TY - JOUR KW - CROHN'S DISEASE KW - inflammatory bowel disease KW - INTESTINAL EPITHELIUM KW - METHYLATION AU - Thomas W. Dennison AU - Rachel D. Edgar AU - Felicity Payne AU - Komal M. Nayak AU - Alexander D. B. Ross AU - Aurelie Cenier AU - Claire Glemas AU - Federica Giachero AU - April R. Foster AU - Rebecca Harris AU - Judith Kraiczy AU - Camilla Salvestrini AU - Georgia Stavrou AU - Franco Torrente AU - Kimberley Brook AU - Claire Trayers AU - Rasa Elmentaite AU - Gehad Youssef AU - Bálint Tél AU - Douglas James Winton AU - Nefeli Skoufou-Papoutsaki AU - Sam Adler AU - Philip Bufler AU - Aline Azabdaftari AU - Andreas Jenke AU - Natasha G AU - Natasha Thomas AU - Erasmo Miele AU - Abdulrahman Al-Mohammad AU - Greta Guarda AU - Subra Kugathasan AU - Suresh Venkateswaran AU - Menna R. Clatworthy AU - Tomas Castro-Dopico AU - Ondrej Suchanek AU - Caterina Strisciuglio AU - Marco Gasparetto AU - Seokjun Lee AU - Xingze Xu AU - Erica Bello AU - Namshik Han AU - Daniel R. Zerbino AU - Sarah A. Teichmann AU - Josquin Nys AU - Robert Heuschkel AU - Francesca Perrone AU - Matthias Zilbauer AB - Objective Epigenetic mechanisms, including DNA methylation (DNAm), have been proposed to play a key role in Crohn’s disease (CD) pathogenesis. However, the specific cell types and pathways affected as well as their potential impact on disease phenotype and outcome remain unknown. We set out to investigate the role of intestinal epithelial DNAm in CD pathogenesis. Design We generated 312 intestinal epithelial organoids (IEOs) from mucosal biopsies of 168 patients with CD (n=72), UC (n=23) and healthy controls (n=73). We performed genome-wide molecular profiling including DNAm, bulk as well as single-cell RNA sequencing. Organoids were subjected to gene editing and the functional consequences of DNAm changes evaluated using an organoid-lymphocyte coculture and a nucleotide-binding oligomerisation domain, leucine-rich repeat and CARD domain containing 5 (NLRC5) dextran sulphate sodium (DSS) colitis knock-out mouse model. Results We identified highly stable, CD-associated loss of DNAm at major histocompatibility complex (MHC) class 1 loci including NLRC5 and cognate gene upregulation. Single-cell RNA sequencing of primary mucosal tissue and IEOs confirmed the role of NLRC5 as transcriptional transactivator in the intestinal epithelium. Increased mucosal MHC-I and NLRC5 expression in adult and paediatric patients with CD was validated in additional cohorts and the functional role of MHC-I highlighted by demonstrating a relative protection from DSS-mediated mucosal inflammation in NLRC5-deficient mice. MHC-I DNAm in IEOs showed a significant correlation with CD disease phenotype and outcomes. Application of machine learning approaches enabled the development of a disease prognostic epigenetic molecular signature. Conclusions Our study has identified epigenetically regulated intestinal epithelial MHC-I as a novel mechanism in CD pathogenesis. BT - Gut DA - 2024/09/01 DO - 10.1136/gutjnl-2024-332043 LA - en N2 - Objective Epigenetic mechanisms, including DNA methylation (DNAm), have been proposed to play a key role in Crohn’s disease (CD) pathogenesis. However, the specific cell types and pathways affected as well as their potential impact on disease phenotype and outcome remain unknown. We set out to investigate the role of intestinal epithelial DNAm in CD pathogenesis. Design We generated 312 intestinal epithelial organoids (IEOs) from mucosal biopsies of 168 patients with CD (n=72), UC (n=23) and healthy controls (n=73). We performed genome-wide molecular profiling including DNAm, bulk as well as single-cell RNA sequencing. Organoids were subjected to gene editing and the functional consequences of DNAm changes evaluated using an organoid-lymphocyte coculture and a nucleotide-binding oligomerisation domain, leucine-rich repeat and CARD domain containing 5 (NLRC5) dextran sulphate sodium (DSS) colitis knock-out mouse model. Results We identified highly stable, CD-associated loss of DNAm at major histocompatibility complex (MHC) class 1 loci including NLRC5 and cognate gene upregulation. Single-cell RNA sequencing of primary mucosal tissue and IEOs confirmed the role of NLRC5 as transcriptional transactivator in the intestinal epithelium. Increased mucosal MHC-I and NLRC5 expression in adult and paediatric patients with CD was validated in additional cohorts and the functional role of MHC-I highlighted by demonstrating a relative protection from DSS-mediated mucosal inflammation in NLRC5-deficient mice. MHC-I DNAm in IEOs showed a significant correlation with CD disease phenotype and outcomes. Application of machine learning approaches enabled the development of a disease prognostic epigenetic molecular signature. Conclusions Our study has identified epigenetically regulated intestinal epithelial MHC-I as a novel mechanism in CD pathogenesis. PY - 2024 SP - 1464 EP - 1477 T2 - Gut TI - Patient-derived organoid biobank identifies epigenetic dysregulation of intestinal epithelial MHC-I as a novel mechanism in severe Crohn’s Disease UR - https://gut.bmj.com/content/73/9/1464 VL - 73 Y2 - 2024-08-13 SN - 0017-5749, 1468-3288 ER -