic autoantibodies directly correlate with anti-tTG amounts and with mucosal damage. In addition, competition assays with recombinant tTG showed a drastically reduction of anti-hypothalamic serum reactivity. Finally, ghrelin levels are increased in CD patients and correlated with anti-tTG autoantibodies and anti-hypothalamus autoantibodies. This study demonstrates for the first time the presence of anti-hypothalamus antibodies and their correlation with the severity of the CD. It also allows us to hypothesize the role of tTG as a putative autoantigen expressed by hypothalamic neurons.


Subject(s)
Autoantibodies , Celiac Disease , Ghrelin , Hypothalamus , Animals , Humans , Autoantibodies/blood , Celiac Disease/blood , Celiac Disease/immunology , Enzyme-Linked Immunosorbent Assay/methods , Immunoglobulin A , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases , Hypothalamus/immunology
14.
Cell Rep ; 39(11): 110956, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35705047

ABSTRACT

Celiac disease (CD) is a multisystem disease in which different organs may be affected. We investigate whether circulating innate lymphoid cells (ILCs) contribute to the CD peripheral inflammatory status. We find that the CD cytokine profile is characterized by high concentrations of IL-12p40, IL-18, and IFN-γ, paralleled by an expansion of ILC precursors (ILCPs). In the presence of the gliadin peptides p31-43 and pα-9, ILCPs from CD patients increase transglutaminase 2 (TG2) expression, produce IL-18 and IFN-γ, and stimulate CD4+ T lymphocytes. IFN-γ is also produced upon stimulation with IL-12p40 and IL-18 and is inhibited by the addition of vitamin D. Low levels of blood vitamin D correlate with high IFN-γ and ILCP presence and mark the CD population mostly affected by extraintestinal symptoms. Dietary vitamin D supplementation appears to be an interesting therapeutic approach to dampen ILCP-mediated IFN-γ production.


Subject(s)
Celiac Disease , Immunity, Innate , Celiac Disease/immunology , Celiac Disease/metabolism , Gliadin/metabolism , Gliadin/pharmacology , Humans , Interleukin-12 Subunit p40/metabolism , Interleukin-18/metabolism , Intestinal Mucosa/metabolism , Lymphocytes/metabolism , Vitamin D/metabolism , Vitamin D/pharmacology
15.
Nat Commun ; 13(1): 888, 2022 02 16.
Article in English | MEDLINE | ID: mdl-35173144

ABSTRACT

Celiac disease (CeD) is an autoimmune disorder induced by consuming gluten proteins from wheat, barley, and rye. Glutens resist gastrointestinal proteolysis, resulting in peptides that elicit inflammation in patients with CeD. Despite well-established connections between glutens and CeD, chemically defined, bioavailable peptides produced from dietary proteins have never been identified from humans in an unbiased manner. This is largely attributable to technical challenges, impeding our knowledge of potentially diverse peptide species that encounter the immune system. Here, we develop a liquid chromatographic-mass spectrometric workflow for untargeted sequence analysis of the urinary peptidome. We detect over 600 distinct dietary peptides, of which ~35% have a CeD-relevant T cell epitope and ~5% are known to stimulate innate immune responses. Remarkably, gluten peptides from patients with CeD qualitatively and quantitatively differ from controls. Our results provide a new foundation for understanding gluten immunogenicity, improving CeD management, and characterizing the dietary and urinary peptidomes.


Subject(s)
Celiac Disease/immunology , Glutens/analysis , Proteome/analysis , Urine/chemistry , Amino Acid Sequence , Celiac Disease/pathology , Chromatography, Liquid , Epitopes, T-Lymphocyte/immunology , Glutens/immunology , Glutens/metabolism , Hordeum/chemistry , Humans , Mass Spectrometry , Secale/chemistry , T-Lymphocytes/immunology , Triticum/chemistry
16.
J Biol Chem ; 298(3): 101619, 2022 03.
Article in English | MEDLINE | ID: mdl-35065967

ABSTRACT

Celiac disease is a T cell-mediated chronic inflammatory condition often characterized by human leukocyte antigen (HLA)-DQ2.5 molecules presenting gluten epitopes derived from wheat, barley, and rye. Although some T cells exhibit cross-reactivity toward distinct gluten epitopes, the structural basis underpinning such cross-reactivity is unclear. Here, we investigated the T-cell receptor specificity and cross-reactivity of two immunodominant wheat gluten epitopes, DQ2.5-glia-α1a (PFPQPELPY) and DQ2.5-glia-ω1 (PFPQPEQPF). We show by surface plasmon resonance that a T-cell receptor alpha variable (TRAV) 4+-T-cell receptor beta variable (TRBV) 29-1+ TCR bound to HLA-DQ2.5-glia-α1a and HLA-DQ2.5-glia-ω1 with similar affinity, whereas a TRAV4- (TRAV9-2+) TCR recognized HLA-DQ2.5-glia-ω1 only. We further determined the crystal structures of the TRAV4+-TRBV29-1+ TCR bound to HLA-DQ2.5-glia-α1a and HLA-DQ2.5-glia-ω1, as well as the structure of an epitope-specific TRAV9-2+-TRBV7-3+ TCR-HLA-DQ2.5-glia-ω1 complex. We found that position 7 (p7) of the DQ2.5-glia-α1a and DQ2.5-glia-ω1 epitopes made very limited contacts with the TRAV4+ TCR, thereby explaining the TCR cross-reactivity across these two epitopes. In contrast, within the TRAV9-2+ TCR-HLA-DQ2.5-glia-ω1 ternary complex, the p7-Gln was situated in an electrostatic pocket formed by the hypervariable CDR3ß loop of the TCR and Arg70ß from HLA-DQ2.5, a polar network which would not be supported by the p7-Leu residue of DQ2.5-glia-α1a. In conclusion, we provide additional insights into the molecular determinants of TCR specificity and cross-reactivity to two closely-related epitopes in celiac disease.


Subject(s)
Celiac Disease , Glutens , HLA-DQ Antigens , CD4-Positive T-Lymphocytes/immunology , Celiac Disease/immunology , Complementarity Determining Regions/metabolism , Cross Reactions/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Glutens/immunology , HLA-DQ Antigens/chemistry , HLA-DQ Antigens/immunology , Humans , Immunodominant Epitopes/metabolism , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , T-Cell Antigen Receptor Specificity/immunology
17.
Am J Surg Pathol ; 46(1): e43-e54, 2022 01 01.
Article in English | MEDLINE | ID: mdl-33739793

ABSTRACT

Celiac disease is a chronic, immune-mediated enteropathy driven by dietary gluten found in genetically susceptible hosts. It has a worldwide distribution, is one of the most common autoimmune disorders globally, and is the only autoimmune condition for which the trigger is known. Despite advances in characterizing mechanisms of disease, gaps in understanding of celiac disease pathogenesis remain. A "frontier" concept is considering what moves an HLA-DQ2 or DQ8-positive individual from asymptomatic gluten tolerance to celiac disease manifestation. In this arena, environmental triggers, including age at the time of initial gluten exposure, the occurrence of usual childhood viral infections, and microbiome alterations have emerged as key events in triggering the symptomatic disease. Pathologists play a major role in frontier aspects of celiac disease. This includes the discovery that duodenal mucosal histology in follow-up biopsies does not correlate with ongoing patient symptoms, antitissue transglutaminase antibody titers and diet adherence in celiac disease patients. Further, in light of recent evidence that the detection of monoclonal T-cell populations in formalin-fixed biopsies is not specific for type II refractory celiac disease, pathologists should resist performing such analyses until common causes of "apparent" refractoriness are excluded. The promise of therapies in celiac disease has led to clinical trials targeting many steps in the inflammatory cascade, which depend upon a pathologist's confirmation of the initial diagnosis and evaluation of responses to therapies. As pathologists continue to be active participants in celiac disease research, partnering with other stakeholders, we will continue to impact this important autoimmune disease.


Subject(s)
Autoimmunity , Celiac Disease/immunology , Duodenum/immunology , Environment , T-Lymphocyte Subsets/immunology , Animals , Bacteria/immunology , Bacteria/pathogenicity , Biopsy , Celiac Disease/microbiology , Celiac Disease/pathology , Celiac Disease/therapy , Duodenum/microbiology , Duodenum/pathology , Dysbiosis , Gastrointestinal Microbiome , Host-Pathogen Interactions , Humans , Phenotype , Predictive Value of Tests , Prognosis , Risk Assessment , Risk Factors
18.
Dig Liver Dis ; 54(1): 3-9, 2022 01.
Article in English | MEDLINE | ID: mdl-34561158

ABSTRACT

BACKGROUND AND AIM: How symptoms and antibodies related to SARS-CoV-2 infection develop in patients with celiac disease (CD) is unclear. We aimed to investigate the impact of SARS-CoV-2 infection in CD patients. METHODS: CD patients were interviewed about the development of COVID-19 symptoms, compliance with anti-virus measures and adherence to a gluten-free diet (GFD). The presence of anti-SARS-CoV-2 IgG and IgA (anti-RBD and N proteins) was compared to that in non-CD subjects. Expression of the duodenal ACE2 receptor was investigated. When available, data on duodenal histology, anti-tissue transglutaminase IgA (tTGA), comorbidities and GFD adherence were analyzed. RESULTS: Of 362 CD patients, 42 (12%) reported COVID-19 symptoms and 21% of these symptomatic patients presented anti-SARS-CoV-2 Ig. Overall, 18% of CD patients showed anti-SARS-CoV-2 Ig versus 25% of controls (p = 0.18). CD patients had significantly lower levels of anti-N IgA. tTGA, duodenal atrophy, GFD adherence or other comorbidities did not influence symptoms and/or antibodies. The ACE2 receptor was detected in the non-atrophic duodenal mucosa of patients; atrophy was associated with lower expression of the ACE2 receptor. CONCLUSION: CD patients have an anti-SARS-CoV-2 Ig profile similar to non-celiac controls, except for anti-N IgA. No risk factors were identified among CD parameters and GFD adherence.


Subject(s)
COVID-19/immunology , Celiac Disease/immunology , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Adult , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/prevention & control , Celiac Disease/diet therapy , Diet, Gluten-Free , Duodenum/metabolism , Female , Humans , Incidence , Italy , Male , Patient Compliance , SARS-CoV-2/immunology
19.
Mucosal Immunol ; 15(1): 51-63, 2022 01.
Article in English | MEDLINE | ID: mdl-34531547

ABSTRACT

Single-cell analysis is a powerful technology that has found widespread use in recent years. For diseases with involvement of adaptive immunity, single-cell analysis of antigen-specific T cells and B cells is particularly informative. In autoimmune diseases, the adaptive immune system is obviously at play, yet the ability to identify the culprit T and B cells recognizing disease-relevant antigen can be difficult. Celiac disease, a widespread disorder with autoimmune components, is unique in that disease-relevant antigens for both T cells and B cells are well defined. Furthermore, the celiac disease gut lesion is readily accessible allowing for sampling of tissue-resident cells. Thus, disease-relevant T cells and B cells from the gut and blood can be studied at the level of single cells. Here we review single-cell studies providing information on such adaptive immune cells and outline some future perspectives in the area of single-cell analysis in autoimmune diseases.


Subject(s)
Autoantigens/immunology , Autoimmune Diseases/immunology , B-Lymphocytes/immunology , Celiac Disease/immunology , Single-Cell Analysis/methods , Adaptive Immunity , Animals , Autoimmunity , Humans , Immunity, Humoral
20.
Expert Rev Clin Immunol ; 18(1): 75-91, 2022 01.
Article in English | MEDLINE | ID: mdl-34767744

ABSTRACT

INTRODUCTION: Recent patient studies have shown that gluten-free diet is less effective in treating celiac disease than previously believed, and additionally patients remain vulnerable to gluten-induced acute symptoms and systemic cytokine release. Safe and effective pharmacological adjuncts to gluten-free diet are in preclinical and clinical development. Clear understanding of the pathogenesis of celiac disease is critical for drug target identification, establishing efficacy endpoints and to develop noninvasive biomarkers suitable to monitor and potentially diagnose celiac disease. AREAS COVERED: The role and clinical effects of CD4+ T cells directed against deamidated gluten in the context of an 'adaptive immune paradigm' are reviewed. Alternative hypotheses of gluten toxicity are discussed and contrasted. In the context of recent patient studies, implications of the adaptive immune paradigm for future strategies to prevent, diagnose, and treat celiac disease are outlined. EXPERT OPINION: Effective therapeutics for celiac disease are likely to be approved and necessitate a variety of new clinical instruments and tests to stratify patient need, monitor remission, and confirm diagnosis in uncertain cases. Sensitive assessments of CD4+ T cells specific for deamidated gluten are likely to play a central role in clinical management, and to facilitate research and pharmaceutical development.


Subject(s)
Celiac Disease , CD4-Positive T-Lymphocytes , Celiac Disease/diagnosis , Celiac Disease/immunology , Celiac Disease/therapy , Diet, Gluten-Free , Glutens , Humans , Immunotherapy
SELECTION OF CITATIONS
SEARCH DETAIL
...