1
|
Turpin W, Lee SH, Croitoru K. Gut Microbiome Signature in Predisease Phase of Inflammatory Bowel Disease: Prediction to Pathogenesis to Prevention. Gastroenterology 2025; 168:902-913. [PMID: 39914464 DOI: 10.1053/j.gastro.2025.01.004] [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: 07/05/2024] [Revised: 01/02/2025] [Accepted: 01/08/2025] [Indexed: 03/23/2025]
Abstract
Advances in understanding the pathogenesis of inflammatory bowel disease (IBD) point toward a key role of the gut microbiome. We review the data describing the changes in the gut microbiome from IBD case-control studies and compare these findings with emerging data from studies of the preclinical phase of IBD. What is apparent is that assessing changes in the composition and function of the gut microbiome during the preclinical phase helps address confounding factors, such as disease activity and drug therapy, which can directly influence the gut microbiome. Understanding these changes in the predisease phase provides a means of predicting IBD in high-risk populations and offers insights into possible mechanisms involved in disease pathogenesis. Finally, we discuss strategies to use this information to design interventions aimed at modulating the microbiome as a means of preventing or delaying the onset of IBD.
Collapse
Affiliation(s)
- Williams Turpin
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Sun-Ho Lee
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Kenneth Croitoru
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Kotagiri P, Rae WM, Bergamaschi L, Pombal D, Lee JY, Noor NM, Sojwal RS, Rubin SJS, Unger LW, Tolmeijer SH, Manferrari G, Bashford-Rogers RJM, Bingham DB, Stift A, Rogalla S, Gubatan J, Lee JC, Smith KGC, McKinney EF, Boyd SD, Lyons PA. Disease-specific B cell clones are shared between patients with Crohn's disease. Nat Commun 2025; 16:3689. [PMID: 40246842 PMCID: PMC12006383 DOI: 10.1038/s41467-025-58977-y] [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: 01/25/2023] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
B cells have important functions in gut homeostasis, and dysregulated B cell populations are frequently observed in patients with inflammatory bowel diseases, including both ulcerative colitis (UC) and Crohn's disease (CD). How these B cell perturbations contribute to disease remains largely unknown. Here, we perform deep sequencing of the B cell receptor (BCR) repertoire in four cohorts of patients with CD, together with healthy controls and patients with UC. We identify BCR clones that are shared between patients with CD but not found in healthy individuals nor in patients with UC, indicating CD-associated B cell immune responses. Shared clones are present in the inflamed gut mucosa, draining intestinal lymph nodes and blood, suggesting the presence of common CD-associated antigens that drive B cell responses in CD patients.
Collapse
Affiliation(s)
- Prasanti Kotagiri
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK.
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA.
| | - William M Rae
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Discovery Sciences, AstraZeneca, Cambridge Biomedical Campus, Cambridge, UK
| | - Laura Bergamaschi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Diana Pombal
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Ji-Yeun Lee
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Nurulamin M Noor
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Raoul S Sojwal
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - Samuel J S Rubin
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - Lukas W Unger
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Sofie H Tolmeijer
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Giulia Manferrari
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Rachael J M Bashford-Rogers
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Department of Biochemistry, South Parks Road, University of Oxford, Oxford, OX1 3QU, UK
| | - David B Bingham
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Anton Stift
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Stephan Rogalla
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - John Gubatan
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - James C Lee
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- The Francis Crick Institute and UCL Institute of Liver and Digestive Health, Division of Medicine, Royal Free Campus, London, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Eoin F McKinney
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Scott D Boyd
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK.
| |
Collapse
|
3
|
Direksunthorn T, T Ahmed A, Pluetrattanabha N, Uthirapathy S, Ballal S, Singh A, Al-Hetty HRAK, Devi A, Sharma GC, Yumashev A. Ferroptosis in immune chaos: Unraveling its impact on disease and therapeutic potential. J Physiol Biochem 2025:10.1007/s13105-025-01078-7. [PMID: 40237936 DOI: 10.1007/s13105-025-01078-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025]
Abstract
Since its introduction in 2012, ferroptosis has garnered significant attention from researchers over the past decade. Unlike autophagy and apoptosis, ferroptosis is an atypical iron-dependent programmed cell death that falls under necrosis. It is regulated by various cellular metabolic and signaling processes, which encompass amino acid, lipid, iron, and mitochondrial metabolism. The initiation of ferroptosis occurs through iron-dependent phospholipid peroxidation. Notably, ferroptosis exhibits a dual effect and is associated with various diseases. A significant challenge lies in managing autoimmune disorders with unknown origins that stem from the reactivation of the immune system. Two contributing factors to autoimmunity are the aberrant stimulation of cell death and the inadequate clearance of dead cells, which can expose or release intracellular components that activate the immune response. Ferroptosis is distinct from other forms of cell death, such as apoptosis, necroptosis, autophagy, and pyroptosis, due to its unique morphological, biochemical, and genetic characteristics and specific relationship with cellular iron levels. Recent studies indicate that immune cells can both induce and undergo ferroptosis. To better understand how ferroptosis influences immune responses and its imbalance in disease, a molecular understanding of the relationship between ferroptosis and immunity is essential. Consequently, further research is needed to develop immunotherapeutics that target ferroptosis. This review primarily focuses on the role of ferroptosis in immune-related disorders.
Collapse
Affiliation(s)
| | | | | | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | | | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
4
|
Zhou Z, Su J, van Os BW, Plug LG, de Jonge-Muller ESM, Brands L, Janson SGT, van de Beek LM, van der Meulen-de Jong AE, Hawinkels LJAC, Barnhoorn MC. Stromal Cell Subsets Show Model-Dependent Changes in Experimental Colitis and Affect Epithelial Tissue Repair and Immune Cell Activation. Inflamm Bowel Dis 2025; 31:1051-1066. [PMID: 40100003 PMCID: PMC11985400 DOI: 10.1093/ibd/izae255] [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: 04/04/2024] [Indexed: 03/20/2025]
Abstract
BACKGROUND Previous work on inflammatory bowel disease (IBD) revealed changes in the abundance of colonic stromal subsets during intestinal inflammation. However, it is currently unknown whether these stromal cell subset changes are also reflected in different IBD mouse models and how commonly used IBD therapies affect stromal cell subset composition. METHODS Stromal subset markers CD55, C-X-C motif chemokine 12 (CXCL12), podoplanin (PDPN), CD90, and CD73 were analyzed by flow cytometry in 3 mouse models for IBD, namely interleukin (IL)-10 knockout (KO), dextran sulfate sodium-induced, and T-cell transfer model for colitis. Next, the effects of IBD therapies on the stromal subset composition were studied. In vitro experiments were performed to study the interaction between stromal cell subsets and epithelial/immune cells. RESULTS The colitis-induced changes in the abundance of stromal cell subsets differed considerably between the 3 colitis mouse models. Interestingly, treatment with IBD medication affected specific stromal subsets in a therapy and model-specific manner. In vitro experiments showed that specific stromal subsets affected epithelial wound healing and/or T-cell activation. CONCLUSIONS The relative abundance changes of stromal cell subsets during experimental colitis differ between 3 established colitis models. Treatment with IBD therapies influences stromal subset abundance, indicating their importance in IBD pathogenesis, possibly through affecting epithelial migration, and T-cell activation.
Collapse
Affiliation(s)
- Zhou Zhou
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Jie Su
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Bram W van Os
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Leonie G Plug
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | | | | | - Stef G T Janson
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | | | - Andrea E van der Meulen-de Jong
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Lukas J A C Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Marieke C Barnhoorn
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| |
Collapse
|
5
|
Golob JL, Hou G, Swanson BJ, Berinstein JA, Bishu S, Grasberger H, Zataari ME, Lee A, Kao JY, Kamada N, Bishu S. Inflammation-Induced Th17 Cells Synergize with the Inflammation-Trained Microbiota to Mediate Host Resiliency Against Intestinal Injury. Inflamm Bowel Dis 2025; 31:1082-1094. [PMID: 39851236 DOI: 10.1093/ibd/izae293] [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: 06/06/2024] [Indexed: 01/26/2025]
Abstract
BACKGROUND AND AIMS Inflammation can generate pathogenic Th17 cells and cause an inflammatory dysbiosis. In the context of inflammatory bowel disease (IBD), these inflammatory Th17 cells and dysbiotic microbiota may perpetuate injury to intestinal epithelial cells. However, many models of IBD like T-cell transfer colitis and IL-10-/- mice rely on the absence of regulatory pathways, so it is difficult to tell if inflammation can also induce protective Th17 cells. METHODS We subjected C57BL6, RAG1-/-, or JH-/- mice to systemic or gastrointestinal (GI) Citrobacter rodentium (Cr). Mice were then subjected to 2.5% dextran sodium sulfate (DSS) to cause epithelial injury. Fecal microbiota transfer was performed by bedding transfer and co-housing. Flow cytometry, qPCR, and histology were used to assess mucosal and systemic immune responses, cytokines, and tissue inflammation. 16s sequencing was used to assess gut bacterial taxonomy. RESULTS Transient inflammation with GI but not systemic Cr was protective against subsequent intestinal injury. This was replicated with sequential DSS collectively indicating that transient inflammation provides tissue-specific protection. Inflammatory Th17 cells that have a tissue-resident memory (TRM) signature expanded in the intestine. Experiments with reconstituted RAG1-/-, JH-/- mice, and cell trafficking inhibitors showed that inflammation-induced Th17 cells were required for protection. Fecal microbiota transfer showed that the inflammation-trained microbiota was necessary for protection, likely by maintaining protective Th17 cells in situ. CONCLUSION Inflammation can generate protective Th17 cells that synergize with the inflammation-trained microbiota to provide host resiliency against subsequent injury, indicating that inflammation-induced Th17 TRM T cells are heterogenous and contain protective subsets.
Collapse
Affiliation(s)
- Jonathan L Golob
- Division of Infectious Diseases, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Guoqing Hou
- Division of Gastroenterology, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Benjamin J Swanson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 42 and Emile, Omaha, NE 68198, USA
| | - Jeffrey A Berinstein
- Division of Gastroenterology, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Shreenath Bishu
- Laboratory and Pathology Diagnostics LLC, 1220 Hobson Road, Suite 244, Naperville, IL 60540, USA
| | - Helmut Grasberger
- Division of Gastroenterology, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Mohamed El Zataari
- Division of Gastroenterology, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Allen Lee
- Division of Infectious Diseases, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - John Y Kao
- Division of Gastroenterology, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Nobuhiko Kamada
- Division of Gastroenterology, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Shrinivas Bishu
- Division of Gastroenterology, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
Aljabri A, Soliman GM, Ramadan YN, Medhat MA, Hetta HF. Biosimilars versus biological therapy in inflammatory bowel disease: challenges and targeting strategies using drug delivery systems. Clin Exp Med 2025; 25:107. [PMID: 40186719 PMCID: PMC11972199 DOI: 10.1007/s10238-025-01558-6] [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: 10/02/2024] [Accepted: 01/03/2025] [Indexed: 04/07/2025]
Abstract
Inflammatory bowel disease (IBD) is a multifactorial illness with a climbing prevalence worldwide. While biologics are commonly prescribed especially for severe cases, they may worsen patients' outcomes due to financial burden. Consequently, there has been an increased focus on biosimilars to improve overall disease outcomes by maintaining similar efficacy and safety while minimizing the cost of therapy. Infliximab-dyyb was the first biosimilar approved by US-FDA for IBD. Since that, the US-FDA approved 14 biosimilars with different mechanisms of action and different routes of administration for IBD patients (four infliximab biosimilars, nine adalimumab biosimilars, and most recently one ustekinumab biosimilar). It should be noted that more biologics are in the pipeline as golimumab and natalizumab patents are set to expire in the near future, and biosimilars are now in pre-clinical to phase 3 trials. Different studies have evaluated biologics' effectiveness and safety and concluded that the majority of available biosimilars are efficacious and have similar adverse effect profiles compared to their reference biologics. It is worth mentioningthat post-marketing surveillance reports revealed some risks associated with biosimilars which should be taken into consideration in future research and clinical trials to avoid health hazards. Most biologics and biosimilars are administered parenterally which results in several drawbacks such as raised risk of infections, hypersensitivity, autoimmunity, development of malignancies, liver toxicity as well as worsening of heart failure. Several drug delivery systems based on passive and active targeting mechanisms are under active investigation to overcome these limitations. This review sheds light on the emergence of biologics and biosimilars as alternatives in IBD management, the differences between them, challenges and risks, and future perspectives in IBD therapy and new trends in drug delivery systems.
Collapse
Affiliation(s)
- Ahmed Aljabri
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Ghareb M Soliman
- Department of Pharmaceutics, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Yasmin N Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut, 71515, Egypt.
| | - Mohammed A Medhat
- Department of Tropical Medicine and Gastroenterology, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Helal F Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| |
Collapse
|
7
|
Raimondi D, Verplaetse N, Passemiers A, Jans DS, Cleynen I, Moreau Y. Genomic prediction with kinship-based multiple kernel learning produces hypothesis on the underlying inheritance mechanisms of phenotypic traits. Genome Biol 2025; 26:84. [PMID: 40181452 PMCID: PMC11969835 DOI: 10.1186/s13059-025-03544-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 03/17/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Genomic prediction encompasses the techniques used in agricultural technology to predict the genetic merit of individuals towards valuable phenotypic traits. It is related to Genome Interpretation in humans, which models the individual risk of developing disease traits. Genomic prediction is dominated by linear mixed models, such as the Genomic Best Linear Unbiased Prediction (GBLUP), which computes kinship matrices from SNP array data, while Genome Interpretation applications to clinical genetics rely mainly on Polygenic Risk Scores. RESULTS In this article, we exploit the positive semidefinite characteristics of the kinship matrices that are conventionally used in GBLUP to propose a novel Genomic Multiple Kernel Learning method (GMKL), in which the multiple kinship matrices corresponding to Additive, Dominant, and Epistatic Inheritance Mechanisms are used as kernels in support vector machines, and we apply it to both worlds. We benchmark GMKL on simulated cattle phenotypes, showing that it outperforms the classical GBLUP predictors for genomic prediction. Moreover, we show that GMKL ranks the kinship kernels representing different inheritance mechanisms according to their compatibility with the observed data, allowing it to produce hypotheses on the normally unknown inheritance mechanisms generating the target phenotypes. We then apply GMKL to the prediction of two inflammatory bowel disease cohorts with more than 6500 samples in total, consistently obtaining results suggesting that epistasis might have a relevant, although underestimated role in inflammatory bowel disease (IBD). CONCLUSIONS We show that GMKL performs similarly to GBLUP, but it can formulate biological hypotheses about inheritance mechanisms, such as suggesting that epistasis influences IBD.
Collapse
Affiliation(s)
- Daniele Raimondi
- Institut de Génétique Moléculaire de Montpellier (IGMM), CNRS-UMR5535, Université de Montpellier, Montpellier, 34293, France.
- ESAT-STADIUS, KU Leuven, Leuven, 3001, Belgium.
| | | | | | | | | | - Yves Moreau
- ESAT-STADIUS, KU Leuven, Leuven, 3001, Belgium
| |
Collapse
|
8
|
Yu Y, Tong K, Deng J, Wu J, Guo C. Causal effects of various particulate matter on inflammatory bowel disease and its subtypes: insights from Mendelian randomization. INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2025; 69:849-860. [PMID: 39918593 DOI: 10.1007/s00484-025-02862-2] [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: 11/05/2024] [Revised: 01/02/2025] [Accepted: 01/26/2025] [Indexed: 03/27/2025]
Abstract
The role of particulate matter (PM) on genetic susceptibility to inflammatory bowel disease (IBD) is a controversial topic. The study aims to evaluate the causal effect of PM on IBD using Mendelian randomization (MR). PM and IBD datasets were screened for common single nucleotide polymorphisms (SNPs) based on the MR basic assumptions. Subsequently, the effect of these SNPs on genetic susceptibility to IBD was analyzed using inverse variance weighted. Finally, the heterogeneity and robustness of the results were assessed using Cochran's Q and leave-one-out sensitivity analysis, respectively. MR analysis revealed that PM2.5 was linked to a heightened genetic predisposition to (odds ratio [OR] 1.530, 95% confidence interval [CI] 1.100-2.128, p = 0.011) and ulcerative colitis (UC) (OR 1.675, 95% CI 1.129-2.485, p = 0.010), but not to Crohn's disease (CD) (OR 1.685, 95% CI 0.883-3.216, p = 0.114). PM10 was not associated with increased genetic susceptibility to IBD (OR 1.164, 95% CI 0.650-2.083, p = 0.610), UC (OR 1.439, 95% CI 0.691-2.996, p = 0.331), or CD (OR 0.825, 95% CI 0.265-2.564, p = 0.739). MR-Egger intercept did not indicate any horizontal pleiotropy (p > 0.05). Cochran's Q revealed no evidence of heterogeneity (p > 0.05). Leave-one-out sensitivity analysis confirmed the robustness of the results. The MR analysis demonstrated that PM2.5 increased genetic susceptibility to UC, while no such association was observed for CD. Furthermore, PM10 showed no association with genetic susceptibility to IBD. Therefore, implementing protective measures, such as air purifiers and anti-haze masks, may help reduce the risk of UC.
Collapse
Affiliation(s)
- Yunfeng Yu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Keke Tong
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Juan Deng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jingyi Wu
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310005, China
| | - Chenlu Guo
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China.
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
9
|
Ronca V, Gerussi A, Collins P, Parente A, Oo YH, Invernizzi P. The liver as a central "hub" of the immune system: pathophysiological implications. Physiol Rev 2025; 105:493-539. [PMID: 39297676 DOI: 10.1152/physrev.00004.2023] [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: 05/18/2023] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 01/16/2025] Open
Abstract
The purpose of this review is to describe the immune function of the liver, guiding the reader from the homeostatic tolerogenic status to the aberrant activation demonstrated in chronic liver disease. An extensive description of the pathways behind the inflammatory modulation of the healthy liver will be provided focusing on the complex immune cell network residing within the liver. The limit of tolerance will be presented in the context of organ transplantation, seizing the limits of homeostatic mechanisms that fail in accepting the graft, progressing eventually toward rejection. The triggers and mechanisms behind chronic activation in metabolic liver conditions and viral hepatitis will be discussed. The last part of the review will be dedicated to one of the greatest paradoxes for a tolerogenic organ, developing autoimmunity. Through the description of the three most common autoimmune liver diseases, the autoimmune reaction against hepatocytes and biliary epithelial cells will be dissected.
Collapse
Affiliation(s)
- Vincenzo Ronca
- Centre for Liver and Gastro Research and National Institute for Health and Care Research (NIHR) Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Liver Unit, Queen Elizabeth Hospital University Hospital Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Rare Diseases, European Reference Network Centre-Rare Liver, Birmingham, United Kingdom
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Milan, Italy
| | - Alessio Gerussi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), IRCCS Fondazione San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Paul Collins
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Alessandro Parente
- Liver Unit, Queen Elizabeth Hospital University Hospital Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Ye Htun Oo
- Centre for Liver and Gastro Research and National Institute for Health and Care Research (NIHR) Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Liver Unit, Queen Elizabeth Hospital University Hospital Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Rare Diseases, European Reference Network Centre-Rare Liver, Birmingham, United Kingdom
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), IRCCS Fondazione San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
10
|
Kaden T, Alonso‐Román R, Stallhofer J, Gresnigt MS, Hube B, Mosig AS. Leveraging Organ-on-Chip Models to Investigate Host-Microbiota Dynamics and Targeted Therapies for Inflammatory Bowel Disease. Adv Healthc Mater 2025; 14:e2402756. [PMID: 39491534 PMCID: PMC12004439 DOI: 10.1002/adhm.202402756] [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: 07/25/2024] [Revised: 09/29/2024] [Indexed: 11/05/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic gastrointestinal disease with drastically increasing incidence rates. Due to its multifactorial etiology, a precise investigation of the pathogenesis is extremely difficult. Although reductionist cell culture models and more complex disease models in animals have clarified the understanding of individual disease mechanisms and contributing factors of IBD in the past, it remains challenging to bridge research and clinical practice. Conventional 2D cell culture models cannot replicate complex host-microbiota interactions and stable long-term microbial culture. Further, extrapolating data from animal models to patients remains challenging due to genetic and environmental diversity leading to differences in immune responses. Human intestine organ-on-chip (OoC) models have emerged as an alternative in vitro model approach to investigate IBD. OoC models not only recapitulate the human intestinal microenvironment more accurately than 2D cultures yet may also be advantageous for the identification of important disease-driving factors and pharmacological interventions targets due to the possibility of emulating different complexities. The predispositions and biological hallmarks of IBD focusing on host-microbiota interactions at the intestinal mucosal barrier are elucidated here. Additionally, the potential of OoCs to explore microbiota-related therapies and personalized medicine for IBD treatment is discussed.
Collapse
Affiliation(s)
- Tim Kaden
- Dynamic42 GmbH07745JenaGermany
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
| | - Raquel Alonso‐Román
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | | | - Mark S. Gresnigt
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | - Bernhard Hube
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Institute of MicrobiologyFaculty of Biological SciencesFriedrich Schiller University07743JenaGermany
| | - Alexander S. Mosig
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
| |
Collapse
|
11
|
Shao X, Chen J, Zhu C, Li J, Huang C. Analysis of "Inflammatory Markers at Birth and Risk of Early-Onset Inflammatory Bowel Disease". Gastroenterology 2025; 168:833-834. [PMID: 39710339 DOI: 10.1053/j.gastro.2024.10.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 12/24/2024]
Affiliation(s)
- Xi Shao
- Department of Anorectal, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Jun Chen
- Department of Radiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Cuirong Zhu
- Department of Anorectal, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Jun Li
- Department of Anorectal, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Chengzi Huang
- Department of Anorectal, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China; Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
12
|
Sobhani I, De Oliveira Alves N, Sadeghi M, Charpy C, Bergsten E, Amiot A, Barau C, Brunetti F, Vaysse A, Tournigand C, Chamaillard M, Khashayarsha K, Mestivier D. Poor prognosis in IBD-complicated colon cancer through gut dysbiosis-related immune response failure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645177. [PMID: 40196693 PMCID: PMC11974894 DOI: 10.1101/2025.03.25.645177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background Colorectal cancer (CRC) results from the accumulation of mutations and epigenetic changes in gut epithelial cells likely due to gut microbiota dysbiosis. However, limited research has been done to explore the link between host tumour dysbiosis and disease outcome. Methods The mechanisms influencing outcomes of 97 colorectal cancer (CRC) patients, including 13 with Lynch syndrome, 20 with inflammatory bowel disease (IBD), and 64 sporadic cases, were analyzed using a multiomics approach. These patients were categorized into two groups: "disease-free/stable disease" and "progression disease" survival outcomes. The analysis included tumor adherent microbiota composition (16S rRNA), somatic gene mutations (WES), gene expression (RNAseq), immune markers (RNAscope), and immune infiltrate cells (immunohistochemistry). Results IBD-CRC patients had worse outcomes than those with Lynch or sporadic CRC, regardless of TNM staging or treatment. Symbiotic bacteria like Lactococcus lactis were significantly reduced in IBD-CRC tissues. Patient outcomes were influenced by the abundance of virulent ( Escherichia coli ) relative to beneficial bacteria ( Lactococcus lactis ). Although no significant increase in deleterious somatic mutations was found in IBD-CRC. 16sRNA revealed increased virulent- and decreased anti-inflammatory symbiotic-bacteria correlating with the upregulation of oncogenes and downregulation of anti-oncogenes like PHLPP1. The multiplex in situ hybridization of CD8, IFNγ and PHLPP1 an anti-oncogene revealed significant decrease of immune cells with detectable PHLPP1 expression in IBD-CRC tumour tissues as compared to sporadic CRCs. Conclusion The poor outcomes in IBD-CRC patients are likely due to gut dysbiosis and immune cell alterations, possibly triggered by microbiota-related epigenetic pathways. What You Need to Know BACKGROUND AND CONTEXT: Colorectal cancer (CRC) is associated with gut microbiota dysbiosis. Inflammatory bowel disease-related CRC (IBD-CRC) is classified as an environment-related condition.NEW FINDINGS: In relation with patient outcomes, tumour tissues from three types of CRC (Sporadic-, IBD-, and Lynch syndrome-CRC) were analyzed using a multiomic approach. This included examining tissue adherent virulent bacteria, gene analyses, and quantifying immune cell infiltration in the mucosa. IBD-CRC patients had the worst outcomes, associated with the down regulation of PHLPP1 gene, virulent/symbiotic imbalance, and immune response failure.LIMITATIONS: Lack of animal experiments using FMT of fresh stool from IBD-CRC patients.CLINICAL AND TRANSLATIONAL RESEARCH RELEVANCE: Among the different types of CRC, IBD-CRC patients showed a greater imbalance between harmful and beneficial bacteria, along with immune response failure.Lay summary: This study compares the pathological and clinical characteristics of patients with colorectal cancer (CRC) across three distinct etiologies: sporadic CRC, inflammatory bowel disease (IBD)-associated CRC, and Lynch syndrome-associated CRC (LS-CRC). Distinct differences in tumor-adherent microbiota, gene expression and immune response profiles were observed. Notably, IBD-CRC patients demonstrated the poorest prognosis depending on microbe-host gene interaction highlighting potential biomarkers for disease prognosis and treatment strategies.
Collapse
|
13
|
Humeidi R, Oshiro-Rapley N, Gu X, An JS, Ananthakrishnan AN, Creasey EA, Daly MJ, Schreiber SL, Graham DB, Seyedsayamdost MR, Xavier RJ. The Ulcerative Colitis-Associated Gene NXPE1 Catalyzes Glycan Modifications on Colonic Mucin. J Am Chem Soc 2025; 147:10618-10628. [PMID: 40067145 DOI: 10.1021/jacs.5c00769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Colonic mucus forms a first line of defense against bacterial invasion while providing nutrition to support coinhabiting microbes in the gut. Mucus is composed of polymeric networks of mucin proteins, which are heavily modified post-translationally. The full compendium of enzymes responsible for these modifications and their roles in health and disease remain incompletely understood. Herein, we determine the biochemical function of NXPE1, a gene implicated in ulcerative colitis (UC), and demonstrate that it encodes an acetyltransferase that modifies mucin glycans. Specifically, NXPE1 utilizes acetyl-CoA to regioselectively modify the mucus sialic acid, 5-N-acetylneuraminic acid (Neu5Ac), at the 9-OH group to generate 9-O-acetylated Neu5Ac (Neu5,9Ac2). We further demonstrate that colonic organoids derived from donors harboring the missense variant NXPE1 G353R, which is protective against UC, exhibit severely impaired acetylation of Neu5Ac on mucins. Together, our findings support a model in which NXPE1 masks the alcohols of mucus sialoglycans via acetylation, which is important for modulating mucus barrier properties that limit interactions with commensal microbes.
Collapse
Affiliation(s)
- Ranad Humeidi
- Program for Chemistry & Chemical Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, United States
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Noriko Oshiro-Rapley
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, United States
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Xiebin Gu
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, United States
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Joon Soo An
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Ashwin N Ananthakrishnan
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114, United States
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Elizabeth A Creasey
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Mark J Daly
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, United States
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Stuart L Schreiber
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, United States
- Department of Chemistry & Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Daniel B Graham
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, United States
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
- Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Mohammad R Seyedsayamdost
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, United States
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
- Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
14
|
Hume DA, Summers KM, O'Brien C, Pavli P. The Relationship Between CSF1R Signaling, Monocyte-Macrophage Differentiation, and Susceptibility to Inflammatory Bowel Disease. Cell Mol Gastroenterol Hepatol 2025:101510. [PMID: 40154882 DOI: 10.1016/j.jcmgh.2025.101510] [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: 10/04/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
More than 300 genomic loci have been associated with increased susceptibility to inflammatory bowel disease (IBD) through genome-wide association studies. A major challenge in the translation of genome-wide association studies to mechanistic insights lies in connecting noncoding variants to function. For example, single-nucleotide variants (SNVs) in the vicinity of the gene encoding the transcription factor ETS2 on human chromosome 21 are associated with the risk of developing IBD in Europeans. The peak of SNV association lies within a distal enhancer that may regulate ETS2 transcription. The interpretation of this and many other SNV associations with IBD depends on a model linking variation in transcriptional regulation to the likelihood of developing chronic intestinal inflammation. One model for the ETS2 locus is that overexpression in monocytes is causally associated with the risk allele, which in turn leads to a hyperinflammatory state. Here we summarize evidence for an alternative mechanism focused on negative regulators of monocyte-macrophage activation. We argue that IBD susceptibility arises from dysregulation of monocyte adaptation in the intestinal milieu to form resident intestinal macrophages that are anergic to inflammatory stimuli. This process depends on signals initiated by macrophage colony-stimulating factor (CSF1) binding to its receptor (CSF1R). Within this framework, ETS2 is a myeloid-specific transcription factor, expressed in pluripotent and committed progenitors and monocytes, and is down-regulated by CSF1, in common with many genes associated with IBD susceptibility, including NOD2. ETS2 is also both a downstream target and a mediator of the CSF1/CSF1R signaling pathway. Therapeutic targeting of ETS2 and its upstream regulators has the potential to prevent CSF1-dependent monocyte differentiation toward a prorepair resident macrophage phenotype and consequently exacerbate intestinal inflammation.
Collapse
Affiliation(s)
- David A Hume
- Mater Research Institute, University of Queensland, Woolloongabba, Brisbane, Australia.
| | - Kim M Summers
- Mater Research Institute, University of Queensland, Woolloongabba, Brisbane, Australia
| | - Claire O'Brien
- Centre for Research in Therapeutics Solutions, Faculty of Science and Technology, University of Canberra, Canberra, Australian Capital Territory, Australia
| | - Paul Pavli
- School of Medicine and Psychology, The Australian National University, Canberra, Australian Capital Territory, Australia; Gastroenterology and Hepatology Unit, Canberra Hospital, Canberra, Australian Capital Territory, Australia.
| |
Collapse
|
15
|
Wang Q, Zhang W, Liu J, Qin W, Cai J. Exopolysaccharide of Levilactobacillus brevis M-10 Improved Physiological and Biochemical Indicators and Gut Microbiota in DSS-Induced Colitis Mice. Curr Microbiol 2025; 82:204. [PMID: 40126646 DOI: 10.1007/s00284-025-04190-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 03/13/2025] [Indexed: 03/26/2025]
Abstract
Ulcerative colitis (UC) is a typical type of inflammatory bowel disease, which is often recurrent and directly related with colorectal cancer. Therefore, early prevention and treatment for UC is very necessary. Therefore, it is necessary to find efficient substances to treat the UC with less side effects than drugs. Exopolysaccharides (EPSs) are important bioactive constituents of lactic acid bacteria. The study evaluated the effects of EPS1 produced by Levilactobacillus brevis M-10 on UC by determining the weight, the disease activity index (DAI) and the physiological and biochemical indexes. The pathological structures of colon were observed. The gut microbiota and the short-chain fatty acids were analyzed. The results demonstrated high dose (HD) of EPS1 (400 mg/kg-BW) that had the best effects on UC mice. The HD group restored body weight, decreased DAI and alleviated shortening of the length of the colon, recovered liver tissue, declined lipopolysaccharide, and myeloperoxidase. Also the HD group showed that the expression of tight junction proteins increased, IL-10 up-regulated, IL-6, IL-1β, and TNF-α down-regulated, and the gut microbiota dysbiosis balanced. The HD group markedly elevated the relative abundance of Lachnospiraceae_NK4A136_group, Unclassified-Lachnospiraceae, and Unclassified- Muribaculaceae. Acetic acid, propionic acid, and n-butyric acid were significantly increased in the HD group (P < 0.05). The study could provide a theoretical basis and material support for the exploration of safe functional food in alleviating and preventing UC.
Collapse
Affiliation(s)
- Qi Wang
- School of Life Science, Shanxi University, Taiyuan, 030006, Shanxi, China.
| | - Wenwen Zhang
- School of Life Science, Shanxi University, Taiyuan, 030006, Shanxi, China
| | - Jiaqin Liu
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, 010110, Inner Mongolia, China
| | - Wenjun Qin
- Nutritional department, Shanxi Hospital of Traditional Chinese Medicine, Taiyuan, 030012, Shanxi, China
| | - Jin Cai
- Institute of Applied Chemistry, Shanxi University, Taiyuan, 030006, Shanxi, China
| |
Collapse
|
16
|
Gabryel M, Zakerska-Banaszak O, Ladziak K, Hubert KA, Baturo A, Suszynska-Zajczyk J, Hryhorowicz M, Dobrowolska A, Skrzypczak-Zielinska M. Is a rare CXCL8 gene variant a new possible cause or curse factor of inflammatory bowel disease? Front Immunol 2025; 16:1562618. [PMID: 40176809 PMCID: PMC11961448 DOI: 10.3389/fimmu.2025.1562618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/26/2025] [Indexed: 04/04/2025] Open
Abstract
Introduction The pathogenesis of inflammatory bowel diseases (IBD) involves genetic, environmental, immunological, and microbial factors; however, it remains unclear. Pro-inflammatory interleukin 8 (IL-8), encoded by the CXCL8 gene, assumes a crucial chemotactic role in leukocyte migration. Methods This study aimed to investigate whether an association exists between IBD and two CXCL8 variants, namely, c.-251A>T (rs4073) and c.91G>T (rs188378669), and IL-8 concentration. We analyzed the distribution of both variants among 353 Polish IBD patients and 200 population subjects using pyrosequencing, competitive allele-specific PCR and Sanger sequencing. Results The c.91T stop-gained allele was significantly more frequent in IBD patients (2.12%) than in controls (0.25%) (p = 0.0121), while the c.-251T allele frequencies were similar (54% vs. 51.5%, p = 0.4955). Serum IL-8 concentrations, measured using ELISA, were higher in IBD patients with the c.91 GG genotype compared to healthy controls (mean, 70.02 vs. 51.5 pg/ml, p<0.01) and patients with c.91 GT (mean, 61.73 pg/ml). Moreover, clinical data indicated that carriers of the c.91T variant need more often corticosteroids and surgical treatment of the disease than GG homozygous IBD patients. Conclusion This suggest that the CXCL8 c.91T allele may influence IBD manifestation and the course of the disorders in Polish patients, potentially serving as a novel target for future studies and therapeutic approaches.
Collapse
Affiliation(s)
- Marcin Gabryel
- Department of Gastroenterology, Dietetics and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Karolina Ladziak
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | - Alina Baturo
- Department of Gastroenterology, Dietetics and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Joanna Suszynska-Zajczyk
- Department of Biochemistry and Biotechnology, Poznan University of Life Sciences, Poznan, Poland
| | - Magdalena Hryhorowicz
- Department of Biochemistry and Biotechnology, Poznan University of Life Sciences, Poznan, Poland
| | - Agnieszka Dobrowolska
- Department of Gastroenterology, Dietetics and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | | |
Collapse
|
17
|
Hébert-Milette I, Lévesque C, Paquette J, Rivard MÈ, Villeneuve L, Boucher G, Goyette P, Charron G, Rioux JD. Inflammatory bowel disease risk gene C1ORF106 regulates actin dynamics in intestinal epithelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643205. [PMID: 40161582 PMCID: PMC11952551 DOI: 10.1101/2025.03.14.643205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background and aims C1ORF106 has previously been associated with inflammatory bowel diseases (IBD) via large-scale genetic studies. Increased intestinal permeability is a hallmark of IBD and is observed in at-risk individuals prior to the appearance of clinical symptoms. C1ORF106 was previously shown to regulate intestinal barrier permeability through the regulation of adherens junction stability and through the formation of tight junctions, which impacted actin assembly. However, the downstream impact and molecular mechanisms involved in actin regulation by C1ORF106 haven't been explored. Our study aimed at identifying which pathways involved in intestinal epithelial barrier regulation and F-actin regulation are impacted by C1ORF106 and its IBD-associated variant. Methods We knocked down (KD) the expression of C1ORF106 in human colonic epithelial cells and characterized the function of the 333F variant in intestinal epithelial spheroid cultures obtained from patient-derived human induced pluripotent stem cell (hiPSC). We measured barrier permeability and characterized spheroid formation, actin regulation and cell migration though immunofluorescence, western blots and permeability assays. Results C1ORF106 KD leads to impaired cortical actin belt dynamics and regulation of stress fiber formation, resulting in increased cell constriction, impaired barrier permeability, cell polarity and cell migration. Moreover, we demonstrated that an inhibition of ROCK rescues the actin belt and cell polarity phenotypes in C1ORF106 KD cells, demonstrating that C1ORF106 regulates these phenotypes through a ROCK-dependent mechanism. We also observed an altered nmMYO2-P localization in C1ORF106 KD cells associated with the formation of Vacuolar Apical Compartments (VACs), which are important for 3D epithelial spheroid formation. We observed a similar impact on cell polarity in intestinal epithelial spheroids obtained from hiPSC carrying the 333F variant, providing additional support that this pathway is involved in disease development. Conclusion We provide insights into the molecular mechanisms by which C1ORF106 controls actin dynamics to regulate intestinal epithelial integrity.
Collapse
Affiliation(s)
- Isabelle Hébert-Milette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Chloé Lévesque
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - Jean Paquette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - Marie-Ève Rivard
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - Louis Villeneuve
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - Gabrielle Boucher
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - Philippe Goyette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - Guy Charron
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - John D. Rioux
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
18
|
Li Y, Ascui G, Dicker M, Riffelmacher T, Chandra V, Schmiedel B, Chou TF, Vijayanand P, Kronenberg M. Crohn's Disease-associated variant in laccase domain containing 1 (LACC1) modulates T cell gene expression, metabolism and T cell function. Nat Commun 2025; 16:2577. [PMID: 40089498 PMCID: PMC11910630 DOI: 10.1038/s41467-025-57744-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Genome wide association studies (GWAS) identify many risks for Crohn's disease (CD), including a site near the metabolism gene laccase domain containing 1 (LACC1). We previously found this site near LACC1 was associated with decreased LACC1 expression in T lymphocytes, yet the mechanism affecting gene expression and its links to T cell function and inflammatory disease were unknown. Here we identify variants in the promoter region that influence transcription of LACC1. Direct association of disease-risk variants with lower LACC1 pre-mRNA in human CD4+ T cells is confirmed by comparing transcripts from each allele from donors heterozygous for the LACC1 CD-risk allele. Using gene editing, we validate the function of this promoter region in LACC1 expression in T cells. Human CD4+ T cells with LACC1 gene knockdown show altered metabolism, including reduced oxygen consumption rate, and reduced in vitro regulatory T cell differentiation. Therefore, our study provides a mechanism linking these specific LACC1 variants to colitis by attributing promoter region variants to changes in T cell metabolism and function.
Collapse
Affiliation(s)
- Yingcong Li
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Gabriel Ascui
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | | | - Vivek Chandra
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | | | - Pandurangan Vijayanand
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Molecular and Clinical Cancer Medicine and NIHR and CRUK Liverpool Experimental Cancer Medicine Center, University of Liverpool, Liverpool, UK.
| | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
19
|
Chen HH, Highland HM, Frankel EG, Scartozzi AC, Zhang X, Roshani R, Sharma P, Kar A, Buchanan VL, Polikowsky HG, Petty LE, Seo J, Anwar MY, Kim D, Graff M, Young KL, Zhu W, Karastergiou K, Shaw DM, Justice AE, Fernández-Rhodes L, Krishnan M, Gutierrez A, McCormick PJ, Aguilar-Salinas CA, Tusié-Luna MT, Muñoz-Hernandez LL, Herrera-Hernandez M, Lee M, Gamazon ER, Cox NJ, Pajukanta P, Fried SK, Gordon-Larsen P, Shah RV, Fisher-Hoch SP, McCormick JB, North KE, Below JE. Multiomics reveal key inflammatory drivers of severe obesity: IL4R, LILRA5, and OSM. CELL GENOMICS 2025; 5:100784. [PMID: 40043711 PMCID: PMC11960538 DOI: 10.1016/j.xgen.2025.100784] [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: 04/25/2024] [Revised: 10/08/2024] [Accepted: 02/06/2025] [Indexed: 03/15/2025]
Abstract
Polygenic severe obesity (body mass index [BMI] ≥40 kg/m2) has increased, especially in Hispanic/Latino populations, yet we know little about the underlying mechanistic pathways. We analyzed whole-blood multiomics data to identify genes differentially regulated in severe obesity in Mexican Americans from the Cameron County Hispanic Cohort. Our RNA sequencing analysis identified 124 genes significantly differentially expressed between severe obesity cases (BMI ≥40 kg/m2) and controls (BMI <25 kg/m2); 33% replicated in an independent sample from the same population. Our integrative approach identified inflammatory genes, including IL4R, ZNF438, and LILRA5. Several genes displayed transcriptomic effects on severe obesity in subcutaneous adipose tissue. We further showed that the genetic regulation of these genes is associated with several traits in a large biobank, including bone fractures, obstructive sleep apnea, and hyperaldosteronism, illuminating potential risk mechanisms. Our findings furnish a molecular architecture of the severe obesity phenotype across multiple molecular domains.
Collapse
Affiliation(s)
- Hung-Hsin Chen
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Academia Sinica, Institute of Biomedical Sciences, Taipei, Taiwan
| | - Heather M Highland
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elizabeth G Frankel
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alyssa C Scartozzi
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xinruo Zhang
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rashedeh Roshani
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Priya Sharma
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Asha Kar
- Department of Human Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA
| | - Victoria L Buchanan
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hannah G Polikowsky
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lauren E Petty
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jungkyun Seo
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of MetaBiohealth, Sungkyunkwan University, Suwon, Republic of Korea
| | - Mohammad Yaser Anwar
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daeeun Kim
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kristin L Young
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wanying Zhu
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kalypso Karastergiou
- Obesity Research Center, Boston University School of Medicine, Boston, MA, USA; Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Douglas M Shaw
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anne E Justice
- Department of Population Health Services, Geisinger Health, Danville, PA, USA
| | | | - Mohanraj Krishnan
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Absalon Gutierrez
- Department of Internal Medicine, Division of Endocrinology, Diabetes, and Metabolism, Houston, TX, USA
| | - Peter J McCormick
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| | - Carlos A Aguilar-Salinas
- Unidad de Investigación de Enfermedades Metabólicas and Research Direction of the Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México; Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, México City, México
| | - Maria Teresa Tusié-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto de Investigaciones Biomédicas UNAM Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
| | - Linda Liliana Muñoz-Hernandez
- Unidad de Investigación de Enfermedades Metabólicas del Instituto Nacional de Ciencias, Médicas, y Nutrición Salvador Zubirán, México City, México
| | - Miguel Herrera-Hernandez
- Surgery Direction of the Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
| | - Miryoung Lee
- Department of Epidemiology, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX, USA
| | - Eric R Gamazon
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Nancy J Cox
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA; Institute for Precision Health at University of California, Los Angeles, Los Angeles, CA, USA
| | - Susan K Fried
- Obesity Research Center, Boston University School of Medicine, Boston, MA, USA; Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Penny Gordon-Larsen
- Department of Nutrition, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Susan P Fisher-Hoch
- Department of Epidemiology, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX, USA
| | - Joseph B McCormick
- Department of Epidemiology, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Jennifer E Below
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
20
|
Flanagan K, Gassner K, Lang M, Ozelyte J, Hausmann B, Crepaz D, Pjevac P, Gasche C, Berry D, Vesely C, Pereira FC. Human-derived microRNA 21 regulates indole and L-tryptophan biosynthesis transcripts in the gut commensal Bacteroides thetaiotaomicron. mBio 2025; 16:e0392824. [PMID: 39878512 PMCID: PMC11898669 DOI: 10.1128/mbio.03928-24] [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: 12/17/2024] [Accepted: 01/08/2025] [Indexed: 01/31/2025] Open
Abstract
In the gut, microRNAs (miRNAs) produced by intestinal epithelial cells are secreted into the lumen and can shape the composition and function of the gut microbiome. Crosstalk between gut microbes and the host plays a key role in irritable bowel syndrome (IBS) and inflammatory bowel diseases, yet little is known about how the miRNA-gut microbiome axis contributes to the pathogenesis of these conditions. Here, we investigate the ability of miR-21, a miRNA that we found decreased in fecal samples from IBS patients, to associate with and regulate gut microbiome function. When incubated with the human fecal microbiota, miR-21 revealed a rapid internalization or binding to microbial cells, which varied in extent across different donor samples. Fluorescence-activated cell sorting and sequencing of microbial cells incubated with fluorescently labeled miR-21 identified organisms belonging to the genera Bacteroides, Limosilactobacillus, Ruminococcus, or Coprococcus, which predominantly interacted with miR-21. Surprisingly, these and other genera also interacted with a miRNA scramble control, suggesting that physical interaction and/or uptake of these miRNAs by gut microbiota is not sequence-dependent. Nevertheless, transcriptomic analysis of the gut commensal Bacteroides thetaiotaomicron revealed a miRNA sequence-specific effect on bacterial transcript levels. Supplementation of miR-21, but not of small RNA controls, resulted in significantly altered levels of many cellular transcripts and increased transcription of a biosynthetic operon for indole and L-tryptophan, metabolites known to regulate host inflammation and colonic motility. Our study identifies a novel putative miR-21-dependent pathway of regulation of intestinal function through the gut microbiome with implications for gastrointestinal conditions. IMPORTANCE The mammalian gut represents one of the largest and most dynamic host-microbe interfaces. Host-derived microRNAs (miRNAs), released from the gut epithelium into the lumen, have emerged as important contributors to host-microbe crosstalk. Levels of several miRNAs are altered in the stool of patients with irritable bowel syndrome or inflammatory bowel disease. Understanding how miRNAs interact with and shape gut microbiota function is crucial as it may enable the development of new targeted treatments for intestinal diseases. This study provides evidence that the miRNA miR-21 can rapidly associate with diverse microbial cells form the gut and increase levels of transcripts involved in tryptophan synthesis in a ubiquitous gut microbe. Tryptophan catabolites regulate key functions, such as gut immune response or permeability. Therefore, this mechanism represents an unexpected host-microbe interaction and suggests that host-derived miR-21 may help regulate gut function via the gut microbiota.
Collapse
Affiliation(s)
- Kayla Flanagan
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Kirsten Gassner
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Michaela Lang
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Jurgita Ozelyte
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Bela Hausmann
- Joint Microbiome Facility, Medical University of Vienna and University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Division of Clinical Microbiology, Medical University of Vienna, Vienna, Austria
| | - Daniel Crepaz
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Petra Pjevac
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Joint Microbiome Facility, Medical University of Vienna and University of Vienna, Vienna, Austria
| | - Christoph Gasche
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - David Berry
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Joint Microbiome Facility, Medical University of Vienna and University of Vienna, Vienna, Austria
| | - Cornelia Vesely
- Center of Anatomy and Cell Biology, Division of Cell and Developmental Biology, Medical University of Vienna, Vienna, Austria
| | - Fatima C. Pereira
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
21
|
Noble A, Adams A, Nowak J, Cheng G, Nayak K, Quinn A, Kristiansen M, Kalla R, Ventham NT, Giachero F, Jayamanne C, Hansen R, Hold GL, El-Omar E, Croft NM, Wilson D, Beattie RM, Ashton JJ, Zilbauer M, Ennis S, Uhlig HH, Satsangi J. The Circulating Methylome in Childhood-Onset Inflammatory Bowel Disease. J Crohns Colitis 2025; 19:jjae157. [PMID: 39365013 PMCID: PMC11945304 DOI: 10.1093/ecco-jcc/jjae157] [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: 04/08/2024] [Revised: 07/16/2024] [Accepted: 10/02/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND The genetic contribution to inflammatory bowel disease (IBD), encompassing both Crohn's disease (CD) and ulcerative colitis (UC), accounts for around 20% of disease variance, highlighting the need to characterize environmental and epigenetic influences. Recently, considerable progress has been made in characterizing the adult methylome in epigenome-wide association studies. METHODS We report detailed analysis of the circulating methylome in 86 patients with childhood-onset CD and UC and 30 controls using the Illumina Infinium Human MethylationEPIC platform. RESULTS We derived and validated a 4-probe methylation biomarker (RPS6KA2, VMP1, CFI, and ARHGEF3), with specificity and high diagnostic accuracy for pediatric IBD in UK and North American cohorts (area under the curve: 0.90-0.94). Significant epigenetic age acceleration is present at diagnosis, with the greatest observed in CD patients. Cis-methylation quantitative trait loci (meQTL) analysis identifies genetic determinants underlying epigenetic alterations notably within the HLA 6p22.1-p21.33 region. Passive smoking exposure is associated with the development of UC rather than CD, contrary to previous findings. CONCLUSIONS These data provide new insights into epigenetic alterations in IBD and illustrate the reproducibility and translational potential of epigenome-wide association studies in complex diseases.
Collapse
Affiliation(s)
- Alexandra Noble
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, UK
| | - Alex Adams
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, UK
- Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Jan Nowak
- Department of Paediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Guo Cheng
- Department of Human Genetics and Genomic Medicine, University of Southampton, Southampton, UK
| | - Komal Nayak
- Department of Paediatrics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | - Aisling Quinn
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, UK
| | - Mark Kristiansen
- UCL Genomics, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Rahul Kalla
- Medical Research Council Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Nicholas T Ventham
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Federica Giachero
- Department of Paediatrics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Cambridge University Hospitals (CUH), Addenbrooke’s Hospital, Cambridge, UK
| | - Chamara Jayamanne
- Department of Paediatrics, John Radcliffe Hospital, Oxford University Hospital NHS Trust, Oxford, UK
| | - Richard Hansen
- Department of Child Health, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Georgina L Hold
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, New South Wales, Australia
| | - Emad El-Omar
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, New South Wales, Australia
| | - Nicholas M Croft
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - David Wilson
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, UK
- Department of Child Life and Health, University of Edinburgh, Edinburgh, UK
| | - R Mark Beattie
- Department of Paediatric Gastroenterology, Southampton Children’s Hospital, Southampton, UK
| | - James J Ashton
- Department of Human Genetics and Genomic Medicine, University of Southampton, Southampton, UK
- Department of Paediatric Gastroenterology, Southampton Children’s Hospital, Southampton, UK
| | - Matthias Zilbauer
- Department of Paediatrics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Cambridge University Hospitals (CUH), Addenbrooke’s Hospital, Cambridge, UK
| | - Sarah Ennis
- Department of Human Genetics and Genomic Medicine, University of Southampton, Southampton, UK
| | - Holm H Uhlig
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, UK
- Biomedical Research Centre, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Jack Satsangi
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
22
|
Fan X, He A, Li K, Zhang M, Zhang Q, Xiao W, Liu G. The causal impact of genetically predicted inflammatory bowel disease on extraintestinal manifestations: a mendelian randomization study. BMC Gastroenterol 2025; 25:135. [PMID: 40038580 DOI: 10.1186/s12876-024-03566-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 12/13/2024] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Extraintestinal manifestations (EIMs) significantly affect the life quality of people with inflammatory bowel disease (IBD) and are crucial factors impacting occurrence rates and mortality among IBD patients. This study performed a Mendelian randomization (MR) analysis to investigate the causal relationships between genetically predicted IBD and the development of EIMs, including erythema nodosum (EN), episcleritis, scleritis, uveitis, primary sclerosing cholangitis (PSC), and spondyloarthritis. To further investigate differences between subtypes, separate analyses were conducted for ulcerative colitis (UC) and Crohn's disease (CD). METHODS The study was conducted based on genome-wide association studies (GWAS) data. We carefully selected SNPs associated with both exposure and outcome by comparing and integrating data from GWAS and relevant literature, and prioritizing studies with large sample sizes, high quality, and as much population homogeneity as possible. The SNPs associated with IBD, UC and CD were extracted from the International Inflammatory Bowel Disease Genetics Consortium. And the SNPs associated with EIMs were extracted from the UK Biobank, the International PSC Study Group and the FinnGen study. A series of quality control steps were taken in our analysis to select eligible instrumental SNPs which were strongly associated with exposure. The causal effects were estimated using a primary analysis that employed inverse-variance weighting (IVW) and complementary analysis that utilized MR-Egger weighted by the median. A sensitivity analysis was conducted using the Cochran Q statistic, a funnel plot, the MR-Egger intercept, and a leave-one-out approach. Reverse causality analysis was also performed to ensure the robustness of the findings. Furthermore, a fixed-effects meta-analysis was employed to combine MR outcomes from various data origins, bolstering the strength and dependability of our findings. RESULTS Our findings indicated that genetically predicted IBD had a robust causal relationship with an increased risk of specific conditions, including EN (OR, 1.20; 95% CI, 1.09-1.32; p < 0.01), uveitis (OR, 1.15; 95% CI, 1.11-1.20; p < 0.01), PSC (OR, 1.21; 95% CI, 1.13-1.28; p < 0.01), and spondyloarthritis (OR, 1.19; 95% CI, 1.14-1.23; p < 0.01). In subgroup analyses, the causal effects of both UC and CD on EN, uveitis, PSC, and spondyloarthritis were also significant and robust. Additionally, no significant evidence of causality was observed between genetically predicted IBD, UC, and CD, and the occurrence of both episcleritis and scleritis. The results of reverse causality analysis indicated a robust causal association between genetically predicted PSC and the elevated risk of IBD (OR, 1.21; 95% CI, 1.15-1.29; p < 0.01), UC (OR, 1.27; 95% CI, 1.17-1.37; p < 0.01), and CD (OR, 1.10; 95% CI, 1.02-1.20; p < 0.01). Additionally, spondyloarthritis had a causal relationship with an increased risk of both IBD (OR, 1.03; 95% CI, 1.01-1.06; p < 0.01) and UC (OR, 1.05; 95% CI, 1.02-1.08; p < 0.01).
Collapse
Affiliation(s)
- Xingcan Fan
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Anqi He
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Kaiyu Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Maorun Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qi Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Wanyi Xiao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Gang Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
23
|
Verstockt S, Hannes L, Jans DS, Deman S, Souche E, van der Werf I, Vandermeulen L, Lobaton T, Laukens D, Verstockt B, Van Houdt J, Hoischen A, Vermeire S, Cleynen I. MIP4IBD: An Easy and Rapid Genotyping-by-Sequencing Assay for the Inflammatory Bowel Diseases Risk Loci. Inflamm Bowel Dis 2025; 31:786-799. [PMID: 39657915 DOI: 10.1093/ibd/izae289] [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: 09/23/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Inflammatory bowel diseases (IBD) are polygenic, with many genetic variants contributing to disease risk. Knowing the genotype of specific variants or calculating a combined genetic risk score benefits translational and functional research. To address this, we developed MIP4IBD, a flexible and cost-effective genotyping-by-sequencing assay using molecular inversion probes (MIPs). METHODS The assay targets 463 IBD risk variants, and 77 additional relevant variants. Molecular inversion probes capture and library preparation were optimized using 15 IBD DNA samples, comparing genotypes with immunochip. A custom GitHub pipeline was created for data processing, performance testing, and genotype calling. The final design was validated on a larger scale (149 IBD patients, 104 non-IBD controls, and 3 external cell lines), incorporating post hoc quality control criteria. RESULTS The assay achieved a 3.5-day turnaround time at €15 per sample with optimal sample throughput, demonstrating a 92.6% success rate in variant capture and genotype concordance rates of 99.3% and 99.6% with Infinium Global Screening Array24 BeadChip and WGS, respectively. A downstream application involved the calculation of a weighted IBD polygenic risk score (PRS), which was significantly higher in IBD patients than controls (mean 0.42 vs -0.49, P = 1.95E-11). Individuals in the highest PRS quartile had a 15.7-fold (95% CI: 6.5-38.3) risk of developing IBD and an earlier age of onset (26 vs 37 years, P = 0.02), compared to the lowest quartile. CONCLUSIONS MIP4IBD is a validated, scalable genotyping assay targeting IBD risk loci, with an integrated bioinformatics pipeline from sequencing data to genotypes and PRS calculation. Its cost-effectiveness and flexibility for additional variants make it particularly appealing for translational and clinical applications.
Collapse
Affiliation(s)
- Sare Verstockt
- Department of Chronic Diseases and Metabolism (CHROMETA), University of Leuven, Herestraat 49 Box 701, 3000 Leuven, Belgium
| | - Laurens Hannes
- Department of Human Genetics, University of Leuven, KU Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
- Center for Human Genetics, University Hospitals Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
| | - Deborah Sarah Jans
- Department of Human Genetics, Laboratory for Complex Genetics Leuven, University of Leuven, Herestraat 49 Box 604, 3000 Leuven, Belgium
| | - Stephanie Deman
- Department of Human Genetics, University of Leuven, KU Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
- Genomics Core, University Hospitals Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
| | - Erika Souche
- Department of Human Genetics, University of Leuven, KU Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
- Genomics Core, University Hospitals Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
| | - Ilse van der Werf
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Liv Vandermeulen
- Department of Gastroenterology, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Triana Lobaton
- Department of Gastroenterology, University Hospital of Ghent, Corneel Heymanslaan 10, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Debby Laukens
- Department of Internal Medicine and Pediatrics, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Bram Verstockt
- Department of Chronic Diseases and Metabolism (CHROMETA), University of Leuven, Herestraat 49 Box 701, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Jeroen Van Houdt
- J&J Innovative Medicine, Antwerpseweg 15-17, 2340 Beerse, Belgium
| | - Alexander Hoischen
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Department of Human Genetics, Heyendaalseweg 135 Box 9010, 6525 AJ Nijmegen, The Netherlands
| | - Séverine Vermeire
- Department of Chronic Diseases and Metabolism (CHROMETA), University of Leuven, Herestraat 49 Box 701, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Isabelle Cleynen
- Department of Human Genetics, Laboratory for Complex Genetics Leuven, University of Leuven, Herestraat 49 Box 604, 3000 Leuven, Belgium
| |
Collapse
|
24
|
Yu Y, Ba X, Li T, Xu W, Zhao J, Zhang N, Zhao Y, Wang T, Zhang X, Wang X, Bai B, Wang B. PTPN22 and the pathogenesis of ulcerative colitis: Insights into T cell differentiation and the JAK/STAT signaling pathway. Cell Signal 2025; 127:111551. [PMID: 39643025 DOI: 10.1016/j.cellsig.2024.111551] [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: 04/30/2024] [Revised: 10/20/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
70 % of the ulcerative colitis (UC) linked gene loci are associated with other autoimmune or immunodeficient diseases. The phosphatase activity of PTPN22 can regulate the development of T cells and contribute to regulate the level of inflammation in autoimmune diseases. We produced PTPN22-CS thymus-specific transgenic mice, which suppressed PTPN22 enzyme activity in the thymocytes. Overexpressed PTPN22-CS facilitated the development of the thymocytes towards CD4+T cells and resulted in an increased proportion of the Th1 and Treg cells in the UC mesenteric lymph nodes. PTPN22-CS promoted the activation of the JAK/STAT signaling pathway in the Th1 and Treg cells that localized in the colon, resulting in an excessive production of inflammatory mediators such as IL-2 and IFN-γ. Consequently, PTPN22-CS contributes to the inflammatory response of ulcerative colitis. In summary, the tyrosine phosphatase activity of PTPN22 plays a role in modulating UC by regulating T cell differentiation and modulating the JAK/STAT signaling pathway, thereby influencing the inflammatory response in colonic. These findings provide new insight into the association between PTPN22 and the pathogenesis of UC.
Collapse
Affiliation(s)
- Yang Yu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Xinlei Ba
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Tong Li
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Wenying Xu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Jiahui Zhao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China
| | - Na Zhang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Yanjiao Zhao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China
| | - Tao Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Xiaonan Zhang
- Department of Pathophysiology, Bengbu Medical University, Longzihu, Bengbu, 233030, Anhui, PR China..
| | - Xipeng Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China
| | - Bin Bai
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Bing Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| |
Collapse
|
25
|
Ravenhill BJ, Oliveira M, Wood G, Di Y, Kite J, Wang X, Davies CTR, Lu Y, Antrobus R, Elliott G, Irigoyen N, Hughes DJ, Lyons PA, Chung B, Borner GHH, Weekes MP. Spatial proteomics identifies a CRTC-dependent viral signaling pathway that stimulates production of interleukin-11. Cell Rep 2025; 44:115263. [PMID: 39921859 DOI: 10.1016/j.celrep.2025.115263] [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: 05/02/2024] [Revised: 12/09/2024] [Accepted: 01/12/2025] [Indexed: 02/10/2025] Open
Abstract
Appropriate cellular recognition of viruses is essential for the generation of an effective innate and adaptive immune response. Viral sensors and their downstream signaling components thus provide a crucial first line of host defense. Many of them exhibit subcellular relocalization upon activation, resulting in the expression of interferon and antiviral genes. To comprehensively identify signaling factors, we analyzed protein relocalization on a global scale during viral infection. cAMP-responsive element-binding protein (CREB)-regulated transcription coactivators 2 and 3 (CRTC2/3) exhibited early cytoplasmic-to-nuclear translocation upon infection with multiple viruses in diverse cell types. This movement was dependent on mitochondrial antiviral signaling protein (MAVS), cyclo-oxygenase proteins, and protein kinase A. A key effect of CRTC2/3 translocation is transcription of the fibro-inflammatory cytokine interleukin (IL)-11. This may be important clinically in viral infections associated with fibrosis, including SARS-CoV-2. Nuclear translocation of CRTC2/3 is, therefore, identified as an important pathway in the context of viral infection.
Collapse
Affiliation(s)
- Benjamin J Ravenhill
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Marisa Oliveira
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - George Wood
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ying Di
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Joanne Kite
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Xinyue Wang
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Colin T R Davies
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Yongxu Lu
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Gill Elliott
- Department of Microbial Sciences, School of Biosciences, University of Surrey, Guildford, UK
| | - Nerea Irigoyen
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - David J Hughes
- School of Biology, University of St. Andrews, St. Andrews, UK
| | - Paul A Lyons
- Department of Medicine, University of Cambridge, Cambridge, UK; Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Betty Chung
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Georg H H Borner
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, Germany
| | - Michael P Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
26
|
Luo C, Liu L, Zhu D, Ge Z, Chen Y, Chen F. Risk of stroke in patients with inflammatory bowel disease: a systematic review and meta-analysis. BMC Gastroenterol 2025; 25:114. [PMID: 40000943 PMCID: PMC11853978 DOI: 10.1186/s12876-025-03702-8] [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: 07/11/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Current studies suggest a potential link between inflammatory bowel disease (IBD), comprising Crohn's disease (CD) and ulcerative colitis (UC), and cardiovascular diseases, such as stroke. This study aimed to assess the risk of stroke in IBD patients compared to general population. METHODS Systematic search was done in PubMed, Embase, CENTRAL, Scopus, and CINAHL databases for studies published till September 2023. Using a random-effects model, the hazard ratios (HRs) with 95% confidence intervals (CIs) for stroke occurrence were calculated. Subgroup analyses were done to estimate pooled HR with 95%CI for CD, UC, and overall IBD cases separately. Publication bias assessment was done by Begg's and Egger's tests. RESULTS Thirteen studies with 2,802,955 participants were included. IBD patients in general had significantly higher risk of stroke, with HR of 1.30 [95% CI 1.21-1.39]. Subgroup analysis demonstrated an HR of 1.35 [95% CI 1.22-1.49] for CD and 1.15 [95% CI 1.09-1.22] for UC. Substantial heterogeneity was detected across studies, with no substantial publication bias. Sensitivity analyses affirmed the stability of findings. CONCLUSION IBD in general, and Crohn's disease in particular are associated with significantly higher risk of stroke. Our findings further emphasize the importance of cardiovascular risk assessment and management strategies in IBD care. PROTOCOL REGISTRATION PROSPERO, CRD42023470602.
Collapse
Affiliation(s)
- Chao Luo
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, China
| | - Lingpei Liu
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, China
| | - Di Zhu
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, China
| | - Zuanmin Ge
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, China
| | - Yuehua Chen
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, China
| | - Feng Chen
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, China.
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, No. 365, Renmin East Road, Jinhua City, Zhejiang Province, 321000, China.
| |
Collapse
|
27
|
Galan C, Lu G, Gill R, Li D, Liu Y, Huh JR, Hang S. RTF1 mediates epigenetic control of Th17 cell differentiation via H2B monoubiquitination. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkae043. [PMID: 40073106 PMCID: PMC11952878 DOI: 10.1093/jimmun/vkae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/02/2024] [Indexed: 03/14/2025]
Abstract
A gene encoding the transcription factor RTF1 has been associated with an increased risk of ulcerative colitis (UC). In this study, we investigated its function in modulating T cells expressing interleukin-17A (Th17 cells), a cardinal cell type promoting intestinal inflammation. Our results indicate that Rtf1 deficiency disrupts the differentiation of Th17 cells, while leaving regulatory T cells (Treg) unaffected. Mechanistically, RTF1 facilitates histone H2B monoubiquitination (H2Bub1), which requires its histone modification domain (HMD), for supporting Th17 cell function. Impaired Th17 differentiation was also observed in cells lacking the H2Bub1 E3 ligase subunit RNF40, an enzyme known to physically interact with RTF1. Thus, our study underscores the essential role of RTF1 in H2Bub1-mediated epigenetic regulation of Th17 cell differentiation. Understanding this process will likely provide valuable insights into addressing Th17-associated inflammatory disorders. (Images were created with BioRender).
Collapse
Affiliation(s)
| | - Guangqing Lu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, United States
| | - Richard Gill
- Genentech, Inc, South San Francisco, CA, United States
| | - Dun Li
- Genentech, Inc, South San Francisco, CA, United States
| | - Yifang Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, United States
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, United States
- Bio2Q, Keio University, Tokyo, Japan
| | - Saiyu Hang
- Genentech, Inc, South San Francisco, CA, United States
| |
Collapse
|
28
|
Sun Z, Ye J, Sun W, Jiang L, Shan B, Zhang M, Xu J, Li W, Liu J, Jing H, Zhang T, Hou M, Xie C, Wu R, Pan H, Yuan J. Cooperation of TRADD- and RIPK1-dependent cell death pathways in maintaining intestinal homeostasis. Nat Commun 2025; 16:1890. [PMID: 39987261 PMCID: PMC11846980 DOI: 10.1038/s41467-025-57211-z] [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: 09/11/2024] [Accepted: 02/12/2025] [Indexed: 02/24/2025] Open
Abstract
Dysfunctional NF-κB signaling is critically involved in inflammatory bowel disease (IBD). We investigated the mechanism by which RIPK1 and TRADD, two key mediators of NF-κB signaling, in mediating intestinal pathology using TAK1 IEC deficient model. We show that phosphorylation of TRADD by TAK1 modulates RIPK1-dependent apoptosis. TRADD and RIPK1 act cooperatively to mediate cell death regulated by TNF and TLR signaling. We demonstrate the pathological evolution from RIPK1-dependent ileitis to RIPK1- and TRADD-co-dependent colitis in TAK1 IEC deficient condition. Combined RIPK1 inhibition and TRADD knockout completely protect against intestinal pathology and lethality in TAK1 IEC KO mice. Furthermore, we identify distinctive microbiota dysbiosis biomarkers for RIPK1-dependent ileitis and TRADD-dependent colitis. These findings reveal the cooperation between RIPK1 and TRADD in mediating cell death and inflammation in IBD with NF-κB deficiency and suggest the possibility of combined inhibition of RIPK1 kinase and TRADD as a new therapeutic strategy for IBD.
Collapse
Affiliation(s)
- Ziyu Sun
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shanghai Key Laboratory of Aging Studies, Shanghai, 201210, China
| | - Jianyu Ye
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shanghai Key Laboratory of Aging Studies, Shanghai, 201210, China
| | - Weimin Sun
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Libo Jiang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Bing Shan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Mengmeng Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Jingyi Xu
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Wanjin Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Jianping Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Hongyang Jing
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Tian Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Meiling Hou
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Rongling Wu
- Beijing Key Laboratory of Topological Statistics and Applications for Complex Systems, Beijing Institute of Mathematical Sciences and Applications, Beijing, 101408, China
- Yau Mathematical Sciences Center, Tsinghua University, Beijing, 100084, China
- Shanghai Institute for Mathematics and Interdisciplinary Sciences, Shanghai, 200433, China
| | - Heling Pan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
- Shanghai Key Laboratory of Aging Studies, Shanghai, 201210, China
| | - Junying Yuan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China.
- Shanghai Key Laboratory of Aging Studies, Shanghai, 201210, China.
| |
Collapse
|
29
|
Chen S, Putnik R, Li X, Diwaker A, Vasconcelos M, Liu S, Gondi S, Zhou J, Guo L, Xu L, Temme S, Bersch K, Hyland S, Yin J, Burstein E, Bahnson BJ, Gildersleeve JC, Grimes CL, Reinecker HC. PGLYRP1-mediated intracellular peptidoglycan detection promotes intestinal mucosal protection. Nat Commun 2025; 16:1864. [PMID: 39984444 PMCID: PMC11845746 DOI: 10.1038/s41467-025-57126-9] [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: 10/02/2024] [Accepted: 02/10/2025] [Indexed: 02/23/2025] Open
Abstract
Peptidoglycan recognition proteins (PGLYRPs) are implicated in the control of the intestinal microbiota; however, molecular requirements for peptidoglycan (PGN) binding and receptor signaling mechanisms remain poorly understood. Here we show that PGLYRP1 is a receptor for the disaccharide motif of lysine N-acetylglucosamine N-acetylmuramic tripeptide (GMTriP-K). PGLYRP1 is required for innate immune activation by GMTriP-K but not muramyl dipeptide (MDP). In macrophages, intracellular PGLYRP1 complexes with NOD2 and GEF-H1, both of which are required for GMTriP-K-regulated gene expression. PGLYRP1 localizes to the endoplasmic reticulum and interacts at the Golgi with NOD2 upon GMTriP-K stimulation. PGLYRP1 and dependent gene expression signatures are induced in both mouse intestinal inflammation and human ulcerative colitis. Importantly, PGLYRP1 activation by GMTriP-K can result in the protection of mice from TNBS-induced colitis. Mammalian PGLYRPs can function as intracellular pattern recognition receptors for the control of host defense responses in the intestine.
Collapse
Affiliation(s)
- Shuyuan Chen
- Division of Digestive and Liver Diseases, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rachel Putnik
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Xi Li
- Division of Digestive and Liver Diseases, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alka Diwaker
- Division of Digestive and Liver Diseases, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marina Vasconcelos
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Shuzhen Liu
- Division of Digestive and Liver Diseases, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sudershan Gondi
- Division of Digestive and Liver Diseases, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Junhui Zhou
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Lei Guo
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sebastian Temme
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Klare Bersch
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Stephen Hyland
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Jianyi Yin
- Division of Digestive and Liver Diseases, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ezra Burstein
- Division of Digestive and Liver Diseases, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Brian J Bahnson
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Jeffrey C Gildersleeve
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | | | - Hans-Christian Reinecker
- Division of Digestive and Liver Diseases, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
30
|
Wils P, Habibi Kavashkohie MR, Sélos Guerra F, Landais S, Rubio M, Mehta H, Sarfati M, Chapuy L. Single-Cell Transcriptomic Profile of Innate Cell Populations in Mesenteric Lymph Nodes of Inflammatory Bowel Disease Patients. Inflamm Bowel Dis 2025:izaf017. [PMID: 39982469 DOI: 10.1093/ibd/izaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Indexed: 02/22/2025]
Abstract
BACKGROUND AND AIMS Innate immune cells, including dendritic cells (DCs), monocytes (Mono), macrophages (Mac), natural killer (NK), and innate lymphoid cells (ILC), contribute to chronic inflammation in lymphoid tissues. Here, we characterized the innate immune cell landscape in inflamed mesenteric lymph nodes (MLNs) of patients with inflammatory bowel diseases (IBD) at the single-cell level. METHODS Surgically resected colonic MLNs were obtained from patients with Crohn's disease (CD; n = 3), ulcerative colitis (UC; n = 3), non-inflamed UC (n = 1), and non-IBD (n = 2). CD45+CD3-CD19- non-T/non-B cells were FACS-sorted to capture rare innate immune cells. Cellular Indexing of Transcriptomes and Epitopes by Sequencing (CITE-seq) was performed on the BD Rhapsody platform alongside multiparameter flow cytometry staining. RESULTS CITE-seq analysis unveiled the molecular signature of 11 Mono/Mac/DC (MMDC) and 7 NK/ILC enriched clusters in human MLNs. DC clusters included 3 newly characterized DC clusters such as CD1c/CD163/VCAN/CD64-expressing DC3; AXL-expressing DCs; and a CD103+ DC subset, expressing LTB, S100B, and IL22RA2 (encoding IL22BP). Mono/Mac clusters comprised inflammatory monocytes, which accumulated in IBD compared to non-IBD MLNs. Among NK/ILC clusters, we identified a cytotoxic ILC subset (IL7R, KLRD1, GNLY), previously not reported in MLNs, reminiscent of cytotoxic ILC1-like cells found in inflamed gut mucosa. CONCLUSION CITE-seq and flow-cytometry analyses of colonic MLNs from patients with active IBD reveal the molecular signature and cell distribution of previously uncharacterized DC and ILC subpopulations in human MLNs. These findings expand our understanding of immune responses during chronic inflammation in IBD.
Collapse
Affiliation(s)
- Pauline Wils
- Hepato-Gastroenterology Department, Claude Huriez Hospital, University of Lille 2, 59000 Lille, France
- INFINITE, University of Lille, INSERM U1286-Institute for Translational Research in Inflammation, 59000 Lille, France
| | | | - Fabiana Sélos Guerra
- Department of Pediatrics, Centre de Recherche du CHU Sainte-Justine, Université de Montréal, H3T 1C5 Montréal, Québec, Canada
| | - Séverine Landais
- Department of Pediatrics, Centre de Recherche du CHU Sainte-Justine, Université de Montréal, H3T 1C5 Montréal, Québec, Canada
| | - Manuel Rubio
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM), Université de Montréal, H2X 0A9 Montréal, Québec, Canada
| | - Heena Mehta
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM), Université de Montréal, H2X 0A9 Montréal, Québec, Canada
| | - Marika Sarfati
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM), Université de Montréal, H2X 0A9 Montréal, Québec, Canada
| | - Laurence Chapuy
- Department of Pediatrics, Centre de Recherche du CHU Sainte-Justine, Université de Montréal, H3T 1C5 Montréal, Québec, Canada
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM), Université de Montréal, H2X 0A9 Montréal, Québec, Canada
- Department of Pediatrics, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, H4A 3J1 Montreal, Quebec, Canada
| |
Collapse
|
31
|
Li J, Jacobse J, Pilat JM, Kaur H, Gu W, Kang SW, Rusznak M, Huang HI, Barrera J, Oloo PA, Roland JT, Hawkins CV, Pahnke AP, Khalil M, Washington MK, Wilson KT, Williams CS, Peebles RS, Konnikova L, Choksi YA, Hammer GE, Lau KS, Goettel JA. Interleukin-10 production by innate lymphoid cells restricts intestinal inflammation in mice. Mucosal Immunol 2025:S1933-0219(25)00023-6. [PMID: 39988202 DOI: 10.1016/j.mucimm.2025.02.005] [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: 03/19/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Interleukin-10 (IL-10) is an immunomodulatory cytokine critical for intestinal immune homeostasis. IL-10 is produced by various immune cells but IL-10 receptor signaling in intestinal CX3CR1+ mononuclear phagocytes is necessary to prevent spontaneous colitis in mice. Here, we utilized fluorescent protein reporters and cell-specific targeting and found that Rorc-expressing innate lymphoid cells (ILCs) produce IL-10 in response to anti-CD40-mediated intestinal inflammation. Deletion of Il10 specifically in Rorc-expressing ILCs led to phenotypic changes in intestinal macrophages and exacerbated both innate and adaptive immune-mediated models of experimental colitis. The population of IL-10+ producing ILCs shared markers with both ILC2 and ILC3 with nearly all ILC3s being of the NCR+ subtype. Interestingly, Ccl26 was enriched in IL-10+ ILCs and was markedly reduced in IL-10-deficient ILC3s. Since CCL26 is a ligand for CX3CR1, we employed RNA in situ hybridization and observed increased numbers of ILCs in close proximity to Cx3cr1-expressing cells under inflammatory conditions. Finally, we generated transgenic RorctdTomato reporter mice that faithfully marked RORγt+ cells that could rescue disease pathology and aberrant macrophage phenotype following adoptive transfer into mice with selective Il10 deficiency in ILC3s. These results demonstrate that IL-10 production by a population of ILCs functions to promote immune homeostasis in the intestine possibly via direct effects on intestinal macrophages.
Collapse
Affiliation(s)
- Jing Li
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Justin Jacobse
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212
| | - Jennifer M Pilat
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Harsimran Kaur
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Weihong Gu
- Department of Pediatrics, Yale Medical School, New Haven, CT 06520, USA
| | - Seung Woo Kang
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Mark Rusznak
- Department of Internal Medicine Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hsin-I Huang
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Julio Barrera
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Pauline A Oloo
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Joseph T Roland
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Internal Medicine Vanderbilt University Medical Center, Nashville, TN, USA
| | - Caroline V Hawkins
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrew P Pahnke
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Marian Khalil
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Keith T Wilson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christopher S Williams
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - R Stokes Peebles
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212; Department of Internal Medicine Vanderbilt University Medical Center, Nashville, TN, USA
| | - Liza Konnikova
- Department of Pediatrics, Yale Medical School, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA; Center for Systems and Engineering Immunology, Yale School of Medicine, New Haven, CT 06520, USA; Human and Translational Immunology Program, Yale School of Medicine, New Haven, CT 06520, USA; Program in Translational Biomedicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Obstetrics Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yash A Choksi
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gianna Elena Hammer
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Ken S Lau
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA; Center for Computational Systems Biology, Vanderbilt University, Nashville, TN, USA
| | - Jeremy A Goettel
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
32
|
Du X, Yu W, Chen F, Jin X, Xue L, Zhang Y, Wu Q, Tong H. HDAC inhibitors and IBD: Charting new approaches in disease management. Int Immunopharmacol 2025; 148:114193. [PMID: 39892171 DOI: 10.1016/j.intimp.2025.114193] [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: 09/08/2024] [Revised: 12/14/2024] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Inflammatory bowel disease (IBD) represents a group of chronic inflammatory disorders of the gastrointestinal tract. Despite substantial advances in our understanding of IBD pathogenesis, the currently available therapeutic options remain limited in their efficacy and often come with significant side effects. Therefore, there is an urgent need to explore novel approaches for the management of IBD. One promising avenue of investigation revolves around the use of histone deacetylase (HDAC) inhibitors, which have garnered considerable attention for their potential in modulating gene expression and curbing inflammatory responses. This review emphasizes the pressing need for innovative drugs in the treatment of IBD, and drawing from a wealth of preclinical studies and clinical trials, we underscore the multifaceted roles and the therapeutic effects of HDAC inhibitors in IBD models and patients. This review aims to contribute significantly to the understanding of HDAC inhibitors' importance and prospects in the management of IBD, ultimately paving the way for improved therapeutic strategies in this challenging clinical landscape.
Collapse
Affiliation(s)
- Xueting Du
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, China
| | - Weilai Yu
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Fangyu Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, China
| | - Xiaosheng Jin
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Liwei Xue
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Ya Zhang
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, China; Hepatology Diagnosis and Treatment Center & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Qifang Wu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, China.
| | - Haibin Tong
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, China.
| |
Collapse
|
33
|
Wu W, Tong H, Li Y, Cui J. Diabetes mellitus, metformin's target gene AMPK, and inflammatory bowel disease: A Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41532. [PMID: 39960958 PMCID: PMC11835072 DOI: 10.1097/md.0000000000041532] [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: 05/17/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
The causal relationship between inflammatory bowel disease (IBD) and diabetes mellitus remains unclear. The aim of this study was to delve into this association and investigate the correlation between AMP-activated protein kinase (AMPK), a target gene of metformin, and the risk of developing IBD. Researchers conducted a bidirectional two-sample Mendelian randomization analysis to examine causal relationships between IBD, including ulcerative colitis and Crohn disease (CD), and diabetes mellitus, encompassing both type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). Additionally, this study utilized AMPK-related variants associated with HbA1c (%) as instrumental variables for the metformin target gene AMPK to further investigate their association with the risk of IBD. The inverse variance weighted method was used as the primary analytical approach. Mendelian randomization analysis revealed a suggestive association between IBD and T1DM (P = .024). CD was associated with an increased risk of T1DM (P = .011). In the reverse analysis, T1DM also increased the risk of IBD (P = .043). No causal relationship was found between IBD and T2DM in either the forward or reverse analyses. In addition, this study did not find any significant effect of AMPK on IBD. In conclusion, this study suggests a bidirectional association between IBD and T1DM, in which CD may increase the risk of T1DM. However, no causal relationship was found between IBD and T2DM. Furthermore, our findings revealed that the metformin's target gene AMPK had no significant effect on the onset of IBD.
Collapse
Affiliation(s)
- Wei Wu
- Department of Geriatrics, Chun’an First People’s Hospital (Chun’an Branch of Zhejiang Provincial People’s Hospital), Hangzhou, Zhejiang Province, China
| | - Huomu Tong
- Department of Endocrinology, Chun’an First People’s Hospital (Chun’an Branch of Zhejiang Provincial People’s Hospital), Hangzhou, Zhejiang Province, China
| | - Yunsheng Li
- Department of Geriatrics, Chun’an First People’s Hospital (Chun’an Branch of Zhejiang Provincial People’s Hospital), Hangzhou, Zhejiang Province, China
| | - Jia Cui
- Department of Endocrinology, Chun’an First People’s Hospital (Chun’an Branch of Zhejiang Provincial People’s Hospital), Hangzhou, Zhejiang Province, China
| |
Collapse
|
34
|
Tamburri S, Zucchelli C, Matafora V, Zapparoli E, Jevtic Z, Farris F, Iannelli F, Musco G, Bachi A. SP140 represses specific loci by recruiting polycomb repressive complex 2 and NuRD complex. Nucleic Acids Res 2025; 53:gkae1215. [PMID: 39718989 DOI: 10.1093/nar/gkae1215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/26/2024] Open
Abstract
SP140, a lymphocytic-restricted protein, is an epigenetic reader working as a corepressor of genes implicated in inflammation and orchestrating macrophage transcriptional programs to maintain cellular identity. Reduced SP140 expression is associated both to autoimmune diseases and blood cancers. However, the molecular mechanisms that link SP140 altered protein levels to detrimental effects on the immune response and cellular growth, as well as the interactors through which SP140 promotes gene silencing, remain elusive. In this work, we have applied a multi-omics approach (i.e. interactomics, ChIP-seq and proteomics) in two Burkitt lymphoma cell lines to identify both interactors and target genes of endogenous SP140. We found that SP140 interacts with the PRC2 and NuRD complexes, and we showed that these interactions are functional as SP140 directs H3K27me3 deposition and NuRD binding on a set of target genes implicated in cellular growth and leukemia progression.
Collapse
Affiliation(s)
- Simone Tamburri
- IFOM ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 16039 Milano, Italy
| | - Chiara Zucchelli
- Biomolecular NMR Laboratory, Division of Genetics and Cell biology, IRCCS Ospedale San Raffaele, Via Olgettina 58, 20132 Milano, Italy
| | - Vittoria Matafora
- IFOM ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 16039 Milano, Italy
| | - Ettore Zapparoli
- Center for Omics Sciences, IRCCS Ospedale San Raffaele, Via Olgettina 58, 20132 Milano, Italy
| | - Zivojin Jevtic
- IFOM ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 16039 Milano, Italy
| | - Francesco Farris
- IFOM ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 16039 Milano, Italy
| | - Fabio Iannelli
- IFOM ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 16039 Milano, Italy
| | - Giovanna Musco
- Biomolecular NMR Laboratory, Division of Genetics and Cell biology, IRCCS Ospedale San Raffaele, Via Olgettina 58, 20132 Milano, Italy
| | - Angela Bachi
- IFOM ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 16039 Milano, Italy
| |
Collapse
|
35
|
de Vries MH, Meddens CA, Hijma HJ, Berrens AC, Jansen SA, Kooiman BA, Snapper S, Clevers H, Mokry M, Kuijk EW, Nieuwenhuis EE. Human colon stem cells are the principal epithelial responders to bacterial antigens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637053. [PMID: 39975165 PMCID: PMC11839077 DOI: 10.1101/2025.02.07.637053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Intestinal epithelial cells (IECs) are capable of mounting an adequate antimicrobial inflammatory response to pathogens while tolerating commensals. The underlying regulatory mechanisms of immune sensitivity remain incompletely understood, particularly in the context of human IECs. To enhance our understanding of the immune response of IECs to bacterial epithelial barrier breach, we investigated whether epithelial responsiveness is contingent on cell identity and cell polarization. We exposed human intestinal organoids to bacterial antigens to study their immune responses. Notable discrepancies were observed in the specific reactions exhibited by intestinal stem cells (ISCs) and enterocytes. It was determined that basolateral exposure of IECs to bacterial antigens resulted in a robust response, whereas apical exposure elicited a significantly more modest response. We identified ISCs as the responders, while the reaction of enterocytes was found to be attenuated. The regulation of bacterial responsiveness in enterocytes occurs at multiple levels, including the modulation of NFκB activation and post-transcriptional control of mRNA stability. Our findings demonstrate that differentiated non-responsive enterocytes can be sensitized to bacterial antigens through the activation of the WNT pathway. These findings extend the crucial role of WNT signaling for intestinal epithelial homeostasis and regulation of stem cell maintenance, proliferation, differentiation, and tissue architecture in the gut. Additionally, they reveal a new function of WNT signaling in regulating microbial responses within the intestinal environment.
Collapse
Affiliation(s)
- Maaike H. de Vries
- Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Claartje A. Meddens
- Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Hemme J. Hijma
- Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Anne-Claire Berrens
- Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Suze A. Jansen
- Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Berend A.P. Kooiman
- Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Scott Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Hans Clevers
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Utrecht, the Netherlands
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
- Present address: Pharma, Research and Early Development, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Michal Mokry
- Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy, University Medical Center Utrecht, University Utrecht, The Netherlands
| | - Ewart W. Kuijk
- Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- Department of Pediatric Pulmonary, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Edward E.S. Nieuwenhuis
- Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- Present address: Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| |
Collapse
|
36
|
Wei J, Resztak JA, Ranjbaran A, Alazizi A, Mair-Meijers HE, Slatcher RB, Zilioli S, Wen X, Luca F, Pique-Regi R. Functional characterization of eQTLs and asthma risk loci with scATAC-seq across immune cell types and contexts. Am J Hum Genet 2025; 112:301-317. [PMID: 39814021 DOI: 10.1016/j.ajhg.2024.12.017] [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: 02/27/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/18/2025] Open
Abstract
cis-regulatory elements (CREs) control gene transcription dynamics across cell types and in response to the environment. In asthma, multiple immune cell types play an important role in the inflammatory process. Genetic variants in CREs can also affect gene expression response dynamics and contribute to asthma risk. However, the regulatory mechanisms underlying control of transcriptional dynamics across different environmental contexts and cell types at single-cell resolution remain to be elucidated. To resolve this question, we performed single-cell ATAC-seq (scATAC-seq) in peripheral blood mononuclear cells (PBMCs) from 16 children with asthma. PBMCs were activated with phytohemagglutinin (PHA) or lipopolysaccharide (LPS) and treated with dexamethasone (DEX), an anti-inflammatory glucocorticoid. We analyzed changes in chromatin accessibility, measured transcription factor motif activity, and identified treatment- and cell-type-specific transcription factors that drive changes in both gene expression mean and variability. We observed a strong positive linear dependence between motif response and their target gene expression changes but a negative relationship with changes in target gene expression variability. This result suggests that an increase of transcription factor binding tightens the variability of gene expression around the mean. We then annotated genetic variants in chromatin accessibility peaks and response motifs, followed by computational fine-mapping of expression quantitative trait loci (eQTL) from a pediatric asthma cohort. We found that eQTLs were 5-fold enriched in peaks with response motifs and refined the credible set for 410 asthma risk genes, with 191 having the causal variant in response motifs. In conclusion, scATAC-seq enhances the understanding of molecular mechanisms for asthma risk variants mediated by gene expression.
Collapse
Affiliation(s)
- Julong Wei
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Justyna A Resztak
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Ali Ranjbaran
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Adnan Alazizi
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | | | | | - Samuele Zilioli
- Department of Psychology, Wayne State University, Detroit, MI, USA; Department of Family Medicine and Public Health Sciences, Wayne State University, Detroit, MI, USA
| | - Xiaoquan Wen
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Francesca Luca
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA.
| | - Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
37
|
Kayali S, Fantasia S, Gaiani F, Cavallaro LG, de’Angelis GL, Laghi L. NOD2 and Crohn's Disease Clinical Practice: From Epidemiology to Diagnosis and Therapy, Rewired. Inflamm Bowel Dis 2025; 31:552-562. [PMID: 38582044 PMCID: PMC11808579 DOI: 10.1093/ibd/izae075] [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: 08/03/2023] [Indexed: 04/08/2024]
Abstract
Crohn's disease (CD) is a chronic inflammatory bowel disease with a multifactorial pathogenesis involving environmental and genetic factors. Since the late 20th century, the discovery of the first susceptibility gene (NOD2, previously referred to as CARD15) for CD has paved the way for further investigations into the correlations between clinical features and genetics, and its potential impact on clinical practice has fueled the research in the last 2 decades. Recent therapeutic advancements involving novel biologic drugs and small molecules have shifted inflammatory bowel disease management from a disease-centered to a patient-centric approach. To date, the role of NOD2 has not been fully understood yet. Recent data suggest that its clinical impact may be greater than currently recognized. This review overviews the most common NOD2 variants' role in real-life clinical practice. These genetic variants increase the risk of developing the disease and can aid in tailoring diagnosis and treatment. They are associated with the stricturing phenotype and ileal involvement and increase the risk of steroid refractoriness. In the meantime, limited and inconclusive evidence exists regarding their predictive role in response to azathioprine, biologic drugs, and small molecules. Eventually, their role in increasing the risk for surgery is evident, especially in those with the L1007fs variant. If further trials will support the initial evidence reported so far, NOD2 genetic variants will emerge as possible candidates for developing precision medicine in CD.
Collapse
Affiliation(s)
- Stefano Kayali
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Stefano Fantasia
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Federica Gaiani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Gastroenterology and Endoscopy Unit, University Hospital of Parma, Parma, Italy
| | | | | | - Luigi Laghi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Centre, Rozzano, Italy
| |
Collapse
|
38
|
Gao Y, Cao J, Han B, Sun D. Preliminary exploration of mRNA, lncRNA, and miRNA expressions in the bovine jejunum unveils novel aspects of Mycobacterium avium subspecies paratuberculosis infections. BMC Genomics 2025; 26:108. [PMID: 39905315 PMCID: PMC11796175 DOI: 10.1186/s12864-025-11299-1] [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: 08/21/2024] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Paratuberculosis (Johne's disease, JD) is a chronic and enteric disease in a range of ruminants, often caused by Mycobacterium avium subspecies paratuberculosis (MAP) infection, leading to substantial economic losses worldwide. Yet, the molecular underpinning of paratuberculosis remains elusive. Here we performed RNA sequencing (RNA-seq) and small RNA sequencing (sRNA-seq) of the jejunum tissues from the Holstein cows with three distinct statuses of paratuberculosis, i.e., healthy, subclinical, and clinical to screen potential genes, lncRNAs, and miRNAs associated with the resistance or susceptibility to MAP infection and build ceRNA regulatory networks via miRNAs. RESULTS We applied whole transcriptome sequencing analysis to examine the jejunum tissue in nine Holstein cows. Starting with 19,994 expressed genes, 13,529 lncRNAs, and 735 miRNAs, we screened out differentially expressed genes (DEGs), lncRNAs, and miRNAs of three comparison groups, i.e., clinical vs. healthy, subclinical vs. healthy, and clinical vs. subclinical, subsequently identifying ceRNA pairs. Ultimately, we detected 76, 74, and 24 DEGs, 19, 39, and 10 lncRNAs, as well as 28, 61, and 20 miRNAs across the three comparison groups, respectively. Through integrating these DEGs with functional annotation, previously reported QTLs, and GWAS results, we proposed eight genes (LYZ, LYZ1, BOLA-DQB, BOLA-DQA1, TAP, CATD, VNN1, and PPARG), six lncRNAs, 48 miRNAs, and 107 ceRNA pairs implying their potential associations with susceptibility to MAP infection. CONCLUSION The present study provided a global view of the dynamics in transcriptomes of the bovine jejunum tissues in terms of JD status. Our results demonstrated that not only mRNAs but also lncRNAs and miRNAs played important roles in regulating MAP infection in dairy cattle. This study provided a valuable resource for understanding the molecular basis of JD, potentially contributing to the genetic improvement of JD resistance in dairy cattle.
Collapse
Affiliation(s)
- Yahui Gao
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jie Cao
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Bo Han
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Dongxiao Sun
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
39
|
Jurickova I, Dreskin BW, Angerman E, Bonkowski E, Nguyen J, Villarreal R, Tominaga K, Iwasawa K, Braun T, Takebe T, Helmrath MA, Haberman Y, Wells JM, Denson LA. Eicosatetraynoic Acid Regulates Profibrotic Pathways in an Induced Pluripotent Stem Cell-Derived Macrophage-Human Intestinal Organoid Model of Crohn's Disease. J Crohns Colitis 2025; 19:jjae139. [PMID: 39212594 PMCID: PMC11836882 DOI: 10.1093/ecco-jcc/jjae139] [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: 12/05/2023] [Revised: 07/19/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND AIMS We previously identified small molecules predicted to reverse an ileal gene signature for future Crohn's Disease (CD) strictures. Here we used a new human intestinal organoid (HIO) model system containing macrophages to test a lead candidate, eicosatetraynoic acid (ETYA). METHODS Induced pluripotent stem cell lines (iPSC) were derived from CD patients and differentiated into macrophages and HIOs. Macrophages and macrophage-HIO cocultures were exposed to lipopolysaccharide (LPS) with and without ETYA pretreatment. Cytospin and flow cytometry characterized macrophage morphology and activation markers, and RNA sequencing defined the global pattern of macrophage gene expression. TaqMan low-density array, Luminex multiplex assay, immunohistologic staining, and sirius red polarized light microscopy were performed to measure macrophage cytokine production and HIO profibrotic gene expression and collagen content. RESULTS Induced PSC-derived macrophages exhibited morphology similar to primary macrophages and expressed inflammatory macrophage cell surface markers including CD64 and CD68. LPS-stimulated macrophages expressed a global pattern of gene expression enriched in CD ileal inflammatory macrophages and matrisome-secreted products and produced cytokines and chemokines including CCL2, IL1B, and OSM implicated in refractory disease. ETYA suppressed CD64 abundance and profibrotic gene expression pathways in LPS-stimulated macrophages. Coculture of LPS-primed macrophages with HIO led to upregulation of fibroblast activation genes including ACTA2 and COL1A1, and an increase in HIO collagen content. ETYA pretreatment prevented profibrotic effects of LPS-primed macrophages. CONCLUSIONS ETYA inhibits profibrotic effects of LPS-primed macrophages upon cocultured HIO. This model may be used in future untargeted screens for small molecules to treat refractory CD.
Collapse
Affiliation(s)
- Ingrid Jurickova
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Benjamin W Dreskin
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Elizabeth Angerman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Erin Bonkowski
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jack Nguyen
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Richard Villarreal
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kentaro Tominaga
- Division of Gastroenterology and Hepatology, Niigata University, Niigata, Japan
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kentaro Iwasawa
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tzipi Braun
- Department of Pediatrics, Sheba Medical Center, Tel-HaShomer, Affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Takanori Takebe
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Institute of Research, Tokyo Medical and Dental University, Tokyo, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), and Division of Stem Cell and Organoid Medicine, Osaka University, Suita, Osaka, Japan
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael A Helmrath
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Pediatric General and Thoracic Surgery, Department of Surgery, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Yael Haberman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, Sheba Medical Center, Tel-HaShomer, Affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - James M Wells
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lee A Denson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
40
|
Khan A, Azzam MA. Inflammatory Bowel Disease and Stroke: Exploring Hidden Vascular Risks. Cureus 2025; 17:e79304. [PMID: 40125129 PMCID: PMC11927930 DOI: 10.7759/cureus.79304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease and ulcerative colitis, is primarily known for its gastrointestinal manifestations. However, emerging evidence suggests a potential link between IBD and an increased risk of stroke, likely mediated by chronic systemic inflammation, endothelial dysfunction, and a prothrombotic state. Despite this growing recognition, the exact mechanisms and extent of this association remain unclear, highlighting a critical knowledge gap. This review aims to systematically analyze the association between IBD and stroke, exploring the underlying vascular mechanisms and identifying potential risk factors contributing to cerebrovascular events in IBD patients. A comprehensive literature search was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines across PubMed, Scopus, and Google Scholar using keywords such as "IBD," "Stroke," "Chronic inflammation," "Cerebrovascular risk," and "Gut-brain axis." After screening 150 studies and applying inclusion and exclusion criteria, six studies were included in the final synthesis. The findings suggest that chronic inflammation in IBD plays a key role in increasing stroke risk through endothelial dysfunction and a heightened prothrombotic state, with additional risk factors such as atrial fibrillation during active IBD flares further contributing to cerebrovascular events. While biologic therapies, including tumor necrosis factor (TNF)-alpha inhibitors, are effective in reducing systemic inflammation, their impact on mitigating stroke risk remains inconclusive. Given the potential role of IBD as an independent risk factor for stroke, a multidisciplinary approach to management is crucial. Addressing modifiable risk factors through pharmacologic interventions such as biologics, statins, and antiplatelet agents, alongside lifestyle modifications, could help reduce cerebrovascular complications in IBD patients. Further research is needed to explore personalized therapeutic strategies and establish clearer preventive guidelines for this at-risk population.
Collapse
Affiliation(s)
- Abdallah Khan
- Internal Medicine, RAK Medical and Health Sciences University, Ras Al Khaimah, ARE
| | - Maysoon A Azzam
- Internal Medicine, RAK Medical and Health Sciences University, Ras Al Khaimah, ARE
| |
Collapse
|
41
|
Loya H, Kalantzis G, Cooper F, Palamara PF. A scalable variational inference approach for increased mixed-model association power. Nat Genet 2025; 57:461-468. [PMID: 39789286 PMCID: PMC11821521 DOI: 10.1038/s41588-024-02044-7] [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: 10/10/2023] [Accepted: 11/27/2024] [Indexed: 01/12/2025]
Abstract
The rapid growth of modern biobanks is creating new opportunities for large-scale genome-wide association studies (GWASs) and the analysis of complex traits. However, performing GWASs on millions of samples often leads to trade-offs between computational efficiency and statistical power, reducing the benefits of large-scale data collection efforts. We developed Quickdraws, a method that increases association power in quantitative and binary traits without sacrificing computational efficiency, leveraging a spike-and-slab prior on variant effects, stochastic variational inference and graphics processing unit acceleration. We applied Quickdraws to 79 quantitative and 50 binary traits in 405,088 UK Biobank samples, identifying 4.97% and 3.25% more associations than REGENIE and 22.71% and 7.07% more than FastGWA. Quickdraws had costs comparable to REGENIE, FastGWA and SAIGE on the UK Biobank Research Analysis Platform service, while being substantially faster than BOLT-LMM. These results highlight the promise of leveraging machine learning techniques for scalable GWASs without sacrificing power or robustness.
Collapse
Affiliation(s)
- Hrushikesh Loya
- Department of Statistics, University of Oxford, Oxford, UK
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Georgios Kalantzis
- Department of Statistics, University of Oxford, Oxford, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Fergus Cooper
- Doctoral Training Centre, University of Oxford, Oxford, UK
| | - Pier Francesco Palamara
- Department of Statistics, University of Oxford, Oxford, UK.
- Centre for Human Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
42
|
Schwab AD, Wyatt TA, Schanze OW, Nelson AJ, Gleason AM, Duryee MJ, Mosley DD, Thiele GM, Mikuls TR, Poole JA. Lung-delivered IL-10 mitigates Lung inflammation induced by repeated endotoxin exposures in male mice. Physiol Rep 2025; 13:e70253. [PMID: 39980189 PMCID: PMC11842461 DOI: 10.14814/phy2.70253] [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: 12/16/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
Therapies capable of resolving inflammatory lung disease resulting from high-consequence occupational/environmental hazards are lacking. This study seeks to determine the therapeutic potential of direct lung-delivered interleukin (IL)-10 following repeated lipopolysaccharide exposures. C57BL/6 mice were intratracheally instilled with LPS (10 μg) and treated with IL-10 (1 μg) or vehicle control for 3 days. Lung cell infiltrates were enumerated by flow cytometry. Lung sections were stained for myeloperoxidase (MPO), CCR2, vimentin, and post-translational protein citrullination (CIT) and malondialdehyde-acetaldehyde (MAA) modifications. Lung function testing and longitudinal in vivo micro-CT imaging were performed. Whole lungs were profiled using bulk RNA sequencing. IL-10 treatment reduced LPS-induced weight loss, pentraxin-2, and IL-6 serum levels. LPS-induced lung proinflammatory and wound repair mediators (i.e., TNF-α, IL-6, CXCL1, CCL2, MMP-8, MMP-9, TIMP-1, fibronectin) were decreased with IL-10. IL-10 reduced LPS-induced influx of lung neutrophils, CD8+ T cells, NK cells, recruited monocyte-macrophages, monocytes, and tissue expression of CCR2+ monocytes-macrophages, MPO+ neutrophils, vimentin, CIT, and MAA. IL-10 reduced LPS-induced airway hyperresponsiveness and improved lung compliance. Micro-CT imaging confirmed the reduction in LPS-induced lung density by IL-10. Lung-delivered IL-10 therapy administered after daily repeated endotoxin exposures strikingly reduces lung inflammatory and wound repair processes to decrease lung pathologic changes and mitigate airway dysfunction.
Collapse
Affiliation(s)
- Aaron D. Schwab
- Division of Allergy & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Todd A. Wyatt
- Division of Pulmonary, Critical Care & SleepUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Veterans Affairs Nebraska‐Western Iowa Health Care SystemResearch ServiceOmahaNebraskaUSA
- Department of Environmental, Agricultural and Occupational Health, College of Public HealthUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Oliver W. Schanze
- Division of Allergy & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Amy J. Nelson
- Division of Allergy & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Angela M. Gleason
- Division of Allergy & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Michael J. Duryee
- Veterans Affairs Nebraska‐Western Iowa Health Care SystemResearch ServiceOmahaNebraskaUSA
- Division of Rheumatology & Immunology, Department of Internal Medicine, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Deanna D. Mosley
- Division of Pulmonary, Critical Care & SleepUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Veterans Affairs Nebraska‐Western Iowa Health Care SystemResearch ServiceOmahaNebraskaUSA
| | - Geoffrey M. Thiele
- Veterans Affairs Nebraska‐Western Iowa Health Care SystemResearch ServiceOmahaNebraskaUSA
- Division of Rheumatology & Immunology, Department of Internal Medicine, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Ted R. Mikuls
- Veterans Affairs Nebraska‐Western Iowa Health Care SystemResearch ServiceOmahaNebraskaUSA
- Division of Rheumatology & Immunology, Department of Internal Medicine, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Jill A. Poole
- Division of Allergy & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
43
|
Ghahramani Almanghadim H, Karimi B, Valizadeh S, Ghaedi K. Biological functions and affected signaling pathways by Long Non-Coding RNAs in the immune system. Noncoding RNA Res 2025; 10:70-90. [PMID: 39315339 PMCID: PMC11417496 DOI: 10.1016/j.ncrna.2024.09.001] [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] [Received: 04/14/2024] [Revised: 08/14/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024] Open
Abstract
Recently, the various regulative functions of long non-coding RNAs (LncRNAs) have been well determined. Recently, the vital role of LncRNAs as gene regulators has been identified in the immune system, especially in the inflammatory response. All cells of the immune system are governed by a complex and ever-changing gene expression program that is regulated through both transcriptional and post-transcriptional processes. LncRNAs regulate gene expression within the cell nucleus by influencing transcription or through post-transcriptional processes that affect the splicing, stability, or translation of messenger RNAs (mRNAs). Recent studies in immunology have revealed substantial alterations in the expression of lncRNAs during the activation of the innate immune system as well as the development, differentiation, and activation of T cells. These lncRNAs regulate key aspects of immune function, including the manufacturing of inflammatory molecules, cellular distinction, and cell movement. They do this by modulating protein-protein interactions or through base pairing with RNA and DNA. Here we review the current understanding of the mechanism of action of lncRNAs as novel immune-related regulators and their impact on physiological and pathological processes related to the immune system, including autoimmune diseases. We also highlight the emerging pattern of gene expression control in important research areas at the intersection between immunology and lncRNA biology.
Collapse
Affiliation(s)
| | - Bahareh Karimi
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Sepehr Valizadeh
- Department of Internal Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
44
|
Ananthakrishnan AN, Whelan K, Allegretti JR, Sokol H. Diet and Microbiome-Directed Therapy 2.0 for IBD. Clin Gastroenterol Hepatol 2025; 23:406-418. [PMID: 38992408 DOI: 10.1016/j.cgh.2024.05.049] [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: 03/08/2024] [Revised: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024]
Abstract
Inflammatory bowel disease (IBD) comprises chronic and relapsing disorders of the gastrointestinal tract, characterized by dysregulated immune responses to the gut microbiome. The gut microbiome and diet are key environmental factors that influence the onset and progression of IBD and can be leveraged for treatment. In this review, we summarize the current evidence on the role of the gut microbiome and diet in IBD pathogenesis, and the potential of microbiome-directed therapies and dietary interventions to improve IBD outcomes. We discuss available data and the advantages and drawbacks of the different approaches to manipulate the gut microbiome, such as fecal microbiota transplantation, next-generation and conventional probiotics, and postbiotics. We also review the use of diet as a therapeutic tool in IBD, including the effects in induction and maintenance, special diets, and exclusive enteral nutrition. Finally, we highlight the challenges and opportunities for the translation of diet and microbiome interventions into clinical practice, such as the need for personalization, manufacturing and regulatory hurdles, and the specificity to take into account for clinical trial design.
Collapse
Affiliation(s)
- Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts.
| | - Kevin Whelan
- Department of Nutritional Sciences, King's College London, London, United Kingdom
| | - Jessica R Allegretti
- Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Harry Sokol
- Gastroenterology Department, Centre de Recherche Saint-Antoine, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France; Paris Center for Microbiome Medicine, Fédération Hospitalo-Univeresitaire, Paris, France; Micalis Institute, AgroParisTech, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
45
|
Ekhtiar M, Ghasemi-Dehnoo M, Azadegan-Dehkordi F, Bagheri N. Evaluation of Anti-Inflammatory and Antioxidant Effects of Ferulic Acid and Quinic Acid on Acetic Acid-Induced Ulcerative Colitis in Rats. J Biochem Mol Toxicol 2025; 39:e70169. [PMID: 39957712 DOI: 10.1002/jbt.70169] [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: 11/17/2024] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 02/18/2025]
Abstract
Ulcerative colitis is a chronic inflammatory disease characterized by oxidative stress and the production of pro-inflammatory cytokines. Ferulic acid and quinic acid, two phenolic compounds, are thought to have potent antioxidant and anti-inflammatory properties. This study aimed to investigate the anti-inflammatory and antioxidant effects of ferulic acid and quinic acid in rats with acetic acid (AA)-induced ulcerative colitis. To this end, 64 Wistar rats were randomly divided into eight groups, each consisting of eight rats. AA was administered intrarectally to induce ulcerative colitis. Ferulic acid (20, 40, and 60 mg/kg), quinic acid (10, 30, 60, and 100 mg/kg), and dexamethasone (2 mg/kg) were received daily for five consecutive days. Then, the macroscopic and histopathological changes in the colon tissue were examined. Finally, the tissue levels of heme oxygenase 1 (HO1), nuclear factor erythroid 2-related factor 2 (NRF2), and NAD(P)H quinone dehydrogenase 1 (NQO1) mRNA expression and pro-inflammatory cytokine tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1β) were measured using the quantitative real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA) methods, respectively. AA-induced ulcerative colitis in rats was associated with edema and severe damage to the epithelium, infiltration of inflammatory cells, and the presence of ulcers in the colon tissue. The results showed that rats who were administered AA showed a decrease in the expression of HO-1, Nrf2, and NQO1 and increased protein levels of TNF-α and IL-1β than the control group. Rats were administered ferulic acid, quinic acid and, dexamethasone significantly improved histopathological indices. The expression of HO-1, Nrf2, and NQO1 were upregulated by 60 mg/kg of ferulic acid, 60 and100 mg/kg of quinic acid and, 2 mg/kg of dexamethasone treatment compared to the ulcerative colitis group. The protein levels of TNF-α and IL-1β dose-dependently decreased by ferulic acid and quinic acid treatment compared to the ulcerative colitis group. Ferulic acid and quinic acid effectively reduce inflammation and mucosal damage in rats with ulcerative colitis, especially when administered in high doses. The possible mechanism of anti-inflammatory response by ferulic acid and quinic acid may involve the activating of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Mahsa Ekhtiar
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Maryam Ghasemi-Dehnoo
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Fatemeh Azadegan-Dehkordi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
46
|
Kaur G, Kushwah AS. Sodium orthovanadate protects against ulcerative colitis and associated liver damage in mice: insights into modulations of Nrf2/Keap1 and NF-κB pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1557-1574. [PMID: 39120720 DOI: 10.1007/s00210-024-03335-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024]
Abstract
Ulcerative colitis (UC) is a prominent category of disease that is associated with bowel inflammation, it can occur at any period of life and is prevalently rising on a global scale. Dextran sulfate sodium (DSS) has been extensively used to develop colitis due to its ability to mimic human UC, providing consistent and reproducible inflammation, ulceration, and disruption of the epithelial barrier in the colon. Chronic inflammation in the gut can lead to alterations in the gut-liver axis, potentially impacting liver function over time, while direct evidence linking diversion colitis to liver damage is limited. Thus, the present study aims to assess the gut and liver damage against DSS and the possible molecular mechanisms. Forty-seven animals were randomly assigned to six groups. Ulcerative colitis was induced using 2.5% w/v DSS in three alternate cycles, each lasting 7 days, with 1-week remission periods in between. SOV (5 and 10 mg/kg, orally) and the standard drug 5-aminosalicylic acid (100 mg/kg, orally) were administered from the start of the 2nd DSS cycle until the end of the experiment. Biochemical parameters, ELISA, histopathological, and immunohistochemical analyses have been conducted to assess damage in the colon and liver. SOV significantly reduced colitis severity by lowering the DAI score, oxidative stress markers (LPS, IL-1β, MPO, nitrite), and restoring liver biomarkers (SGPT, SGOT). Histopathological findings supported these protective benefits in the liver and gut. Moreover, immunohistochemical analysis showed SOV enhanced the expression of the cytoprotective mediator Nrf2/Keap-1 and reduced the expression of inflammatory mediators NF-κB and IL-6. Present findings concluded that SOV demonstrated a dose-dependent effect against UC through anti-inflammatory and antioxidant pathways, with the highest dose of SOV 10 mg/kg having more significant (p < 0.001) results than the low dose of 5 mg/kg.
Collapse
Affiliation(s)
- Gurpreet Kaur
- IK Gujral Punjab Technical University, Kapurthala, 144601, Jalandhar, Punjab, India
- Department of Pharmacology, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy (An Autonomous College), Bela, 140111, Ropar, Punjab, India
| | - Ajay Singh Kushwah
- Department of Pharmacology, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy (An Autonomous College), Bela, 140111, Ropar, Punjab, India.
| |
Collapse
|
47
|
Calafat M, Suria C, Mesonero F, de Francisco R, Yagüe Caballero C, de la Peña L, Hernández-Camba A, Marcé A, Gallego B, Martín-Vicente N, Rivero M, Iborra M, Guerra I, Carrillo-Palau M, Madero L, Burgueño B, Monfort D, Torres G, Teller M, Ferrer Rosique JÁ, Vega Villaamil P, Roig C, Ponferrada-Diaz A, Betoré Glaría E, Zabana Y, Gisbert JP, Busquets D, Alcaide N, Camps B, Legido J, González-Vivo M, Bosca-Watts MM, Pérez-Martínez I, Casas Deza D, Guardiola J, Arranz Hernández L, Navarro M, Gargallo-Puyuelo CJ, Cañete F, Mañosa M, Domènech E. HIV Infection Is Associated With a Less Aggressive Phenotype of Inflammatory Bowel Disease: A Multicenter Study of the ENEIDA Registry. Am J Gastroenterol 2025; 120:431-439. [PMID: 39888687 DOI: 10.14309/ajg.0000000000002965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/12/2024] [Indexed: 07/17/2024]
Abstract
INTRODUCTION The coexistence of HIV infection and inflammatory bowel disease (IBD) is uncommon. Data on the impact of HIV on IBD course and its management are scarce. The aim of this study was to describe the IBD phenotype, therapeutic requirements, and prevalence of opportunistic infections (OIs) in IBD patients with a coexistent HIV infection. METHODS Case-control, retrospective study includes all HIV-positive patients diagnosed with IBD in the Nationwide study on genetic and environmental determinants of inflammatory bowel disease registry. Patients with positive HIV serology (HIV-IBD) were compared with controls (HIV seronegative), matched 1:3 by year of IBD diagnosis, age, sex, and type of IBD. RESULTS A total of 364 patients (91 HIV-IBD and 273 IBD controls) were included. In the whole cohort, 58% had ulcerative colitis, 35% had Crohn's disease (CD), and 7% were IBD unclassified. The HIV-IBD group presented a significantly higher proportion of proctitis in ulcerative colitis and colonic location in CD but fewer extraintestinal manifestations than controls. Regarding treatments, nonbiological therapies (37.4% vs 57.9%; P = 0.001) and biologicals (26.4% vs 42.1%; P = 0.007), were used less frequently among patients in the HIV-IBD group. Conversely, patients with HIV-IBD developed more OI than controls, regardless of nonbiological therapy use. In the multivariate analysis, HIV infection (odds ratio 4.765, 95% confidence interval (CI) 2.48-9.14; P < 0.001) and having ≥1 comorbidity (OR 2.445, 95% CI 1.23-4.85; P = 0.010) were risk factors for developing OI, while CD was protective (OR 0.372, 95% CI 0.18-0.78; P = 0.009). DISCUSSION HIV infection seems to be associated with a less aggressive phenotype of IBD and a lesser use of nonbiological therapies and biologicals but entails a greater risk of developing OI.
Collapse
Affiliation(s)
- Margalida Calafat
- Gastroenterology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Gastroenterology Department, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Carles Suria
- Gastroenterology Department, Hospital Clínic Universitari de València, Universitat de València, València, Spain
| | - Francisco Mesonero
- Gastroenterology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Ruth de Francisco
- Gastroenterology Department, Hospital Universitario Central de Asturias (Oviedo), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Carmen Yagüe Caballero
- Gastroenterology Department, Hospital Universitario Miguel Servet (Zaragoza) and Instituto de Investigación Sanitaria de Aragón (IISA), Zaragoza, Spain
| | - Luisa de la Peña
- Gastroenterology Department, Hospital Universitari de Bellvitge (L'Hospitalet de Llobregat), Barcelona, Spain
| | | | - Ainhoa Marcé
- Gastroenterology Department, Hospital Universitari Moisès Broggi, Sant Joan Despí, Spain
| | - Beatriz Gallego
- Gastroenterology Department, Hospital Clínico Universitario «Lozano Blesa» (Zaragoza), Instituto de Investigación Sanitaria, IIS Aragón, Zaragoza, Spain
| | | | - Montserrat Rivero
- Gastroenterology Department, Hospital Universitario Marqués de Valdecilla (Santander), Instituto de Investigación Marqués de Valdecilla IDIVAL, Santander, Spain
| | - Marisa Iborra
- Gastroenterology Department, Hospital Universitari i Politècnic la Fe de València, València, Spain
| | - Iván Guerra
- Gastroenterology Department, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain
| | - Marta Carrillo-Palau
- Gastroenterology Department, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife, Spain
| | - Lucía Madero
- Gastroenterology Department, Hospital General Universitario Dr Balmis de Alicante (Alicante), ISABIAL, Alicante, Spain
| | - Beatriz Burgueño
- Gastroenterology Department, Hospital Universitario Rio Hortega, Valladolid, Spain
| | - David Monfort
- Gastroenterology Department, Consorci Sanitari de Terrassa, Terrassa, Spain
| | - Gisela Torres
- Gastroenterology Department, Hospital Universitari Arnau de Vilanova de Lleida, Lleida, Spain
| | - Marta Teller
- Gastroenterology Department, Althaia, Xarxa Assistencial Universitària de Manresa, Manresa, Spain
| | | | - Pablo Vega Villaamil
- Gastroenterology Department, Complexo Hospitalario Universitario de Ourense, Ourense, Spain
| | - Cristina Roig
- Gastroenterology Department, Hospital Universitari de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Elena Betoré Glaría
- Gastroenterology Department, Hospital Universitario San Jorge, Huesca, Spain
| | - Yamile Zabana
- Gastroenterology Department, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Gastroenterology Department, Hospital Universitari Mútua de Terrassa, Terrassa, Spain
| | - Javier P Gisbert
- Gastroenterology Department, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Gastroenterology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - David Busquets
- Gastroenterology Department, Hospital Universitari Dr. Trueta de Girona, Girona, Spain
| | - Noelia Alcaide
- Gastroenterology Department, Gastroenterology Department, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Blau Camps
- Gastroenterology Department, Hospital de Granollers, Granollers, Spain
| | - Jesús Legido
- Gastroenterology Department, Complejo Asistencial de Segovia, Segovia, Spain
| | | | - Marta Maia Bosca-Watts
- Gastroenterology Department, Hospital Clínic Universitari de València, Universitat de València, València, Spain
| | - Isabel Pérez-Martínez
- Gastroenterology Department, Hospital Universitario Central de Asturias (Oviedo), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Diego Casas Deza
- Gastroenterology Department, Hospital Universitario Miguel Servet (Zaragoza) and Instituto de Investigación Sanitaria de Aragón (IISA), Zaragoza, Spain
| | - Jordi Guardiola
- Gastroenterology Department, Hospital Universitari de Bellvitge (L'Hospitalet de Llobregat), Barcelona, Spain
| | - Laura Arranz Hernández
- Gastroenterology Department, Hospital Universitario Nuestra Señora de Candelaria, Tenerife, Spain
| | - Mercè Navarro
- Gastroenterology Department, Hospital Universitari Moisès Broggi, Sant Joan Despí, Spain
| | - Carla J Gargallo-Puyuelo
- Gastroenterology Department, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Gastroenterology Department, Hospital Clínico Universitario «Lozano Blesa» (Zaragoza), Instituto de Investigación Sanitaria, IIS Aragón, Zaragoza, Spain
| | - Fiorella Cañete
- Gastroenterology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Gastroenterology Department, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Míriam Mañosa
- Gastroenterology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Gastroenterology Department, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Eugeni Domènech
- Gastroenterology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Gastroenterology Department, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
48
|
Fioretti MT, Gianolio L, Armstrong K, Rabone RM, Henderson P, Wilson DC, Russell RK. A decade of real-world clinical experience with 8-week azithromycin-metronidazole combined therapy in paediatric Crohn's disease. J Pediatr Gastroenterol Nutr 2025; 80:300-307. [PMID: 39648957 DOI: 10.1002/jpn3.12430] [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: 09/17/2024] [Revised: 11/01/2024] [Accepted: 11/11/2024] [Indexed: 12/10/2024]
Abstract
OBJECTIVES The aim of our study was to assess the effectiveness and side-effect profile of a combination of azithromycin and metronidazole (CD AZCRO) as alternative induction therapy for 8 weeks in mild to moderately active paediatric Crohn's disease (CD). METHODS We performed a retrospective cohort study (November 2012 to July 2023) of a regional paediatric inflammatory bowel disease service. Disease activity, faecal calprotectin (FC), C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), haematological parameters and albumin were collected at baseline, 8 and 16 weeks. At Week 8, patients were divided based on (paediatric Crohn's disease activity index) score and inflammatory markers (blood and stool) into: Group 1 clinical remission and Group 2 non-remission. RESULTS A total of 48 patients were initially identified of whom 44 were included in the intention-to-treat analysis. After 8 weeks, the overall remission rate was 64%. Of the 38 patients who completed the CD AZCRO course, 28 patients (74%) entered remission (Group 1) and 10 (26%) did not (Group 2). At baseline a shorter disease duration, low weight z score and higher inflammatory burden (ESR, platelets and FC levels) were observed in Group 2. After 8 weeks, Group 1 showed improved CRP levels and higher albumin and haemoglobin levels than Group 2. Median FC declined significantly from 650 mcg/g at baseline to 190 mcg/g at Week 8 in Group 1 (p < 0.001). At 16 weeks, 23/28 patients (82%) continued in clinical remission. Nausea and vomiting were reported in 4/44 patients. CONCLUSIONS Our real-world data demonstrate that CD AZCRO represents an alternative induction therapy for mild to moderate paediatric CD.
Collapse
Affiliation(s)
- Maria Teresa Fioretti
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, UK
| | - Laura Gianolio
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, UK
| | - Katherine Armstrong
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, UK
| | - Rosalind M Rabone
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, UK
| | - Paul Henderson
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, UK
- Child Life and Health, University of Edinburgh, Royal Hospital for Children and Young People, Edinburgh, UK
| | - David C Wilson
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, UK
- Child Life and Health, University of Edinburgh, Royal Hospital for Children and Young People, Edinburgh, UK
| | - Richard K Russell
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, UK
- Child Life and Health, University of Edinburgh, Royal Hospital for Children and Young People, Edinburgh, UK
| |
Collapse
|
49
|
Wu D, Huang Q, Xu Y, Cao R, Yang M, Xie J, Zhang D. Clinical efficacy and future application of indigo naturalis in the treatment of ulcerative colitis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118782. [PMID: 39236777 DOI: 10.1016/j.jep.2024.118782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/03/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by non-specific inflammation. Managing UC presents significant challenges due to its chronic nature and high recurrence rates. Indigo naturalis has emerged as a potential therapeutic agent in clinical UC treatment, demonstrating advantages in alleviating refractory UC and maintaining remission periods compared to other therapeutic approaches. AIM OF REVIEW This review aims to elucidate the potential mechanisms underlying the therapeutic effects of indigo naturalis in UC treatment, assess its clinical efficacy, advantages, and limitations, and provide insights into methods and strategies for utilizing indigo naturalis in UC management. MATERIALS AND METHODS Comprehensive data on indigo naturalis were collected from reputable online databases including PubMed, GreenMedical, Web of Science, Google Scholar, China National Knowledge Infrastructure Database, and National Intellectual Property Administration. RESULTS Clinical studies have demonstrated that indigo naturalis, either alone or in combination with other drugs, yields favorable outcomes in UC treatment. Its mechanisms of action involve modulation of the AHR receptor, anti-inflammatory properties, regulation of intestinal flora, restoration of the intestinal barrier, and modulation of immunity. Despite its efficacy in managing refractory UC and prolonging remission periods, indigo naturalis treatment is associated with adverse reactions, quality variations, and inadequate pharmacokinetic investigations. CONCLUSION The therapeutic effects of indigo naturalis in UC treatment are closely linked to its ability to regulate the AHR receptor, exert anti-inflammatory effects, mcodulate intestinal flora, restore the intestinal barrier, and regulate immunity. Addressing the current shortcomings, including adverse reactions, quality control issues, and insufficient pharmacokinetic data, is crucial for optimizing the clinical utility of indigo naturalis in UC management. By refining patient-centered treatment strategies, indigo naturalis holds promise for broader application in UC treatment, thereby alleviating the suffering of UC patients.
Collapse
Affiliation(s)
- Dianzhen Wu
- Sichuan Medical Products Administration, Chengdu, 610017, China
| | - Qi Huang
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yingbi Xu
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ruiyi Cao
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ming Yang
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Jin Xie
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Dingkun Zhang
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Sichuan Provincial Engineering Research Center of Innovative Re-development of Famous Classical Formulas, Tianfu TCM Innovation Harbour, Chengdu University of Traditional Chinese Medicine, Chengdu, 611930, China.
| |
Collapse
|
50
|
Attauabi M, Madsen GR, Bendtsen F, Burisch J, Seidelin JB. The Role of Environmental Factors Prior to Diagnosis on the Activity and Severity of Inflammatory Bowel Diseases-Results From the Prospective Population-Based Copenhagen Inflammatory Bowel Disease Inception Cohort. United European Gastroenterol J 2025. [PMID: 39878314 DOI: 10.1002/ueg2.12737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/30/2024] [Accepted: 10/21/2024] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND The influence of environmental factors on the severity of early inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), is unclear. Herein, we aimed to investigate the role of environmental factors in the initial phenotype, activity, and severity of IBD. METHODS Copenhagen IBD Inception Cohort is a prospective population-based cohort of patients with newly diagnosed IBD between May 2021 and May 2023. Data on environmental factors were captured at IBD diagnosis using International Organisation of IBD (IOIBD) and HeartDiet questionnaires. Environmental factors' influence on outcome was analyzed and odds ratios (aOR) were adjusted for age, gender, and disease characteristics (adjusted OR, aOR [95% confidence interval]). RESULTS In total, 208 and 128 patients with incident UC and CD, respectively, were included. Active smoking was associated with increased risk of CD-related hospitalization (aOR = 2.84 [1.03; 7.88]) and stricturing phenotype (aOR = 5.28 [1.76; 15.85]) but lower risk of severe UC course (aOR = 0.28 [0.08; 0.95]). Further, previous smoking was not associated with negative effects in patients with CD in terms of early need for biologics, surgery, or hospitalization. In terms of diets, daily consumption of fruits (aOR = 0.27 [0.07; 0.99]) or vegetables (aOR = 0.27 [0.09; 0.80]) was inversely associated with stricturing CD, whereas whole meal bread was associated with reduced risk of severe CD activity (aOR = 0.40 [0.16; 0.98]). CONCLUSIONS This prospective population-based study highlighted several environmental factors associated with the initial severity and activity of IBD, emphasizing their pivotal role in the initial disease burden and giving guidance to personalized patient counseling.
Collapse
Affiliation(s)
- Mohamed Attauabi
- Department of Gastroenterology and Hepatology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- Gastrounit, Medical Section, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents, and Adults, Hvidovre Hospital, Hvidovre, Denmark
| | - Gorm Roager Madsen
- Gastrounit, Medical Section, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents, and Adults, Hvidovre Hospital, Hvidovre, Denmark
| | - Flemming Bendtsen
- Gastrounit, Medical Section, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents, and Adults, Hvidovre Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johan Burisch
- Gastrounit, Medical Section, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents, and Adults, Hvidovre Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Benedict Seidelin
- Department of Gastroenterology and Hepatology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|