Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.582
Filter
1.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273359

ABSTRACT

Celiac disease (CD) is an immune-mediated enteropathy triggered by the ingestion of proline- and glutamine-rich proteins, widely termed "gluten", in genetically susceptible individuals. CD induces an altered immune response that leads to chronic inflammation and duodenal mucosal damage. Currently, there are no specific tests for the accurate diagnosis of CD, and no drugs are available to treat this condition. The only available treatment strategy is lifelong adherence to a gluten-free diet. However, some studies have investigated the involvement of microRNAs (miRNAs) in CD pathogenesis. miRNAs are small noncoding ribonucleic acid molecules that regulate gene expression. Despite the growing number of studies on the role of miRNAs in autoimmune disorders, data on miRNAs and CD are scarce. Therefore, this study aimed to perform a literature review to summarize CD, miRNAs, and the potential interactions between miRNAs and CD in adults. This review shows that miRNA expression can suppress or stimulate pathways related to CD pathogenesis by regulating cell proliferation and differentiation, regulatory T-cell development, innate immune response, activation of the inflammatory cascade, focal adhesion, T-cell commitment, tissue transglutaminase synthesis, and cell cycle. Thus, identifying miRNAs and their related effects on CD could open new possibilities for diagnosis, prognosis, and follow-up of biomarkers.


Subject(s)
Celiac Disease , MicroRNAs , Celiac Disease/genetics , Celiac Disease/immunology , Celiac Disease/metabolism , Humans , MicroRNAs/genetics , Adult , Gene Expression Regulation , Biomarkers
2.
Immun Inflamm Dis ; 12(8): e1354, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39150219

ABSTRACT

BACKGROUND: In mammals, amino acid metabolism has evolved to control immune responses. Tryptophan (Trp) is the rarest essential amino acid found in food and its metabolism has evolved to be a primary regulatory node in the control of immune responses. Celiac disease (CeD) is a developed immunological condition caused by gluten intolerance and is linked to chronic small intestine enteropathy in genetically predisposed individuals. Dendritic cells (DCs), serving as the bridge between innate and adaptive immunities, can influence immunological responses in CeD through phenotypic alterations. OBJECTIVE: This review aims to highlight the connection between Trp metabolism and tolerogenic DCs, and the significance of this interaction in the pathogenesis of CeD. RESULTS: It is been recognized that various DC subtypes contribute to the pathogenesis of CeD. Tolerogenic DCs, in particular, are instrumental in inducing immune tolerance, leading to T-reg differentiation that helps maintain intestinal immune tolerance against inflammatory responses in CeD patients and those with other autoimmune disorders. T-regs, a subset of T-cells, play a crucial role in maintaining intestinal immunological homeostasis by regulating the activities of other immune cells. Notably, Trp metabolism, essential for T-reg function, facilitates T-reg differentiation through microbiota-mediated degradation and the kynurenine pathway. CONCLUSION: Therefore, alterations in Trp metabolism could potentially influence the immune response in CeD, affecting both the development of the disease and the persistence of symptoms despite adherence to a gluten-free diet.


Subject(s)
Celiac Disease , Dendritic Cells , Immune Tolerance , Tryptophan , Humans , Dendritic Cells/immunology , Dendritic Cells/metabolism , Tryptophan/metabolism , Celiac Disease/immunology , Celiac Disease/metabolism , Animals , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
3.
Food Chem ; 461: 140924, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39181042

ABSTRACT

High-sensitivity 4D label-free proteomic technology was used to identify protein components related to gluten quality and celiac disease (CD) in strong-gluten wheat cultivar KX 3302 and medium-gluten wheat cultivar BN 207. The highly expressed storage protein components in KX3302 were high-molecular-weight-glutenin-subunits (HMW-GSs), α-gliadin, and globulin, whereas those in BN207 were γ-gliadin, low-molecular-weight-glutenin-subunits (LMW-GSs) and avenin-like proteins. In addition, BN207 had more upregulated metabolic proteins than KX3302. The abundance of storage proteins increased during dough formation. After heat treatment, the upregulated proteins accounted for 57.53 % of the total proteins, but the downregulated storage proteins accounted for 79.34 % of the total storage proteins. In cultivar KX3302, CD proteins mainly included α-gliadin and HMW-GSs, whereas in BN207, they were mainly γ-gliadin and LMW-GSs. Thermal treatment significantly reduces the expression levels of CD-related proteins. These findings provide a new perspective on reducing the content of CD-related proteins in wheat products.


Subject(s)
Bread , Celiac Disease , Flour , Glutens , Hot Temperature , Proteomics , Triticum , Triticum/chemistry , Triticum/metabolism , Celiac Disease/diet therapy , Celiac Disease/metabolism , Flour/analysis , Glutens/analysis , Glutens/metabolism , Humans , Bread/analysis , Plant Proteins/analysis , Plant Proteins/metabolism , Plant Proteins/genetics , Gliadin/analysis , Gliadin/metabolism , Gliadin/chemistry
4.
Nutrients ; 16(16)2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39203879

ABSTRACT

Sulforaphane is considered the bioactive metabolite of glucoraphanin after dietary consumption of broccoli sprouts. Although both molecules pass through the gut lumen to the large intestine in stable form, their biological impact on the first intestinal tract is poorly described. In celiac patients, the function of the small intestine is affected by celiac disease (CD), whose severe outcomes are controlled by gluten-free dietary protocols. Nevertheless, pathological signs of inflammation and oxidative stress may persist. The aim of this study was to compare the biological activity of sulforaphane with its precursor glucoraphanin in a cellular model of gliadin-induced inflammation. Human intestinal epithelial cells (CaCo-2) were stimulated with a pro-inflammatory combination of cytokines (IFN-γ, IL-1ß) and in-vitro-digested gliadin, while oxidative stress was induced by H2O2. LC-MS/MS analysis confirmed that sulforaphane from broccoli sprouts was stable after simulated gastrointestinal digestion. It inhibited the release of all chemokines selected as inflammatory read-outs, with a more potent effect against MCP-1 (IC50 = 7.81 µM). On the contrary, glucoraphanin (50 µM) was inactive. The molecules were unable to counteract the oxidative damage to DNA (γ-H2AX) and catalase levels; however, the activity of NF-κB and Nrf-2 was modulated by both molecules. The impact on epithelial permeability (TEER) was also evaluated in a Transwell® model. In the context of a pro-inflammatory combination including gliadin, TEER values were recovered by neither sulforaphane nor glucoraphanin. Conversely, in the context of co-culture with activated macrophages (THP-1), sulforaphane inhibited the release of MCP-1 (IC50 = 20.60 µM) and IL-1ß (IC50 = 1.50 µM) only, but both molecules restored epithelial integrity at 50 µM. Our work suggests that glucoraphanin should not merely be considered as just an inert precursor at the small intestine level, thus suggesting a potential interest in the framework of CD. Its biological activity might imply, at least in part, molecular mechanisms different from sulforaphane.


Subject(s)
Brassica , Celiac Disease , Glucosinolates , Imidoesters , Isothiocyanates , Oxidative Stress , Oximes , Sulfoxides , Humans , Isothiocyanates/pharmacology , Sulfoxides/pharmacology , Glucosinolates/pharmacology , Glucosinolates/metabolism , Celiac Disease/drug therapy , Celiac Disease/diet therapy , Celiac Disease/metabolism , Caco-2 Cells , Oximes/pharmacology , Oxidative Stress/drug effects , Imidoesters/pharmacology , Brassica/chemistry , Gliadin/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Interleukin-1beta/metabolism , Chemokine CCL2/metabolism , Cytokines/metabolism , Interferon-gamma/metabolism
5.
Front Immunol ; 15: 1405344, 2024.
Article in English | MEDLINE | ID: mdl-39034995

ABSTRACT

Background: Exposure to antigens is crucial for child immune system development, aiding disease prevention and promoting infant health. Some common food antigen proteins are found in human breast milk. However, it is unclear whether gluten antigens linked to celiac disease (CD) are transmitted through breast milk, potentially impacting the development of the infant's immune system. Objective: This study aimed to analyze the passage of gluten immunogenic peptides (GIP) into human breast milk. We evaluated the dynamics of GIP secretion after lactating mothers adopted a controlled gluten-rich diet. Methods: We prospectively enrolled 96 non-CD and 23 CD lactating mothers, assessing total proteins and casein in breast milk, and GIP levels in breast milk and urine. Subsequently, a longitudinal study was conducted in a subgroup of 12 non-CD lactating mothers who adopted a controlled gluten-rich diet. GIP levels in breast milk and urine samples were assayed by multiple sample collections over 96 hours. Results: Analysis of a single sample revealed that 24% of non-CD lactating mothers on a regular unrestricted diet tested positive for GIP in breast milk, and 90% tested positive in urine, with significantly lower concentrations in breast milk. Nevertheless, on a controlled gluten-rich diet and the collection of multiple samples, GIP were detected in 75% and 100% of non-CD participants in breast milk and urine, respectively. The transfer dynamics in breast milk samples were long-enduring and GIP secretion persisted from 0 to 72 h. In contrast, GIP secretion in urine samples was limited to the first 24 h, with inter-individual variations. In the cohort of CD mothers, 82.6% and 87% tested negative for GIP in breast milk and urine, respectively. Conclusions: This study definitively established the presence of GIP in breast milk, with substantial inter-individual variations in secretion dynamics. Our findings provide insights into distinct GIP kinetics observed in sequentially collected breast milk and urine samples, suggesting differential gluten metabolism patterns depending on the organ or system involved. Future research is essential to understand whether GIP functions as sensitizing or tolerogenic agents in the immune system of breastfed infants.


Subject(s)
Celiac Disease , Glutens , Lactation , Milk, Human , Humans , Milk, Human/immunology , Milk, Human/chemistry , Milk, Human/metabolism , Celiac Disease/immunology , Celiac Disease/metabolism , Glutens/immunology , Female , Adult , Prospective Studies , Longitudinal Studies , Peptides/immunology , Peptides/urine , Infant , Kinetics
6.
Proc Natl Acad Sci U S A ; 121(28): e2407066121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38959038

ABSTRACT

Mammalian transglutaminases, a family of Ca2+-dependent proteins, are implicated in a variety of diseases. For example, celiac disease (CeD) is an autoimmune disorder whose pathogenesis requires transglutaminase 2 (TG2) to deamidate select glutamine residues in diet-derived gluten peptides. Deamidation involves the formation of transient γ-glutamyl thioester intermediates. Recent studies have revealed that in addition to the deamidated gluten peptides themselves, their corresponding thioester intermediates are also pathogenically relevant. A mechanistic understanding of this relevance is hindered by the absence of any structure of Ca2+-bound TG2. We report the X-ray crystallographic structure of human TG2 bound to an inhibitory gluten peptidomimetic and two Ca2+ ions in sites previously designated as S1 and S3. Together with additional structure-guided experiments, this structure provides a mechanistic explanation for how S1 regulates formation of an inhibitory disulfide bond in TG2, while also establishing that S3 is essential for γ-glutamyl thioester formation. Furthermore, our crystallographic findings and associated analyses have revealed that i) two interacting residues, H305 and E363, play a critical role in resolving the thioester intermediate into an isopeptide bond (transamidation) but not in thioester hydrolysis (deamidation); and ii) residues N333 and K176 stabilize preferred TG2 substrates and inhibitors via hydrogen bonding to nonreactive backbone atoms. Overall, the intermediate-state conformer of TG2 reported here represents a superior model to previously characterized conformers for both transition states of the TG2-catalyzed reaction.


Subject(s)
Calcium , GTP-Binding Proteins , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases , Transglutaminases/metabolism , Transglutaminases/chemistry , Protein Glutamine gamma Glutamyltransferase 2/metabolism , Humans , Calcium/metabolism , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/chemistry , Crystallography, X-Ray , Glutens/metabolism , Glutens/chemistry , Models, Molecular , Protein Conformation , Celiac Disease/metabolism , Protein Binding
7.
Nature ; 632(8024): 401-410, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39048815

ABSTRACT

In vitro models of autoimmunity are constrained by an inability to culture affected epithelium alongside the complex tissue-resident immune microenvironment. Coeliac disease (CeD) is an autoimmune disease in which dietary gluten-derived peptides bind to the major histocompatibility complex (MHC) class II human leukocyte antigen molecules (HLA)-DQ2 or HLA-DQ8 to initiate immune-mediated duodenal mucosal injury1-4. Here, we generated air-liquid interface (ALI) duodenal organoids from intact fragments of endoscopic biopsies that preserve epithelium alongside native mesenchyme and tissue-resident immune cells as a unit without requiring reconstitution. The immune diversity of ALI organoids spanned T cells, B and plasma cells, natural killer (NK) cells and myeloid cells, with extensive T-cell and B-cell receptor repertoires. HLA-DQ2.5-restricted gluten peptides selectively instigated epithelial destruction in HLA-DQ2.5-expressing organoids derived from CeD patients, and this was antagonized by blocking MHC-II or NKG2C/D. Gluten epitopes stimulated a CeD organoid immune network response in lymphoid and myeloid subsets alongside anti-transglutaminase 2 (TG2) autoantibody production. Functional studies in CeD organoids revealed that interleukin-7 (IL-7) is a gluten-inducible pathogenic modulator that regulates CD8+ T-cell NKG2C/D expression and is necessary and sufficient for epithelial destruction. Furthermore, endogenous IL-7 was markedly upregulated in patient biopsies from active CeD compared with remission disease from gluten-free diets, predominantly in lamina propria mesenchyme. By preserving the epithelium alongside diverse immune populations, this human in vitro CeD model recapitulates gluten-dependent pathology, enables mechanistic investigation and establishes a proof of principle for the organoid modelling of autoimmunity.


Subject(s)
Celiac Disease , Duodenum , Interleukin-7 , Intestinal Mucosa , Models, Biological , Organoids , Humans , Autoantibodies/immunology , Autoimmunity , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biopsy , Celiac Disease/immunology , Celiac Disease/pathology , Celiac Disease/metabolism , Duodenum/immunology , Duodenum/pathology , Duodenum/metabolism , Epitopes/immunology , Glutens/immunology , Glutens/metabolism , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/immunology , HLA-DQ Antigens/immunology , HLA-DQ Antigens/metabolism , Interleukin-7/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Organoids/immunology , Organoids/metabolism , Organoids/pathology , Protein Glutamine gamma Glutamyltransferase 2/immunology , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
8.
Microbiol Spectr ; 12(7): e0352423, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38860826

ABSTRACT

Gluten possesses unique properties that render it only partially digestible. Consequently, it exerts detrimental effects on a part of the worldwide population who are afflicted with celiac disease (1%) or related disorders (5%), particularly due to the potential for cross-contamination even when adhering to a gluten-free diet (GFD). Finding solutions to break down gluten during digestion has a high nutritional and social impact. Here, a randomized double-blind placebo-controlled in vivo challenge investigated the gluten-degrading activity of a novel probiotic preparation comprising lactobacilli and their cytoplasmic extracts, Bacillus sp., and bacterial protease. In our clinical trial, we collected feces from 70 healthy volunteers at specific time intervals. Probiotic/placebo administration lasted 32 days, followed by 10 days of wash-out. After preliminary GFD to eliminate residual gluten from feces, increasing amounts of gluten (50 mg-10 g) were administered, each one for 4 consecutive days. Compared to placebo, the feces of volunteers fed with probiotics showed much lower amounts of residual gluten, mainly with increased intakes. Probiotics also regulate the intestinal microbial communities, improving the abundance of genera pivotal to maintaining homeostasis. Quantitative PCR confirmed that all probiotics persisted during the intervention, some also during wash-out. Probiotics promoted a fecal metabolome with potential immunomodulating activity, mainly related to derivatives of branched-chain amino acids and short-chain fatty acids. IMPORTANCE: The untapped potential of gluten-degrading bacteria and their application in addressing the recognized limitations of gluten-related disorder management and the ongoing risk of cross-contamination even when people follow a gluten-free diet (GFD) emphasizes the significance of the work. Because gluten, a common protein found in many cereals, must be strictly avoided to stop autoimmune reactions and related health problems, celiac disease and gluten sensitivity present difficult hurdles. However, because of the hidden presence of gluten in many food products and the constant danger of cross-contamination during food preparation and processing, total avoidance is frequently challenging. Our study presents a novel probiotic preparation suitable for people suffering from gluten-related disorders during GFD and for healthy individuals because it enhances gluten digestion and promotes gut microbiota functionality.


Subject(s)
Feces , Gastrointestinal Microbiome , Glutens , Probiotics , Humans , Probiotics/administration & dosage , Glutens/metabolism , Gastrointestinal Microbiome/drug effects , Feces/microbiology , Feces/chemistry , Double-Blind Method , Adult , Male , Female , Lactobacillus/metabolism , Celiac Disease/microbiology , Celiac Disease/metabolism , Celiac Disease/diet therapy , Diet, Gluten-Free , Bacillus/metabolism , Middle Aged , Young Adult
9.
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
10.
Clin Immunol ; 263: 110202, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38575045

ABSTRACT

Celiac disease (CD) is an immune-driven disease characterized by tissue damage in the small intestine of genetically-susceptible individuals. We evaluated here a crucial immune regulatory pathway involving TYRO3, AXL, and MERTK (TAM) receptors and their ligands PROS1 and GAS6 in duodenal biopsies of controls and CD patients. We found increased GAS6 expression associated with downregulation of PROS1 and variable TAM receptors levels in duodenum tissue of CD patients. Interestingly, CD3+ lymphocytes, CD68+, CD11c+ myeloid and epithelial cells, showed differential expressions of TAM components comparing CD vs controls. Principal component analysis revealed a clear segregation of two groups of CD patients based on TAM components and IFN signaling. In vitro validation demonstrated that monocytes, T lymphocytes and epithelial cells upregulated TAM components in response to IFN stimulation. Our findings highlight a dysregulated TAM axis in CD related to IFN signaling and contribute to a deeper understanding of the pathophysiology of CD.


Subject(s)
Axl Receptor Tyrosine Kinase , Celiac Disease , Duodenum , Intercellular Signaling Peptides and Proteins , Intestinal Mucosa , Protein S , Receptor Protein-Tyrosine Kinases , c-Mer Tyrosine Kinase , Female , Humans , Male , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/metabolism , Celiac Disease/immunology , Celiac Disease/metabolism , Celiac Disease/genetics , Duodenum/metabolism , Duodenum/immunology , Duodenum/pathology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Interferons/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Protein S/metabolism , Protein S/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/immunology , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
11.
Gastroenterology ; 167(3): 493-504.e10, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38467384

ABSTRACT

BACKGROUND & AIMS: Histologic evaluation of gut biopsies is a cornerstone for diagnosis and management of celiac disease (CeD). Despite its wide use, the method depends on proper biopsy orientation, and it suffers from interobserver variability. Biopsy proteome measurement reporting on the tissue state can be obtained by mass spectrometry analysis of formalin-fixed paraffin-embedded tissue. Here we aimed to transform biopsy proteome data into numerical scores that give observer-independent measures of mucosal remodeling in CeD. METHODS: A pipeline using glass-mounted formalin-fixed paraffin-embedded sections for mass spectrometry-based proteome analysis was established. Proteome data were converted to numerical scores using 2 complementary approaches: a rank-based enrichment score and a score based on machine learning using logistic regression. The 2 scoring approaches were compared with each other and with histology analyzing 18 patients with CeD with biopsies collected before and after treatment with a gluten-free diet as well as biopsies from patients with CeD with varying degree of remission (n = 22). Biopsies from individuals without CeD (n = 32) were also analyzed. RESULTS: The method yielded reliable proteome scoring of both unstained and H&E-stained glass-mounted sections. The scores of the 2 approaches were highly correlated, reflecting that both approaches pick up proteome changes in the same biological pathways. The proteome scores correlated with villus height-to-crypt depth ratio. Thus, the method is able to score biopsies with poor orientation. CONCLUSIONS: Biopsy proteome scores give reliable observer and orientation-independent measures of mucosal remodeling in CeD. The proteomic method can readily be implemented by nonexpert laboratories in parallel to histology assessment and easily scaled for clinical trial settings.


Subject(s)
Celiac Disease , Diet, Gluten-Free , Intestinal Mucosa , Proteome , Proteomics , Celiac Disease/pathology , Celiac Disease/metabolism , Celiac Disease/diagnosis , Humans , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Biopsy , Proteome/analysis , Proteomics/methods , Female , Male , Adult , Machine Learning , Middle Aged , Mass Spectrometry , Observer Variation , Predictive Value of Tests , Paraffin Embedding , Reproducibility of Results , Case-Control Studies
12.
Angew Chem Int Ed Engl ; 63(21): e202317552, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38497459

ABSTRACT

Celiac disease (CeD) is an autoimmune disorder triggered by gluten proteins, affecting approximately 1 % of the global population. The 33-mer deamidated gliadin peptide (DGP) is a metabolically modified wheat-gluten superantigen for CeD. Here, we demonstrate that the 33-mer DGP spontaneously assembles into oligomers with a diameter of approximately 24 nm. The 33-mer DGP oligomers present two main secondary structural motifs-a major polyproline II helix and a minor ß-sheet structure. Importantly, in the presence of 33-mer DGP oligomers, there is a statistically significant increase in the permeability in the gut epithelial cell model Caco-2, accompanied by the redistribution of zonula occludens-1, a master tight junction protein. These findings provide novel molecular and supramolecular insights into the impact of 33-mer DGP in CeD and highlight the relevance of gliadin peptide oligomerization.


Subject(s)
Celiac Disease , Enterocytes , Gliadin , Humans , Celiac Disease/metabolism , Celiac Disease/pathology , Caco-2 Cells , Gliadin/chemistry , Gliadin/metabolism , Enterocytes/metabolism , Superantigens/chemistry , Superantigens/metabolism , Permeability
13.
J Mol Histol ; 55(1): 15-24, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38165564

ABSTRACT

Intestinal epithelium is a dynamic cellular layer that lines the small-bowel and makes a relatively impenetrable barrier to macromolecules. Intestinal epithelial cell polarity is crucial in coordinating signalling pathways within cells and mainly regulated by three conserved polarity protein complexes, the Crumbs (Crb) complex, partitioning defective (PAR) complex, and Scribble (Scrib) complex. Polarity proteins regulate the proper establishment of the intercellular junctional complexes including tight junctions (TJs), adherence junctions (AJs), and desmosomes which hold epithelial cells together and play a major role in maintaining intestinal barrier integrity. Impaired intestinal epithelial cell polarity and barrier integrity result in irreversible immune responses, the host- microbial imbalance and intestinal inflammatory disorders. Disassembling the epithelial tight junction and augmented paracellular permeability is a conspicuous hallmark of celiac disease (CD) pathogenesis. There are several dietary components that can improve intestinal integrity and function. The aim of this review article is to summarize current information about the association of polarity proteins and AJC damages with pathogenesis of CD.


Subject(s)
Celiac Disease , Humans , Celiac Disease/metabolism , Celiac Disease/pathology , Intestinal Mucosa/metabolism , Epithelial Cells/metabolism , Intestines , Tight Junctions/metabolism
14.
Biol Sex Differ ; 14(1): 86, 2023 12 11.
Article in English | MEDLINE | ID: mdl-38072919

ABSTRACT

BACKGROUND: Celiac disease (CeD) is an immune-mediated disorder that develops in genetically predisposed individuals upon gluten consumption. HLA risk alleles explain 40% of the genetic component of CeD, so there have been continuing efforts to uncover non-HLA loci that can explain the remaining heritability. As in most autoimmune disorders, the prevalence of CeD is significantly higher in women. Here, we investigated the possible involvement of the X chromosome on the sex bias of CeD. METHODS: We performed a X chromosome-wide association study (XWAS) and a gene-based association study in women from the CeD Immunochip (7062 cases, 5446 controls). We also constructed a database of X chromosome cis-expression quantitative trait loci (eQTLs) in monocytes from unstimulated (n = 226) and lipopolysaccharide (LPS)-stimulated (n = 130) female donors and performed a Summary-data-based MR (SMR) analysis to integrate XWAS and eQTL information. We interrogated the expression of the potentially causal gene (TMEM187) in peripheral blood mononuclear cells (PBMCs) from celiac patients at onset, on a gluten-free diet, potential celiac patients and non-celiac controls. RESULTS: The XWAS and gene-based analyses identified 13 SNPs and 25 genes, respectively, 22 of which had not been previously associated with CeD. The X chromosome cis-eQTL analysis found 18 genes with at least one cis-eQTL in naïve female monocytes and 8 genes in LPS-stimulated female monocytes, 2 of which were common to both situations and 6 were unique to LPS stimulation. SMR identified a potentially causal association of TMEM187 expression in naïve monocytes with CeD in women, regulated by CeD-associated, eQTL-SNPs rs7350355 and rs5945386. The CeD-risk alleles were correlated with lower TMEM187 expression. These results were replicated using eQTLs from LPS-stimulated monocytes. We observed higher levels of TMEM187 expression in PBMCs from female CeD patients at onset compared to female non-celiac controls, but not in male CeD individuals. CONCLUSION: Using X chromosome genotypes and gene expression data from female monocytes, SMR has identified TMEM187 as a potentially causal candidate in CeD. Further studies are needed to understand the implication of the X chromosome in the higher prevalence of CeD in women.


Celiac disease (CeD) is an immune-related condition triggered by gluten consumption in genetically susceptible individuals. Women present higher prevalence of CeD than men, but the biological explanation of such difference has not been elucidated. In this study, we investigated whether specific genetic variations on the X chromosome were associated with CeD in each sex. Surprisingly, we found 13 genetic variants and 25 genes significantly linked to CeD in women, but not in men. Additionally, we identified genetic variants on the X chromosome associated with gene expression of monocytes, a type of immune cells that is activated in CeD after gluten intake. Integrating these data with our previous findings, we found that lower expression of a gene termed TMEM187 might be associated with a potential increase in CeD risk in women. Finally, validation experiments confirmed higher TMEM187 levels in blood cells from female CeD patients compared to non-celiac women, while no such difference was seen in males. In summary, our study suggests that the X-chromosome gene TMEM187 may play a key role in CeD development, providing insights into the higher prevalence of CeD in females.


Subject(s)
Celiac Disease , Quantitative Trait Loci , Humans , Male , Female , Celiac Disease/genetics , Celiac Disease/metabolism , Monocytes/metabolism , Leukocytes, Mononuclear , Sexism , Lipopolysaccharides , Membrane Proteins/genetics
15.
Sci Rep ; 13(1): 21180, 2023 12 01.
Article in English | MEDLINE | ID: mdl-38040898

ABSTRACT

Enzyme therapy can be an appropriate treatment option for celiac disease (CeD). Here, we developed Bromelain-Loaded Nanocomposites (BLNCs) to improve the stability and retention of bromelain enzyme activity. After the characterization of BLNCs, the cytotoxicity of BLNCs was determined on the Caco-2 cell line. The effect of BLNCs on gliadin degradation and the production of pro-inflammatory cytokines and anti-inflammatory molecules in peripheral blood mononuclear cells (PBMCs) obtained from celiac patients were assessed. Furthermore, the expression of CXCR3 and CCR5 genes was measured in CaCo-2 cells treated with gliadin, gliadin-digested with BLNCs, and bromelain. Our study demonstrated that the Bromelain entrapment efficiency in these nanoparticles was acceptable, and BLNCs have no toxic effect on cells. SDS-PAGE confirmed the digestion effect of bromelain released from nanocomposites. When Caco-2 cells were treated with gliadin digested by free bromelain and BLNCs, the expression of CXCR3 and CCR5 genes was significantly decreased. PBMCs of celiac patients treated with Bromelain and BLNCs decreased inflammatory cytokines (IL-1ß, IL-6, TNF-α, and IFN-γ) production compared to untreated PBMCs. This treatment also increased IL-10 and CTLA-4 in PBMCs of CeD patients. According to the promising results of this study, we can hope for the therapeutic potential of BLNCs for CeD.


Subject(s)
Celiac Disease , Gliadin , Humans , Caco-2 Cells , Gliadin/metabolism , Leukocytes, Mononuclear/metabolism , Bromelains/pharmacology , Cytokines/metabolism , Celiac Disease/drug therapy , Celiac Disease/metabolism
16.
Trends Pharmacol Sci ; 44(12): 949-962, 2023 12.
Article in English | MEDLINE | ID: mdl-37839914

ABSTRACT

Celiac disease (CeD) is a widespread, gluten-induced, autoimmune disorder that lacks any medicinal therapy. Towards the goal of developing non-dietary treatments for CeD, research has focused on elucidating its molecular and cellular etiology. A model of pathogenesis has emerged centered on interactions between three molecular families: specific class II MHC proteins on antigen-presenting cells (APCs), deamidated gluten-derived peptides, and T cell receptors (TCRs) on inflammatory CD4+ T cells. Growing evidence suggests that this pathogenic axis can be pharmacologically targeted to protect patients from some of the adverse effects of dietary gluten. Further studies have revealed the existence of additional host and environmental contributors to disease initiation and tissue damage. This review summarizes our current understanding of CeD pathogenesis and how it is being harnessed for therapeutic design and development.


Subject(s)
Celiac Disease , Humans , Celiac Disease/therapy , Celiac Disease/metabolism , Glutens/metabolism , T-Lymphocytes , Receptors, Antigen, T-Cell , Antigen-Presenting Cells
17.
Trends Immunol ; 44(10): 745-747, 2023 10.
Article in English | MEDLINE | ID: mdl-37591713

ABSTRACT

Celiac disease (CeD) is an immune disorder characterized by gluten intolerance that can be unleashed by enteric viral infections in mice. However, Sanchez-Medina et al. recently identified a murine commensal protist, Tritrichomonas arnold, that protects against reovirus-induced intolerance to dietary protein by counteracting virus-induced epithelial stress and proinflammatory dendritic cell (DC) activation.


Subject(s)
Celiac Disease , Virus Diseases , Animals , Mice , Celiac Disease/metabolism , Immune Tolerance
18.
Nutrients ; 15(12)2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37375673

ABSTRACT

BACKGROUND: Non-celiac wheat sensitivity (NCWS) is a poorly understood gluten-related disorder (GRD) and its prominent symptoms can be ameliorated by gluten avoidance. This study aimed to determine the effectiveness of a probiotic mixture in hydrolyzing gliadin peptides (toxic components of gluten) and suppressing gliadin-induced inflammatory responses in Caco-2 cells. METHODS: Wheat dough was fermented with a probiotic mix for 0, 2, 4, and 6 h. The effect of the probiotic mix on gliadin degradation was monitored by SDS-PAGE. The expression levels of IL-6, IL-17A, INF-γ, IL-10, and TGF-ß were evaluated using ELISA and qRT-PCR methods. RESULTS: According to our findings, fermenting wheat dough with a mix of B. longum, L. acidophilus, and L. plantarum for 6 h was effective in gliadin degradation. This process also reduced levels of IL-6 (p = 0.004), IL-17A (p = 0.004), and IFN-γ (p = 0.01) mRNA, as well as decreased IL-6 (p = 0.006) and IFN-γ (p = 0.0009) protein secretion. 4 h fermentation led to a significant decrease in IL-17A (p = 0.001) and IFN-γ (p = 0.003) mRNA, as well as reduced levels of IL-6 (p = 0.002) and IFN-γ (p < 0.0001) protein secretion. This process was also observed to increase the expression levels of IL-10 (p < 0.0001) and TGF-ß (p < 0.0001) mRNA. CONCLUSIONS: 4 h fermentation of wheat flour with the proposed probiotic mix might be a good strategy to develop an affordable gluten-free wheat dough for NCWS and probably other GRD patients.


Subject(s)
Celiac Disease , Gliadin , Humans , Gliadin/adverse effects , Caco-2 Cells , Hydrolysis , Interleukin-10 , Interleukin-17 , Celiac Disease/metabolism , Interleukin-6 , Flour , Triticum/metabolism , Glutens/adverse effects , Lactobacillus acidophilus , Transforming Growth Factor beta
19.
World J Gastroenterol ; 29(13): 1994-2000, 2023 Apr 07.
Article in English | MEDLINE | ID: mdl-37155522

ABSTRACT

BACKGROUND: Celiac disease (CeD) is a multisystem immune-mediated multifactorial condition strongly associated with the intestinal microbiota. AIM: To evaluate the predictive power of the gut microbiota in the diagnosis of CeD and to search for important taxa that may help to distinguish CeD patients from controls. METHODS: Microbial DNA from bacteria, viruses, and fungi, was isolated from mucosal and fecal samples of 40 children with CeD and 39 controls. All samples were sequenced using the HiSeq platform, the data were analyzed, and abundance and diversities were assessed. For this analysis, the predictive power of the microbiota was evaluated by calculating the area under the curve (AUC) using data for the entire microbiome. The Kruskal-Wallis test was used to evaluate the significance of the difference between AUCs. The Boruta logarithm, a wrapper built around the random forest classification algorithm, was used to identify important bacterial biomarkers for CeD. RESULTS: In fecal samples, AUCs for bacterial, viral, and fungal microbiota were 52%, 58%, and 67.7% respectively, suggesting weak performance in predicting CeD. However, the combination of fecal bacteria and viruses showed a higher AUC of 81.8 %, indicating stronger predictive power in the diagnosis of CeD. In mucosal samples, AUCs for bacterial, viral, and fungal microbiota were 81.2%, 58.6%, and 35%, respectively, indicating that mucosal bacteria alone had the highest predictive power. Two bacteria, Bacteroides intestinalis and Burkholderiales bacterium 1-1-47, in fecal samples and one virus, Human_endogenous _retrovirus_K, in mucosal samples are predicted to be "important" biomarkers, differentiating celiac from nonceliac disease groups. Bacteroides intestinalis is known to degrade complex arabinoxylans and xylan which have a protective role in the intestinal mucosa. Similarly, several Burkholderiales species have been reported to produce peptidases that hydrolyze gluten peptides, with the potential to reduce the gluten content of food. Finally, a role for Human_endogenous _retrovirus_K in immune-mediated disease such as CeD has been reported. CONCLUSION: The excellent predictive power of the combination of the fecal bacterial and viral microbiota with mucosal bacteria alone indicates a potential role in the diagnosis of difficult cases of CeD. Bacteroides intestinalis and Burkholderiales bacterium 1-1-47, which were found to be deficient in CeD, have a potential protective role in the development of prophylactic modalities. Further studies on the role of the microbiota in general and Human_endogenous _retrovirus_K in particular are needed.


Subject(s)
Celiac Disease , Gastrointestinal Microbiome , Humans , Child , Celiac Disease/diagnosis , Celiac Disease/epidemiology , Celiac Disease/metabolism , Saudi Arabia/epidemiology , Glutens , Biomarkers/metabolism , Bacteria/genetics , Bacteria/metabolism
20.
Biochem Genet ; 61(6): 2457-2480, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37103600

ABSTRACT

Immunogenicity of gliadin peptides in celiac disease (CD) is majorly determined by the pattern of molecular interactions with HLA-DQ and T-cell receptors (TCR). Investigation of the interactions between immune-dominant gliadin peptides, DQ protein, and TCR are warranted to unravel the basis of immunogenicity and variability contributed by the genetic polymorphisms. Homology modeling of HLA and TCR done using Swiss Model and iTASSER, respectively. Molecular interactions of eight common deamidated immune-dominant gliadin with HLA-DQ allotypes and specific TCR gene pairs were evaluated. Docking of the three structures was performed with ClusPro2.0 and ProDiGY was used to predict binding energies. Effects of known allelic polymorphisms and reported susceptibility SNPs were predicted on protein-protein interactions. CD susceptible allele, HLA-DQ2.5 was shown to have considerable binding affinity to 33-mer gliadin (ΔG = - 13.9; Kd = 1.5E - 10) in the presence of TRAV26/TRBV7. Higher binding affinity was predicted (ΔG = - 14.3, Kd = 8.9E - 11) when TRBV28 was replaced with TRBV20 paired with TRAV4 suggesting its role in CD predisposition. SNP rs12722069 at HLA-DQ8 that codes Arg76α forms three H-bonds with Glu12 and two H-bonds with Asn13 of DQ2 restricted gliadin in the presence of TRAV8-3/TRBV6. None of the HLA-DQ polymorphisms was found to be in linkage disequilibrium with reported CD susceptibility markers. Haplotypic presentations of rs12722069-G, rs1130392-C, rs3188043-C and rs4193-A with CD reported SNPs were observed in sub-ethnic groups. Highly polymorphic sites of HLA alleles and TCR variable regions could be utilized for better risk prediction models in CD. Therapeutic strategies by identifying inhibitors or blockers targeting specific gliadin:HLA-DQ:TCR binding sites could be investigated.


Subject(s)
Celiac Disease , Humans , Celiac Disease/genetics , Celiac Disease/metabolism , Gliadin/genetics , Gliadin/chemistry , HLA-DQ Antigens/genetics , HLA-DQ Antigens/chemistry , HLA-DQ Antigens/metabolism , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Polymorphism, Genetic , Peptides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL