1
|
Chen L, Wang X, Xie N, Zhang Z, Xu X, Xue M, Yang Y, Liu L, Su L, Bjaanæs M, Karlsson A, Planck M, Staaf J, Helland Å, Esteller M, Christiani DC, Chen F, Zhang R. A two-phase epigenome-wide four-way gene-smoking interaction study of overall survival for early-stage non-small cell lung cancer. Mol Oncol 2024. [PMID: 39630602 DOI: 10.1002/1878-0261.13766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/05/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
High-order interactions associated with non-small cell lung cancer (NSCLC) survival may elucidate underlying molecular mechanisms and identify potential therapeutic targets. Our previous work has identified a three-way interaction among pack-year of smoking (the number of packs of cigarettes smoked per day multiplied by the number of years the person has smoked) and two DNA methylation probes (cg05293407TRIM27 and cg00060500KIAA0226). However, whether a four-way interaction exists remains unclear. Therefore, we adopted a two-phase design to identify the four-way gene-smoking interactions by a hill-climbing strategy on the basis of the previously detected three-way interaction. One CpG probe, cg16658473SHISA9, was identified with FDR-q ≤ 0.05 in the discovery phase and P ≤ 0.05 in the validation phase. Meanwhile, the four-way interaction improved the discrimination ability for the prognostic prediction model, as indicated by the area under the receiver operating characteristic curve (AUC) for both 3- and 5-year survival. In summary, we identified a four-way interaction associated with NSCLC survival among pack-year of smoking, cg05293407TRIM27, cg00060500KIAA0226 and g16658473SHISA9, providing novel insights into the complex mechanisms underlying NSCLC progression.
Collapse
Affiliation(s)
- Leyi Chen
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, China
| | - Xiang Wang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, China
| | - Ning Xie
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, China
| | - Zhongwen Zhang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, China
| | - Xiaowen Xu
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, China
| | - Maojie Xue
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, China
- Department of Health Inspection and Quarantine, Center for Global Health, School of Public Health, Nanjing Medical University, China
| | - Yuqing Yang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, China
| | - Liya Liu
- School of Public Health, Health Science Center, Ningbo University, China
| | - Li Su
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Pulmonary and Critical Care Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Maria Bjaanæs
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Norway
| | - Anna Karlsson
- Division of Oncology, Department of Clinical Sciences Lund and CREATE Health Strategic Center for Translational Cancer Research, Lund University, Sweden
| | - Maria Planck
- Division of Oncology, Department of Clinical Sciences Lund and CREATE Health Strategic Center for Translational Cancer Research, Lund University, Sweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund and CREATE Health Strategic Center for Translational Cancer Research, Lund University, Sweden
| | - Åslaug Helland
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Norway
- Institute of Clinical Medicine, University of Oslo, Norway
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
- Centro de Investigacion Biomedica en Red Cancer, Madrid, Spain
- Institucio Catalana de Recerca i Estudis Avançats, Barcelona, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona, Spain
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Pulmonary and Critical Care Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Feng Chen
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, China
| | - Ruyang Zhang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, China
- China International Cooperation Center for Environment and Human Health, Nanjing Medical University, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
- Information Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
2
|
Zheng J, Sun Q, Zhang M, Liu C, Su Q, Zhang L, Xu Z, Lu W, Ching J, Tang W, Cheung CP, Hamilton AL, Wilson O'Brien AL, Wei SC, Bernstein CN, Rubin DT, Chang EB, Morrison M, Kamm MA, Chan FKL, Zhang J, Ng SC. Noninvasive, microbiome-based diagnosis of inflammatory bowel disease. Nat Med 2024; 30:3555-3567. [PMID: 39367251 PMCID: PMC11645270 DOI: 10.1038/s41591-024-03280-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 08/29/2024] [Indexed: 10/06/2024]
Abstract
Despite recent progress in our understanding of the association between the gut microbiome and inflammatory bowel disease (IBD), the role of microbiome biomarkers in IBD diagnosis remains underexplored. Here we developed a microbiome-based diagnostic test for IBD. By utilization of metagenomic data from 5,979 fecal samples with and without IBD from different geographies and ethnicities, we identified microbiota alterations in IBD and selected ten and nine bacterial species for construction of diagnostic models for ulcerative colitis and Crohn's disease, respectively. These diagnostic models achieved areas under the curve >0.90 for distinguishing IBD from controls in the discovery cohort, and maintained satisfactory performance in transethnic validation cohorts from eight populations. We further developed a multiplex droplet digital polymerase chain reaction test targeting selected IBD-associated bacterial species, and models based on this test showed numerically higher performance than fecal calprotectin in discriminating ulcerative colitis and Crohn's disease from controls. Here we discovered universal IBD-associated bacteria and show the potential applicability of a multibacteria biomarker panel as a noninvasive tool for IBD diagnosis.
Collapse
Affiliation(s)
- Jiaying Zheng
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Qianru Sun
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Chengyu Liu
- Microbiota I-Center (MagIC), Hong Kong, China
| | - Qi Su
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Lin Zhang
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhilu Xu
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Wenqi Lu
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Jessica Ching
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Whitney Tang
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Chun Pan Cheung
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Amy L Hamilton
- Department of Gastroenterology, St Vincent's Hospital, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
| | - Amy L Wilson O'Brien
- Department of Gastroenterology, St Vincent's Hospital, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
| | - Shu Chen Wei
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Charles N Bernstein
- Department of Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David T Rubin
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Eugene B Chang
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Mark Morrison
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Michael A Kamm
- Department of Gastroenterology, St Vincent's Hospital, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
| | - Francis K L Chan
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong, China
| | - Jingwan Zhang
- Microbiota I-Center (MagIC), Hong Kong, China.
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China.
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| | - Siew C Ng
- Microbiota I-Center (MagIC), Hong Kong, China.
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China.
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong, China.
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
3
|
Chen J, Zhang H, Fu T, Zhao J, Nowak JK, Kalla R, Wellens J, Yuan S, Noble A, Ventham NT, Dunlop MG, Halfvarson J, Mao R, Theodoratou E, Satsangi J, Li X. Exposure to air pollution increases susceptibility to ulcerative colitis through epigenetic alterations in CXCR2 and MHC class III region. EBioMedicine 2024; 110:105443. [PMID: 39536393 PMCID: PMC11605448 DOI: 10.1016/j.ebiom.2024.105443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND This study aims to confirm the associations of air pollution with ulcerative colitis (UC) and Crohn's disease (CD); to explore interactions with genetics and lifestyle; and to characterize potential epigenetic mechanisms. METHODS We identified over 450,000 individuals from the UK Biobank and investigated the relationship between air pollution and incident inflammatory bowel disease (IBD). Cox regression was utilized to calculate hazard ratios (HRs), while also exploring potential interactions with genetics and lifestyle factors. Additionally, we conducted epigenetic Mendelian randomization (MR) analyses to examine the association between air pollution-related DNA methylation and UC. Finally, our findings were validated through genome-wide DNA methylation analysis of UC, as well as co-localization and gene expression analyses. FINDINGS Higher exposures to NOx (HR = 1.20, 95% CI 1.05-1.38), NO2 (HR = 1.19, 95% CI = 1.03-1.36), PM2.5 (HR = 1.19, 95% CI = 1.05-1.36) and combined air pollution score (HR = 1.26, 95% CI = 1.11-1.45) were associated with incident UC but not CD. Interactions with genetic risk score and lifestyle were observed. In MR analysis, we found five and 22 methylated CpG sites related to PM2.5 and NO2 exposure to be significantly associated with UC. DNA methylation alterations at CXCR2 and sites within the MHC class III region, were validated in genome-wide DNA methylation analysis, co-localization analysis and analysis of colonic tissue. INTERPRETATION We report a potential causal association between air pollution and UC, modified by lifestyle and genetic influences. Biological pathways implicated include epigenetic alterations in key genetic loci, including CXCR2 and susceptible loci within MHC class III region. FUNDING Xue Li was supported by the Natural Science Fund for Distinguished Young Scholars of Zhejiang Province (LR22H260001) and the National Nature Science Foundation of China (No. 82204019). ET was supported by the CRUK Career Development Fellowship (C31250/A22804) and the Research Foundation Flanders (FWO). JW was supported by Belgium by a PhD Fellowship strategic basic research (SB) grant (1S06023N). JKN was supported by the National Science Center, Poland (No. 2020/39/D/NZ5/02720). The IBD Character was supported by the European Union's Seventh Framework Programme [FP7] grant IBD Character (No. 2858546).
Collapse
Affiliation(s)
- Jie Chen
- The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Han Zhang
- The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tian Fu
- The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China; Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University Medical College, Hangzhou, China
| | - Jianhui Zhao
- The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jan Krzysztof Nowak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 60572, Poznan, Poland
| | - Rahul Kalla
- Medical Research Council Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Judith Wellens
- KU Leuven Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Leuven, Belgium; Translational Gastro-Intestinal Unit, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, UK
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Alexandra Noble
- Translational Gastro-Intestinal Unit, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, UK
| | - Nicholas T Ventham
- Medical Research Council Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Malcolm G Dunlop
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Jonas Halfvarson
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Evropi Theodoratou
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK; Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom.
| | - Jack Satsangi
- Translational Gastro-Intestinal Unit, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, UK.
| | - Xue Li
- The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Danielewski M, Walkowiak J, Wielgus K, Nowak JK. Stability of Blood DNA Methylation Across Two Timepoints in Three Cohorts. Biomedicines 2024; 12:2557. [PMID: 39595122 PMCID: PMC11592101 DOI: 10.3390/biomedicines12112557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
Background: DNA methylation mediates the gene-environment interactions, with implications for health and disease. Studies with sampling at more than one timepoint revealed the considerable variability of the blood methylome, but comprehensive resources on genome-wide methylation stability are still lacking. We aimed to identify methylation sites that remain the most stable across two timepoints in human whole blood. Methods: Publicly available blood DNA methylation data from three cohorts were analysed, which included methylation profiles at two timepoints >1 year apart. The cohorts included pre-/post-pubertal children (Illumina 450k array), the elderly (Illumina 450k array), and middle-aged adults with obesity (Illumina EPIC array). Two metrics were used for the stability assessment: the mean absolute difference (MAD) of beta values between two measurements and the intraclass correlation coefficient (ICC). We searched for probes demonstrating high stability (low MAD and high ICC) across the three cohorts. Data from 51 children, 86 elderly adults, and 120 middle-aged participants were re-analysed. Results: The median interquartile range (IQR) of the maximum (from three datasets) MAD was 2.1% (1.5-2.9%), and the median of the minimum ICC agreement coefficient was 0.053 (-0.077-0.304). The Pearson's correlation coefficient for the ICC vs. maximum MAD was low (r = 0.34, p < 2.2 × 10-16). We found only 239 probes that were highly stable based on both the maximum MAD (<5th percentile, <0.01) and ICC criterion (>95th percentile, >0.74). Conclusions: The whole-blood DNA methylation profile, as measured using microarrays, is dynamic over >1 year, but contains a fraction of stable probes, most of which are related to genomic variation. A resource describing probe stability is made publicly available, with the intention to support biomarker studies and the investigation of early epigenetic programming. The absolute error and correlation are two complementary facets of probe stability that may be considered in further research, especially to determine the stability of probes in health and disease across different tissues and populations.
Collapse
Affiliation(s)
| | | | | | - Jan Krzysztof Nowak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, ul. Szpitalna 27/33, 60-572 Poznan, Poland; (M.D.); (J.W.); (K.W.)
| |
Collapse
|
5
|
Tovo PA, Ribaldone DG, Galliano I, Caviglia GP, Dini M, Veglio V, Calvi C, Montanari P, Pitoni D, Frara S, Tribocco E, Poshnjari A, Bergallo M. Enhanced Transcription of Human Endogenous Retroviruses and TRIM28 Downregulation in Patients with Inflammatory Bowel Disease. Viruses 2024; 16:1570. [PMID: 39459904 PMCID: PMC11512322 DOI: 10.3390/v16101570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Inflammatory bowel disease (IBD) includes patients affected by Crohn's disease or ulcerative colitis. IBD is thought to be a chronic immune-mediated disease, but its origin remains elusive, and this limits new therapeutic approaches. Human endogenous retroviruses (HERVs) originate from ancestral infections and represent 8% of the human genome. HERVs are no longer infectious, but some retroviral sequences can be activated, and their aberrant expressions have been implicated in inflammatory and autoimmune disorders. HERV transcription is regulated by TRIM28 and SETDB1, which are also directly involved in epigenetic processes and modulation of the immune response. Using a PCR real-time Taqman amplification assay, we assessed, for the first time, the transcription levels of pol genes of HERV-H, -K, and -W families of env genes of syncytin 1 (SYN1), SYN2, and HERV-W, as well as of TRIM28 and SETDB1 in the whole blood of 48 patients with Crohn's disease (CD), 20 with ulcerative colitis (UC), and in healthy controls (HC) of comparable age. The transcriptional levels of HERV-H-pol (p = 0.0003) and HERV-K-pol (p = 0.001) were significantly higher in IBD patients compared with HC, with no differences between patients with CD and UC. No significant differences were found for the remaining HERVs between IBD patients and HC. The transcript levels of TRIM28 were significantly downregulated in IBD patients (p < 0.001), without differences between CD and UC, while the SETDB1 levels were preserved. The enhanced transcription of HERV-H-pol and HERV-K-pol, as well as the impaired activation of TRIM28, were not influenced by clinical disease activity and type of treatment. The overexpression of HERVs and impaired transcription of TRIM28 in patients affected by CD or UC suggest that they might be the main actors in the pathophysiology of IBD, opening the way to innovative targeted interventions.
Collapse
Affiliation(s)
- Pier-Angelo Tovo
- Department of Public Health and Pediatric Sciences, University of Turin, Piazza Polonia 94, 10126 Turin, Italy;
| | - Davide Giuseppe Ribaldone
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, 10123 Turin, Italy; (D.G.R.); (G.P.C.); (V.V.); (D.P.); (S.F.); (E.T.); (A.P.)
| | - Ilaria Galliano
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy; (M.D.); (C.C.); (P.M.); (M.B.)
| | - Gian Paolo Caviglia
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, 10123 Turin, Italy; (D.G.R.); (G.P.C.); (V.V.); (D.P.); (S.F.); (E.T.); (A.P.)
| | - Maddalena Dini
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy; (M.D.); (C.C.); (P.M.); (M.B.)
| | - Valentina Veglio
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, 10123 Turin, Italy; (D.G.R.); (G.P.C.); (V.V.); (D.P.); (S.F.); (E.T.); (A.P.)
| | - Cristina Calvi
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy; (M.D.); (C.C.); (P.M.); (M.B.)
| | - Paola Montanari
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy; (M.D.); (C.C.); (P.M.); (M.B.)
| | - Demis Pitoni
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, 10123 Turin, Italy; (D.G.R.); (G.P.C.); (V.V.); (D.P.); (S.F.); (E.T.); (A.P.)
| | - Simone Frara
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, 10123 Turin, Italy; (D.G.R.); (G.P.C.); (V.V.); (D.P.); (S.F.); (E.T.); (A.P.)
| | - Elisa Tribocco
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, 10123 Turin, Italy; (D.G.R.); (G.P.C.); (V.V.); (D.P.); (S.F.); (E.T.); (A.P.)
| | - Anxhela Poshnjari
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, 10123 Turin, Italy; (D.G.R.); (G.P.C.); (V.V.); (D.P.); (S.F.); (E.T.); (A.P.)
| | - Massimiliano Bergallo
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy; (M.D.); (C.C.); (P.M.); (M.B.)
| |
Collapse
|
6
|
Du X, Xu J, Mei F, Shen J, Zhou B, Zhu Z, Li Z, Su X, Li J, Schlüter D, Ruan J, Wang X. Deubiquitination of RIPK2 by OTUB2 augments NOD2 signalling and protective effects in intestinal inflammation. Clin Transl Med 2024; 14:e70038. [PMID: 39358938 PMCID: PMC11446981 DOI: 10.1002/ctm2.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/02/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract, but the molecular mechanisms underlying IBD are incompletely understood. In this study, we explored the role and regulating mechanism of otubain 2 (OTUB2), a deubiquitinating enzyme, in IBD. METHODS To study the function of OTUB2 in IBD, we generated Otub2-/- mice and treated them with dextran sulfate sodium (DSS) to induce experimental colitis. Bone marrow transplantation was performed to identify the cell populations that were affected by OTUB2 in colitis. The molecular mechanism of OTUB2 in signal transduction was studied by various biochemical methods. RESULTS OTUB2 was highly expressed in colon-infiltrating macrophages in both humans with IBD and mice with DSS-induced experimental colitis. Colitis was significantly aggravated in Otub2-/- mice and bone marrow chimeric mice receiving Otub2-/- bone marrow. OTUB2-deficiency impaired the production of cytokines and chemokines in macrophages in response to the NOD2 agonist muramyl dipeptide (MDP). Upon MDP stimulation, OTUB2 promoted NOD2 signaling by stabilizing RIPK2. Mechanistically, OTUB2 inhibited the proteasomal degradation of RIPK2 by removing K48-linked polyubiquitination on RIPK2, which was mediated by the active C51 residue in OTUB2. In mice, OTUB2 ablation abolished the protective effects of MDP administration in colitis. CONCLUSION This study identified OTUB2 as a novel regulator of intestinal inflammation.
Collapse
Affiliation(s)
- Xue Du
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Jun Xu
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Fuqi Mei
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Jiangyun Shen
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Bincheng Zhou
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Zhenhu Zhu
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Zhongding Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Xian Su
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Jianmin Li
- Department of PathologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Dirk Schlüter
- Hannover Medical SchoolInstitute of Medical Microbiology and Hospital EpidemiologyHannoverGermany
| | - Jing Ruan
- Department of PathologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xu Wang
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| |
Collapse
|
7
|
Lin D, Jin Y, Shao X, Xu Y, Ma G, Jiang Y, Xu Y, Jiang Y, Hu D. Global, regional, and national burden of inflammatory bowel disease, 1990-2021: Insights from the global burden of disease 2021. Int J Colorectal Dis 2024; 39:139. [PMID: 39243331 PMCID: PMC11380638 DOI: 10.1007/s00384-024-04711-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
PURPOSE The prevalence of inflammatory bowel disease (IBD) is on the rise worldwide. We utilizes data from the Global Burden of Diseases (GBD) 2021 to analyze the national-level burden of IBD, trends in disease incidence, and epidemiological characteristics. METHODS Detailed information on IBD was gathered from 204 countries and territories spanning 1990 to 2021, sourced from the GBD 2021. Calculations were performed for incidence rates, mortality rates, disease-adjusted life years (DALYs), and estimated annual percentage changes (EAPCs). These trends were analyzed based on region, nationality, age, gender, and World Bank income level stratifications. RESULTS The global age-standardised incident rate (ASIR) of IBD increased from 4.22 per 100000 in 1990 to 4.45 per 100000 in 2021. However, the age-standardised mortality rate (ASMR) decreased from 0.60 per 100000 in 1990 to 0.52 per 100000 in 2021. Similarly, the age-standardised DALYs rate decreased from 21.55 per 100000 in 1990 to 18.07 per 100000 in 2021. Gender comparisons showed negligible differences in disease burden. The greatest increase in IBD-associated ASIR and ASMR occurred in World Bank upper-middle income region (EAPCs, 1.25) and World Bank high-income region (EAPCs, 1.00), respectively. Regionally, East Asia experienced the largest increase in ASIR (EAPCs, 2.89). Among 204 countries, China had the greatest increases in ASIR (EAPCs, 2.93), Netherlands had the highest ASMR in 2021 (2.21 per 100000). CONCLUSIONS Global incidence rate of IBD have been increasing from 1990 to 2021, while the DALYs and mortality have been decreasing. The escalating incident rates in select Asian regions deserves further attention.
Collapse
Affiliation(s)
- Daopo Lin
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yang Jin
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xiaoxiao Shao
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yuan Xu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Guolong Ma
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yi Jiang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yinghe Xu
- Department of Critical Care Medicine, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang Province, China.
| | - Yongpo Jiang
- Department of Critical Care Medicine, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang Province, China.
| | - Dingyuan Hu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
8
|
Zeng X, Tang J, Zhang Q, Wang C, Qi J, Wei Y, Xu J, Yang K, Zhou Z, Wu H, Luo J, Jiang Y, Song Z, Wu J, Wu J. CircHIPK2 Contributes Cell Growth in Intestinal Epithelial of Colitis and Colorectal Cancer through Promoting TAZ Translation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401588. [PMID: 38981023 PMCID: PMC11425914 DOI: 10.1002/advs.202401588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/13/2024] [Indexed: 07/11/2024]
Abstract
Colorectal cancer (CRC) and inflammatory bowel disease (IBD) are escalating global health concerns. Despite their distinct clinical presentations, both disorders share intricate genetic and molecular interactions. The Hippo signaling pathway plays a crucial role in regulating cell processes and is implicated in the pathogenesis of IBD and CRC. Circular RNAs (circRNAs) have gained attention for their roles in various diseases, including IBD and CRC. However, a comprehensive understanding of specific circRNAs involved in both IBD and CRC, and their functional roles is lacking. Here, it is found that circHIPK2 (hsa_circRNA_104507) is a bona fide circRNA consistently upregulated in both IBD and CRC suggesting its potential as a biomarker. Furthermore, silencing of circHIPK2 suppressed the growth of CRC cells in vitro and in vivo. Interestingly, decreased circHipk2 potentiated dextran sulfate sodium (DSS)-induced colitis but alleviated colitis-associated tumorigenesis. Most significantly, mechanistic investigations further unveil that circHIPK2, mediated by FUS, interacting with EIF4A3 to promote the translation of TAZ, ultimately increasing the transcription of downstream target genes CCN1 and CCN2. Taken together, circHIPK2 emerges as a key player in the shared mechanisms of IBD and CRC, modulating the Hippo signaling pathway. CircHIPK2-EIF4A3 axis contributes to cell growth in intestinal epithelial of colitis and CRC by enhancing TAZ translation.
Collapse
Affiliation(s)
- Xixi Zeng
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
- The Joint Innovation Center for Health and Medicine, Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, Zhejiang, 324000, China
| | - Jielin Tang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, 325035, China
| | - Qian Zhang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Chenxing Wang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children of Wenzhou Medical University, Zhejiang, 325003, China
| | - Ji Qi
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Yusi Wei
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Jiali Xu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Kaiyuan Yang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Zuolin Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Hao Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Jiarong Luo
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - Yi Jiang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children of Wenzhou Medical University, Zhejiang, 325003, China
| | - Zengqiang Song
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, 325035, China
| | - Jinyu Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
| | - Jianmin Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, 325035, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Zhejiang, 325035, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315302, China
| |
Collapse
|
9
|
Verstockt B. Epigenetic Fingerprints in IBD: From Methylation Patterns to Clinical Implications. J Crohns Colitis 2024; 18:1175-1176. [PMID: 38885255 DOI: 10.1093/ecco-jcc/jjae086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Affiliation(s)
- Bram Verstockt
- University Hospitals Leuven, Department of Gastroenterology and Hepatology, KU Leuven, Leuven, Belgium
- KU Leuven, Department of Chronic Diseases and Metabolism, Leuven, Belgium
| |
Collapse
|
10
|
Sæterstad S, Østvik AE, Hansen MD, Bruland T, van Beelen Granlund A. The effect of rs2910686 on ERAP2 expression in IBD and epithelial inflammatory response. J Transl Med 2024; 22:750. [PMID: 39123229 PMCID: PMC11316291 DOI: 10.1186/s12967-024-05532-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND ERAP2 is an aminopeptidase involved in antigen processing and presentation, and harbor genetic variants linked to several inflammatory diseases such as Inflammatory Bowel Disease (IBD). The lack of an ERAP2 gene homologue in mice has hampered functional studies, and most human studies have focused on cells of hematopoietic origin. Using an IBD biobank as vantage point, this study explores how genetic variation in ERAP2 affects gene expression in human-derived epithelial organoids upon proinflammatory stimulation. METHODS An IBD patient cohort was genotyped with regards to two single nucleotide polymorphisms (SNP) (rs2910686/rs2248374) associated with ERAP2 expression levels, and we examined the correlation between colon gene expression and genotype, specifically aiming to establish a relationship with ERAP2 expression proficiency. Human-derived colon organoids (colonoids) with known ERAP2 genotype were established and used to explore differences in whole genome gene expression between ERAP2-deficient (n = 4) and -proficient (n = 4) donors upon pro-inflammatory encounter. RESULTS When taking rs2910686 genotype into account, ERAP2 gene expression is upregulated in the inflamed colon of IBD patients. Colonoids upregulate ERAP2 upon IFNɣ stimulation, and ERAP2 expression proficiency is dependent on rs2910686 genotype. Colonoid genotyping confirms that mechanisms independent of the frequently studied SNP rs2248374 can cause ERAP2-deficiency. A total of 586 genes involved in various molecular mechanisms are differentially expressed between ERAP2 proficient- and deficient colonoids upon proinflammatory stimulation, including genes encoding proteins with the following molecular function: catalytic activity (AOC1, CPE, ANPEP and MEP1A), regulator activity (TNFSF9, MDK, GDF15, ILR6A, LGALS3 and FLNA), transmembrane transporter activity (SLC40A1 and SLC5A1), and extracellular matrix structural constituents (FGL2, HMCN2, and MUC17). CONCLUSIONS ERAP2 is upregulated in the inflamed IBD colon mucosa, and expression proficiency is highly correlated with genotype of rs2910686. While the SNP rs2248374 is commonly used to determine ERAP2 expressional proficiency, our data confirms that mechanisms independent of this SNP can lead to ERAP2 deficiency. Our data demonstrates that epithelial ERAP2 presence affects the inflammatory response in colonoids, suggesting a pleiotropic role of ERAP2 beyond MHC class I antigen processing.
Collapse
Affiliation(s)
- Siri Sæterstad
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ann Elisabeth Østvik
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Marianne Doré Hansen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Torunn Bruland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Atle van Beelen Granlund
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway.
- Department of Pathology, St. Olav's University Hospital, Trondheim, Norway.
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| |
Collapse
|
11
|
Minea H, Singeap AM, Minea M, Juncu S, Muzica C, Sfarti CV, Girleanu I, Chiriac S, Miftode ID, Stanciu C, Trifan A. The Contribution of Genetic and Epigenetic Factors: An Emerging Concept in the Assessment and Prognosis of Inflammatory Bowel Diseases. Int J Mol Sci 2024; 25:8420. [PMID: 39125988 PMCID: PMC11313574 DOI: 10.3390/ijms25158420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Inflammatory bowel disease (IBD) represents heterogeneous and relapsing intestinal conditions with a severe impact on the quality of life of individuals and a continuously increasing prevalence. In recent years, the development of sequencing technology has provided new means of exploring the complex pathogenesis of IBD. An ideal solution is represented by the approach of precision medicine that investigates multiple cellular and molecular interactions, which are tools that perform a holistic, systematic, and impartial analysis of the genomic, transcriptomic, proteomic, metabolomic, and microbiomics sets. Hence, it has led to the orientation of current research towards the identification of new biomarkers that could be successfully used in the management of IBD patients. Multi-omics explores the dimension of variation in the characteristics of these diseases, offering the advantage of understanding the cellular and molecular mechanisms that affect intestinal homeostasis for a much better prediction of disease development and choice of treatment. This review focuses on the progress made in the field of prognostic and predictive biomarkers, highlighting the limitations, challenges, and also the opportunities associated with the application of genomics and epigenomics technologies in clinical practice.
Collapse
Affiliation(s)
- Horia Minea
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (H.M.); (S.J.); (C.V.S.); (I.G.); (S.C.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Ana-Maria Singeap
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (H.M.); (S.J.); (C.V.S.); (I.G.); (S.C.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Manuela Minea
- Department of Microbiology, The National Institute of Public Health, 700464 Iasi, Romania;
| | - Simona Juncu
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (H.M.); (S.J.); (C.V.S.); (I.G.); (S.C.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Cristina Muzica
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (H.M.); (S.J.); (C.V.S.); (I.G.); (S.C.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Catalin Victor Sfarti
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (H.M.); (S.J.); (C.V.S.); (I.G.); (S.C.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Irina Girleanu
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (H.M.); (S.J.); (C.V.S.); (I.G.); (S.C.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Stefan Chiriac
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (H.M.); (S.J.); (C.V.S.); (I.G.); (S.C.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Ioana Diandra Miftode
- Department of Radiology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Department of Radiology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Carol Stanciu
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (H.M.); (S.J.); (C.V.S.); (I.G.); (S.C.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Anca Trifan
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (H.M.); (S.J.); (C.V.S.); (I.G.); (S.C.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| |
Collapse
|
12
|
Santos EA, Silva JL, Leocádio PCL, Andrade MER, Queiroz-Junior CM, Oliveira NSS, Alves JL, Oliveira JS, Aguilar EC, Boujour K, Cogliati B, Cardoso VN, Fernandes SO, Faria AMC, Alvarez-Leite JI. Cutaneous Application of Capsaicin Cream Reduces Clinical Signs of Experimental Colitis and Repairs Intestinal Barrier Integrity by Modulating the Gut Microbiota and Tight Junction Proteins. ACS Pharmacol Transl Sci 2024; 7:2143-2153. [PMID: 39022369 PMCID: PMC11249629 DOI: 10.1021/acsptsci.4c00207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/25/2024] [Accepted: 05/30/2024] [Indexed: 07/20/2024]
Abstract
Capsaicin, a pungent compound in chili peppers, is described as having potent anti-inflammatory, antioxidant, and antimicrobial properties. It is also described as a potential modulator of the immune system and intestinal microbiota. Oral or rectal administration of capsaicin has been studied to treat or prevent colitis. However, those vias are often not well accepted due to the burning sensation that capsaicin can cause. Our objective was to evaluate whether the application of capsaicin skin creams (0.075%) would be effective in improving inflammation and epithelial barrier function as well as the composition of the gut microbiota in a model of mild colitis induced by dextran sulfate sodium (1.5%). The results showed that the cutaneous application of capsaicin reversed weight loss and decreased colon shortening and diarrhea, all typical signs of colitis. There was also an improvement in the intestinal epithelial barrier, preserving proteins from tight junctions. We also evaluated the biodistribution of 99mtechnetium-radiolabeled capsaicin (99mTc-CAPS) applied to the back skin of the animals. We found significant concentrations of 99 mTc-Cap in the colon and small intestine after 2 and 4 h of administration. In addition, there was an increased expression of capsaicin receptor TRPV1 in the colon. Moreover, animals with colitis receiving cutaneous capsaicin presented a better short-chain fatty acid profile and increased levels of SIgA, suggesting increased microbiota diversity. In conclusion, our work opens avenues for further studies to better understand capsaicin's potential benefits and mechanisms in addressing colitis through cutaneous application.
Collapse
Affiliation(s)
- Elandia A. Santos
- Departamento
de Bioquímica e Imunologia—Instituto de Ciências
Biológicas, Universidade Federal
de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| | - Janayne L. Silva
- Departamento
de Bioquímica e Imunologia—Instituto de Ciências
Biológicas, Universidade Federal
de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| | - Paola C. L. Leocádio
- Departamento
de Bioquímica e Imunologia—Instituto de Ciências
Biológicas, Universidade Federal
de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| | - Maria Emilia R. Andrade
- Departamento
de Análises Clínicas e Toxicológicas, Faculdade de Farmácia da UFMG, Belo Horizonte 31270-901, Brazil
| | - Celso M. Queiroz-Junior
- Departamento
de Morfologia, Instituto de Ciências
Biológicas—(UFMG), Belo Horizonte 31270-901, Brazil
| | - Nathan S. S. Oliveira
- Departamento
de Bioquímica e Imunologia—Instituto de Ciências
Biológicas, Universidade Federal
de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| | - Juliana L. Alves
- Departamento
de Bioquímica e Imunologia—Instituto de Ciências
Biológicas, Universidade Federal
de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| | - Jamil S. Oliveira
- Departamento
de Bioquímica e Imunologia—Instituto de Ciências
Biológicas, Universidade Federal
de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| | - Edenil C. Aguilar
- Icahn School
of Medicine at Mount Sinai, New
York, New York 10029, United States
| | - Kennedy Boujour
- Departamento
de Patologia Animal, Universidade de São
Paulo (USP), São Paulo 05508-220, Brazil
- Department
of Cellular Biology and Infection, Unity of Biochemistry Membrane
and Transport, Institut Pasteur, Paris 75724, France
| | - Bruno Cogliati
- Departamento
de Patologia Animal, Universidade de São
Paulo (USP), São Paulo 05508-220, Brazil
| | - Valbert N. Cardoso
- Departamento
de Análises Clínicas e Toxicológicas, Faculdade de Farmácia da UFMG, Belo Horizonte 31270-901, Brazil
| | - Simone Odilia
A. Fernandes
- Departamento
de Análises Clínicas e Toxicológicas, Faculdade de Farmácia da UFMG, Belo Horizonte 31270-901, Brazil
| | - Ana Maria C. Faria
- Departamento
de Bioquímica e Imunologia—Instituto de Ciências
Biológicas, Universidade Federal
de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| | - Jacqueline I. Alvarez-Leite
- Departamento
de Bioquímica e Imunologia—Instituto de Ciências
Biológicas, Universidade Federal
de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| |
Collapse
|
13
|
Sandys O, Stokkers PCF, Te Velde AA. DAMP-ing IBD: Extinguish the Fire and Prevent Smoldering. Dig Dis Sci 2024:10.1007/s10620-024-08523-5. [PMID: 38963463 DOI: 10.1007/s10620-024-08523-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024]
Abstract
In inflammatory bowel diseases (IBD), the most promising therapies targeting cytokines or immune cell trafficking demonstrate around 40% efficacy. As IBD is a multifactorial inflammation of the intestinal tract, a single-target approach is unlikely to solve this problem, necessitating an alternative strategy that addresses its variability. One approach often overlooked by the pharmaceutically driven therapeutic options is to address the impact of environmental factors. This is somewhat surprising considering that IBD is increasingly viewed as a condition heavily influenced by such factors, including diet, stress, and environmental pollution-often referred to as the "Western lifestyle". In IBD, intestinal responses result from a complex interplay among the genetic background of the patient, molecules, cells, and the local inflammatory microenvironment where danger- and microbe-associated molecular patterns (D/MAMPs) provide an adjuvant-rich environment. Through activating DAMP receptors, this array of pro-inflammatory factors can stimulate, for example, the NLRP3 inflammasome-a major amplifier of the inflammatory response in IBD, and various immune cells via non-specific bystander activation of myeloid cells (e.g., macrophages) and lymphocytes (e.g., tissue-resident memory T cells). Current single-target biological treatment approaches can dampen the immune response, but without reducing exposure to environmental factors of IBD, e.g., by changing diet (reducing ultra-processed foods), the adjuvant-rich landscape is never resolved and continues to drive intestinal mucosal dysregulation. Thus, such treatment approaches are not enough to put out the inflammatory fire. The resultant smoldering, low-grade inflammation diminishes physiological resilience of the intestinal (micro)environment, perpetuating the state of chronic disease. Therefore, our hypothesis posits that successful interventions for IBD must address the complexity of the disease by simultaneously targeting all modifiable aspects: innate immunity cytokines and microbiota, adaptive immunity cells and cytokines, and factors that relate to the (micro)environment. Thus the disease can be comprehensively treated across the nano-, meso-, and microscales, rather than with a focus on single targets. A broader perspective on IBD treatment that also includes options to adapt the DAMPing (micro)environment is warranted.
Collapse
Affiliation(s)
- Oliver Sandys
- Tytgat Institute for Liver and Intestinal Research, AmsterdamUMC, AGEM, University of Amsterdam, Amsterdam, The Netherlands
| | - Pieter C F Stokkers
- Department of Gastroenterology and Hepatology, OLVG West, Amsterdam, The Netherlands
| | - Anje A Te Velde
- Tytgat Institute for Liver and Intestinal Research, AmsterdamUMC, AGEM, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
14
|
Jian M, Lu X, Tang M, Ouyang Z, Lai Z, Zhuang J, Qian R. Umbrella review of risk factors for inflammatory bowel disease: a study protocol. BMJ Open 2024; 14:e077267. [PMID: 38925703 PMCID: PMC11202652 DOI: 10.1136/bmjopen-2023-077267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
INTRODUCTION Inflammatory bowel disease (IBD) is a chronic idiopathic inflammatory disorder that arises from complex interactions between genetics, environment and gut microbiota. It encompasses Crohn's disease, ulcerative colitis and IBD-unclassified. The protracted course of IBD imposes a significant burden on patients' quality of life, economic productivity, social functioning, as well as treatment, hospitalisation and surgery. This study aims to conduct an umbrella review of meta-analyses to systematically evaluate the methodology's quality, potential biases and validity of all epidemiological evidence focused on risk factors for IBD while providing an overview of the evidence concerning IBD risk factors. METHODS AND ANALYSIS We will systematically search, extract and analyse data from reported systematic reviews and meta-analyses that specifically focus on the risk factors of IBD, following the guidelines outlined in Preferred Reporting Items for Overviews of Reviews. Our search will encompass PubMed, Embase, Web of Science and the Cochrane Database of Systematic Reviews from the initial period up until April 2023 (last update), targeting systematic reviews and meta-analyses based on non-interventional studies. Inclusion criteria allow for systematic reviews and meta-analyses evaluating IBD risk factors across all countries and settings, regardless of ethnicity or sex. The identified risk factors will be categorised according to the health ecological model into innate personal traits, behavioural lifestyles, interpersonal networks, socioeconomic status and macroenvironments. To assess methodological quality for each meta-analysis included in our study, two authors will employ a measurement tool to assess the methodological quality of systematic reviews (AMSTAR)-2, Grading of Recommendations, Assessment, Development and Evaluation (GRADE) criteria along with evidence classification criteria. ETHICS AND DISSEMINATION Ethical approval is not required for this umbrella review. We will seek to submit the results for publication in a peer-reviewed journal or present it at conferences. PROSPERO REGISTRATION NUMBER CRD42023417175.
Collapse
Affiliation(s)
- Mingwei Jian
- Shenzhen Bao'an Traditional Chinese Medicine Hospital,Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xiang Lu
- Shenzhen Bao'an Traditional Chinese Medicine Hospital,Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Min Tang
- Shenzhen Bao'an Traditional Chinese Medicine Hospital,Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zichen Ouyang
- Shenzhen Bao'an Traditional Chinese Medicine Hospital,Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhiming Lai
- Shenzhen Bao'an Traditional Chinese Medicine Hospital,Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jiamei Zhuang
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Rui Qian
- Shenzhen Bao'an Traditional Chinese Medicine Hospital,Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
15
|
Hammerhøj A, Chakravarti D, Sato T, Jensen KB, Nielsen OH. Organoids as regenerative medicine for inflammatory bowel disease. iScience 2024; 27:110118. [PMID: 38947526 PMCID: PMC11214415 DOI: 10.1016/j.isci.2024.110118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic disorder with an increasing global prevalence. Managing disease activity relies on various pharmacological options. However, the effectiveness of current therapeutics is limited and not universally applicable to all patients and circumstances. Consequently, developing new management strategies is necessary. Recent advances in endoscopically obtained intestinal biopsy specimens have highlighted the potential of intestinal epithelial organoid transplantation as a novel therapeutic approach. Experimental studies using murine and human organoid transplantations have shown promising outcomes, including tissue regeneration and functional recovery. Human trials with organoid therapy have commenced; thus, this article provides readers with insights into the necessity and potential of intestinal organoid transplantation as a new regenerative therapeutic option in clinical settings and explores its associated challenges.
Collapse
Affiliation(s)
- Alexander Hammerhøj
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Deepavali Chakravarti
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kim Bak Jensen
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ole Haagen Nielsen
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| |
Collapse
|
16
|
Yan Ang Q, Plichta D, Kim S, Hyun-A Kim I, Gregory S, Xia Y, Lau H, Xavier R, Ananthakrishnan AN. Differential Impact of Smoking on Methylome and Transcriptome in Crohn's Disease and Ulcerative Colitis. Inflamm Bowel Dis 2024; 30:981-991. [PMID: 38001042 PMCID: PMC11145016 DOI: 10.1093/ibd/izad268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND Smoking is an environmental factor that differentially impacts Crohn's disease (CD) and ulcerative colitis (UC). The mechanism of impact of smoking on disease risk and clinical outcomes remains to be established. METHODS This study used a prospective cohort of patients with CD or UC. Self-reported smoking status was validated using serum cotinine measurement. We profiled methylation changes in peripheral blood using the Illumina Methylation BeadChip. Transcriptomic profiling was performed on ileal and colonic tissue using an Illumina TruSeq platform. We compared the methylation and transcriptional changes in current, former, and never smokers stratified by disease type. RESULTS Our cohort included 200 patients with CD or UC with methylation profiles and 160 with transcriptomic data. The mean serum cotinine level was higher in current compared with former or never smokers. Epigenetic changes common to both CD and UC included hypomethylation at AHRR. Smoking-associated MGAT3 hypomethylation was associated with severe disease course only in UC, while IER3 hypomethylation was associated with worse course only in CD. Smoking downregulated several inflammatory pathways in UC. Current smoking in CD but not in UC was associated with upregulation of several genes mediating Paneth cell function. Genes with opposite direction of effects in CD and UC include HSD3B2 and GSTA1. CONCLUSIONS Our findings suggest both common and differential effects of cigarette smoking on CD and UC. Paneth cell dysfunction may mediate adverse impact of smoking on CD. Bile acid and oxidative stress pathways may be relevant for the differential effect of smoking on CD and UC.
Collapse
Affiliation(s)
| | | | - Sean Kim
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Iris Hyun-A Kim
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Sadie Gregory
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Yan Xia
- Broad Institute, Cambridge, MA, USA
| | - Helena Lau
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Ramnik Xavier
- Broad Institute, Cambridge, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Wang J, Zhou Y, Zhang M, Wu Y, Wu Q, Su W, Xu M, Wu J, Zhang M, Shuai J, Tang W, Lv J, Wu M, Xia Z. YTHDF1-CLOCK axis contributes to pathogenesis of allergic airway inflammation through LLPS. Cell Rep 2024; 43:113947. [PMID: 38492220 DOI: 10.1016/j.celrep.2024.113947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/23/2024] [Accepted: 02/27/2024] [Indexed: 03/18/2024] Open
Abstract
N6-methyladenosine (m6A) modification has been implicated in many cell processes and diseases. YTHDF1, a translation-facilitating m6A reader, has not been previously shown to be related to allergic airway inflammation. Here, we report that YTHDF1 is highly expressed in allergic airway epithelial cells and asthmatic patients and that it influences proinflammatory responses. CLOCK, a subunit of the circadian clock pathway, is the direct target of YTHDF1. YTHDF1 augments CLOCK translation in an m6A-dependent manner. Allergens enhance the liquid-liquid phase separation (LLPS) of YTHDF1 and drive the formation of a complex comprising dimeric YTHDF1 and CLOCK mRNA, which is distributed to stress granules. Moreover, YTHDF1 strongly activates NLRP3 inflammasome production and interleukin-1β secretion leading to airway inflammatory responses, but these phenotypes are abolished by deleting CLOCK. These findings demonstrate that YTHDF1 is an important regulator of asthmatic airway inflammation, suggesting a potential therapeutic target for allergic airway inflammation.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Zhou
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng Zhang
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yujiao Wu
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qun Wu
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Su
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinhong Wu
- Department of Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Zhang
- Department of Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianwei Shuai
- Joint Research Centre on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Wei Tang
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jiajia Lv
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Min Wu
- Joint Research Centre on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China.
| | - Zhenwei Xia
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
18
|
Liu C, Zhang JX, Li JT, Wei YP, Zhen JH, Wu YR, He HD, Chen Y, Sun JY, Tan C, Wang S, Xiong QT, Liao F, Yang XC, An P, Liu ZC, Jiang CQ, Shi J, Wu KC, Dong WG. Geographic differences in psychological symptoms, sleep quality, and quality of life in patients with inflammatory bowel disease: A multicenter study in China. J Dig Dis 2024; 25:109-122. [PMID: 38503514 DOI: 10.1111/1751-2980.13259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 02/09/2024] [Accepted: 02/16/2024] [Indexed: 03/21/2024]
Abstract
OBJECTIVE We aimed to explore the geographic differences in psychological symptoms, sleep quality, and quality of life (QoL) among adult patients with inflammatory bowel disease (IBD). METHODS A unified questionnaire was developed to collect data on psychological status and QoL of IBD patients from 42 hospitals across 22 provinces, municipalities, and autonomous regions in China's mainland from September 2021 to May 2022. RESULTS A total of 2478 patients with IBD were surveyed. The proportions of patients with anxiety (28.5% vs 23.1%), depression (32.3% vs 27.8%), and poor QoL (44.8% vs 32.2%) were significantly higher in patients from the northern region compared to the southern region (all P < 0.05). In the western region, the proportions of patients with anxiety (31.9% vs 23.0%), depression (37.7% vs 26.7%), sleep disturbances (64.5% vs 58.5%), and poor QoL (44.9% vs 34.8%) were significantly higher than in the eastern and central regions (all P < 0.01). Patients from inland regions had significantly higher rates of anxiety (27.1% vs 23.3%), depression (32.5% vs 26.0%), sleep disturbance (62.0% vs 57.7%), and poor QoL (43.5% vs 29.9%) compared to those from coastal regions (all P < 0.05). In economically underdeveloped areas, the proportions of patients with depression (33.1% vs 28.5%) and poor QoL (52.0% vs 32.4%) were significantly higher than in economically (relatively) developed areas (both P < 0.05). CONCLUSION There are significant geographic differences in psychological symptoms, sleep quality, and QoL among Chinese patients with IBD, which might provide valuable insights for global IBD research and clinical practice.
Collapse
Affiliation(s)
- Chuan Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Ji Xiang Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Jin Ting Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yu Ping Wei
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Jun Hai Zhen
- Department of General Practice, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yan Rui Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Hao Dong He
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Ying Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Jia Yi Sun
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Cheng Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Shuo Wang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Qiu Tang Xiong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Fei Liao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Wuhan University Shenzhen Research Institute, Shenzhen, Guangdong Province, China
| | - Xiao Cui Yang
- Department of Gastroenterology, Ankang Central Hospital, Ankang, Shaanxi Province, China
| | - Ping An
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhong Chun Liu
- Center for Mental Health, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Chang Qing Jiang
- Department of Clinical Psychology, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Jie Shi
- Department of Medical Psychology, Chinese People's Liberation Army Rocket Army Characteristic Medical Center, Beijing, China
| | - Kai Chun Wu
- Department of Gastroenterology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Wei Guo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
19
|
Bertin B, Foligne B, Ley D, Lesage J, Beghin L, Morcel J, Gottrand F, Hermann E. An Overview of the Influence of Breastfeeding on the Development of Inflammatory Bowel Disease. Nutrients 2023; 15:5103. [PMID: 38140362 PMCID: PMC10745409 DOI: 10.3390/nu15245103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The first 1000 days of life is a critical period that contributes significantly to the programming of an individual's future health. Among the many changes that occur during this period early in life, there is growing evidence that the establishment of healthy gut microbiota plays an important role in the prevention of both short- and long-term health problems. Numerous publications suggest that the quality of the gut microbiota colonisation depends on several dietary factors, including breastfeeding. In this respect, a relationship between breastfeeding and the risk of inflammatory bowel disease (IBD) has been suggested. IBDs are chronic intestinal diseases, and perinatal factors may be partly responsible for their onset. We review the existence of links between breastfeeding and IBD based on experimental and clinical studies. Overall, despite encouraging experimental data in rodents, the association between breastfeeding and the development of IBD remains controversial in humans, partly due to the considerable heterogeneity between clinical studies. The duration of exclusive breastfeeding is probably decisive for its lasting effect on IBD. Thus, specific improvements in our knowledge could support dietary interventions targeting the gut microbiome, such as the early use of prebiotics, probiotics or postbiotics, in order to prevent the disease.
Collapse
Affiliation(s)
- Benjamin Bertin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Benoit Foligne
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Delphine Ley
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Jean Lesage
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Laurent Beghin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Jules Morcel
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Frédéric Gottrand
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Emmanuel Hermann
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| |
Collapse
|
20
|
Prame Kumar K, Ooi JD, Goldberg R. The interplay between the microbiota, diet and T regulatory cells in the preservation of the gut barrier in inflammatory bowel disease. Front Microbiol 2023; 14:1291724. [PMID: 38107848 PMCID: PMC10722198 DOI: 10.3389/fmicb.2023.1291724] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023] Open
Abstract
Inflammatory bowel disease (IBD) is becoming more common in the Western world due to changes in diet-related microbial dysbiosis, genetics and lifestyle. Incidences of gut permeability can predate IBD and continued gut barrier disruptions increase the exposure of bacterial antigens to the immune system thereby perpetuating chronic inflammation. Currently, most of the approved IBD therapies target individual pro-inflammatory cytokines and pathways. However, they fail in approximately 50% of patients due to their inability to overcome the redundant pro inflammatory immune responses. There is increasing interest in the therapeutic potential of T regulatory cells (Tregs) in inflammatory conditions due to their widespread capability to dampen inflammation, promote tolerance of intestinal bacteria, facilitate healing of the mucosal barrier and ability to be engineered for more targeted therapy. Intestinal Treg populations are inherently shaped by dietary molecules and gut microbiota-derived metabolites. Thus, understanding how these molecules influence Treg-mediated preservation of the intestinal barrier will provide insights into immune tolerance-mediated mucosal homeostasis. This review comprehensively explores the interplay between diet, gut microbiota, and immune system in influencing the intestinal barrier function to attenuate the progression of colitis.
Collapse
Affiliation(s)
- Kathryn Prame Kumar
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash Medical Centre, Monash University, Clayton, VIC, Australia
| | | | | |
Collapse
|
21
|
Fiocchi C. Omics and Multi-Omics in IBD: No Integration, No Breakthroughs. Int J Mol Sci 2023; 24:14912. [PMID: 37834360 PMCID: PMC10573814 DOI: 10.3390/ijms241914912] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/27/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
The recent advent of sophisticated technologies like sequencing and mass spectroscopy platforms combined with artificial intelligence-powered analytic tools has initiated a new era of "big data" research in various complex diseases of still-undetermined cause and mechanisms. The investigation of these diseases was, until recently, limited to traditional in vitro and in vivo biological experimentation, but a clear switch to in silico methodologies is now under way. This review tries to provide a comprehensive assessment of state-of-the-art knowledge on omes, omics and multi-omics in inflammatory bowel disease (IBD). The notion and importance of omes, omics and multi-omics in both health and complex diseases like IBD is introduced, followed by a discussion of the various omics believed to be relevant to IBD pathogenesis, and how multi-omics "big data" can generate new insights translatable into useful clinical tools in IBD such as biomarker identification, prediction of remission and relapse, response to therapy, and precision medicine. The pitfalls and limitations of current IBD multi-omics studies are critically analyzed, revealing that, regardless of the types of omes being analyzed, the majority of current reports are still based on simple associations of descriptive retrospective data from cross-sectional patient cohorts rather than more powerful longitudinally collected prospective datasets. Given this limitation, some suggestions are provided on how IBD multi-omics data may be optimized for greater clinical and therapeutic benefit. The review concludes by forecasting the upcoming incorporation of multi-omics analyses in the routine management of IBD.
Collapse
Affiliation(s)
- Claudio Fiocchi
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland, OH 44195, USA;
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
22
|
Kamal S, Parkash N, Beattie W, Christensen B, Segal JP. Are We Ready to Reclassify Crohn's Disease Using Molecular Classification? J Clin Med 2023; 12:5786. [PMID: 37762727 PMCID: PMC10532006 DOI: 10.3390/jcm12185786] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/21/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Crohn's disease (CD) is a type of inflammatory bowel disease. The number of IBD cases worldwide was estimated to be 4.9 million in 2019. CD exhibits heterogeneity in clinical presentation, anatomical involvement, disease behaviour, clinical course and response to treatment. The classical description of CD involves transmural inflammation with skip lesions anywhere along the entire gastrointestinal tract. The complexity and heterogeneity of Crohn's disease is not currently reflected in the conventional classification system. Though the knowledge of Crohn's pathophysiology remains far from understood, the established complex interplay of the omics-genomics, transcriptomics, proteomics, epigenomics, metagenomics, metabolomics, lipidomics and immunophenomics-provides numerous targets for potential molecular markers of disease. Advancing technology has enabled identification of small molecules within these omics, which can be extrapolated to differentiate types of Crohn's disease. The multi-omic future of Crohn's disease is promising, with potential for advancements in understanding of its pathogenesis and implementation of personalised medicine.
Collapse
Affiliation(s)
- Shahed Kamal
- Department of Gastroenterology, Northern Hospital, Epping, Melbourne VIC 3076, Australia
| | - Nikita Parkash
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, Melbourne VIC 3052, Australia
| | - William Beattie
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, Melbourne VIC 3052, Australia
| | - Britt Christensen
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, Melbourne VIC 3052, Australia
- Department of Gastroenterology, The University of Melbourne, Parkville, Melbourne VIC 3010, Australia
| | - Jonathan P. Segal
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, Melbourne VIC 3052, Australia
- Department of Gastroenterology, The University of Melbourne, Parkville, Melbourne VIC 3010, Australia
| |
Collapse
|
23
|
Ventham NT, Kennedy NA, Kalla R, Adams AT, Noble A, Ennis H, Mowat C, Dunlop MG, Satsangi J. Genome-Wide Methylation Profiling in 229 Patients With Crohn's Disease Requiring Intestinal Resection: Epigenetic Analysis of the Trial of Prevention of Post-operative Crohn's Disease (TOPPIC). Cell Mol Gastroenterol Hepatol 2023; 16:431-450. [PMID: 37331566 PMCID: PMC10372903 DOI: 10.1016/j.jcmgh.2023.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND & AIMS DNA methylation alterations may provide important insights into gene-environment interaction in cancer, aging, and complex diseases, such as inflammatory bowel disease (IBD). We aim first to determine whether the circulating DNA methylome in patients requiring surgery may predict Crohn's disease (CD) recurrence following intestinal resection; and second to compare the circulating methylome seen in patients with established CD with that we had reported in a series of inception cohorts. METHODS TOPPIC was a placebo-controlled, randomized controlled trial of 6-mercaptopurine at 29 UK centers in patients with CD undergoing ileocolic resection between 2008 and 2012. Genomic DNA was extracted from whole blood samples from 229 of the 240 patients taken before intestinal surgery and analyzed using 450KHumanMethylation and Infinium Omni Express Exome arrays (Illumina, San Diego, CA). Coprimary objectives were to determine whether methylation alterations may predict clinical disease recurrence; and to assess whether the epigenetic alterations previously reported in newly diagnosed IBD were present in the patients with CD recruited into the TOPPIC study. Differential methylation and variance analysis was performed comparing patients with and without clinical evidence of recurrence. Secondary analyses included investigation of methylation associations with smoking, genotype (MeQTLs), and chronologic age. Validation of our previously published case-control observation of the methylome was performed using historical control data (CD, n = 123; Control, n = 198). RESULTS CD recurrence in patients following surgery is associated with 5 differentially methylated positions (Holm P < .05), including probes mapping to WHSC1 (P = 4.1 × 10-9, Holm P = .002) and EFNA3 (P = 4.9 × 10-8, Holm P = .02). Five differentially variable positions are demonstrated in the group of patients with evidence of disease recurrence including a probe mapping to MAD1L1 (P = 6.4 × 10-5). DNA methylation clock analyses demonstrated significant age acceleration in CD compared with control subjects (GrimAge + 2 years; 95% confidence interval, 1.2-2.7 years), with some evidence for accelerated aging in patients with CD with disease recurrence following surgery (GrimAge +1.04 years; 95% confidence interval, -0.04 to 2.22). Significant methylation differences between CD cases and control subjects were seen by comparing this cohort in conjunction with previously published control data, including validation of our previously described differentially methylated positions (RPS6KA2 P = 1.2 × 10-19, SBNO2 = 1.2 × 10-11) and regions (TXK [false discovery rate, P = 3.6 × 10-14], WRAP73 [false discovery rate, P = 1.9 × 10-9], VMP1 [false discovery rate, P = 1.7 × 10-7], and ITGB2 [false discovery rate, P = 1.4 × 10-7]). CONCLUSIONS We demonstrate differential methylation and differentially variable methylation in patients developing clinical recurrence within 3 years of surgery. Moreover, we report replication of the CD-associated methylome, previously characterized only in adult and pediatric inception cohorts, in patients with medically refractory disease needing surgery.
Collapse
Affiliation(s)
- Nicholas T Ventham
- Centre for Genomic and Experimental Medicine, The University of Edinburgh, Edinburgh, Midlothian, United Kingdom.
| | - Nicholas A Kennedy
- Centre for Genomic and Experimental Medicine, The University of Edinburgh, Edinburgh, Midlothian, United Kingdom
| | - Rahul Kalla
- Centre for Genomic and Experimental Medicine, The University of Edinburgh, Edinburgh, Midlothian, United Kingdom
| | - Alex T Adams
- Centre for Genomic and Experimental Medicine, The University of Edinburgh, Edinburgh, Midlothian, United Kingdom
| | - Alexandra Noble
- Centre for Genomic and Experimental Medicine, The University of Edinburgh, Edinburgh, Midlothian, United Kingdom
| | - Holly Ennis
- Centre for Genomic and Experimental Medicine, The University of Edinburgh, Edinburgh, Midlothian, United Kingdom
| | - Craig Mowat
- Centre for Genomic and Experimental Medicine, The University of Edinburgh, Edinburgh, Midlothian, United Kingdom
| | - Malcolm G Dunlop
- Centre for Genomic and Experimental Medicine, The University of Edinburgh, Edinburgh, Midlothian, United Kingdom
| | - Jack Satsangi
- Centre for Genomic and Experimental Medicine, The University of Edinburgh, Edinburgh, Midlothian, United Kingdom
| |
Collapse
|
24
|
Kurolap A, Baris Feldman H. Special issue: the genetics of early onset inflammatory bowel disease (IBD) and diarrheal disorders. Hum Genet 2023; 142:595-597. [PMID: 37106128 DOI: 10.1007/s00439-023-02566-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Affiliation(s)
- Alina Kurolap
- The Genetics Institute and Genomics Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.
| | - Hagit Baris Feldman
- The Genetics Institute and Genomics Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|