Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.054
Filtrar
1.
Cytokine ; 182: 156722, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39116536

RESUMO

We have previously shown the ability of transamidated gluten (spf) to modulate both innate and adaptive intestinal immunity elicited by wheat gliadin in HLA-DQ8 transgenic mice (DQ8 mice), a model of gluten sensitivity. Herein, we evaluated the influence of spf when administered intragastrically on the immune response to native gliadin in DQ8 mice. To address the issue, we analysed three regimens of antigen administration: before immunisation (pre-treatment), during immunisation (co-treatment) and through breast milk during the lactating phase (suckling treatment). Mice were immunised mucosally by intranasal delivery of digested wheat gliadin along with cholera toxin in multiple doses. After sacrifice, isolated spleen and mesenteric lymph node (MLN) cells were challenged in vitro and the cytokine profile of culture supernatants assessed by ELISA and multiparametric assay. We found that only pre-treatment with spf was effective in down-regulating the gliadin-specific IFN-γ response and only in spleen cells. Interestingly, spf pre-treatment also induced systemic IL-6, IL-17A and TNF-α. By contrast, we found that spf pre-treatment upregulated INF-γ in MLN but also significantly decreased IL-2. In conclusion, our data provide evidence that the preventive intragastric administration of transamidated gluten is able to interfere with the classical cytokine profile induced by gliadin via mucosal immunisation in a transgenic model expressing one of the HLA molecules associated with coeliac disease.


Assuntos
Gliadina , Antígenos HLA-DQ , Camundongos Transgênicos , Triticum , Animais , Gliadina/imunologia , Antígenos HLA-DQ/imunologia , Camundongos , Triticum/imunologia , Feminino , Citocinas/metabolismo , Baço/imunologia , Doença Celíaca/imunologia , Humanos , Toxina da Cólera/farmacologia , Toxina da Cólera/imunologia , Toxina da Cólera/administração & dosagem , Interferon gama/metabolismo , Intestinos/imunologia , Linfonodos/imunologia , Linfonodos/efeitos dos fármacos , Imunização/métodos , Glutens/imunologia , Glutens/administração & dosagem , Fator de Necrose Tumoral alfa/metabolismo , Interleucina-17/metabolismo
2.
Front Immunol ; 15: 1433304, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39161759

RESUMO

Introduction: Understanding intestinal permeability is paramount for elucidating gastrointestinal health and pathology. The size and nature of the molecule traversing the intestinal barrier offer crucial insights into various acute and chronic diseases, as well as the evolution of some conditions. This study aims to assess the urinary excretion kinetics of gluten immunogenic peptides (u-GIP), a unique class of dietary peptides detectable in urine, in volunteers under controlled dietary conditions. This evaluation should be compared to established probes like lactulose, a non-digestible disaccharide indicative of paracellular permeability, and mannitol, reflecting transcellular permeability. Methods: Fifteen participants underwent simultaneous ingestion of standardized doses of gluten (10 g), lactulose (10 g), and mannitol (1 g) under fasting conditions for at least 8 hours pre-ingestion and during 6 hours post-ingestion period. Urine samples were collected over specified time intervals. Excretion patterns were analyzed, and correlations between the lactulose-to-mannitol ratio (LMR) and u-GIP parameters were assessed. Results: The majority of u-GIP were detected within the first 12 hours post-ingestion. Analysis of the variability in cumulative excretion across two sample collection ranges demonstrated that lactulose and u-GIP exhibited similar onset and excretion dynamics, although GIP reached its maximum peak earlier than either lactulose or mannitol. Additionally, a moderate correlation was observed between the LMR and u-GIP parameters within the longest urine collection interval, indicating potential shared characteristics among permeability pathways. These findings suggest that extending urine collection beyond 6 hours may enhance data reliability. Discussion: This study sheds light on the temporal dynamics of u-GIP in comparison to lactulose and mannitol, established probes for assessing intestinal permeability. The resemblance between u-GIP and lactulose excretion patterns aligns with the anticipated paracellular permeability pathway. The capacity to detect antigenic food protein fragments in urine opens novel avenues for studying protein metabolism and monitoring pathologies related to the digestive and intestinal systems.


Assuntos
Jejum , Glutens , Voluntários Saudáveis , Lactulose , Manitol , Humanos , Glutens/urina , Glutens/imunologia , Masculino , Adulto , Feminino , Jejum/urina , Lactulose/urina , Manitol/urina , Adulto Jovem , Peptídeos/urina , Peptídeos/imunologia , Permeabilidade , Biomarcadores/urina , Mucosa Intestinal/metabolismo , Mucosa Intestinal/imunologia , Pessoa de Meia-Idade
3.
World J Gastroenterol ; 30(26): 3201-3205, 2024 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-39086640

RESUMO

In our editorial, we want to comment on the article by Stefanolo et al titled "Effect of Aspergillus niger prolyl endopeptidase in patients with celiac disease on a long-term gluten-free diet". Celiac disease is an immune-mediated disorder triggered by dietary gluten in genetically predisposed individuals. Although avoiding gluten can permit patients to live symptom-free, ongoing voluntary or involuntary exposure to gluten is common and associated with persistent villous atrophy in small bowel mucosa. As villous atrophy predisposes patients to life threatening complications, such as osteoporotic fractures or malignancies, therapeutic adjuncts to gluten-free diet become important to improve patients' quality of life and, if these adjuncts can be shown to improve villous atrophy, avoid complications. Oral administration of enzyme preparations, such as endopeptidases that digest gluten and mitigate its antigenicity to trigger inflammation, is one clinical strategy under investigation. The article is about the utility of one endopeptidase isolated from Aspergillus niger. We critique findings of this clinical trial and also summarize endopeptidase-based as well as other strategies and how they can complement gluten-free diet in the management of celiac disease.


Assuntos
Aspergillus niger , Doença Celíaca , Dieta Livre de Glúten , Glutens , Prolil Oligopeptidases , Humanos , Doença Celíaca/dietoterapia , Doença Celíaca/imunologia , Aspergillus niger/enzimologia , Glutens/imunologia , Glutens/efeitos adversos , Glutens/administração & dosagem , Administração Oral , Mucosa Intestinal/imunologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/enzimologia , Qualidade de Vida , Endopeptidases/metabolismo , Serina Endopeptidases/metabolismo , Serina Endopeptidases/imunologia , Resultado do Tratamento
4.
Front Immunol ; 15: 1405344, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39034995

RESUMO

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.


Assuntos
Doença Celíaca , Glutens , Lactação , Leite Humano , Humanos , Leite Humano/imunologia , Leite Humano/química , Leite Humano/metabolismo , Doença Celíaca/imunologia , Doença Celíaca/metabolismo , Glutens/imunologia , Feminino , Adulto , Estudos Prospectivos , Estudos Longitudinais , Peptídeos/imunologia , Peptídeos/urina , Lactente , Cinética
5.
Int J Mol Sci ; 25(13)2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-39000431

RESUMO

Wheat allergy is a major type of food allergy with the potential for life-threatening anaphylactic reactions. Common wheat, Triticum aestivum (hexaploid, AABBDD genome), was developed using tetraploid wheat (AABB genome) and the ancient diploid wheat progenitor (DD genome)-Aegilops tauschii. The potential allergenicity of gluten from ancient diploid wheat is unknown. In this study, using a novel adjuvant-free gluten allergy mouse model, we tested the hypothesis that the glutenin extract from this ancient wheat progenitor will be intrinsically allergenic in this model. The ancient wheat was grown, and wheat berries were used to extract the glutenin for testing. A plant protein-free colony of Balb/c mice was established and used in this study. The intrinsic allergic sensitization potential of the glutenin was determined by measuring IgE response upon transdermal exposure without the use of an adjuvant. Clinical sensitization for eliciting systemic anaphylaxis (SA) was determined by quantifying the hypothermic shock response (HSR) and the mucosal mast cell response (MMCR) upon intraperitoneal injection. Glutenin extract elicited a robust and specific IgE response. Life-threatening SA associated and a significant MMCR were induced by the glutenin challenge. Furthermore, proteomic analysis of the spleen tissue revealed evidence of in vivo Th2 pathway activation. In addition, using a recently published fold-change analysis method, several immune markers positively and negatively associated with SA were identified. These results demonstrate for the first time that the glutenin from the ancient wheat progenitor is intrinsically allergenic, as it has the capacity to elicit clinical sensitization for anaphylaxis via activation of the Th2 pathway in vivo in mice.


Assuntos
Alérgenos , Anafilaxia , Glutens , Camundongos Endogâmicos BALB C , Células Th2 , Triticum , Hipersensibilidade a Trigo , Animais , Anafilaxia/imunologia , Células Th2/imunologia , Células Th2/metabolismo , Camundongos , Triticum/imunologia , Triticum/química , Glutens/imunologia , Hipersensibilidade a Trigo/imunologia , Alérgenos/imunologia , Imunoglobulina E/imunologia , Imunoglobulina E/sangue , Modelos Animais de Doenças , Feminino , Mastócitos/imunologia , Mastócitos/metabolismo , Mastócitos/efeitos dos fármacos , Proteômica/métodos
6.
World J Gastroenterol ; 30(24): 3044-3047, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38983964

RESUMO

We comment here on the article by Stefanolo et al entitled "Effect of Aspergillus niger prolyl endopeptidase in patients with celiac disease on a long-term gluten-free diet", published in the World Journal of Gastroenterology. Celiac disease is a well-recognized systemic autoimmune disorder. In genetically susceptible people, the most evident damage is located in the small intestine, and is caused and worsened by the ingestion of gluten. For that reason, celiac patients adopt a gluten-free diet (GFD), but it has some limitations, and it does not prevent re-exposure to gluten. Research aims to develop adjuvant therapies, and one of the most studied alternatives is supplementation with Aspergillus niger prolyl endopeptidase protease (AN-PEP), which is able to degrade gluten in the stomach, reducing its concentration in the small intestine. The study found a high adherence to the GFD, but did not address AN-PEP as a gluten immunogenic peptide reducer, as it was only tested in patients following a GFD and not in gluten-exposing conditions. This study opens up new research perspectives in this area and shows that further study is needed to clarify the points that are still in doubt.


Assuntos
Aspergillus niger , Doença Celíaca , Dieta Livre de Glúten , Glutens , Prolil Oligopeptidases , Serina Endopeptidases , Doença Celíaca/imunologia , Doença Celíaca/microbiologia , Doença Celíaca/enzimologia , Humanos , Aspergillus niger/enzimologia , Serina Endopeptidases/metabolismo , Glutens/imunologia , Glutens/metabolismo , Glutens/efeitos adversos , Intestino Delgado/microbiologia , Intestino Delgado/enzimologia , Resultado do Tratamento
7.
Nature ; 632(8024): 401-410, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39048815

RESUMO

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.


Assuntos
Doença Celíaca , Duodeno , Interleucina-7 , Mucosa Intestinal , Modelos Biológicos , Organoides , Humanos , Autoanticorpos/imunologia , Autoimunidade , Linfócitos B/imunologia , Linfócitos B/metabolismo , Biópsia , Doença Celíaca/imunologia , Doença Celíaca/patologia , Doença Celíaca/metabolismo , Duodeno/imunologia , Duodeno/patologia , Duodeno/metabolismo , Epitopos/imunologia , Glutens/imunologia , Glutens/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/imunologia , Antígenos HLA-DQ/imunologia , Antígenos HLA-DQ/metabolismo , Interleucina-7/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Células Matadoras Naturais/imunologia , Células Mieloides/imunologia , Organoides/imunologia , Organoides/metabolismo , Organoides/patologia , Proteína 2 Glutamina gama-Glutamiltransferase/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
8.
Nutrients ; 16(14)2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39064736

RESUMO

BACKGROUND: The differential diagnosis between patients with celiac disease (CD) and non-celiac gluten sensitivity (NCGS) is difficult when a gluten-free diet (GFD) has been initiated before the diagnostic work-up. Isolated increases in TCRγδ+ and celiac lymphogram (increased TCRγδ+ plus decreased CD3-) may enable differential diagnosis in this challenging clinical setting. This study evaluated: (1) the accuracy of %TCRγδ+ and celiac lymphogram for diagnosing CD before and after GFD and for differentiation with NCGS; (2) TCRγδ+ kinetics at baseline and after starting GFD in both CD and NCGS. METHODS: The inclusion criteria were patients with CD (n = 104), NCGS (n = 37), and healthy volunteers (n = 18). An intestinal biopsy for intraepithelial lymphogram by flow cytometry was performed at baseline and after GFD. The optimal cutoff for CD diagnostic accuracy was established by maximizing the Youden index and via logistic regression. RESULTS: %TCRγδ+ showed better diagnostic accuracy than celiac lymphogram for identifying CD before and after GFD initiation. With a cutoff > 13.31, the accuracy for diagnosing CD in patients under GFD was 0.88 [0.80-0.93], whereas the accuracy for diagnosing NCGS (%TCRγδ+ ≤ 13.31) was 0.84 [0.76-0.89]. The percentage of TCRγδ+ cells showed differential kinetics between CD (baseline 22.7% [IQR, 16.4-33.6] vs. after GFD 26.4% [IQR, 17.8-36.8]; p = 0.026) and NCGS (baseline 9.4% [IQR, 4.1-14.6] vs. after GFD 6.4% [IQR, 3.2-11]; p = 0.022). CONCLUSION: TCRγδ+ T cell assessment accurately diagnoses CD before and after a GFD. Increased TCRγδ+ was maintained in the long term after GFD in CD but not in NCGS. Altogether, this suggests the potential usefulness of this marker for the differential diagnosis of these two entities in patients on a GFD.


Assuntos
Biomarcadores , Doença Celíaca , Dieta Livre de Glúten , Glutens , Receptores de Antígenos de Linfócitos T gama-delta , Humanos , Doença Celíaca/diagnóstico , Doença Celíaca/dietoterapia , Doença Celíaca/imunologia , Feminino , Diagnóstico Diferencial , Masculino , Adulto , Glutens/imunologia , Pessoa de Meia-Idade , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/imunologia , Adulto Jovem , Linfócitos Intraepiteliais/imunologia
9.
JAMA ; 332(3): 249-250, 2024 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-38922595

RESUMO

This JAMA Clinical Guidelines Synopsis summarizes the American College of Gastroenterology's 2023 guideline update on diagnosis and management of celiac disease.


Assuntos
Doença Celíaca , Dieta Livre de Glúten , Doença Celíaca/diagnóstico , Doença Celíaca/terapia , Doença Celíaca/dietoterapia , Humanos , Glutens/imunologia , Glutens/efeitos adversos
10.
Nutrients ; 16(11)2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38892641

RESUMO

Potential celiac disease (PCD) is a clinical condition characterised by the presence of a positive CD-specific serology and a normal intestinal architecture. Asymptomatic PCD patients are generally advised to continue on a gluten-containing diet (GCD), but long-term risks of this approach have never been explored. In the present study, we aimed to investigate nutritional and autoimmune complications possibly developing overtime in a cohort of asymptomatic PCD children on a GCD. We compared children's parameters of growth, nutritional status, and autoimmunity between the time of diagnosis and on the occasion of their last medical check, after a long-term gluten-containing diet. Altogether, we collected data from 171 PCD children with a mean follow-up time of 3 years (range 0.35-15.3 years). During follow-up, although patients did not reduce their amount of daily gluten intake, their anti-tissue transglutaminase (anti-TG2) antibodies spontaneously and significantly decreased. Most parameters analysed had not changed during follow-up (height centile, ferritin, albumin, cholesterol, calcium, alkaline phosphatase, parathormone, and vitamin D) or even improved significantly (weight and BMI centile, haemoglobin, blood iron, HDL, glycaemia, and HbA1C, p < 0.05), always remaining within the limit of normality. Equally, autoantibodies for other concomitant autoimmune disorders did not increase overtime. Similar results were obtained excluding from analysis patients who had stopped producing anti-TG2 and those with a follow-up time < 3 years. Our pilot study has provided reassuring results regarding the maintenance of a gluten-containing diet in asymptomatic PCD children, even when long-term follow-up was considered.


Assuntos
Autoanticorpos , Doença Celíaca , Dieta Livre de Glúten , Estado Nutricional , Humanos , Doença Celíaca/dietoterapia , Doença Celíaca/imunologia , Criança , Masculino , Feminino , Pré-Escolar , Adolescente , Autoanticorpos/sangue , Proteína 2 Glutamina gama-Glutamiltransferase , Proteínas de Ligação ao GTP/imunologia , Transglutaminases/imunologia , Glutens/efeitos adversos , Glutens/imunologia , Nível de Saúde , Lactente , Seguimentos , Autoimunidade
11.
Nutrients ; 16(12)2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38931237

RESUMO

Celiac disease (CeD) is an autoimmune disease with a strong association with human leukocyte antigen (HLA), characterized by the production of specific autoantibodies and immune-mediated enterocyte killing. CeD is a unique autoimmune condition, as it is the only one in which the environmental trigger is known: gluten, a storage protein present in wheat, barley, and rye. How and when the loss of tolerance of the intestinal mucosa to gluten occurs is still unknown. This event, through the activation of adaptive immune responses, enhances epithelial cell death, increases the permeability of the epithelial barrier, and induces secretion of pro-inflammatory cytokines, resulting in the transition from genetic predisposition to the actual onset of the disease. While the role of gastrointestinal infections as a possible trigger has been considered on the basis of a possible mechanism of antigen mimicry, a more likely alternative mechanism appears to involve a complex disruption of the gastrointestinal microbiota ecosystem triggered by infections, rather than the specific effect of a single pathogen on intestinal mucosal homeostasis. Several lines of evidence show the existence of intestinal dysbiosis that precedes the onset of CeD in genetically at-risk subjects, characterized by the loss of protective bacterial elements that both epigenetically and functionally can influence the response of the intestinal epithelium leading to the loss of gluten tolerance. We have conducted a literature review in order to summarize the current knowledge about the complex and in part still unraveled dysbiosis that precedes and accompanies CeD and present some exciting new data on how this dysbiosis might be prevented and/or counteracted. The literature search was conducted on PubMed.gov in the time frame 2010 to March 2024 utilizing the terms "celiac disease and microbiota", "celiac disease and microbiome", and "celiac disease and probiotics" and restricting the search to the following article types: Clinical Trials, Meta-Analysis, Review, and Systematic Review. A total of 364 papers were identified and reviewed. The main conclusions of this review can be outlined as follows: (1) quantitative and qualitative changes in gut microbiota have been clearly documented in CeD patients; (2) intestinal microbiota's extensive and variable interactions with enterocytes, viral and bacterial pathogens and even gluten combine to impact the inflammatory immune response to gluten and the loss of gluten tolerance, ultimately affecting the pathogenesis, progression, and clinical expression of CeD; (3) gluten-free diet fails to restore the eubiosis of the digestive tract in CeD patients, and also negatively affects microbial homeostasis; (4) new tools allowing targeted microbiota therapy, such as the use of probiotics (a good example being precision probiotics like the novel strain of B. vulgatus (20220303-A2) begin to show exciting potential applications.


Assuntos
Doença Celíaca , Disbiose , Microbioma Gastrointestinal , Glutens , Doença Celíaca/imunologia , Doença Celíaca/microbiologia , Humanos , Glutens/imunologia , Glutens/efeitos adversos , Disbiose/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/metabolismo
12.
Nat Immunol ; 25(7): 1218-1230, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38914866

RESUMO

Transglutaminase 2 (TG2) plays a pivotal role in the pathogenesis of celiac disease (CeD) by deamidating dietary gluten peptides, which facilitates antigenic presentation and a strong anti-gluten T cell response. Here, we elucidate the molecular mechanisms underlying the efficacy of the TG2 inhibitor ZED1227 by performing transcriptional analysis of duodenal biopsies from individuals with CeD on a long-term gluten-free diet before and after a 6-week gluten challenge combined with 100 mg per day ZED1227 or placebo. At the transcriptome level, orally administered ZED1227 effectively prevented gluten-induced intestinal damage and inflammation, providing molecular-level evidence that TG2 inhibition is an effective strategy for treating CeD. ZED1227 treatment preserved transcriptome signatures associated with mucosal morphology, inflammation, cell differentiation and nutrient absorption to the level of the gluten-free diet group. Nearly half of the gluten-induced gene expression changes in CeD were associated with the epithelial interferon-γ response. Moreover, data suggest that deamidated gluten-induced adaptive immunity is a sufficient step to set the stage for CeD pathogenesis. Our results, with the limited sample size, also suggest that individuals with CeD might benefit from an HLA-DQ2/HLA-DQ8 stratification based on gene doses to maximally eliminate the interferon-γ-induced mucosal damage triggered by gluten.


Assuntos
Doença Celíaca , Dieta Livre de Glúten , Proteínas de Ligação ao GTP , Perfilação da Expressão Gênica , Glutens , Mucosa Intestinal , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases , Doença Celíaca/imunologia , Humanos , Glutens/imunologia , Transglutaminases/metabolismo , Transglutaminases/antagonistas & inibidores , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/antagonistas & inibidores , Proteínas de Ligação ao GTP/genética , Mucosa Intestinal/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/efeitos dos fármacos , Feminino , Masculino , Adulto , Transcriptoma , Duodeno/patologia , Duodeno/imunologia , Duodeno/metabolismo , Interferon gama/metabolismo , Pessoa de Meia-Idade , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/imunologia , Adulto Jovem , Imunidade Adaptativa/efeitos dos fármacos
13.
Clin Immunol ; 264: 110259, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38768856

RESUMO

The gluten-free diet for celiac disease (CeD) is restrictive and often fails to induce complete symptom and/or mucosal disease remission. Central to CeD pathogenesis is the gluten-specific CD4+ T cell that is restricted by HLA-DQ2.5 in over 85% of CeD patients, making HLA-DQ2.5 an attractive target for suppressing gluten-dependent immunity. Recently, a novel anti-HLA-DQ2.5 antibody that specifically recognizes the complexes of HLA-DQ2.5 and multiple gluten epitopes was developed (DONQ52). OBJECTIVE: To assess the ability of DONQ52 to inhibit CeD patient-derived T-cell responses to the most immunogenic gluten peptides that encompass immunodominant T cell epitopes. METHODS: We employed an in vivo gluten challenge model in patients with CeD that affords a quantitative readout of disease-relevant gluten-specific T-cell responses. HLA-DQ2.5+ CeD patients consumed food containing wheat, barley, or rye for 3 days with collection of blood before (D1) and 6 days after (D6) commencing the challenge. Peripheral blood mononuclear cells were isolated and assessed in an interferon (IFN)-γ enzyme-linked immunosorbent spot assay (ELISpot) testing responses to gluten peptides encompassing a series of immunodominant T cell epitopes. The inhibitory effect of DONQ52 (4 or 40 µg/mL) was assessed and compared to pan-HLA-DQ blockade (SPVL3 antibody). RESULTS: In HLA-DQ2.5+ CeD patients, DONQ52 reduced T cell responses to all wheat gluten peptides to an equivalent or more effective degree than pan-HLA-DQ antibody blockade. It reduced T cell responses to a cocktail of the most immunodominant wheat epitopes by a median of 87% (IQR 72-92). Notably, DONQ52 also substantially reduced T-cell responses to dominant barley hordein and rye secalin derived peptides. DONQ52 had no effect on T-cell responses to non-gluten antigens. CONCLUSION: DONQ52 can significantly block HLA-DQ2.5-restricted T cell responses to the most highly immunogenic gluten peptides in CeD. Our findings support in vitro data that DONQ52 displays selectivity and broad cross-reactivity against multiple gluten peptide:HLA-DQ2.5 complexes. This work provides proof-of-concept multi-specific antibody blockade has the potential to meaningfully inhibit pathogenic gluten-specific T-cell responses in CeD and supports ongoing therapeutic development.


Assuntos
Anticorpos Biespecíficos , Doença Celíaca , Glutens , Antígenos HLA-DQ , Humanos , Doença Celíaca/imunologia , Glutens/imunologia , Antígenos HLA-DQ/imunologia , Anticorpos Biespecíficos/imunologia , Anticorpos Biespecíficos/farmacologia , Feminino , Epitopos de Linfócito T/imunologia , Adulto , Masculino , Linfócitos T CD4-Positivos/imunologia , Peptídeos/imunologia , Pessoa de Meia-Idade , Linfócitos T/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Epitopos Imunodominantes/imunologia , Dieta Livre de Glúten
14.
Dig Dis Sci ; 69(8): 2916-2921, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38769224

RESUMO

BACKGROUND: Patients on a gluten-free diet (GFD) whose celiac disease (CD) status is unknown may undergo gluten challenge (GC) to clarify their diagnosis. Though this is an established diagnostic practice, the proportion of patients undergoing GC who are diagnosed with CD is unknown. AIMS: We aimed to analyze which factors were predictive of having CD in a cohort of patients who underwent GC followed by upper endoscopy with duodenal biopsy. METHODS: We identified adult patients at a CD referral center who had been on a GFD and then underwent GC to determine a diagnosis of CD during the years spanning 2006 to 2020. We compared those patients found to have CD (defined as villus atrophy/Marsh 3) on duodenal biopsy with those who did not, using the chi square and Fischer exact tests. RESULTS: We identified 206 patients who underwent GC. Of these 206, 30 (14%) were diagnosed with CD based on post-GC duodenal biopsy. 176 of the 206 (85%) patients reported various gastrointestinal symptoms, including bloating (39%), though these were more common in those without CD (any GI symptoms: 89% vs 67%, p 0.004; bloating: 43% vs 20%, p 0.019). Serology values, when normalized, including pre- and post-challenge TTG IgA (37% vs 1.7%, p 0.001; 23% versus 2.3%, p 0.001), DGP IgG and IgA (57% vs 2.8%, p 0.001; 37% vs 6.2%, p 0.001) were higher in the group of patients with CD. CONCLUSION: Among patients undergoing GC for diagnostic purposes, only 14% had evidence of villus atrophy corresponding with CD on duodenal biopsy. The presence of any elevated pre-challenge serology was associated with CD. Bloating in combination with low serologies may help risk stratify patients as being less likely to have CD upon GC.


Assuntos
Doença Celíaca , Dieta Livre de Glúten , Duodeno , Glutens , Humanos , Doença Celíaca/dietoterapia , Doença Celíaca/diagnóstico , Masculino , Feminino , Pessoa de Meia-Idade , Adulto , Duodeno/patologia , Glutens/efeitos adversos , Glutens/administração & dosagem , Glutens/imunologia , Biópsia , Idoso , Estudos Retrospectivos
15.
Int Immunopharmacol ; 133: 112020, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38608449

RESUMO

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.


Assuntos
Doença Celíaca , Dieta Livre de Glúten , Humanos , Doença Celíaca/dietoterapia , Doença Celíaca/terapia , Doença Celíaca/imunologia , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Proteína 2 Glutamina gama-Glutamiltransferase , Imunoterapia/métodos , Glutens/imunologia , Transglutaminases/imunologia , Transglutaminases/metabolismo
16.
Gastroenterology ; 167(1): 51-63, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38636679

RESUMO

Views on the clinical presentation and symptomatology of celiac disease have evolved alongside advances in disease detection and understanding of disease pathogenesis. Although historically regarded as a pediatric illness characterized by malabsorption, it is now better viewed as an immune illness of gluten-specific T cells with systemic manifestations affecting all ages. Its broad presentation, including frequent extraintestinal manifestations and asymptomatic disease, contributes to suboptimal disease detection. Adverse symptoms greatly impact patient quality of life and can result from chronic gluten exposure in untreated disease or those poorly responsive to the gluten-free diet. They can also present as acute symptoms after episodic gluten exposure. Functional gastrointestinal disease is a common comorbidity. Biomarkers like interleukin-2 that are highly sensitive and specific for celiac disease highlight a role for gluten-specific T cells in acute gluten symptomatology. A mechanistic understanding of symptoms will inform approaches to better measure and treat them effectively.


Assuntos
Doença Celíaca , Dieta Livre de Glúten , Glutens , Doença Celíaca/diagnóstico , Doença Celíaca/imunologia , Doença Celíaca/dietoterapia , Doença Celíaca/epidemiologia , Humanos , Glutens/imunologia , Glutens/efeitos adversos , Biomarcadores/sangue , Qualidade de Vida , Linfócitos T/imunologia
17.
Gastroenterology ; 167(1): 159-171, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38670279

RESUMO

Although many biomarkers have been proposed, and several are in widespread clinical use, there is no single readout or combination of readouts that correlates tightly with gluten exposure, disease activity, or end-organ damage in treated patients with celiac disease. Challenges to developing and evaluating better biomarkers include significant interindividual variability-related to immune amplification of gluten exposure and how effects of immune activation are manifest. Furthermore, the current "gold standard" for assessment of end-organ damage, small intestinal biopsy, is itself highly imperfect, such that a marker that is a better reflection of the "ground truth" may indeed appear to perform poorly. The goal of this review was to analyze past and present efforts to establish robust noninvasive tools for monitoring treated patients with celiac disease and to highlight emerging tools that may prove to be useful in clinical practice.


Assuntos
Biomarcadores , Doença Celíaca , Glutens , Doença Celíaca/diagnóstico , Doença Celíaca/imunologia , Doença Celíaca/dietoterapia , Humanos , Biomarcadores/análise , Glutens/imunologia , Glutens/efeitos adversos , Biópsia , Dieta Livre de Glúten , Valor Preditivo dos Testes , Índice de Gravidade de Doença
18.
Gastroenterology ; 167(1): 4-22, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38670280

RESUMO

Celiac disease (CeD) is a gluten-induced enteropathy that develops in genetically susceptible individuals upon consumption of cereal gluten proteins. It is a unique and complex immune disorder to study as the driving antigen is known and the tissue targeted by the immune reaction can be interrogated. This review integrates findings gained from genetic, biochemical, and immunologic studies, which together have revealed mechanisms of gluten peptide modification and HLA binding, thereby enabling a maladapted anti-gluten immune response. Observations in human samples combined with experimental mouse models have revealed that the gluten-induced immune response involves CD4+ T cells, cytotoxic CD8+ T cells, and B cells; their cross-talks are critical for the tissue-damaging response. The emergence of high-throughput technologies is increasing our understanding of the phenotype, location, and presumably function of the gluten-specific cells, which are all required to identify novel therapeutic targets and strategies for CeD.


Assuntos
Doença Celíaca , Predisposição Genética para Doença , Glutens , Doença Celíaca/imunologia , Doença Celíaca/genética , Humanos , Glutens/imunologia , Glutens/efeitos adversos , Animais , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia
19.
Cell Rep ; 43(4): 114045, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38578826

RESUMO

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.


Assuntos
Linfócitos B , Doença Celíaca , Proteínas de Ligação ao GTP , Imunoglobulina A , Plasmócitos , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases , Doença Celíaca/imunologia , Doença Celíaca/patologia , Humanos , Transglutaminases/imunologia , Transglutaminases/metabolismo , Imunoglobulina A/imunologia , Imunoglobulina A/metabolismo , Imunoglobulina A/sangue , Linfócitos B/imunologia , Linfócitos B/metabolismo , Plasmócitos/imunologia , Plasmócitos/metabolismo , Proteínas de Ligação ao GTP/imunologia , Proteínas de Ligação ao GTP/metabolismo , Autoanticorpos/imunologia , Autoanticorpos/sangue , Adulto , Masculino , Feminino , Pessoa de Meia-Idade , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Glutens/imunologia
20.
Int J Mol Sci ; 25(8)2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38673984

RESUMO

Gluten hypersensitivity is characterized by the production of IgE antibodies against specific wheat proteins (allergens) and a myriad of clinical allergic symptoms including life-threatening anaphylaxis. Currently, the only recommended treatment for gluten hypersensitivity is the complete avoidance of gluten. There have been extensive efforts to develop dietary-based novel therapeutics for combating this disorder. There were four objectives for this study: (i) to compile the current understanding of the mechanism of gluten hypersensitivity; (ii) to critically evaluate the outcome from preclinical testing of novel therapeutics in animal models; (iii) to determine the potential of novel dietary-based therapeutic approaches under development in humans; and (iv) to synthesize the outcomes from these studies and identify the gaps in research to inform future translational research. We used Google Scholar and PubMed databases with appropriate keywords to retrieve published papers. All material was thoroughly checked to obtain the relevant data to address the objectives. Our findings collectively demonstrate that there are at least five promising dietary-based therapeutic approaches for mitigating gluten hypersensitivity in development. Of these, two have advanced to a limited human clinical trial, and the others are at the preclinical testing level. Further translational research is expected to offer novel dietary-based therapeutic options for patients with gluten hypersensitivity in the future.


Assuntos
Glutens , Humanos , Glutens/imunologia , Animais , Hipersensibilidade Alimentar/dietoterapia , Hipersensibilidade Alimentar/terapia , Hipersensibilidade Alimentar/imunologia , Alérgenos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA