11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      DNA Methylation and Transcription Patterns in Intestinal Epithelial Cells From Pediatric Patients With Inflammatory Bowel Diseases Differentiate Disease Subtypes and Associate With Outcome

      research-article
      1 , 3 , , 1 , , 1 , 1 , 2 , 1 , 4 , 1 , 2 , 1 , 4 , 5 , 5 , 6 , 6 , 2 , 3 , ∗∗ , § , 1 , 2 , 4 , , §
      Gastroenterology
      W.B. Saunders
      Epigenetics, Intestinal Epithelium, Gut Microbiota, Human Intestinal Organoids, AC, ascending colon, AUC, area under the curve, CD, Crohn’s disease, DMP, differentially methylated position, DEG, differentially expressed gene, DMR, differentially methylated region, DNAm, DNA methylation, FDR, false discovery rate, IBD, inflammatory bowel disease, IEC, intestinal epithelial cell, MDS, multidimensional scaling, rDMR, regulatory DMR, ROC, receiver operator characteristic, SC, sigmoid colon, TI, terminal ileum, UC, ulcerative colitis, WGCNA, Weighted Gene Co-expression Network Analysis

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background & Aims

          We analyzed DNA methylation patterns and transcriptomes of primary intestinal epithelial cells (IEC) of children newly diagnosed with inflammatory bowel diseases (IBD) to learn more about pathogenesis.

          Methods

          We obtained mucosal biopsies (N = 236) collected from terminal ileum and ascending and sigmoid colons of children (median age 13 years) newly diagnosed with IBD (43 with Crohn’s disease [CD], 23 with ulcerative colitis [UC]), and 30 children without IBD (controls). Patients were recruited and managed at a hospital in the United Kingdom from 2013 through 2016. We also obtained biopsies collected at later stages from a subset of patients. IECs were purified and analyzed for genome-wide DNA methylation patterns and gene expression profiles. Adjacent microbiota were isolated from biopsies and analyzed by 16S gene sequencing. We generated intestinal organoid cultures from a subset of samples and genome-wide DNA methylation analysis was performed.

          Results

          We found gut segment-specific differences in DNA methylation and transcription profiles of IECs from children with IBD vs controls; some were independent of mucosal inflammation. Changes in gut microbiota between IBD and control groups were not as large and were difficult to assess because of large amounts of intra-individual variation. Only IECs from patients with CD had changes in DNA methylation and transcription patterns in terminal ileum epithelium, compared with controls. Colon epithelium from patients with CD and from patients with ulcerative colitis had distinct changes in DNA methylation and transcription patterns, compared with controls. In IECs from patients with IBD, changes in DNA methylation, compared with controls, were stable over time and were partially retained in ex-vivo organoid cultures. Statistical analyses of epithelial cell profiles allowed us to distinguish children with CD or UC from controls; profiles correlated with disease outcome parameters, such as the requirement for treatment with biologic agents.

          Conclusions

          We identified specific changes in DNA methylation and transcriptome patterns in IECs from pediatric patients with IBD compared with controls. These data indicate that IECs undergo changes during IBD development and could be involved in pathogenesis. Further analyses of primary IECs from patients with IBD could improve our understanding of the large variations in disease progression and outcomes.

          Graphical abstract

          Related collections

          Most cited references20

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Fungal microbiota dysbiosis in IBD

          Objective The bacterial intestinal microbiota plays major roles in human physiology and IBDs. Although some data suggest a role of the fungal microbiota in IBD pathogenesis, the available data are scarce. The aim of our study was to characterise the faecal fungal microbiota in patients with IBD. Design Bacterial and fungal composition of the faecal microbiota of 235 patients with IBD and 38 healthy subjects (HS) was determined using 16S and ITS2 sequencing, respectively. The obtained sequences were analysed using the Qiime pipeline to assess composition and diversity. Bacterial and fungal taxa associated with clinical parameters were identified using multivariate association with linear models. Correlation between bacterial and fungal microbiota was investigated using Spearman's test and distance correlation. Results We observed that fungal microbiota is skewed in IBD, with an increased Basidiomycota/Ascomycota ratio, a decreased proportion of Saccharomyces cerevisiae and an increased proportion of Candida albicans compared with HS. We also identified disease-specific alterations in diversity, indicating that a Crohn's disease-specific gut environment may favour fungi at the expense of bacteria. The concomitant analysis of bacterial and fungal microbiota showed a dense and homogenous correlation network in HS but a dramatically unbalanced network in IBD, suggesting the existence of disease-specific inter-kingdom alterations. Conclusions Besides bacterial dysbiosis, our study identifies a distinct fungal microbiota dysbiosis in IBD characterised by alterations in biodiversity and composition. Moreover, we unravel here disease-specific inter-kingdom network alterations in IBD, suggesting that, beyond bacteria, fungi might also play a role in IBD pathogenesis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease

            Genetic association studies have identified 215 risk loci for inflammatory bowel disease 1–8, which have revealed fundamental aspects of its molecular biology. We performed a genome-wide association study of 25,305 individuals, and meta-analyzed with published summary statistics, yielding a total sample size of 59,957 subjects. We identified 25 new loci, three of which contain integrin genes that encode proteins in pathways identified as important therapeutic targets in inflammatory bowel disease. The associated variants are correlated with expression changes in response to immune stimulus at two of these genes (ITGA4, ITGB8) and at previously implicated loci (ITGAL, ICAM1). In all four cases, the expression increasing allele also increases disease risk. We also identified likely causal missense variants in the primary immune deficiency gene PLCG2 and a negative regulator of inflammation, SLAMF8. Our results demonstrate that new common variant associations continue to identify genes relevant to therapeutic target identification and prioritization.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Inflammatory bowel disease: clinical aspects and established and evolving therapies.

              Crohn's disease and ulcerative colitis are two idiopathic inflammatory bowel disorders. In this paper we discuss the current diagnostic approach, their pathology, natural course, and common complications, the assessment of disease activity, extraintestinal manifestations, and medical and surgical management, and provide diagnostic and therapeutic algorithms. We critically review the evidence for established (5-aminosalicylic acid compounds, corticosteroids, immunomodulators, calcineurin inhibitors) and emerging novel therapies--including biological therapies--directed at cytokines (eg, infliximab, adalimumab, certolizumab pegol) and receptors (eg, visilizumab, abatacept) involved in T-cell activation, selective adhesion molecule blockers (eg, natalizumab, MLN-02, alicaforsen), anti-inflammatory cytokines (eg, interleukin 10), modulation of the intestinal flora (eg, antibiotics, prebiotics, probiotics), leucocyte apheresis and many more monoclonal antibodies, small molecules, recombinant growth factors, and MAP kinase inhibitors targeting various inflammatory cells and pathways. Finally, we summarise the practical aspects of standard therapies including dosing, precautions, and side-effects.
                Bookmark

                Author and article information

                Contributors
                Journal
                Gastroenterology
                Gastroenterology
                Gastroenterology
                W.B. Saunders
                0016-5085
                1528-0012
                1 February 2018
                February 2018
                : 154
                : 3
                : 585-598
                Affiliations
                [1 ]University Department of Paediatrics, University of Cambridge, UK
                [2 ]Department of Paediatric Gastroenterology, University of Cambridge and Addenbrooke’s Hospital, Cambridge, UK
                [3 ]European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
                [4 ]Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
                [5 ]Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
                [6 ]Institute of Clinical Molecular Biology (IKMB), Kiel University, Kiel, Germany
                Author notes
                [] Reprint requests Address requests for reprints to: Matthias Zilbauer, MD, PhD, MRCPCH, University Department of Paediatrics, Level 8 Addenbrooke's Hospital, Box 116, Hills Road, Cambridge, CB2 0QQ, UK. fax: +44 (0) 1223 336996. mz304@ 123456medschl.cam.ac.uk
                [∗∗ ]Oliver Stegle, PhD, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK. fax: + 44 (0) 1223 494 468. stegle@ 123456ebi.ac.uk
                [∗]

                Authors share co-first authorship.

                [§]

                Authors share co-senior authorship.

                Article
                S0016-5085(17)36241-8
                10.1053/j.gastro.2017.10.007
                6381389
                29031501
                90a914b0-09ed-4ab4-b1ac-ffe3ef3267fd
                © 2018 The AGA Institute All rights reserved.

                This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

                History
                : 31 January 2017
                : 2 October 2017
                Categories
                Article

                Gastroenterology & Hepatology
                epigenetics,intestinal epithelium,gut microbiota,human intestinal organoids,ac, ascending colon,auc, area under the curve,cd, crohn’s disease,dmp, differentially methylated position,deg, differentially expressed gene,dmr, differentially methylated region,dnam, dna methylation,fdr, false discovery rate,ibd, inflammatory bowel disease,iec, intestinal epithelial cell,mds, multidimensional scaling,rdmr, regulatory dmr,roc, receiver operator characteristic,sc, sigmoid colon,ti, terminal ileum,uc, ulcerative colitis,wgcna, weighted gene co-expression network analysis

                Comments

                Comment on this article