Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 147
Filtrar
1.
J Autoimmun ; 146: 103241, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38754235

RESUMEN

Many antibody responses induced by infection, vaccination or autoimmunity show signs of convergence across individuals with epitope-dependent selection of particular variable region gene segments and complementarity determining region 3 properties. However, not much is known about the relationship between antigen-specific effector cells and antigen-specific precursors present in the naïve B-cell repertoire. Here, we sought to address this relationship in the context of celiac disease, where there is a stereotyped autoantibody response against the enzyme transglutaminase 2 (TG2). By generating TG2-specific monoclonal antibodies from both duodenal plasma cells and circulating naïve B cells, we demonstrate a discord between the naïve TG2-specific repertoire and the cells that are selected for autoantibody production. Hence, the naïve repertoire does not fully reflect the epitope preference and gene usage observed for memory B cells and plasma cells. Instead, distinct naïve B cells that target particular TG2 epitopes appear to be selectively activated at the expense of TG2-binding B cells targeting other epitopes.

2.
Cell Rep ; 43(4): 114045, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38578826

RESUMEN

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.


Asunto(s)
Linfocitos B , Enfermedad Celíaca , Proteínas de Unión al GTP , Inmunoglobulina A , Células Plasmáticas , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transglutaminasas , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/patología , Humanos , Transglutaminasas/inmunología , Transglutaminasas/metabolismo , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Inmunoglobulina A/sangre , Linfocitos B/inmunología , Linfocitos B/metabolismo , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Proteínas de Unión al GTP/inmunología , Proteínas de Unión al GTP/metabolismo , Autoanticuerpos/inmunología , Autoanticuerpos/sangre , Adulto , Masculino , Femenino , Persona de Mediana Edad , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Glútenes/inmunología
3.
Gastroenterology ; 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38552723

RESUMEN

BACKGROUND & AIMS: The treatment of celiac disease (CeD) with gluten-free diet (GFD) normalizes gut inflammation and disease-specific antibodies. CeD patients have HLA-restricted, gluten-specific T cells persisting in the blood and gut even after decades of GFD, which are reactivated and disease driving upon gluten exposure. Our aim was to examine the transition of activated gluten-specific T cells into a pool of persisting memory T cells concurrent with normalization of clinically relevant biomarkers during the first year of treatment. METHODS: We followed 17 CeD patients during their initial GFD year, leading to disease remission. We assessed activation and frequency of gluten-specific CD4+ blood and gut T cells with HLA-DQ2.5:gluten tetramers and flow cytometry, disease-specific serology, histology, and symptom scores. We assessed gluten-specific blood T cells within the first 3 weeks of GFD in 6 patients and serology in an additional 9 patients. RESULTS: Gluten-specific CD4+ T cells peaked in blood at day 14 while up-regulating Bcl-2 and down-regulating Ki-67 and then decreased in frequency within 10 weeks of GFD. CD38, ICOS, HLA-DR, and Ki-67 decreased in gluten-specific cells within 3 days. PD-1, CD39, and OX40 expression persisted even after 12 months. IgA-transglutaminase 2 decreased significantly within 4 weeks. CONCLUSIONS: GFD induces rapid changes in the phenotype and number of gluten-specific CD4+ blood T cells, including a peak of nonproliferating, nonapoptotic cells at day 14. Subsequent alterations in T-cell phenotype associate with the quiescent but chronic nature of treated CeD. The rapid changes affecting gluten-specific T cells and disease-specific antibodies offer opportunities for clinical trials aiming at developing nondietary treatments for patients with newly diagnosed CeD.

4.
Gastroenterology ; 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38467384

RESUMEN

BACKGROUND & AIMS: Histologic evaluation of gut biopsy specimens 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 biopsy specimens collected before and after treatment with a gluten-free diet as well as biopsy specimens from patients with CeD with varying degree of remission (n = 22). Biopsy specimens 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 biopsy specimens 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.

6.
Gastroenterology ; 2024 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-38286391

RESUMEN

In its conventional form, celiac disease (CeD) is characterized by both positive serology and flat villi in the duodenum, and is well known by gastroenterologists and general practitioners. The aim of this review was to shed light on 2 neglected and not yet well-defined celiac phenotypes, that is, seronegative and ultrashort CeD. Seronegative CeD can be suspected in the presence of flat villi, positive HLA-DQ2 and/or HLA-DQ8, and the absence of CeD antibodies. After ruling out other seronegative enteropathies, the diagnosis can be confirmed by both clinical and histologic improvements after 1 year of a gluten-free diet. Ultrashort CeD is characterized by the finding of flat villi in the duodenal bulb in the absence of mucosal damage in the distal duodenum and with serologic positivity. Data on the prevalence, clinical manifestations, histologic lesions, genetic features, and outcome of seronegative and ultrashort CeD are inconclusive due to the few studies available and the small number of patients diagnosed. Some additional diagnostic tools have been developed recently, such as assessing intestinal transglutaminase 2 deposits, flow cytometry technique, microRNA detection, or proteomic analysis, and they seem to be useful in the identification of complex cases. Further cooperative studies are highly desirable to improve the knowledge of these 2 still-obscure variants of CeD.

7.
Gastroenterology ; 166(1): 88-102, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37704112

RESUMEN

BACKGROUND & AIMS: There is a need to develop safe and effective pharmacologic options for the treatment of celiac disease (CeD); however, consensus on the appropriate design and configuration of randomized controlled trials (RCTs) in this population is lacking. METHODS: A 2-round modified Research and Development/University of California Los Angeles Appropriateness Method study was conducted. Eighteen gastroenterologists (adult and pediatric) and gastrointestinal pathologists voted on statements pertaining to the configuration of CeD RCTs, inclusion and exclusion criteria, gluten challenge, and trial outcomes. Two RCT designs were considered, representing the following distinct clinical scenarios for which pharmacotherapy may be used: trials incorporating a gluten challenge to simulate exposure; and trials evaluating reversal of histologic changes, despite attempted adherence to a gluten-free diet. Each statement was rated as appropriate, uncertain, or inappropriate, using a 9-point Likert scale. RESULTS: For trials evaluating prevention of relapse after gluten challenge, participants adherent to a gluten-free diet for 12 months or more with normal or near-normal-sized villi should be enrolled. Gluten challenge should be FODMAPS (fermentable oligosaccharides, disaccharides, monosaccharides, and polyols) free, and efficacy evaluated using histology with a secondary patient-reported outcome measure. For trials evaluating reversal of villus atrophy, the panel voted it appropriate to enroll participants with a baseline villus height to crypt depth ratio ≤2 and measure efficacy using a primary histologic end point. Guidance for measuring histologic, endoscopic, and patient-reported outcomes in adult and pediatric patients with CeD are provided, along with recommendations regarding the merits and limitations of different end points. CONCLUSIONS: We developed standardized recommendations for clinical trial design, eligibility criteria, outcome measures, gluten challenge, and disease evaluations for RCTs in patients with CeD.


Asunto(s)
Enfermedad Celíaca , Adulto , Humanos , Niño , Enfermedad Celíaca/patología , Recurrencia Local de Neoplasia , Ensayos Clínicos Controlados Aleatorios como Asunto , Glútenes/efectos adversos , Dieta Sin Gluten
8.
Nat Rev Gastroenterol Hepatol ; 21(3): 198-215, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38110546

RESUMEN

Coeliac disease (CeD) is an immunological disease triggered by the consumption of gluten contained in food in individuals with a genetic predisposition. Diagnosis is based on the presence of small bowel mucosal atrophy and circulating autoantibodies (anti-type 2 transglutaminase antibodies). After diagnosis, patients follow a strict, life-long gluten-free diet. Although the criteria for diagnosis of this disease are well defined, the monitoring phase has been studied less and there is a lack of specific guidelines for this phase. To develop a set of clinical guidelines for CeD monitoring, we followed the Grading of Recommendations Assessment, Development and Evaluation methodology. Statements and recommendations with the level of evidence were developed and approved by the working group, which comprised gastroenterologists, pathologists, dieticians and biostatisticians. The proposed guidelines, endorsed by the North American and European coeliac disease scientific societies, make recommendations for best practices in monitoring patients with CeD based on the available evidence. The evidence level is low for many topics, suggesting that further research in specific aspects of CeD would be valuable. In conclusion, the present guidelines support clinicians in improving CeD treatment and follow-up and highlight novel issues that should be considered in future studies.


Asunto(s)
Enfermedad Celíaca , Gastroenterólogos , Adulto , Humanos , Enfermedad Celíaca/diagnóstico , Autoanticuerpos , Dieta Sin Gluten , Predisposición Genética a la Enfermedad
9.
Nature ; 624(7992): 653-662, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37993717

RESUMEN

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.


Asunto(s)
Amelogénesis Imperfecta , Autoanticuerpos , Enfermedad Celíaca , Poliendocrinopatías Autoinmunes , Humanos , Amelogénesis Imperfecta/complicaciones , Amelogénesis Imperfecta/inmunología , Autoanticuerpos/inmunología , Enfermedad Celíaca/complicaciones , Enfermedad Celíaca/inmunología , Inmunoglobulina A/inmunología , Poliendocrinopatías Autoinmunes/complicaciones , Poliendocrinopatías Autoinmunes/inmunología , Proteínas/inmunología , Proteínas/metabolismo , Ameloblastos/metabolismo , Esmalte Dental/inmunología , Esmalte Dental/metabolismo , Proteína AIRE/deficiencia , Antígenos/inmunología , Antígenos/metabolismo , Intestinos/inmunología , Intestinos/metabolismo
11.
Adv Sci (Weinh) ; 10(25): e2300401, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37424036

RESUMEN

Dermatitis herpetiformis (DH) is an inflammatory skin disorder often considered as an extra intestinal manifestation of celiac disease (CeD). Hallmarks of CeD and DH are auto-antibodies to transglutaminase 2 (TG2) and transglutaminase 3 (TG3), respectively. DH patients have auto-antibodies reactive with both transglutaminase enzymes. Here it is reported that in DH both gut plasma cells and serum auto-antibodies are specific for either TG2 or TG3 with no TG2-TG3 cross reactivity. By generating monoclonal antibodies from TG3-specific duodenal plasma cells of DH patients, three conformational epitope groups are defined. Both TG2-specific and TG3-specific gut plasma cells have few immunoglobulin (Ig) mutations, and the two transglutaminase-reactive populations show distinct selection of certain heavy and light chain V-genes. Mass spectrometry analysis of TG3-specific serum IgA corroborates preferential usage of IGHV2-5 in combination with IGKV4-1. Collectively, these results demonstrate parallel induction of anti-TG2 and anti-TG3 auto-antibody responses involving separate B-cell populations in DH patients.


Asunto(s)
Enfermedad Celíaca , Dermatitis Herpetiforme , Humanos , Inmunoglobulina A , Células Plasmáticas , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transglutaminasas
12.
Br J Nutr ; 130(12): 2061-2075, 2023 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-37272479

RESUMEN

Individuals with coeliac disease (CeD) often experience gastrointestinal symptoms despite adherence to a gluten-free diet (GFD). While we recently showed that a diet low in fermentable oligo-, di-, monosaccharides and polyols (FODMAP) successfully provided symptom relief in GFD-treated CeD patients, there have been concerns that the low FODMAP diet (LFD) could adversely affect the gut microbiota. Our main objective was therefore to investigate whether the LFD affects the faecal microbiota and related variables of gut health. In a randomised controlled trial GFD-treated CeD adults, having persistent gastrointestinal symptoms, were randomised to either consume a combined LFD and GFD (n 39) for 4 weeks or continue with GFD (controls, n 36). Compared with the control group, the LFD group displayed greater changes in the overall faecal microbiota profile (16S rRNA gene sequencing) from baseline to follow-up (within-subject ß-diversity, P < 0·001), characterised by lower and higher follow-up abundances (%) of genus Anaerostipes (Pgroup < 0·001) and class Erysipelotrichia (Pgroup = 0·02), respectively. Compared with the control group, the LFD led to lower follow-up concentrations of faecal propionic and valeric acid (GC-FID) in participants with high concentrations at baseline (Pinteraction ≤ 0·009). No differences were found in faecal bacterial α-diversity (Pgroup ≥ 0·20) or in faecal neutrophil gelatinase-associated lipocalin (ELISA), a biomarker of gut integrity and inflammation (Pgroup = 0·74), between the groups at follow-up. The modest effects of the LFD on the gut microbiota and related variables in the CeD patients of the present study are encouraging given the beneficial effects of the LFD strategy to treat functional GI symptoms (Registered at clinicaltrials.gov as NCT03678935).


Asunto(s)
Enfermedad Celíaca , Microbioma Gastrointestinal , Síndrome del Colon Irritable , Adulto , Humanos , Dieta Baja en Carbohidratos , Dieta FODMAP , ARN Ribosómico 16S/genética , Dieta , Monosacáridos , Dieta Sin Gluten , Síndrome del Colon Irritable/diagnóstico , Fermentación , Oligosacáridos
13.
PLoS One ; 18(6): e0287662, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37368893

RESUMEN

BACKGROUND: Formation of complexes between transglutaminase 2 (TG2) and gluten can mechanistically explain why TG2 serves both as B-cell autoantigen and as an enzyme that creates deamidated gluten epitopes in coeliac disease (CeD). A model has been proposed where TG2 released from shed epithelial cells encounters high concentrations of dietary gluten peptides to form these TG2:gluten complexes. In this work we have characterised TG2 protein expression in gut epithelial cells in humans. METHODS: Western blot analysis, immunofluorescence staining and mass spectrometry in combination with laser capture microdissection to gain spatial resolution were used to characterise TG2 expression in the epithelial cell layer of healthy and coeliac disease affected duodenum. FINDINGS: TG2 is expressed in human duodenal epithelial cells, including cells in the apical region that are shed into the gut lumen. In untreated CeD the apical expression of TG2 is doubled. Enzymatically active TG2 is readily released from isolated human intestinal epithelial cells. CONCLUSION: Shed epithelial cells are a plausible source of pathogenic TG2 enzyme in CeD. Increased epithelial TG2 expression and increased epithelial shedding in active CeD may reinforce action of luminal TG2 in this condition.


Asunto(s)
Enfermedad Celíaca , Proteína Glutamina Gamma Glutamiltransferasa 2 , Humanos , Autoanticuerpos , Células Epiteliales/metabolismo , Glútenes/metabolismo , Transglutaminasas/metabolismo
14.
J Allergy Clin Immunol ; 152(1): 278-289.e6, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36893861

RESUMEN

BACKGROUND: Regulatory T (Treg) CD4 cells in mouse gut are mainly specific for intestinal antigens and play an important role in the suppression of immune responses against harmless dietary antigens and members of the microbiota. However, information about the phenotype and function of Treg cells in the human gut is limited. OBJECTIVE: We performed a detailed characterization of Foxp3+ CD4 Treg cells in human normal small intestine (SI) as well as from transplanted duodenum and celiac disease lesions. METHODS: Treg cells and conventional CD4 T cells derived from SI were subjected to extensive immunophenotyping and their suppressive activity and ability to produce cytokines assessed. RESULTS: SI Foxp3+ CD4 T cells were CD45RA-CD127-CTLA-4+ and suppressed proliferation of autologous T cells. Approximately 60% of Treg cells expressed the transcription factor Helios. When stimulated, Helios-negative Treg cells produced IL-17, IFN-γ, and IL-10, whereas Helios-positive Treg cells produced very low levels of these cytokines. By sampling mucosal tissue from transplanted human duodenum, we demonstrated that donor Helios-negative Treg cells persisted for at least 1 year after transplantation. In normal SI, Foxp3+ Treg cells constituted only 2% of all CD4 T cells, while in active celiac disease, both Helios-negative and Helios-positive subsets expanded 5- to 10-fold. CONCLUSION: The SI contains 2 subsets of Treg cells with different phenotypes and functional capacities. Both subsets are scarce in healthy gut but increase dramatically in active celiac disease.


Asunto(s)
Enfermedad Celíaca , Linfocitos T Reguladores , Humanos , Animales , Ratones , Citocinas , Intestino Delgado , Factores de Transcripción Forkhead , Subgrupos de Linfocitos T
15.
Clin Nutr ESPEN ; 54: 251-257, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36963870

RESUMEN

BACKGROUND & AIMS: The nutritional quality of a gluten-free diet is debated because of the elimination of grains that are important sources of nutrients. The aim of this cross-sectional study was to perform a nutritional assessment in treated women with celiac disease and ongoing symptoms, and compare dietary intake with a healthy reference group (Norkost 3). METHODS: Celiac disease patients with biopsy confirmed mucosal healing, but persistent gastrointestinal symptoms, were included from an ongoing clinical trial. Nutritional assessment included anthropometrics, blood samples and dietary intake obtained by two 24 h recalls. Dietary intake in celiac women was compared with dietary intake in healthy women (Norkost 3). Two sample t-test was used for comparison of CeD and Norkost 3 women. Adjustment for age, BMI, education and smoking, by use of multiple linear regression analysis, did not change the results. RESULTS: In total, 59 women with celiac disease and 925 women that participated in Norkost 3 were included, with a mean age of 45 years in both groups. Women with celiac disease had a higher proportion of energy (E%) from fat (39 vs 34%, P < 0.001) and saturated fat (15 vs 13%, P = 0.01), a lower E% from protein (16 vs 18%, P = 0.01) and a lower intake of dietary fiber (19 vs 22 g, P = 0.002) compared to Norkost 3 women. Women with celiac disease had a lower intake of bread, fruit and milk, and a higher intake of cereals and cheese compared to Norkost 3 women. The average requirement was not met for several micronutrients, but blood analysis revealed few nutritional deficiencies: two women with insufficient vitamin D status and one with insufficient folic acid status. CONCLUSION: The women with celiac disease had an unbalanced diet with a higher intake of total- and saturated fatty acids and a lower intake of fiber compared to the general population. These findings emphasizes the need for nutritional follow-up of celiac patients and development of nutrient dense gluten-free products.


Asunto(s)
Enfermedad Celíaca , Evaluación Nutricional , Humanos , Femenino , Persona de Mediana Edad , Enfermedad Celíaca/epidemiología , Estudios Transversales , Vitaminas , Valor Nutritivo
16.
Rheumatology (Oxford) ; 62(8): 2918-2929, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36688692

RESUMEN

OBJECTIVES: In SSc, gastrointestinal tract (GIT) involvement is a major concern, with no disease-modifying and limited symptomatic therapies available. Faecal microbiota transplantation (FMT) represents a new therapeutic option for GIT-affliction in SSc, showing clinical promise in a recent controlled pilot trial. Here, we aim to investigate effects of FMT on duodenal biopsies collected from SSc patients by immunohistochemistry and transcriptome profiling. METHODS: We analysed duodenal biopsies obtained pre-intervention (week 0) and post-intervention (weeks 2 and 16) from nine SSc patients receiving an intestinal infusion of FMT (n = 5) or placebo (n = 4). The analysis included immunohistochemistry (IHC) with a selected immune function and fibrosis markers, and whole biopsy transcriptome profiling. RESULTS: In patients receiving FMT, the number of podoplanin- and CD64-expressing cells in the mucosa were lower at week 2 compared with baseline. This decline in podoplanin- (r = 0.94) and CD64-positive (r = 0.89) cells correlated with improved patient-reported lower GIT symptoms. Whole biopsy transcriptome profiling from week 2 showed significant enrichment of pathways critical for cellular and endoplasmic reticulum stress responses, microvillus and secretory vesicles, vascular and sodium-dependent transport, and circadian rhythm. At week 16, we found enrichment of pathways mandatory for binding activity of immunoglobulin receptors, T cell receptor complexes, and chemokine receptors, as well as response to zinc-ions. We found that 25 genes, including Matrix metalloproteinase-1 were upregulated at both week 2 and week 16. CONCLUSION: Combining selective IHC and unbiased gene expression analyses, this exploratory study highlights the potential for disease-relevant organ effects of FMT in SSc patients with GIT involvement. TRIAL REGISTRATION: ClinicalTrials.gov, http://clinicaltrials.gov, NCT03444220.


Asunto(s)
Trasplante de Microbiota Fecal , Esclerodermia Sistémica , Humanos , Trasplante de Microbiota Fecal/efectos adversos , Método Doble Ciego , Intestinos , Mucosa Intestinal , Esclerodermia Sistémica/terapia , Esclerodermia Sistémica/etiología , Resultado del Tratamiento
17.
Sci Adv ; 9(4): eade5800, 2023 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-36696493

RESUMEN

CD4+ T cells specific for cereal gluten proteins are key players in celiac disease (CeD) pathogenesis. While several CeD-relevant gluten T cell epitopes have been identified, epitopes recognized by a substantial proportion of gluten-reactive T cells remain unknown. The identification of such CeD-driving gluten epitopes is important for the food industry and in clinical settings. Here, we have combined the knowledge of a distinct phenotype of gluten-reactive T cells and key features of known gluten epitopes for the discovery of unknown epitopes. We tested 42 wheat gluten-reactive T cell clones, isolated on the basis of their distinct phenotype and with no reactivity to known epitopes, against a panel of synthetic peptides bioinformatically identified from a wheat gluten protein database. We were able to assign reactivity to 10 T cell clones and identified a 9-nucleotide oligomer core region of five previously uncharacterized gliadin/glutenin epitopes. This work represents an advance in the effort to identify CeD-driving gluten epitopes.


Asunto(s)
Enfermedad Celíaca , Humanos , Enfermedad Celíaca/metabolismo , Epítopos de Linfocito T , Glútenes , Gliadina/genética , Gliadina/metabolismo , Péptidos/metabolismo
18.
J Allergy Clin Immunol ; 151(3): 767-777, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36220400

RESUMEN

BACKGROUND: A substantial proportion of common variable immunodeficiency (CVID) patients has duodenal inflammation of largely unknown etiology. However, because of its histologic similarities with celiac disease, gluten sensitivity has been proposed as a potential mechanism. OBJECTIVE: We aimed to elucidate the role of the duodenal microenvironment in the pathogenesis of duodenal inflammation in CVID by investigating the transcriptional, proteomic, and microbial signatures of duodenal biopsy samples in CVID. METHODS: DNA, total RNA, and protein were isolated from snap-frozen pieces of duodenal biopsy samples from CVID (with and without duodenal inflammation), healthy controls, and patients with celiac disease (untreated). RNA sequencing, mass spectrometry-based proteomics, and 16S ribosomal DNA sequencing (bacteria) were then performed. RESULTS: CVID separated from controls in regulation of transcriptional response to lipopolysaccharide and cellular immune responses. These differences were independent of mucosal inflammation. Instead, CVID patients with duodenal inflammation displayed alterations in transcription of genes involved in response to viral infections. Four proteins were differently regulated between CVID patients and healthy controls-DBNL, TRMT11, GCHFR, and IGHA2-independent of duodenal inflammation. Despite similar histology, there were major differences in CVID with duodenal inflammation and celiac disease both at the RNA and protein level. No significant difference was observed in the bacterial gut microbial signature between CVID, celiac, and healthy controls. CONCLUSION: Our findings suggest the existence of altered functions of the duodenal epithelium, particularly in response to lipopolysaccharide and viruses. The latter finding was related to duodenal inflammation, suggesting that viruses, not gluten sensitivity, could be related to duodenal inflammation in CVID.


Asunto(s)
Enfermedad Celíaca , Inmunodeficiencia Variable Común , Virus , Humanos , Enfermedad Celíaca/genética , Lipopolisacáridos , Proteómica , Bacterias , Inflamación , Virus/genética , ARN
19.
Scand J Gastroenterol ; 58(2): 142-147, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36049123

RESUMEN

BACKGROUND AND AIMS: SARS-CoV-2 infection and development of the disease COVID-19 is a serious threat to our society. Effective vaccines have now entered the market, but most patient populations were not included in the registration clinical trials. There is evidence that patients with celiac disease (CeD) have reduced effect of vaccines such as the hepatitis B vaccine. Hence, we investigated the humoral response to SARS-CoV-2 vaccines (Chadox1, Comirnaty and Spikevax) in CeD patients and healthy controls. METHODS: CeD patients from a patient registry at Oslo University Hospital were invited to donate serum samples before and after vaccination. We sent out 1537 invitations and received paired samples from 85 individuals. These were compared with similar samples from 238 healthy controls. Sera were analyzed for antibodies to the Spike protein from SARS-CoV2 and the receptor-binding domain. The results where then converted into binding antibody units (BAU)/ml to compare. RESULTS: Prevaccination samples showed that very few patients had been earlier exposed to Sars-CoV2 and the antibody levels were low. Postvaccination analysis showed overlap of antibody levels between CeD and healthy controls. On average, the CeD patient group had 5555.0 BAU/ml (330.1 SD) while the average in healthy controls was 5419 (184.7 SD). CONCLUSION: The humoral response to SARS-CoV-2 vaccines in CeD patients is similar to that observed in healthy controls.


Asunto(s)
COVID-19 , Enfermedad Celíaca , Humanos , Vacunas contra la COVID-19 , COVID-19/prevención & control , ARN Viral , SARS-CoV-2 , Anticuerpos , Vacunación , Inmunidad , Anticuerpos Antivirales
20.
Foods ; 12(23)2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38231857

RESUMEN

BACKGROUND: Whole-grain bread can be an important source of fibre for people with coeliac disease (CeD) who must adhere to a gluten-free diet and avoid consuming wheat, rye and barley. Gluten-free bread frequently has a lower nutritional quality and different texture relative to gluten-containing counterparts. OBJECTIVE: The aim was to investigate experiences with gluten-free bread amongst people with CeD prior to and during a randomised controlled trial (RCT). DESIGN: We conducted individual interviews with 10 people with CeD participating in a RCT that aimed to investigate the effects of fibre-rich gluten-free products on metabolic regulation in people with CeD compared with benchmark gluten-free products. Five participants were in the control group (benchmark gluten-free bread) and five participants in the intervention group (fibre-rich gluten-free bread). The fibre-rich gluten free bread was formulated and prepared by the project group. The benchmark gluten-free bread was commercially available. The RCT lasted for four weeks. Interviews were conducted digitally between October 2021 and January 2022 and were thematically analysed. RESULTS: Participants in both groups appeared to avoid bread prior to the study, primarily due to the poor taste and chewy consistency of the available bread in food stores and bakeries. Participants preferred the fibre-rich intervention bread as opposed to the available bread in the food market. However, participants had to become accustomed to eating the fibre-rich whole-grain bread during the study, since they avoided eating store-bought bread that they experienced chewy and not filling. CONCLUSIONS: Participants asked for fibre-rich gluten-free bread products that are satiating and have a good texture. Palatable gluten-free bread products might be an important source of fibre for people with CeD.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA