Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Article in English | MEDLINE | ID: mdl-38366868

ABSTRACT

BACKGROUND: The pathobiology of the non-destructive inflammatory bowel disease (IBD) lymphocytic colitis (LC) is poorly understood. We aimed to define an LC-specific mucosal transcriptome to gain insight into LC pathology, identify unique genomic signatures, and uncover potentially druggable disease pathways. METHODS: We performed bulk RNA-sequencing of LC and collagenous colitis (CC) colonic mucosa from patients with active disease, and healthy controls (n = 4-10 per cohort). Differential gene expression was analyzed by gene-set enrichment and deconvolution analyses to identify pathologically relevant pathways and cells, respectively, altered in LC. Key findings were validated using reverse transcription quantitative PCR and/or immunohistochemistry. Finally, we compared our data with a previous cohort of ulcerative colitis and Crohn's disease patients (n = 4 per group) to distinguish non-destructive from classic IBD. RESULTS: LC can be subdivided into channelopathic LC, which is governed by organic acid and ion transport dysregulation, and inflammatory LC, which is driven by microbial immune responses. Inflammatory LC displays an innate and adaptive immunity that is limited compared to CC and classic IBD. Conversely, we noted a distinct induction of regulatory non-coding RNA species in inflammatory LC samples. Moreover, compared with CC, water channel and cell adhesion molecule gene expression decreased in channelopathic LC, whereas it was accentuated in inflammatory LC and associated with reduced intestinal epithelial cell proliferation. CONCLUSIONS: We conclude that LC can be subdivided into channelopathic LC and inflammatory LC that could be pathomechanistically distinct subtypes despite their shared clinical presentation. Inflammatory LC exhibits a dampened immune response compared to CC and classic IBDs. Our results point to regulatory micro-RNAs as a potential disease-specific feature that may be amenable to therapeutic intervention.

2.
J Crohns Colitis ; 18(3): 349-359, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37768647

ABSTRACT

BACKGROUND AND AIMS: Microscopic colitis [MC] is currently regarded as an inflammatory bowel disease that manifests as two subtypes: collagenous colitis [CC] and lymphocytic colitis [LC]. Whether these represent a clinical continuum or distinct entities is, however, an open question. Genetic investigations may contribute important insight into their respective pathophysiologies. METHODS: We conducted a genome-wide association study [GWAS] meta-analysis in 1498 CC, 373 LC patients, and 13 487 controls from Europe and the USA, combined with publicly available MC GWAS data from UK Biobank and FinnGen [2599 MC cases and 552 343 controls in total]. Human leukocyte antigen [HLA] alleles and polymorphic residues were imputed and tested for association, including conditional analyses for the identification of key causative variants and residues. Genetic correlations with other traits and diagnoses were also studied. RESULTS: We detected strong HLA association with CC, and conditional analyses highlighted the DRB1*03:01 allele and its residues Y26, N77, and R74 as key to this association (best p = 1.4 × 10-23, odds ratio [OR] = 1.96). Nominally significant genetic correlations were detected between CC and pneumonia [rg = 0.77; p = 0.048] and oesophageal diseases [rg = 0.45, p = 0.023]. An additional locus was identified in MC GWAS analyses near the CLEC16A and RMI2 genes on chromosome 16 [rs35099084, p = 2.0 × 10-8, OR = 1.31]. No significant association was detected for LC. CONCLUSION: Our results suggest CC and LC have distinct pathophysiological underpinnings, characterised by an HLA predisposing role only in CC. This challenges existing classifications, eventually calling for a re-evaluation of the utility of MC umbrella definitions.


Subject(s)
Colitis, Collagenous , Colitis, Lymphocytic , Colitis, Microscopic , Humans , Genome-Wide Association Study , HLA Antigens/genetics , Histocompatibility Antigens Class II , Colitis, Microscopic/genetics , Colitis, Lymphocytic/genetics
3.
J Gastroenterol ; 56(10): 914-927, 2021 10.
Article in English | MEDLINE | ID: mdl-34414506

ABSTRACT

BACKGROUND: Collagenous colitis (CC) is an inflammatory bowel disease where chronic diarrhoea is the main symptom. Diagnostic markers distinguishing between CC and other causes of chronic diarrhoea remain elusive. This study explores neutrophil gelatinase-associated lipocalin (NGAL) and its mRNA lipocalin2 (LCN2) as histological and faecal disease markers in CC. METHODS: NGAL/LCN2 were studied in colonic biopsies from CC patients before and during budesonide treatment using RNA sequencing (n = 9/group), in situ hybridization (ISH) (n = 13-22/group) and immunohistochemistry (IHC) (n = 14-25/group). Faecal samples from CC (n = 3-28/group), irritable bowel syndrome diarrhoea (IBS-D) (n = 14) and healthy controls (HC) (n = 15) were assayed for NGAL and calprotectin. RESULTS: NGAL/LCN2 protein and mRNA expression were upregulated in active CC vs HC, and vs paired samples of treated CC in clinical remission. IHC and ISH localized increased NGAL/LCN2 mainly to epithelium of active CC, compared to almost absence in HC and treated CC. In contrast, calprotectin was solely expressed in immune cells. Despite great individual differences, faecal NGAL was significantly increased in active CC compared to HC, IBS-D and treated CC and had high test sensitivity. Faecal calprotectin levels were variably increased in active CC, but the values remained below usual clinical cut-offs. CONCLUSION: NGAL/LCN2 is upregulated in the epithelium of active CC and reduced during budesonide-induced clinical remission to the level of HC and IBD-S. This was reflected in NGAL faecal concentrations. We propose NGAL as an IHC marker for disease activity in CC and a potential faecal biomarker discriminating CC from HC and IBS-D.


Subject(s)
Biomarkers/analysis , Colitis, Collagenous/diagnosis , Lipocalin-2/analysis , Adult , China/epidemiology , Colitis, Collagenous/blood , Colitis, Collagenous/epidemiology , Enzyme-Linked Immunosorbent Assay/methods , Feces/enzymology , Feces/microbiology , Female , Humans , Male , Middle Aged
4.
Cell Mol Gastroenterol Hepatol ; 12(2): 665-687, 2021.
Article in English | MEDLINE | ID: mdl-33930606

ABSTRACT

BACKGROUND AND AIMS: The pathophysiology of the inflammatory bowel disease collagenous colitis (CC) is poorly described. Our aim was to use RNA sequencing of mucosal samples from patients with active CC, CC in remission, refractory CC, ulcerative colitis (UC), and control subjects to gain insight into CC pathophysiology, identify genetic signatures linked to CC, and uncover potentially druggable disease pathways. METHODS: We performed whole transcriptome sequencing of CC samples from patients before and during treatment with the corticosteroid drug budesonide, CC steroid-refractory patients, UC patients, and healthy control subjects (n = 9-13). Bulk mucosa and laser-captured microdissected intestinal epithelial cell (IEC) gene expression were analyzed by gene set enrichment and gene set variation analyses to identify significant pathways and cells, respectively, altered in CC. Leading genes and cells were validated using reverse-transcription quantitative polymerase chain reaction or immunohistochemistry. RESULTS: We identified an activation of the adaptive immune response to bacteria and viruses in active CC that could be mediated by dendritic cells. Moreover, IECs display hyperproliferation and increased antigen presentation in active CC. Further analysis revealed that genes related to the immune response (DUOX2, PLA2G2A, CXCL9), DNA transcription (CTR9), protein processing (JOSD1, URI1), and ion transport (SLC9A3) remained dysregulated even after budesonide-induced remission. Budesonide-refractory CC patients fail to restore normal gene expression, and displayed a transcriptomic profile close to UC. CONCLUSIONS: Our study confirmed the implication of innate and adaptive immune responses in CC, governed by IECs and dendritic cells, respectively, and identified ongoing epithelial damage. Refractory CC could share pathomechanisms with UC.


Subject(s)
Colitis, Collagenous/genetics , Gene Expression Profiling , Inflammatory Bowel Diseases/genetics , Adolescent , Adult , Aged , Budesonide/pharmacology , Cell Proliferation/drug effects , Colitis, Collagenous/immunology , Colitis, Collagenous/pathology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Collagen/genetics , Collagen/metabolism , Enterocytes/metabolism , Enterocytes/pathology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Gene Expression Regulation , Humans , Immunity/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Male , Middle Aged , Stromal Cells/metabolism , Transcription, Genetic/drug effects , Young Adult
5.
Int Rev Cell Mol Biol ; 358: 133-164, 2021.
Article in English | MEDLINE | ID: mdl-33707053

ABSTRACT

The intestinal epithelium limits host-luminal interactions and maintains gut homeostasis. Breakdown of the epithelial barrier and villous atrophy are hallmarks of coeliac disease. Besides the well characterized immune-mediated epithelial damage induced in coeliac mucosa, constitutional changes and early gluten direct effects disturb intestinal epithelial cells. The subsequent modifications in key epithelial signaling pathways leads to outnumbered immature epithelial cells that, in turn, facilitate epithelial dysfunction, promote crypt hyperplasia, and increase intestinal permeability. Consequently, underlying immune cells have a greater access to gluten, which boosts the proinflammatory immune response against gluten and positively feedback the epithelial damage loop. Gluten-free diet is an indispensable treatment for coeliac disease patients, but additional therapies are under development, including those that reinforce intestinal epithelial healing. In this chapter, we provide an overview of intestinal epithelial cell disturbances that develop during gluten intake in coeliac disease mucosa.


Subject(s)
Celiac Disease/pathology , Celiac Disease/physiopathology , Epithelial Cells/pathology , Animals , Celiac Disease/genetics , Celiac Disease/immunology , Diet , Epigenesis, Genetic , Epithelial Cells/immunology , Gastrointestinal Microbiome , Glutens/adverse effects , Humans
6.
Inflamm Bowel Dis ; 27(9): 1482-1490, 2021 08 19.
Article in English | MEDLINE | ID: mdl-33319252

ABSTRACT

BACKGROUND AND AIM: Increased frequencies of T regulatory (Treg) cells, key players in immune regulation, have been reported in inflammatory bowel diseases, including collagenous colitis (CC). However, traditional Treg identification techniques might have misinterpreted the frequencies of Treg cells in CC. Thus, we investigated the presence of genuine Treg cells in CC. METHODS: Treg cells were analyzed in mucosal and peripheral blood samples of CC patients before and during treatment with the corticosteroid drug budesonide and in healthy controls. Samples were analyzed by flow cytometry by classifying CD3+CD4+ cells as activated FoxP3highCD45RA- Treg cells, resting FoxP3dimCD45RA+ Treg cells, and nonsuppressive FoxP3dimCD45RA- T helper cells. Traditional gating strategies that classified Treg cells as CD25highCD127low, FoxP3+CD127low, and CD4+CD25+FoxP3+ were also used to facilitate comparison with previous studies. RESULTS: Activated and resting Treg cell frequencies did not change in active CC mucosa or peripheral blood and were not affected by budesonide treatment. Instead, nonsuppressive FoxP3dimCD45RA- T helper cells were increased in active CC mucosa, and budesonide helped restore them to normal levels. In contrast, traditional Treg cell gating strategies resulted in increased Treg cell frequencies in active CC mucosa. No alterations were found in peripheral blood samples, independently of patient treatment or gating techniques. CONCLUSION: Previously reported increase of Treg cells is a result of incomplete Treg phenotyping, which included nonsuppressive FoxP3dimCD45RA- T helper cells. Because budesonide did not affect Treg percentage, its therapeutic effect in CC might involve alternative mechanisms.


Subject(s)
Colitis, Collagenous , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Budesonide/therapeutic use , Case-Control Studies , Colitis, Collagenous/drug therapy , Colitis, Collagenous/immunology , Forkhead Transcription Factors/metabolism , Humans , Mucous Membrane
7.
Rev. esp. enferm. dig ; 112(6): 434-439, jun. 2020. tab, graf
Article in Spanish | IBECS | ID: ibc-199790

ABSTRACT

INTRODUCCIÓN: la enfermedad celiaca (EC) presenta un determinado patrón de infiltrado linfocitario en la mucosa duodenal. Gracias a la citometría de flujo como herramienta complementaria al diagnóstico de la EC, se pueden cuantificar y caracterizar los linfocitos intraepiteliales (LIE) a través de lo que comúnmente denominamos linfograma. Con este estudio, pretendemos describir nuestra experiencia con la técnica en el diagnóstico del paciente celiaco adulto. MÉTODOS: se han analizado retrospectivamente los linfogramas realizados en nuestro centro entre 2009 y 2017, que fueron en total 157. Catorce de ellos tenían un diagnóstico previo de EC y seguían una dieta sin gluten, 21 tuvieron un diagnóstico nuevo de EC y el resto fueron considerados no celiacos. Se ha estudiado la asociación de los valores del linfograma (LIE totales, linfocitos CD3- y linfocitos TcRγδ) con el diagnóstico de EC, el cumplimiento de la dieta sin gluten (DSG), el tiempo desde el diagnóstico y el título de inmunoglobulina A antitransglutaminasa tisular. RESULTADOS: el valor de área bajo la curva ROC de los linfocitos TcRγδ para el diagnóstico de EC varía entre 0,86 y 0,86. El porcentaje de linfocitos TcRγδ en pacientes celiacos en DSG es menor 12 (8,5) vs. 20,5 (8,7), p = 0,0153, aunque permanece elevado frente a los no celiacos 12 (8,5) vs. 6,7 (6), p = 0,135. El tiempo desde el diagnóstico y el título de IgA antitransglutaminasa tisular (anti-Tgt) se correlacionan con los valores del linfograma en el paciente celiaco. La infección por Helicobacter pylori y el tratamiento con antagonistas de receptores de angiotensina 2 (ARA2) se asocia a diferencias en el linfograma. CONCLUSIONES: el linfograma duodenal es una herramienta complementaria fiable en el diagnóstico del celiaco adulto. Sin embargo, el cumplimiento y la duración de la DSG, así como otros factores, pueden condicionar su capacidad diagnóstica


No disponible


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Aged , Celiac Disease/diagnosis , Flow Cytometry , Lymphocytes/pathology , Helicobacter pylori , Helicobacter Infections , Retrospective Studies , ROC Curve
8.
Rev Esp Enferm Dig ; 112(6): 434-439, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32022573

ABSTRACT

INTRODUCTION: celiac disease (CD) patients have a specific pattern of lymphocytic infiltrate in the duodenal mucosa. Flow cytometry is a complementary tool for the diagnosis of CD, which allows the quantification and characterization of intraepithelial lymphocytes (IELs) by what is commonly called a lymphogram. Here we describe our experience with this technique in the diagnosis of CD in adult patients. METHODS: lymphograms from 157 patients performed in our center between 2009 and 2017 were retrospectively analyzed. Fourteen patients had a previous diagnosis of CD and followed a gluten-free diet (GFD), 21 had a new diagnosis of CD and the remaining were considered as non-celiac. The association of the lymphogram results (total IELs, CD3- lymphocytes and TcRγδ lymphocytes) with the CD diagnosis, compliance with the GFD, time since diagnosis and IgA anti-TG2 titer were determined. RESULTS: the area under the ROC curve of TcRγδ lymphocytes for CD patients varied between 0.86 and 0.86. The percentage of TcRγδ lymphocytes in GFD-treated patients was lower; 12 (8.5) vs 20.5 (8.7), p = 0.0153. However, it remained high compared to non-CD; 12 (8.5) vs 6.7 (6), p = 0.135. The time since diagnosis and IgA anti-TG2 titer correlated with the lymphogram results. Helicobacter pylori infection and treatment with angiotensin receptor antagonist 2 (ARA2) were associated with differences in the lymphogram results in patients without CD. CONCLUSIONS: the duodenal lymphogram is a reliable complementary tool in adults for the diagnosis of CD. However, compliance and duration of the GFD and other factors may condition its diagnostic capacity.


Subject(s)
Celiac Disease , Helicobacter Infections , Helicobacter pylori , Adult , Celiac Disease/diagnosis , Diet, Gluten-Free , Duodenum/diagnostic imaging , Humans , Intestinal Mucosa , Retrospective Studies
9.
J Crohns Colitis ; 14(7): 962-973, 2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32016376

ABSTRACT

BACKGROUND AND AIMS: Diarrhoea is a common, debilitating symptom of gastrointestinal disorders. Pathomechanisms probably involve defects in trans-epithelial water transport, but the role of aquaporin [AQP] family water channels in diarrhoea-predominant diseases is unknown. We investigated the involvement of AQPs in the pathobiology of collagenous colitis [CC], which features chronic, watery diarrhoea despite overtly normal intestinal epithelial cells [IECs]. METHODS: We assessed the expression of all AQP family members in mucosal samples of CC patients before and during treatment with the corticosteroid drug budesonide, steroid-refractory CC patients and healthy controls. Samples were analysed by genome-wide mRNA sequencing [RNA-seq] and quantitative real-time PCR [qPCR]. In some patients, we performed tissue microdissection followed by RNA-seq to explore the IEC-specific CC transcriptome. We determined changes in the protein levels of the lead candidates in IEC by confocal microscopy. Finally, we investigated the regulation of AQP expression by corticosteroids in model cell lines. RESULTS: Using qPCR and RNA-seq, we identified loss of AQP8 expression as a hallmark of active CC, which was reverted by budesonide treatment in steroid-responsive but not refractory patients. Consistently, decreased AQP8 mRNA and protein levels were observed in IECs of patients with active CC, and steroid drugs increased AQP8 expression in model IECs. Moreover, low APQ8 expression was strongly associated with higher stool frequency in CC patients. CONCLUSION: Down-regulation of epithelial AQP8 may impair water resorption in active CC, resulting in watery diarrhoea. Our results suggest that AQP8 is a potential drug target for the treatment of diarrhoeal disorders.


Subject(s)
Aquaporins/genetics , Aquaporins/metabolism , Colitis, Collagenous/genetics , Colitis, Collagenous/metabolism , Diarrhea/genetics , Diarrhea/metabolism , Adult , Aged , Aged, 80 and over , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Aquaporin 1/genetics , Budesonide/pharmacology , Budesonide/therapeutic use , Caco-2 Cells , Colitis, Collagenous/complications , Colitis, Collagenous/drug therapy , Dexamethasone/pharmacology , Diarrhea/etiology , Down-Regulation/drug effects , Epithelial Cells/metabolism , Female , HT29 Cells , Homeostasis , Humans , Intestinal Mucosa/metabolism , Male , Middle Aged , RNA, Messenger/metabolism , Water/metabolism
10.
Mol Nutr Food Res ; 64(6): e1900989, 2020 03.
Article in English | MEDLINE | ID: mdl-31970917

ABSTRACT

SCOPE: Circulating dendritic cell (DC) and monocyte subsets contribute to the pool of intestinal DC and macrophages in celiac disease (CeD), an autoimmune gut disorder triggered by dietary gluten. Here, this study aims to characterize these circulating subsets in CeD and assess the effect of different gliadin-derived peptides on conventional DC (cDC). METHODS AND RESULTS: Flow cytometry profiling of peripheral blood mononuclear cells reveals a slight decrease in the proportion of plasmacytoid and type 1 cDC in gluten-free diet (GFD)-treated CeD patients. In comparison to healthy donors, DC and monocyte subsets from active and GFD-treated CeD patients display an increased gut-homing profile. Type 2 cDC (cDC2) are sorted and stimulated with the gliadin-derived peptides 8-mer, 19-mer, and 33-mer. All peptides induce cDC2 maturation, although the profile is different. While peptide 8-mer induces a Th1/Th17 pro-inflammatory cytokine profile in active CeD patients, cDC2 primed with peptide 33-mer displays a higher capacity to promote gut-homing CCR9+ expression onto autologous T-cells. CONCLUSION: Distinct gliadin-derived peptides elicit different effects on cDC2 phenotype and function. This effect is compatible with a model where diverse gliadin peptides may cooperate to promote full cDC2 activation and the subsequent T-cell response in genetically predisposed individuals.


Subject(s)
Celiac Disease/pathology , Dendritic Cells/physiology , Gliadin/pharmacology , Adult , Antigens, CD/metabolism , Case-Control Studies , Celiac Disease/diet therapy , Celiac Disease/immunology , Cytokines/metabolism , Dendritic Cells/drug effects , Diet, Gluten-Free , Female , Gastrointestinal Tract/metabolism , Gliadin/chemistry , Gliadin/metabolism , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Peptide Fragments/pharmacology , Receptors, CCR/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
12.
Rev Esp Enferm Dig ; 112(1): 4-6, 2020 01.
Article in English | MEDLINE | ID: mdl-31830796

ABSTRACT

Celiac disease (CeD) and inflammatory bowel disease (IBD) are chronic gastrointestinal disorders of inflammatory origin that develop in response to environmental triggers in genetically predisposed individuals. CeD localizes in the duodenal mucosa, where intolerance develops to dietary gluten from wheat, barley, rye, and some varieties of oats. IBD, in turn, is subdivided primarily into Crohn's disease (CD) and colitis, with ulcerative colitis (UC) being the most thoroughly investigated form.


Subject(s)
Celiac Disease , Colitis, Ulcerative , Crohn Disease , Inflammatory Bowel Diseases , Glutens , Humans
14.
Cytokine ; 99: 73-79, 2017 11.
Article in English | MEDLINE | ID: mdl-28738233

ABSTRACT

Celiac disease is a chronic autoimmune condition triggered by dietary gluten in genetically predisposed individuals and the treatment is a strict gluten-free diet. The major predisposing genes are HLA-DQA1 and HLA-DQB1, but these are not sufficient for disease development. One of the candidate genes worth studying is interleukin (IL)-15 gene, together with its specific receptor, IL-15Rα, as they participate in promoting lymphocyte signaling and survival, and the establishment of appropriate conditions for villous atrophy, then acting as key players in the immunopathogenesis of CD. Here we analyze IL-15 and IL-15Rα genes in samples from the Spanish Consortium for Genetics of Celiac Disease (CEGEC) collection, identifying two regulatory single-nucleotide polymorphisms (SNP) that might be associated with celiac disease: rs4956400 (p-value 0.0112, OR 1.21, 95% CI 1.04-1.40) and rs11100722 (p-value 0.0087, OR 1.24, 95% CI 1.06-1.45), both located upstream the IL15 gene. When the expression of both genes was assessed, these two SNPs were found to be correlated with IL-15 higher protein expression. Besides, rs8177655 from IL15RA was also associated to mRNA IL-15 expression in CD patients. Finally, three SNPs from IL15RA intronic regions, rs2296141, rs3136614 and rs3181148, and another from its 3'UTR region, rs2229135, could be related to the age of diagnosis of celiac disease patients.


Subject(s)
Celiac Disease/genetics , Genetic Predisposition to Disease , Interleukin-15/genetics , Receptors, Interleukin-15/genetics , Adolescent , Adult , Aged , Alleles , Child , Child, Preschool , Female , Gene Expression Regulation , Genotyping Techniques , Humans , Infant , Linkage Disequilibrium/genetics , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Statistics, Nonparametric , Young Adult
15.
Innate Immun ; 23(1): 44-53, 2017 01.
Article in English | MEDLINE | ID: mdl-27794069

ABSTRACT

IL-15 is a pleiotropic cytokine related to IL-2 which acts at a broader level than its counterpart. It is presented through its specific high-affinity receptor, IL-15Rα. Both cytokine and receptor are tightly regulated at multiple levels and are widely distributed. Thus, deregulation of their expression leads to an inflammatory immune response. Variants of splicing of IL-15Rα have been described in immune and barrier cells; however, their presence has not been focused on intestinal epithelial cells. In this study, we describe five new alternative variants of splicing of IL-15Rα in Caco-2 cells. Four of them were expressed into proteins inside Caco-2 cells, but these were unable to bind IL-15 or to follow the secretory pathway. However, the expression of mRNA itself might be relevant to diseases such as celiac disease, inflammatory bowel disease or colorectal cancer.


Subject(s)
Celiac Disease/immunology , Colorectal Neoplasms/immunology , Inflammatory Bowel Diseases/immunology , Interleukin-15 Receptor alpha Subunit/metabolism , Intestinal Mucosa/immunology , Protein Isoforms/metabolism , Caco-2 Cells , DNA Methylation , Epigenesis, Genetic , Exocytosis/genetics , Humans , Interleukin-15/metabolism , Interleukin-15 Receptor alpha Subunit/genetics , Protein Binding/genetics , Protein Isoforms/genetics , Protein Splicing
16.
Curr Gastroenterol Rep ; 18(7): 36, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27216895

ABSTRACT

Celiac disease is the most common oral intolerance in Western countries. It results from an immune response towards gluten proteins from certain cereals in genetically predisposed individuals (HLA-DQ2 and/or HLA-DQ8). Its pathogenesis involves the adaptive (HLA molecules, transglutaminase 2, dendritic cells, and CD4(+) T-cells) and the innate immunity with an IL-15-mediated response elicited in the intraepithelial compartment. At present, the only treatment is a permanent strict gluten-free diet (GFD). Multidisciplinary studies have provided a deeper insight of the genetic and immunological factors and their interaction with the microbiota in the pathogenesis of the disease. Similarly, a better understanding of the composition of the toxic gluten peptides has improved the ways to detect them in food and drinks and how to monitor GFD compliance via non-invasive approaches. This review, therefore, addresses the major findings obtained in the last few years including the re-discovery of non-celiac gluten sensitivity.


Subject(s)
Celiac Disease/genetics , Celiac Disease/immunology , Wheat Hypersensitivity/genetics , Wheat Hypersensitivity/immunology , Celiac Disease/microbiology , Diet, Gluten-Free , Epigenesis, Genetic , Gastrointestinal Microbiome , Glutens/immunology , Humans , Immunogenetic Phenomena , Wheat Hypersensitivity/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...