151
|
Wang M, Zheng C, Zhou F, Ying X, Zhang X, Peng C, Wang L. Iron and Inflammatory Cytokines Synergistically Induce Colonic Epithelial Cell Ferroptosis in Colitis. J Gastroenterol Hepatol 2025; 40:666-676. [PMID: 39586593 DOI: 10.1111/jgh.16826] [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: 01/28/2024] [Revised: 10/05/2024] [Accepted: 11/04/2024] [Indexed: 11/27/2024]
Abstract
Inflammatory bowel disease (IBD) is an inflammatory disease that occurs to the intestinal tract. Many patients with IBD often develop anemia and often receive oral iron supplementation. Many of them develop non-compliance with oral iron therapy, but the mechanisms are not well understood. We interrogated whether colonic epithelial iron overload impacts cell viability and disease severity. We observed increased expression of iron importers and iron accumulation in mature colonocytes in dextran sulfate sodium (DSS)-induced acute colitis and in humans with active colitis. Administration of hepcidin increased epithelial iron overload and aggravated colonic inflammation in DSS-treated mice and IL10-/- mice. Hepcidin-induced iron accumulation increased colonic epithelial death, which was prevented by treatment with Trolox, a vitamin E analog and a scavenger of lipid peroxides. By using cultured Caco-2 cells, we showed that iron and inflammatory cytokines (TNF-α and IL-1β) induced a synergistic increase in the number of necrotic cells. We then showed that the combined treatment by hepcidin and cytokines increased labile iron content and lipid peroxidation in Caco-2 cells. Moreover, liproxstatin-1, a ferroptosis inhibitor, and deferoxamine, an iron chelator, both abolished the hepcidin/cytokines induced death of Caco-2 cells, suggesting ferroptosis. We further elucidated that inflammatory cytokines promote lipid peroxidation and ferroptosis by inducing NOX1-dependent exhaustion of reduced glutathione (GSH). Collectively, our findings demonstrate that the inflammatory context predisposes colonic epithelial cells to iron overload mediated ferroptosis, exacerbating colonic inflammation.
Collapse
Affiliation(s)
- Mo Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Chang Zheng
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Fan Zhou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Xie Ying
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Xiaoqi Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Chunyan Peng
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Lei Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
152
|
Mokaram Doust Delkhah A. Blood transcriptomics identifies FEZ1 as a potential biomarker for inflammatory bowel disease. Comput Biol Med 2025; 187:109742. [PMID: 39894009 DOI: 10.1016/j.compbiomed.2025.109742] [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: 11/19/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/04/2025]
Abstract
INTRODUCTION While the global burden of inflammatory bowel diseases (IBD) is increasing, the identification of novel therapeutic targets and biomarkers is of significant importance. In particular, blood transcriptomes provide a non-invasive source for biomarker discovery. Therefore, this study aimed to identify potential blood markers for IBD. METHODS By employing an integrated transcriptomics approach, four datasets obtained from blood specimens of patients with IBD were analyzed (GSE119600, GSE94648, GSE86434, and GSE71730). After determining differentially expressed genes (DEGs) in IBD, a protein-protein interaction (PPI) network was constructed, and regulatory miRNAs targeting hub genes were identified. Weighted gene co-expression network analysis (WGCNA) was carried out to determine IBD-specific modules. Subsequently, converging results from differential expression analysis and WGCNA were subjected to random forest (RF) decision tree-based and LASSO regression methods. Lastly, the diagnostic efficacy of genes highlighted by both machine learning methods was measured using receiver operating characteristic (ROC) analysis in the integrated dataset, in each individual dataset separately, and in external datasets (GSE276395, GSE169568, GSE112057, GSE100833, GSE33943, and GSE3365). RESULTS Downregulated TNF was identified as the central hub gene of the PPI network, and PRF1 was the only gene identified as a hub gene in a co-expressed gene module enriched in IBD. Following the identification of FEZ1 and NLRC5 among the top 10 genes by both RF and LASSO, ROC analysis demonstrated their acceptable diagnostic efficacy in the integrated data. However, only FEZ1 was considered a potential biomarker based on replication of the results in the external datasets. CONCLUSIONS The results of the present study suggest FEZ1 as a potential blood biomarker for IBD. While autophagy is currently the most convincing explanation for the involvement of FEZ1 in IBD, further investigations are required to elucidate its immunological role.
Collapse
|
153
|
Karra DA, Lidbury JA, Suchodolski JS, Pitropaki M, Newman S, Steiner JM, Xenoulis PG. Fecal and Serum Calprotectin Concentrations in Cats With Chronic Enteropathies Before and During Treatment. J Vet Intern Med 2025; 39:e70067. [PMID: 40110650 PMCID: PMC11923564 DOI: 10.1111/jvim.70067] [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: 10/16/2024] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
ABSTRACTBackgroundChronic enteropathies (CE) are common in cats. Reliable biomarkers that can distinguish different causes and predict or monitor responses to treatment are currently lacking.Hypothesis/ObjectivesEvaluate calprotectin concentrations in serum and feces as potential biomarkers in cats with CE.AnimalsForty‐three cats with either chronic inflammatory enteropathy (CIE; n = 25) or small cell gastrointestinal lymphoma (SCGL; n = 18) and 36 healthy cats were prospectively enrolled.MethodsFecal and serum calprotectin concentrations were determined before and during treatment. Cats with CIE were treated with diet, prednisolone, or diet and prednisolone, and cats with SCGL were treated with prednisolone plus chlorambucil with or without diet.ResultsCompared to controls, fecal calprotectin concentration was significantly higher in cats with CE (median, ≤ 161 ng/g; range, ≤ 161–2827 vs. median, ≤ 161 ng/g; range, ≤ 161–790; p = 0.01). No significant differences were found in fecal (median ≤ 161 ng/g; range, ≤ 161–1920 vs. median, 189 ng/g; range, ≤ 161–2827; p = 0.3) or serum calprotectin (median, ≤ 1291 mg/L; range, ≤ 1291–15 358 vs. median, ≤ 1291 mg/L; range, ≤ 1291–6422; p = 0.99) between cats with CIE and cats with SCGL. Fecal calprotectin was significantly decreased after treatment in cats with CE (median, ≤ 161 ng/g; range, ≤ 161–1897 vs. median, ≤ 161 ng/g; range, ≤ 161–656; p = 0.02).ConclusionsFecal calprotectin concentration might be a good biomarker for diagnosis and treatment monitoring for a subset of cats with CE. Serum calprotectin concentrations do not seem to be useful for diagnosis or treatment monitoring in cats with CE. Neither fecal nor serum calprotectin concentrations could differentiate cats with CIE from cats with SCGL.
Collapse
Affiliation(s)
- Dimitra A Karra
- Clinic of Medicine, Faculty of Veterinary Science, University of Thessaly, Karditsa, Greece
| | - Jonathan A Lidbury
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| | - Jan S Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| | - Matina Pitropaki
- Clinic of Medicine, Faculty of Veterinary Science, University of Thessaly, Karditsa, Greece
| | | | - Jeorg M Steiner
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| | - Panagiotis G Xenoulis
- Clinic of Medicine, Faculty of Veterinary Science, University of Thessaly, Karditsa, Greece
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
154
|
Alhadab A, Almarhoon A, AlAlwan A, Hammo A. Clinical effectiveness and safety of ustekinumab in youth with refractory inflammatory bowel disease: A retrospective cohort study. Saudi J Gastroenterol 2025; 31:59-67. [PMID: 38597337 PMCID: PMC11978247 DOI: 10.4103/sjg.sjg_7_24] [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: 01/04/2024] [Revised: 02/22/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) incidence and prevalence has been increasing worldwide. Limited data exists on the effectiveness of ustekinumab (UST) in children. We aimed to describe the effectiveness and safety of UST in pediatric patients with IBD. METHODS A single-center retrospective study was conducted between January 2017 and February 2022. The study included patients ≤16 years of age who were treated with UST and followed up for ≥1 year. Clinical remission was defined as a score of the Pediatric Crohn's Disease (CD) and Pediatric Ulcerative Colitis (UC) Activity Indices ≤10 at week 52. RESULTS Thirteen patients who had failed anti-tumor necrosis factor-α (anti-TNFα) therapy were included, eight (61.5%) with CD and five (38.5%) with UC. The median age was 13 years (interquartile range [IQR]: 11.5 to 14). UST treatment was initiated at a median age of 3 years (IQR: 2.3 to 7) after diagnosis. Ten patients (76.9%) achieved clinical remission. There were no statistically significant differences in characteristics between patients who achieved and did not achieve clinical remission. Biochemical remission (BioR) was achieved in six patients (46.2%). Body mass index (BMI) significantly improved, C-reactive protein (CRP) significantly decreased, and the need for corticosteroids significantly decreased in the remission group. Endoscopy conducted post-treatment in seven patients confirmed remission in six patients. Adverse events included two cases of infection and one of headache. CONCLUSIONS UST was effective as a secondary biologic therapy for the induction and maintenance of remission in patients with anti-TNFα refractory IBD. At one year, 84% of patients remained on UST with no severe adverse reactions reported.
Collapse
Affiliation(s)
- Abdulhamid Alhadab
- Department of Pediatrics, Division of Pediatric Gastroenterology and Nutrition, McMaster University, Hamilton, Canada
- Department of Pediatrics, Division of Pediatric Gastroenterology and Nutrition, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Amal Almarhoon
- Department of Pediatrics, Division of Pediatric Gastroenterology and Nutrition, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Amena AlAlwan
- Department of Pediatrics, Division of Pediatric Gastroenterology and Nutrition, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - AbdelHai Hammo
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Arkansas for Medical Sciences, USA
| |
Collapse
|
155
|
Schanne G, Vincent A, Chain F, Ruffié P, Carbonne C, Quévrain E, Mathieu E, Balfourier A, Bermúdez-Humarán LG, Langella P, Thenet S, Carrière V, Hammoudi N, Svrcek M, Demignot S, Seksik P, Policar C, Delsuc N. SOD mimics delivered to the gut using lactic acid bacteria mitigate the colitis symptoms in a mouse model of inflammatory bowel diseases. Free Radic Res 2025; 59:262-273. [PMID: 40079422 DOI: 10.1080/10715762.2025.2478121] [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: 09/01/2024] [Revised: 02/06/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
Inflammatory bowel diseases (IBD), which include Crohn's disease and ulcerative colitis, represent a global health issue as a prevalence of 1% is expected in the western world by the end of this decade. These diseases are associated with a high oxidative stress that induces inflammatory pathways and severely damages gut tissues. IBD patients suffer from antioxidant defenses weakening, through, for instance, an impaired activity of superoxide dismutases (SOD)-that catalyze the dismutation of superoxide-or other endogenous antioxidant enzymes including catalase and glutathione peroxidase. Manganese complexes mimicking SOD activity have shown beneficial effects on cells and murine models of IBD. However, efficient SOD mimics are often manganese complexes that can suffer from decoordination and thus inactivation in acidic stomachal pH. To improve their delivery in the gut after oral administration, two SOD mimics Mn1 and Mn1C were loaded into lactic acid bacteria that serve as delivery vectors. When orally administrated to mice suffering from a colitis, these chemically modified bacteria (CMB) showed protective effects on the global health status of mice. In addition, they have shown beneficial effects on lipocalin-2 content and intestinal permeability. Interestingly, mRNA SOD2 content in colon homogenates was significantly decreased upon mice feeding with CMB loaded with Mn1C, suggesting that the beneficial effects observed may be due to the release of the SOD mimic in the gut that complement for this enzyme. These CMB represent new efficient chemically modified antioxidant probiotics for IBD treatment.
Collapse
Affiliation(s)
- Gabrielle Schanne
- Laboratoire Chimie Pysique et Chimie du Vivant, CPCV UMR8228, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris, France
- Centre de Recherche Saint Antoine, INSERM, UMRS 938, Microbiota, Intestine and Inflammation Team, Paris, France
| | - Amandine Vincent
- Laboratoire Chimie Pysique et Chimie du Vivant, CPCV UMR8228, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris, France
| | - Florian Chain
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| | - Pauline Ruffié
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| | - Célia Carbonne
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| | - Elodie Quévrain
- Laboratoire Chimie Pysique et Chimie du Vivant, CPCV UMR8228, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris, France
- Centre de Recherche Saint Antoine, INSERM, UMRS 938, Microbiota, Intestine and Inflammation Team, Paris, France
| | - Emilie Mathieu
- Laboratoire Chimie Pysique et Chimie du Vivant, CPCV UMR8228, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris, France
| | - Alice Balfourier
- Laboratoire Chimie Pysique et Chimie du Vivant, CPCV UMR8228, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris, France
| | | | - Philippe Langella
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| | - Sophie Thenet
- Centre de Recherche Saint Antoine, INSERM, UMRS 938, Microbiota, Intestine and Inflammation Team, Paris, France
- EPHE, PSL University, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, APHP, Paris, France
| | - Véronique Carrière
- Centre de Recherche Saint Antoine, INSERM, UMRS 938, Microbiota, Intestine and Inflammation Team, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, APHP, Paris, France
| | - Nassim Hammoudi
- Department of Gastroenterology, Hôpital Saint-Louis, AP-HP, INSERM U1160, Paris, France
| | - Magali Svrcek
- Centre de Recherche Saint Antoine, INSERM, UMRS 938, Microbiota, Intestine and Inflammation Team, Paris, France
| | - Sylvie Demignot
- Centre de Recherche Saint Antoine, INSERM, UMRS 938, Microbiota, Intestine and Inflammation Team, Paris, France
- EPHE, PSL University, Paris, France
| | - Philippe Seksik
- Centre de Recherche Saint Antoine, INSERM, UMRS 938, Microbiota, Intestine and Inflammation Team, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, APHP, Paris, France
- Gastroenterology Department, Saint-Antoine Hospital, Sorbonne Université, APHP, Paris, France
| | - Clotilde Policar
- Laboratoire Chimie Pysique et Chimie du Vivant, CPCV UMR8228, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris, France
| | - Nicolas Delsuc
- Laboratoire Chimie Pysique et Chimie du Vivant, CPCV UMR8228, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris, France
| |
Collapse
|
156
|
Yang X, Li R. Letter: The Hidden Truth of IBD Risk-From Antibiotics to Environmental Factors. Aliment Pharmacol Ther 2025; 61:1090-1091. [PMID: 39954249 DOI: 10.1111/apt.18517] [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: 12/31/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 02/17/2025]
Affiliation(s)
- Xueneng Yang
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ruijuan Li
- Department of Burn, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
157
|
Däbritz J, Classen M, Krohn K, Krahl A, Buderus S, Lainka E, de Laffolie J, Posovszky C. [Position paper of the Society for Paediatric Gastroenterology and Nutrition (GPGE) on the off-label use of biologics and signal inhibitors in children and adolescents with IBD that have already been approved for adults]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2025; 63:255-268. [PMID: 39961333 PMCID: PMC11893210 DOI: 10.1055/a-2474-3104] [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: 08/09/2024] [Accepted: 11/07/2024] [Indexed: 03/12/2025]
Abstract
Therapy for children and adolescents with chronic inflammatory bowel disease (IBD) is basically no different from that for adult patients. However, of the steadily increasing number of biologics and signalling inhibitors for adults, only two TNFα antibodies are currently approved in Germany for the treatment of IBD from the age of 6. This means that a large proportion of the drugs authorised for adults with IBD are not available for children and adolescents with moderate to severe disease. The small number of approved drugs also makes it difficult to achieve the prognostically important goal of achieving a sustained remission of IBD soon after diagnosis, which is characterised by the patient being free of symptoms and also the objectifiable goal of mucosa healing. This position paper is intended to present the current study situation on the drug treatment of children and adolescents with IBD outside the age limit and to serve as a basis for information and decision-making for the Medical Service in the assessment of individual case applications as well as for the treating physicians, the cost bearers, health policy and social court decision-makers.
Collapse
Affiliation(s)
- Jan Däbritz
- Universitätsmedizin Greifswald Klinik und Poliklinik für Kinder und Jugendmedizin, Greifswald, Deutschland
- Kinder- und Jugendklinik, Klinikum Westbrandenburg, Potsdam, Deutschland
| | - Martin Classen
- Kindergastroenterologische Praxis M. Schacht, Bremen, Deutschland
| | - Kathrin Krohn
- Integriertes Sozialpädiatrisches Zentrum im Dr. von Haunerschen Kinderspital, Ludwig-Maximilians-Universität München, München, Deutschland
| | - Andreas Krahl
- Klinik für Kinder- und Jugendmedizin, Kinder-Gastroenterologie, Sana Klinikum Offenbach GmbH, Offenbach, Deutschland
| | - Stephan Buderus
- GFO Kliniken Bonn Betriebsstätte St. Marien, Bonn, Deutschland
| | - Elke Lainka
- Zentrum für Kinder- und Jugendmedizin, Kinderklinik II, Pädiatrische Gastroenterologie, Hepatologie und Lebertransplantation, Universitätsmedizin Essen, Essen, Deutschland
| | - Jan de Laffolie
- Abteilung Allgemeine Pädiatrie und Neonatologie, Universitätsklinikum Giessen Zentrum für Kinderheilkunde und Jugendmedizin, Giessen, Deutschland
| | - Carsten Posovszky
- Gastroenterologie, Hepatologie und Ernährung, Universitäts-Kinderspital Zürich, Zürich, Schweiz
- Klinik für Kinder- und Jugendmedizin, University Ulm Medical Centre, Ulm, Deutschland
- Universität Zürich, Zürich, Schweiz
| |
Collapse
|
158
|
O'Donnell JEM, Leach ST, Bowcock NL, Chen S, Gupta N, Jiang K, Lopez RN, Messenger R, Nahidi L, Shapiro A, Day AS, Lemberg DA. Daily Vitamin D3 Versus Stoss Vitamin D3 for Correction of 25OHD Deficiency in Children with Inflammatory Bowel Disease, a Randomised Controlled Trial. Dig Dis Sci 2025:10.1007/s10620-025-08913-3. [PMID: 40021606 DOI: 10.1007/s10620-025-08913-3] [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: 07/31/2024] [Accepted: 02/03/2025] [Indexed: 03/03/2025]
Abstract
INTRODUCTION Vitamin D deficiency is common in Paediatric Inflammatory Bowel Disease (PIBD) and has been implicated in disease pathogenesis and disease exacerbation. Current guidelines recommend oral vitamin D supplementation when 25OHD levels are below 50 nmol/L. Supplementation comes in two forms: either a daily supplement of a low dose of vitamin D3 (2000 IU) for several months or a single high dose of oral vitamin D3-termed 'stoss' therapy, with no consensus regarding optimum treatment. METHODS A randomised controlled trial was conducted in children with a prior diagnosis of PIBD with 25OHD deficiency (< 50 nmol/L), comparing 2000 IU oral D3 daily to a stoss protocol (oral D3 dosage 400,000 IU for 3-12 years of age or 800,000 IU for > 12 years). Children were followed for 12 months, with biochemistry (25OHD, calcium, magnesium, phosphate, parathyroid hormone, haemoglobin, haematocrit, platelets, albumin), stool markers (calprotectin, S100A12), anthropometrics (weight, height, body mass index) as well as clinical disease indices (Paediatric Crohn's Disease Activity Index, Paediatric Ulcerative Colitis Activity Index) and medication use collected at 3, 6, 9 and 12 months. RESULTS 74 children aged 5-18 years completed the study. Both 2000 IU daily and stoss protocol significantly increased 25OHD from baseline values at 3, 6, 9 and 12 months. One patient randomised to stoss protocol had a 25OHD level of 263 nmol/L with normal serum calcium. There was no difference in biochemical, stool or clinical markers between groups at any time point, nor was there any correlation between 25OHD level and calprotectin or 25OHD level and clinical disease activity scores. CONCLUSION Stoss protocol was non-inferior to 2000 IU daily vitamin D3 in raising 25OHD levels at 12 months. There was also no difference between 25OHD levels at 3, 6 and 9 months between groups.
Collapse
Affiliation(s)
- Jonathan E M O'Donnell
- Discipline of Paediatrics, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia.
- Department of Gastroenterology, Sydney Children's Hospital, Randwick, Australia.
| | - Steven T Leach
- Discipline of Paediatrics, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia
| | - Nerissa L Bowcock
- Discipline of Paediatrics, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia
| | - Siying Chen
- Discipline of Paediatrics, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia
- Department of Gastroenterology, Sydney Children's Hospital, Randwick, Australia
| | - Nitin Gupta
- Discipline of Paediatrics, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia
- Department of Gastroenterology, Sydney Children's Hospital, Randwick, Australia
| | - Kevin Jiang
- Discipline of Paediatrics, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia
| | - Robert N Lopez
- University of Auckland, Auckland, New Zealand
- Starship Children's Health, Auckland, New Zealand
| | - Rachel Messenger
- Discipline of Paediatrics, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia
| | - Lily Nahidi
- Discipline of Paediatrics, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia
| | - Amanda Shapiro
- Discipline of Paediatrics, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia
- Department of Paediatrics, University of Otago Christchurch, Christchurch, New Zealand
| | - Andrew S Day
- Discipline of Paediatrics, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia
- Department of Gastroenterology, Sydney Children's Hospital, Randwick, Australia
| | - Daniel A Lemberg
- Discipline of Paediatrics, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia
- Department of Gastroenterology, Sydney Children's Hospital, Randwick, Australia
| |
Collapse
|
159
|
Katsumi Y, Nishimura Y, Goto S, Ozawa S, Nishiura T, Kotera A, Kawahara Y, Higashikawa S, Iwasaki R, Toriiminami Y, Asai N, Fujita N. Case Report: HLA-DRB1 04:01 found in a child with adenovirus type 2 -linked hepatitis. Front Immunol 2025; 16:1544622. [PMID: 40079016 PMCID: PMC11896997 DOI: 10.3389/fimmu.2025.1544622] [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: 12/13/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Since 2022, cases of hepatitis of unknown origin have been reported in children worldwide. Adeno-associated virus type 2 (AAV2) was identified as a cause, with most affected children having the HLA-DRB1 04:01 genotype. In this study, we hypothesized that HLA-DRB1 04:01 in the host may also be a potential predisposing factor of acute hepatitis caused by other viruses. We report a case that met the definition of severe hepatitis of unknown cause in a child; adenovirus type 2 (AV2) was detected in her specimens. The patient was a 1-year-old girl who visited a doctor because of fever occurring 1-2 days per week, respiratory symptoms, and diarrhea. One month later, the patient was referred to our hospital because of prolonged elevated liver enzyme concentrations. Two weeks after the initial visit, her aspartate aminotransferase (AST) and alanine aminotransferase (ALT) concentrations increased to 1558 and 1843 IU/L, respectively. The patient's liver enzyme concentrations decreased markedly with only observation and intravenous hydration during hospitalization within a few days. Thereafter, hepatic enzymes were transiently elevated with each common cold, but all recovered spontaneously. The adenovirus (AV) antibody levels increased substantially 2 weeks after admission. The patient's human leukocyte antigen (HLA) was determined to be of the DRB1 04:01 genotype. The presence of HLA-DRB1 04:01 is consistent with that reported in pediatric patients with AAV2 hepatitis in the United Kingdom, indicating that it may have been involved in the host immune response and acute hepatitis in this child. HLA-DRB1 04:01 may predispose children to acute hepatitis from various viruses, including AV2, AAV2, and possibly respiratory viruses, which requires clinical attention.
Collapse
Affiliation(s)
- Yoshiki Katsumi
- Department of Pediatrics, Kyoto Saiseikai Hospital, Kyoto, Japan
| | - Yui Nishimura
- Department of Pediatrics, Kyoto Saiseikai Hospital, Kyoto, Japan
| | - Sachiko Goto
- Department of Pediatrics, Kyoto Saiseikai Hospital, Kyoto, Japan
| | - Seiichiro Ozawa
- Department of Pediatrics, Kyoto Saiseikai Hospital, Kyoto, Japan
| | | | - Akira Kotera
- Kyoto Prefectural Institute of Public Health and Environment, Kyoto, Japan
| | - Yoshiyuki Kawahara
- Kyoto Prefectural Institute of Public Health and Environment, Kyoto, Japan
| | - Shiori Higashikawa
- Kyoto Prefectural Institute of Public Health and Environment, Kyoto, Japan
| | - Rina Iwasaki
- Kyoto Prefectural Institute of Public Health and Environment, Kyoto, Japan
| | - Yutaka Toriiminami
- Kyoto Prefectural Institute of Public Health and Environment, Kyoto, Japan
| | - Norio Asai
- Kyoto Prefectural Institute of Public Health and Environment, Kyoto, Japan
| | - Naohisa Fujita
- Kyoto Prefectural Institute of Public Health and Environment, Kyoto, Japan
| |
Collapse
|
160
|
Stenke E, Alhassan D, Moclair M, Cooper S, Dominika A, Lang C, Quinn S, Broderick A, Fitzpatrick E, Bourke B, Stallard L, Hussey S. Higher-Dose Infliximab Induction Achieves Better Maintenance Trough Levels in a National Pediatric IBD Cohort-A Retrospective Study. Inflamm Bowel Dis 2025:izaf031. [PMID: 40036162 DOI: 10.1093/ibd/izaf031] [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: 11/14/2024] [Indexed: 03/06/2025]
Abstract
BACKGROUND Proactive drug monitoring in our national cohort of children with inflammatory bowel disease suggested that 5 mg/kg dosing of infliximab (IFX) was associated with inadequate trough levels (TLs), thus we commenced empiric 10 mg/kg dosing in 2019. METHODS This was a retrospective study of patients commenced on 5 or 10 mg/kg IFX between 2018 and 2020. The primary outcome was pre-fourth TL ≥5 µg/mL. Data source verification and analysis were undertaken retrospectively. RESULTS During the study period, 122 patients were commenced on IFX and eligible for inclusion. Of these, 72% were male, 76% had Crohn's disease (CD), 23% had ulcerative colitis; mean (standard deviation [SD]) age at diagnosis was 11.8 (2.7) years; 60 commenced 5 mg/kg. Baseline parameters were comparable between groups. Fewer patients in the 5 mg/kg than 10 mg/kg group had pre-fourth TLs ≥5 µg/mL (6/48 [12.5%] vs 28/50 [56%], P < .001; mean [SD] TL 3.5 [6.3] vs 10.0 [9.9], P < .001). The 5 mg/kg group was less likely to have target pre-third TLs (6% vs 80%, P < .001); more likely to have treatment escalation (78% vs 45%, P < .001), and less likely to de-escalate (10% vs 29% P = .008). Proactive therapeutic dose monitoring (TDM) was practiced throughout the study period and mean (SD) IFX exposure per kg/week was similar in both groups by 1 year (1.4 [0.43] vs 1.56 [0.56] mg/kg/week, P = .137), as were infliximab durability and clinical outcomes. CONCLUSIONS Our real-world data show that 5 mg/kg infliximab induction does not achieve target TLs in most children with CD. Higher-dose regimens should be considered, especially if proactive therapeutic drug monitoring is not available.
Collapse
Affiliation(s)
- Emily Stenke
- National Centre for Pediatric Gastroenterology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Dahlal Alhassan
- National Centre for Pediatric Gastroenterology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Molly Moclair
- National Centre for Pediatric Gastroenterology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Sarah Cooper
- National Centre for Pediatric Gastroenterology, Children's Health Ireland at Crumlin, Dublin, Ireland
- The DOCHAS Study, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Anna Dominika
- National Centre for Pediatric Gastroenterology, Children's Health Ireland at Crumlin, Dublin, Ireland
- The DOCHAS Study, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Ciara Lang
- National Centre for Pediatric Gastroenterology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Shoana Quinn
- National Centre for Pediatric Gastroenterology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Annemarie Broderick
- National Centre for Pediatric Gastroenterology, Children's Health Ireland at Crumlin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Emer Fitzpatrick
- National Centre for Pediatric Gastroenterology, Children's Health Ireland at Crumlin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Billy Bourke
- National Centre for Pediatric Gastroenterology, Children's Health Ireland at Crumlin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Lorraine Stallard
- National Centre for Pediatric Gastroenterology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Séamus Hussey
- National Centre for Pediatric Gastroenterology, Children's Health Ireland at Crumlin, Dublin, Ireland
- The DOCHAS Study, Children's Health Ireland at Crumlin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Pediatrics, Royal College of Surgeons of Ireland, Dublin, Ireland
| |
Collapse
|
161
|
Genovese D, Brinch D, Muscarella S, Saladino M, Carrozza L, Cunsolo C, Sanfilippo GL, Amodio E, Cappello M, Ferraro D. Neutralizing Antibody Response to SARS-CoV-2 Variants After Two mRNA COVID-19 Vaccine Doses in a Cohort of Patients with Inflammatory Bowel Disease from a Southern Italy Tertiary Hospital. Healthcare (Basel) 2025; 13:508. [PMID: 40077070 PMCID: PMC11898406 DOI: 10.3390/healthcare13050508] [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: 12/16/2024] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction: Inflammatory bowel diseases (IBDs) require immunosuppressive drugs like biologics. All IBD patients, including those on biological therapy, should be vaccinated against COVID-19, according to the ECCO recommendations. IBD patients on anti-TNF treatment exhibited lower COVID-19 vaccine responses; however, SARS-CoV-2 variant neutralizing antibody titers have been seldom studied. Methods: IBD patients and healthcare professionals (control group) were tested for COVID-19 vaccine immunogenicity by neutralizing antibody titers against Wild-Type SARS-CoV-2 and its variants. IBD patients were assigned to no treatment/mesalamine, anti-TNF biologic therapy, or non-anti-TNF biologic therapy. The study was performed in a tertiary hospital in Palermo, Sicily, from May to July 2021. Results: In total, 107 IBD patients and 41 healthcare workers were enrolled. A total of 46 patients received mesalamine or no medication, 28 received anti-TNF biologics, and 33 received non-anti-TNF biologics. No significant differences were found in age, gender, or timing of blood sampling post vaccination. Omicron neutralizing activity was markedly reduced in all groups (p < 0.001). The group of patients on anti-TNF biologics showed lower neutralizing antibody titers against Alpha, Delta, and Gamma strains than every other group analyzed. Conclusions: IBD patients on anti-TNF drugs have a reduced serological response to the SARS-CoV-2 vaccine, with the Omicron variant not being neutralized. This highlights the necessity for tailored vaccine strategies for these patients.
Collapse
Affiliation(s)
- Dario Genovese
- Hygiene and Preventive Medicine Section, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Daniele Brinch
- Gastroenterology and Hepatology Section, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Stefano Muscarella
- Gastroenterology and Hepatology Section, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Marica Saladino
- Gastroenterology and Hepatology Section, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Lucio Carrozza
- Gastroenterology and Hepatology Section, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Chiara Cunsolo
- Gastroenterology and Hepatology Section, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Giuseppa Luisa Sanfilippo
- Microbiology Section, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Emanuele Amodio
- Hygiene and Preventive Medicine Section, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Maria Cappello
- Gastroenterology and Hepatology Section, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Donatella Ferraro
- Microbiology Section, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
162
|
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
|
163
|
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] [Download PDF] [Figures] [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
|
164
|
Sigall Boneh R, van der Kruk N, Wine E, Verburgt CM, de Meij TGJ, Löwenberg M, Gecse KB, Wierdsma N, Derikx JPM, de Jonge WJ, D’Haens G, Ghiboub M, Van Limbergen JE. Tryptophan metabolites profile predict remission with dietary therapy in pediatric Crohn's disease. Therap Adv Gastroenterol 2025; 18:17562848251323004. [PMID: 40012837 PMCID: PMC11863242 DOI: 10.1177/17562848251323004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
Background Crohn's disease (CD) exclusion diet combined with partial enteral nutrition (CDED + PEN) or exclusive enteral nutrition (EEN) is effective in inducing remission in mild-to-moderate pediatric CD. Although CDED + PEN is better tolerated and has higher compliance compared to EEN, a subset of patients does not achieve remission. Diet-induced remission is shown to be positively associated with specific changes in tryptophan-metabolites. Objectives To investigate whether the abundance of baseline fecal tryptophan-metabolites predicts dietary therapy outcomes in pediatric CD. Design Diagnostic accuracy study and secondary analysis of previously conducted Randomized Controlled Trial (RCT). Methods Twenty-six patients from previously performed RCT of mild-to-moderate pediatric CD were included. The patients were classified as having clinical remission (R) (n = 19 in total; CDED + PEN = 10 and to EEN = 9) or No-Remission (NR) (n = 7 in total; CDED + PEN = 3 and EEN = 4) following 6 weeks of therapy, based on the Pediatric Crohn's Disease Activity Index score (⩽10 = remission). We performed a targeted quantitative analysis of 21 tryptophan-metabolites in baseline (t = 0) fecal samples from both groups, utilizing liquid chromatography coupled with quadrupole mass spectrometry. Receiver operator characteristic curve (ROC) and random forest analysis (RFA) were used to assess the predictive power of fecal tryptophan-metabolites for dietary outcomes at baseline. Ratios of tryptophan-metabolites were compared to investigate different downstream tryptophan pathways. Results Baseline fecal kynurenine level was significantly higher in NR compared to R for CDED + PEN (p = 0.02) and EEN (p = 0.04). ROC analysis highlighted the robust predictive power of kynurenine for CDED + PEN (area under the curve (AUC = 0.97)) and EEN (AUC = 0.88)-induced remission. RFA corroborated these observations. The ratio serotonin/kynurenine was the strongest predictor of CDED + PEN-induced remission (AUC = 1). The ratio 5-hydroxytryptophan/kynurenine (AUC = 0.88) predicted EEN-induced remission. By combining data from CDED + PEN and EEN, kynurenine (AUC = 0.91) and ratios of quinolinic acid/kynurenine (AUC = 0.93) and kynurenine/indole-3-acetic acid (AUC = 0.88) demonstrated strong predictive performance for dietary therapy-induced remission. Conclusion Baseline tryptophan metabolites have the potential to serve as a biomarker for dietary remission in pediatric CD. Some tryptophan metabolite ratios showed the most promising predictive capabilities. If confirmed in validation studies, baseline fecal tryptophan markers may be able to provide much-needed guidance to personalize dietary intervention within the management of pediatric CD. Trial registration NCT01728870.
Collapse
Affiliation(s)
- Rotem Sigall Boneh
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Pediatric Gastroenterology and Nutrition Unit, The E. Wolfson Medical Center, Holon, Israel
| | - Nikki van der Kruk
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Eytan Wine
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Charlotte M. Verburgt
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Tim G. J. de Meij
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Mark Löwenberg
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Krisztina B. Gecse
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Nicolette Wierdsma
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Joep P. M. Derikx
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Wouter J. de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Department of Surgery, University of Bonn, Bonn, Germany
| | - Geert D’Haens
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Mohammed Ghiboub
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Meibergdreef 69, Amsterdam 1105 BK, The Netherlands
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Johan E. Van Limbergen
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
165
|
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
|
166
|
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
|
167
|
Zhao J, Chen R, Luo M, Zhu Q, Zhao Q. Genetic variation in targets of antihyperglycemic drugs and inflammatory bowel disease' risk: A mendelian randomization study. Diabetes Metab Syndr 2025; 19:103204. [PMID: 40023995 DOI: 10.1016/j.dsx.2025.103204] [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: 09/15/2023] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 03/04/2025]
Abstract
AIM Antihyperglycemic drugs have potential therapeutic benefits for inflammatory bowel disease (IBD). We aimed to investigate the association between genetic variations in gene-targeted antihyperglycemic drugs and the risk of IBD. METHODS Summary statistics for HbA1c data were from the UK Biobank including 344,182 participants. Statistics of IBD were obtained from UK Inflammatory Bowel Disease Genetics. Two Mendelian randomization methods were employed to derive the main findings. RESULTS In the SMR analysis, increased expression of genetic variations in SGLT2 inhibitor targets (gene: SLC5A2) was linked to a higher risk of CD (OR: 1.97, P = 0.048). Genetic variation in brain cerebellum tissue of sulfonylurea targets (gene: ABCC8) expression was positively associated with IBD (OR = 1.11, P = 0.000). The genetic variation in the GLP-1RA targets (gene: GLP1R) expression was positively correlated with IBD (OR: 1.45, P = 0.039). The IVW-MR analysis suggested reduced IBD and CD risk with expression of increased genetic variation in the thiazolidinediones targets (gene: PPARG). CONCLUSION Genetic variations in SGLT2 inhibitor targets might be associated with an increased risk of CD. The ABCC8 gene might be linked to IBD, CD, and UC. There might be a positive correlation between genetic variation in the GLP-1RA targets expression and IBD occurrence.
Collapse
Affiliation(s)
- Jiaxi Zhao
- General Practice Ward / International Medical Center Ward General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Mengqi Luo
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Quanjing Zhu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qian Zhao
- General Practice Ward / International Medical Center Ward General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
168
|
Chen Q, Zhang YL, Shi YQ, Zheng L. Mesalazine alleviated the symptoms of spontaneous colitis in interleukin-10 knockout mice by regulating the STAT3/NF-κB signaling pathway. World J Gastroenterol 2025; 31:96459. [PMID: 39991681 PMCID: PMC11755248 DOI: 10.3748/wjg.v31.i7.96459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 12/01/2024] [Accepted: 12/27/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Excessive endoplasmic reticulum (ER) stress in intestinal epithelial cells can lead to damage to the intestinal mucosal barrier, activate the signal transducer and activator of transcription 3 (STAT3)/nuclear factor kappa B (NF-κB) signaling pathway, and exacerbate the inflammatory response, thus participating in the pathogenesis of ulcerative colitis (UC). Mesalazine is a commonly used drug in the clinical treatment of UC. However, further studies are needed to determine whether mesalazine regulates the ER stress of intestinal epithelial cells, down-regulates the STAT3/NF-κB pathway to play a role in the treatment of UC. AIM To study the therapeutic effects of mesalazine on spontaneous colitis in interleukin-10 (IL-10)-/- mice. METHODS The 24-week-old IL-10-/- mice with spontaneous colitis were divided into the model group and the 5-amino salicylic acid group. Littermates of wild-type mice of the same age group served as the control. There were eight mice in each group, four males and four females. The severity of symptoms of spontaneous colitis in IL-10-/- mice was assessed using disease activity index scores. On day 15, the mice were sacrificed. The colon length was measured, and the histopathological changes and ultrastructure of colonic epithelial cells were detected. The protein expressions of STAT3, p-STAT3, NF-κB, IκB, p-IκB, and glucose-regulated protein 78 were identified using Western blotting. The STAT3 and NF-κB mRNA expressions were identified using real-time polymerase chain reaction. The glucose-regulated protein 78 and C/EBP homologous protein expressions in colon sections were detected using immunofluorescence. RESULTS Mesalazine reduced the symptoms of spontaneous colitis in IL-10 knockout mice and the histopathological damage of colonic tissues, and alleviated the ER stress in epithelial cells of colitis mice. Western blotting and quantitative real-time polymerase chain reaction results showed that the STAT3/NF-κB pathway in the colon tissue of model mice was activated, suggesting that this pathway was involved in the pathogenesis of UC and might become a potential therapeutic target. Mesalazine could down-regulate the protein expressions of p-STAT3, NF-κB and p-IκB, and down-regulate the mRNA expression of STAT3 and NF-κB. CONCLUSION Mesalazine may play a protective role in UC by reducing ER stress by regulating the STAT3/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Qian Chen
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ya-Li Zhang
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yong-Quan Shi
- Department of Gastroenterology, Xijing Hospital affiliated to Air Force Medical University, Xi’an 710032, Shaanxi Province, China
| | - Lie Zheng
- Department of Gastroenterology, Traditional Chinese Medicine Hospital of Shaanxi Province, Xi’an 710003, Shaanxi Province, China
| |
Collapse
|
169
|
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
|
170
|
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
|
171
|
Vermeer E, Struys EA, Lin M, van Limbergen JE, de Boer NKH, Bulatović-Ćalasan M, de Meij TGJ, de Jonge R. Erythrocyte Methotrexate-Polyglutamate Concentrations in Pediatric Inflammatory Bowel Disease. Inflamm Bowel Dis 2025:izaf035. [PMID: 39982703 DOI: 10.1093/ibd/izaf035] [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: 11/12/2024] [Indexed: 02/22/2025]
Abstract
BACKGROUND AND AIMS Therapeutic drug monitoring (TDM) of methotrexate (MTX) is challenging due to its pharmacokinetics and short plasma half-life. Intracellular MTX-polyglutamates (PG1-5), which accumulate over time, have not been assessed in pediatric inflammatory bowel disease (IBD). This study aimed to evaluate erythrocyte MTX-PG as a potential TDM tool in pediatric IBD. METHODS In this cross-sectional study, MTX-PG concentrations were measured in erythrocytes of children with IBD on stable low-dose MTX for at least 12 weeks using stable-isotope dilution liquid chromatography-tandem mass spectrometry. The influence of administration route, MTX dosage, and anthropometrics on MTX-PG concentrations was examined. RESULTS Seventy-eight patients were included, showing MTX-PG3 as the predominant subspecies (median 27.0 nmol/L) with a median MTX-PGtotal of 74.8 nmol/L. A higher MTX dose correlated significantly with elevated levels of MTX-PG3, MTX-PG4, MTX-PG5, and MTX-PGtotal (P < .01). Adjusted for body surface area, MTX dose remained significantly associated with higher MTX-PG concentrations (P < .01). However, comparison by administration route was limited due to a few patients on subcutaneous MTX (n = 4). CONCLUSIONS We observed high interindividual variability in the reached erythrocyte MTX-PG concentrations. Body surface adjusted or unadjusted MTX dosage showed a positive linear correlation with erythrocyte MTX-PG concentrations in children with IBD. This is a prerequisite for TDM and provides a strong basis for further research into the relation between TDM of MTX, efficacy, and toxicity.
Collapse
Affiliation(s)
- Eva Vermeer
- Department of Paediatric Gastroenterology, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) Research Institute, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Eduard A Struys
- Laboratory of Specialised Diagnostics and Research, Department of Laboratory Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Marry Lin
- Laboratory of Specialised Diagnostics and Research, Department of Laboratory Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Johan E van Limbergen
- Department of Paediatric Gastroenterology, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) Research Institute, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Nanne K H de Boer
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Maja Bulatović-Ćalasan
- Laboratory of Specialised Diagnostics and Research, Department of Laboratory Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Tim G J de Meij
- Department of Paediatric Gastroenterology, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) Research Institute, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Robert de Jonge
- Laboratory of Specialised Diagnostics and Research, Department of Laboratory Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
172
|
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
|
173
|
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
|
174
|
Lee WS, Chew KS, Huang JG, Tanpowpong P, Mercado KSC, Reodica A, Logarajah V, Hathagoda KLW, Rajindrajith S, Wong YKY, Treepongkaruna S, Aw MM. Disease phenotypic and outcome of very-early onset inflammatory bowel disease in Asian children: an understudied population. Front Pediatr 2025; 13:1487253. [PMID: 40051907 PMCID: PMC11882516 DOI: 10.3389/fped.2025.1487253] [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: 08/27/2024] [Accepted: 02/04/2025] [Indexed: 03/09/2025] Open
Abstract
Background There is a paucity of knowledge on disease phenotype and outcome of very early-onset (VEO) inflammatory bowel disease (VEO-IBD) from recently developed and developing countries, including from Southeast Asia. We studied disease phenotype, clinical characteristics, management and outcome of VEO-IBD in South and Southeast Asian children. Materials and methods We extracted data from a multicentre Asian pediatric (onset <18 years) IBD registry. VEO- and later-onset pediatric (LO-p) IBD were defined as onset of disease <6 years and ≥6 years, respectively. We excluded monogenic IBD. Results Of 440 children with IBD cases; 112 (25.5%) were VEO-IBD; Crohn's disease (CD) 36 (32.1%); ulcerative colitis (UC) 68 (60.7%), and IBD-unspecified 7 (7.1%). UC was more common in VEO-IBD while CD more common in LO-pIBD (CD = 68.9% vs. UC = 25.9%; p < 0.001). Disease location/extent of disease and disease severity were similar in both age groups for both CD and UC. For CD, inflammatory disease behavior was equally common in both age group (77.8% in VEO-IBD vs. 76.6% of LO-pIBD), majority had isolated colonic disease (27.8% VEO-IBD vs. 36.3% LO-pIBD), while stricturing and penetrating diseases were not observed in VEO-CD, but noted in 4.9% and 8.4% of LO-pCD, respectively. Among UC cases, pancolitis was observed in 60.3% of VEO-IBD vs. 65.9% of LO-pIBD. Most UC never had severe disease regardless of age group. Five years after diagnosis, VEO-IBD were more likely to have corticosteroids, immunomodulators or biologics than LO-pIBD. Despite this, inactive/mild disease activity was the predominant outcome at 5 year follow up for both VEO-CD (98.2%) and VEO- UC (96.1%). Bowel surgery rate was 2.4% and 1.7% for VEO- and LO-IBD at 5 years, respectively. Conclusions Despite differences in disease phenotype at diagnosis, disease behaviour, location/extent and disease severity were similar between VEO- and LO-IBD, with a comparable overall clinical remission rates between both age groups at 5 years after diagnosis.
Collapse
Affiliation(s)
- Way-Seah Lee
- Department of Paediatrics, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
- Center of Research for Communicable Disease, M Kandiah Faculty of Medicine and Health Sciences, University Tunku Abdul Rahman, Kajang, Malaysia
| | - Kee-Seang Chew
- Department of Paediatrics, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - James-Guoxian Huang
- KHoo Teck Puat National University Children's Medical Institute, National University Hospital Health System, National University Hospital, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Pornthep Tanpowpong
- Division of Gastroenterology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | - Almida Reodica
- Department of Pediatrics, The Medical City, Pasig, Philippines
| | - Veena Logarajah
- Department of Paediatrics, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - K. L. W. Hathagoda
- Department of Pediatrics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Shaman Rajindrajith
- Department of Pediatrics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Yoko Kin-Yoke Wong
- Epidemiology, Singapore Clinical Research Institute, Singapore, Singapore
| | - Suporn Treepongkaruna
- Division of Gastroenterology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Marion Margaret Aw
- KHoo Teck Puat National University Children's Medical Institute, National University Hospital Health System, National University Hospital, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
175
|
Chen L, Xu Y, Ai F, Shen S, Luo Y, Li X. Dissecting the rising tide of inflammatory bowel disease among youth in a changing world: insights from GBD 2021. Int J Colorectal Dis 2025; 40:44. [PMID: 39964411 PMCID: PMC11836149 DOI: 10.1007/s00384-025-04821-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/03/2025] [Indexed: 02/21/2025]
Abstract
OBJECTIVES This study investigates the alarming epidemiological trends of inflammatory bowel disease (IBD) among children and young adults, highlighting the associated disease burden on global health. MATERIALS AND METHODS Utilizing data from the Global Burden of Disease (GBD) study 2021, we conducted a comprehensive analysis of age-standardized incidence rates (ASIR), age-standardized mortality rates (ASMR), disability-adjusted life years (DALYs), and estimated annual percentage changes (EAPC). Future trends were forecasted using the Bayesian age-period-cohort model. RESULTS From 1990 to 2021, IBD incidence and DALY rates remained persistently high, with a concerning upward trend noted among children and young adults. While men experienced a decline in DALY rates, women faced increasing burdens. In 2021, high-income regions, particularly North America, reported the highest incidence and DALY rates, contrasting sharply with Central Latin America, which exhibited the lowest ASIR. Southeast Asia presented the most favorable DALY rates. A notable negative correlation was identified between DALY rates and socio-demographic index (SDI) at the national level, with high and high-middle SDI countries continuing to bear a substantial burden, while low and middle SDI nations faced rising challenges. CONCLUSIONS The persistent high burden of IBD in children and young adults signifies a critical public health concern. The marked geographical and gender disparities underscore the urgent need for tailored regional and population-based strategies aimed at primary prevention and effective management. This study illuminates the pressing necessity for policy interventions to address the growing epidemic of IBD among vulnerable populations.
Collapse
Affiliation(s)
- Libin Chen
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, Changsha, Hunan, China
| | - Yifu Xu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Feiyan Ai
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, Changsha, Hunan, China
| | - Shourong Shen
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, Changsha, Hunan, China
| | - Yanwei Luo
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Xiayu Li
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, Changsha, Hunan, China.
| |
Collapse
|
176
|
Quaid K, Xing X, Chen YH, Miao Y, Neilson A, Selvamani V, Tran A, Cui X, Hu M, Wang T. iPSCs and iPSC-derived cells as a model of human genetic and epigenetic variation. Nat Commun 2025; 16:1750. [PMID: 39966349 PMCID: PMC11836351 DOI: 10.1038/s41467-025-56569-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Abstract
Understanding the interaction between genetic and epigenetic variation remains a challenge due to confounding environmental factors. We propose that human induced Pluripotent Stem Cells (iPSCs) are an excellent model to study the relationship between genetic and epigenetic variation while controlling for environmental factors. In this study, we have created a comprehensive resource of high-quality genomic, epigenomic, and transcriptomic data from iPSC lines and three iPSC-derived cell types (neural stem cell (NSC), motor neuron, monocyte) from three healthy donors. We find that epigenetic variation is most strongly associated with genetic variation at the iPSC stage, and that relationship weakens as epigenetic variation increases in differentiated cells. Additionally, cell type is a stronger source of epigenetic variation than genetic variation. Further, we elucidate a utility of studying epigenetic variation in iPSCs and their derivatives for identifying important loci for GWAS studies and the cell types in which they may be acting.
Collapse
Affiliation(s)
- Kara Quaid
- Center for Genome Sciences & Systems Biology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA
| | - Xiaoyun Xing
- Center for Genome Sciences & Systems Biology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA
| | - Yi-Hsien Chen
- Genome Engineering & Stem Cell Center (GESC@MGI), Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Yong Miao
- Genome Engineering & Stem Cell Center (GESC@MGI), Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Amber Neilson
- Genome Engineering & Stem Cell Center (GESC@MGI), Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Vijayalingam Selvamani
- Genome Engineering & Stem Cell Center (GESC@MGI), Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Aaron Tran
- Center for Genome Sciences & Systems Biology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA
| | - Xiaoxia Cui
- Genome Engineering & Stem Cell Center (GESC@MGI), Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
| | - Ming Hu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Ting Wang
- Center for Genome Sciences & Systems Biology, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
177
|
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
|
178
|
Rodrigues BL, Pascoal LB, Genaro LM, Warrak LSCA, Rodrigues BAG, Coope A, Camargo MG, Oliveira PDSP, Ayrizono MDLS, Velloso LA, Leal RF. In Vitro Inhibition of Endoplasmic Reticulum Stress: A Promising Therapeutic Strategy for Patients with Crohn's Disease. Cells 2025; 14:270. [PMID: 39996742 PMCID: PMC11853800 DOI: 10.3390/cells14040270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Crohn's disease (CD) is an inflammatory bowel disease marked by an abnormal immune response and excessive pro-inflammatory cytokines, leading to impaired protein processing and endoplasmic reticulum (ER) stress. This stress, caused by the accumulation of misfolded proteins, triggers the unfolded protein response (UPR) through IRE1/Xbp-1, PERK/eIF2α, and ATF6 pathways, which are linked to intestinal inflammation. This study aimed to investigate ER stress in CD patients' intestinal mucosa and evaluate phenylbutyrate (PBA) as an ER stress inhibitor. METHODS Colon biopsies from CD patients and controls were cultured under five conditions, including 4-PBA treatments. Real-time PCR, cytokine level, and immunohistochemistry were performed. RESULTS Immunohistochemistry revealed that ER stress was activated in CD patients' intestinal epithelial cells and lamina propria cells. PERK/eIF2α, but not IRE1/Xbp-1 or ATF6, was upregulated in CD patients compared to controls. UPR-related genes (STC2, CALR, HSPA5, HSP90B1) were also elevated in CD patients. PBA treatment significantly reduced ER stress and UPR markers while decreasing apoptotic markers like DDIT3. Pro-inflammatory cytokines, such as IL-1β, IL-6, IL-17, TNF- α, and sCD40L, were significantly reduced after PBA treatment. CONCLUSION ER stress and UPR pathways are activated in CD colonic mucosa, and PBA reduces these markers, suggesting potential therapeutic benefits for CD-related inflammation.
Collapse
Affiliation(s)
- Bruno Lima Rodrigues
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Lívia Bitencourt Pascoal
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Lívia Moreira Genaro
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Leonardo Saint Clair Assad Warrak
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Beatriz Alves Guerra Rodrigues
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Andressa Coope
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Michel Gardere Camargo
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Priscilla de Sene Portel Oliveira
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Maria de Lourdes Setsuko Ayrizono
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Lício Augusto Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-864, Brazil
| | - Raquel Franco Leal
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| |
Collapse
|
179
|
Wands DIF, Gianolio L, Cameron F, Hansen R, Russell RK, Wilson DC. Pediatric Inflammatory Bowel Disease Type Unclassified: A Nationwide Cohort Study in Scotland With up to 20 Years Follow-up Shows Reclassification in the Majority and Mild Course in Those Whose Diagnosis Is Unchanged. Inflamm Bowel Dis 2025; 31:313-320. [PMID: 39321100 DOI: 10.1093/ibd/izae218] [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: 05/21/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Given the paucity of long-term longitudinal data for inflammatory bowel disease type unclassified (IBDU), we aimed to clarify IBDU disease course and reclassification rate by presenting nationwide data with up to 20 years of follow-up. METHODS We analyzed a prospectively identified 11-year cohort of pediatric patients diagnosed with IBDU between January 1, 2003 and December 31, 2013 at all Scottish pediatric IBD centers and followed up into adult services until December 31, 2022. Data were obtained from electronic medical records at fixed timepoints (5 and 10 years post-diagnosis) and at the final follow-up. RESULTS Overall, 102 patients were included in the analysis (57/102 [56%] male, median [interquartile range {IQR}] age at diagnosis: 11.5 [9.1-13.2] years) with a median (IQR) follow-up length of 10.5 (8.6-14.0) years. A change of diagnosis was made in 61 of 102 patients (60%); of these, 30 patients (29%) were reclassified to Crohn's disease (CD) and 31 patients (30%) to ulcerative colitis (UC). Patients who remained with IBDU had higher 1- to 5-year remission rates (IBDU 30/39 [77%] vs reclassified 16/57 [28%], P < .05), with lower rates of moderate-to-severe disease (IBDU 3/39 [8%] vs reclassified 31/57 [54%], P < .05) and less need for biologics across all timepoints (IBDU vs reclassified: first timepoint 1/39 [3%] vs 17/57 [30%], second timepoint 1/33 [3%] vs 26/56 [46%], third timepoint 0/18 [0%] vs 16/33 [49%]; all P < .05). Higher rates of surgical resections were observed in reclassified patients (reclassified 11/61 [18%] vs IBDU 1/41 [2%], P = .02). CONCLUSIONS In our nationwide pediatric IBDU cohort, 60% of patients were reclassified to either UC or CD over 10.5 years of median follow-up; those who remained with IBDU had a milder disease course.
Collapse
Affiliation(s)
- David I F Wands
- Child Life and Health, University of Edinburgh, Royal Hospital for Children & Young People, Edinburgh, UK
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, UK
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, UK
| | - Laura Gianolio
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, UK
| | - Fiona Cameron
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Alder Hey Children's Hospital, Liverpool, UK
| | - Richard Hansen
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, UK
- Child Health, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Richard K Russell
- Child Life and Health, University of Edinburgh, Royal Hospital for Children & Young People, Edinburgh, UK
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, UK
| | - David C Wilson
- Child Life and Health, University of Edinburgh, Royal Hospital for Children & Young People, Edinburgh, UK
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, UK
| |
Collapse
|
180
|
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 PMCID: PMC11879014 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
|
181
|
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
|
182
|
Yu X, Hu X, Wan X, Zhang Z, Wan X, Cai M, Yu T, Xiao J. A unified framework for cell-type-specific eQTL prioritization by integrating bulk and scRNA-seq data. Am J Hum Genet 2025; 112:332-352. [PMID: 39824189 PMCID: PMC11866979 DOI: 10.1016/j.ajhg.2024.12.018] [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: 08/01/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/20/2025] Open
Abstract
Genome-wide association studies (GWASs) have identified numerous genetic variants associated with complex traits, yet the biological interpretation remains challenging, especially for variants in non-coding regions. Expression quantitative trait locus (eQTL) studies have linked these variations to gene expression, aiding in identifying genes involved in disease mechanisms. Traditional eQTL analyses using bulk RNA sequencing (bulk RNA-seq) provide tissue-level insights but suffer from signal loss and distortion due to unaddressed cellular heterogeneity. Recently, single-cell RNA-seq (scRNA-seq) has provided higher resolution, enabling cell-type-specific eQTL (ct-eQTL) analyses. However, these studies are limited by their smaller sample sizes and technical constraints. In this paper, we present a statistical framework, IBSEP, which integrates bulk RNA-seq and scRNA-seq data for enhanced ct-eQTL prioritization. Our method employs a hierarchical linear model to combine summary statistics from both data types, overcoming the limitations while leveraging the advantages associated with each technique. Through extensive simulations and real data analyses, including peripheral blood mononuclear cells and brain cortex datasets, IBSEP demonstrated superior performance in identifying ct-eQTLs compared to existing methods. Our approach unveils transcriptional regulatory mechanisms specific to cell types, offering deeper insights into the genetic basis of complex diseases at a cellular resolution.
Collapse
Affiliation(s)
- Xinyi Yu
- Shenzhen Research Institute of Big Data, Shenzhen 518172, China; School of Data Science, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Shenzhen 518172, China
| | - Xianghong Hu
- Department of Mathematics, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Xiaomeng Wan
- Department of Mathematics, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Zhiyong Zhang
- Shenzhen Research Institute of Big Data, Shenzhen 518172, China; School of Data Science, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Shenzhen 518172, China
| | - Xiang Wan
- Shenzhen Research Institute of Big Data, Shenzhen 518172, China
| | - Mingxuan Cai
- Department of Biostatistics, City University of Hong Kong, Hong Kong SAR, China
| | - Tianwei Yu
- Shenzhen Research Institute of Big Data, Shenzhen 518172, China; School of Data Science, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Shenzhen 518172, China.
| | - Jiashun Xiao
- Shenzhen Research Institute of Big Data, Shenzhen 518172, China.
| |
Collapse
|
183
|
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 PMCID: PMC11866969 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
|
184
|
Wang Y, Pan CW, Huang Y, Zheng X, Li S, He M, Hashash JG, Farraye FA, Ehrlich AC. Global Epidemiology and Geographic Variations of Pediatric-Onset Inflammatory Bowel Disease: A Comprehensive Analysis of the Global Burden of Disease Study 1990 to 2019. Inflamm Bowel Dis 2025; 31:376-385. [PMID: 38676392 DOI: 10.1093/ibd/izae093] [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: 01/12/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND An increasing incidence of pediatric-onset inflammatory bowel disease (PIBD) has been reported in many countries. However, the global burden and distribution of this disease remain less understood. We aimed to examine the global epidemiology and trends of PIBD from 1990 to 2019. METHODS Data from the 2019 Global Burden of Disease Study, covering 204 countries, were analyzed. We assessed key measures like incidence, prevalence, mortality, and disability-adjusted life years (DALYs) using linear regression to calculate annual percentage changes and assess trends. RESULTS Between 1990 and 2019, the PIBD incidence rate increased and the DALY rate and mortality rate declined. The incidence rate was notably elevated in the high Socio-demographic Index (SDI) quintile, reaching 6.3 per 100 000 person-years, corresponding to 13 914 new cases in 2019. Incidence and prevalence of PIBD positively correlated with the SDI, while higher death and DALY burdens were observed in lower-SDI countries. In 2019, the top 5 countries with the highest PIBD incidence rates were Canada (19.9 per 100 000 population), Denmark (12.4 per 100 000 population), Hungary (8.5 per 100 000 population), Austria (8.1 per 100 000 population), and the United States (7.4 per 100 000 population). Several countries experienced significant increases in incidence rates from 1990 to 2019, led by Taiwan (annual percent change 4.2%), followed by China (2.8%), Japan (2.1%), Australia (1.8%), and Hungary (1.6%). DISCUSSION PIBD incidence has significantly increased since 1990. High-SDI countries face higher incidence, while lower-SDI countries experience higher mortality and DALY burdens. The study underscores the need for ongoing monitoring and research to address this emerging public health issue.
Collapse
Affiliation(s)
- Yichen Wang
- Division of Hospital Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chun-Wei Pan
- Department of Medicine, John H. Stroger, Jr. Hospital of Cook County, Chicago, IL, USA
| | - Yuting Huang
- Division of Gastroenterology and Hepatology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Xin Zheng
- Division of Gastroenterology and Hepatology, Loma Linda University Health, Loma Linda, CA, USA
| | - Si Li
- Department of Medicine, Temple University Hospital, Philadelphia, PA, USA
| | - Mingyue He
- Department of Medicine, Temple University Hospital, Philadelphia, PA, USA
| | - Jana G Hashash
- Division of Gastroenterology and Hepatology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Francis A Farraye
- Division of Gastroenterology and Hepatology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Adam C Ehrlich
- Section of Gastroenterology and Hepatology, Temple University Hospital, Philadelphia, PA, USA
| |
Collapse
|
185
|
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
|
186
|
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
|
187
|
Calcaterra V, Penagini F, Rossi V, Abbattista L, Bianchi A, Turzi M, Cococcioni L, Zuccotti G. Thyroid disorders and inflammatory bowel disease: an association present in adults but also in children and adolescents. Front Endocrinol (Lausanne) 2025; 16:1425241. [PMID: 39968296 PMCID: PMC11832402 DOI: 10.3389/fendo.2025.1425241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Inflammatory bowel diseases (IBD) represent chronic inflammatory multisystemic disorders that primarily involve the gastrointestinal tract. Patients with ulcerative colitis (UC) and Crohn's disease (CD) exhibit a higher prevalence of thyroid disorders compared to the general population. The aim of this review is to summarize the literature on concomitant IBD and thyroid disorders, specifically autoimmune thyroid diseases such as Graves' disease (GD) and Hashimoto's thyroiditis (HT), as well as thyroid cancer, with a focus on children and adolescents. We provide an overview of the age-related differences between children and adults in the prevalence of this association. Literature shows that relatively few studies have been conducted on this subject in pediatric populations. The etiopathogenetic similarities between IBD and autoimmune thyroiditis are undeniable. Nevertheless, current data does not indicate a unanimous association between GD and HT and chronic IBD (both CD and UC). Although evidence suggests a potential association between IBD and thyroid cancer, particularly papillary thyroid cancer, the precise nature of this relationship varies across studies and is influenced by multiple factors. The limited information regarding the relationship between IBD and thyroid disorders in children highlights a significant knowledge gap. Since the thyroid plays a critical role in the pediatric population's development, it is essential to promptly recognize and treat thyroid diseases. A thyroid function monitoring and future research exploring the genetic and immunologic connections are essential to enhance our understanding of the interrelation between IBD and thyroid disorders.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Pediatric and Adolescent Unit, Department of Internal Medicine, University of Pavia, Pavia, Italy
- Pediatric Department, Buzzi Children’s Hospital, Milano, Italy
| | | | - Virginia Rossi
- Pediatric Department, Buzzi Children’s Hospital, Milano, Italy
| | | | - Alice Bianchi
- Pediatric Department, Buzzi Children’s Hospital, Milano, Italy
| | | | | | - Gianvincenzo Zuccotti
- Pediatric Department, Buzzi Children’s Hospital, Milano, Italy
- Department of Biomedical and Clinical Science, University of Milan, Milano, Italy
| |
Collapse
|
188
|
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
|
189
|
Nanì MF, Pagano E, De Cicco P, Lucariello G, Cattaneo F, Tropeano FP, Cicia D, Amico R, Raucci F, Ercolano G, Maione F, Rinaldi MM, Esposito F, Ammendola R, Luglio G, Capasso R, Makriyannis A, Petrosino S, Borrelli F, Romano B, Izzo AA. Pharmacological Inhibition of N-Acylethanolamine Acid Amidase (NAAA) Mitigates Intestinal Fibrosis Through Modulation of Macrophage Activity. J Crohns Colitis 2025; 19:jjae132. [PMID: 39211986 PMCID: PMC11836880 DOI: 10.1093/ecco-jcc/jjae132] [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: 12/21/2023] [Revised: 08/10/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND AIMS Intestinal fibrosis, a frequent complication of inflammatory bowel disease, is characterized by stricture formation with no pharmacological treatment to date. N-acylethanolamine acid amidase (NAAA) is responsible for the hydrolysis of acylethanolamides (AEs, eg, palmitoylethanolamide and oleoylethanolamide). Here, we investigated NAAA and AE signaling in gut fibrosis. METHODS NAAA and AE signaling were evaluated in human intestinal specimens from patients with stenotic Crohn's disease (CD). Gut fibrosis was induced by 2,4,6-trinitrobenzenesulfonic acid, monitored by colonoscopy, and assessed by qRT-PCR, histological analyses, and confocal microscopy. Immune cells in mesenteric lymph nodes were analyzed by FACS. Colonic fibroblasts were cultured in conditioned media derived from polarized or non-polarized bone marrow-derived macrophages (BMDMs). IL-23 signaling was evaluated by qRT-PCR, ELISA, FACS, and western blot in BMDMs and in lamina propria CX3CR1+ cells. RESULTS In ileocolonic human CD strictures, increased transcript expression of NAAA was observed with a decrease in its substrates oleoylethanolamide and palmitoylethanolamide. NAAA inhibition reduced intestinal fibrosis in vivo, as indicated by a decrease in inflammatory parameters, collagen deposition, and fibrosis-related genes, including those involved in epithelial-to-mesenchymal transition. More in-depth studies revealed modulation of the immune response related to IL-23 following NAAA inhibition. The antifibrotic actions of NAAA inhibition are mediated by Mφ and M2 macrophages that indirectly affect fibroblast collagenogenesis. NAAA inhibitor AM9053 normalized IL-23 signaling in BMDMs and in lamina propria CX3CR1+ cells. CONCLUSIONS Our findings provide new insights into the pathophysiological mechanism of intestinal fibrosis and identify NAAA as a promising target for the development of therapeutic treatments to alleviate CD-related fibrosis.
Collapse
Affiliation(s)
- Maria Francesca Nanì
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Ester Pagano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Paola De Cicco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Giuseppe Lucariello
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Fabio Cattaneo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Francesca Paola Tropeano
- Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Donatella Cicia
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Rebecca Amico
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Federica Raucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Giuseppe Ercolano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Maria Michela Rinaldi
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Fabiana Esposito
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Rosario Ammendola
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Gaetano Luglio
- Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
| | - Alexandros Makriyannis
- Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Stefania Petrosino
- Institute of Biomolecular Chemistry, National Research Council, Pozzuoli, Italy
- Epitech Group SpA, Saccolongo, Italy
| | - Francesca Borrelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Barbara Romano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Angelo A Izzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
190
|
Wang X, Zhang G, Bian Z, Chow V, Grimaldi M, Carivenc C, Sirounian S, Li H, Sladekova L, Motta S, Luperi Y, Gong Y, Costello C, Li L, Jachimowicz M, Guo M, Hu S, Wilson D, Balaguer P, Bourguet W, Mani S, Bonati L, Peng H, March J, Wang H, Wang S, Krause HM, Liu J. An abundant ginger compound furanodienone alleviates gut inflammation via the xenobiotic nuclear receptor PXR in mice. Nat Commun 2025; 16:1280. [PMID: 39900639 PMCID: PMC11791082 DOI: 10.1038/s41467-025-56624-0] [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: 08/01/2024] [Accepted: 01/22/2025] [Indexed: 02/05/2025] Open
Abstract
The literature documenting the value of drug-like molecules found in natural products is vast. Although many dietary and herbal remedies have been found to be effective for treating intestinal inflammation, the identification of their active components has lagged behind. In this study, we find that a major ginger component, furanodienone (FDN), is a selective pregnane X receptor (PXR) ligand with agonistic transcriptional outcomes. We show that FDN binds within a sub-pocket of the PXR ligand binding domain (LBD), with subsequent alterations in LBD structure. Using male mice, we show that orally provided FDN has potent PXR-dependant anti-inflammatory outcomes that are colon-specific. Increased affinity and target gene activation in the presence of synergistically acting agonists indicates further opportunities for augmenting FDN activity, efficacy and safety. Collectively, these results support the translational potential of FDN as a therapeutic agent for the treatment and prevention of colonic diseases.
Collapse
Affiliation(s)
- Xiaojuan Wang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Guohui Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, People's Republic of China
| | - Zhiwei Bian
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Vimanda Chow
- Department of Chemistry, York University, Toronto, ON, Canada
| | - Marina Grimaldi
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Coralie Carivenc
- Centre de Biologie Structurale, INSERM, CNRS, Université de Montpellier, Montpellier, France
| | - Savannah Sirounian
- Centre de Biologie Structurale, INSERM, CNRS, Université de Montpellier, Montpellier, France
| | - Hao Li
- Department of Molecular Pharmacology; Department of Genetics; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lucia Sladekova
- Department of Molecular Pharmacology; Department of Genetics; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Stefano Motta
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Milan, Italy
| | - Yulia Luperi
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Milan, Italy
| | - Yufeng Gong
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong, China
| | - Cait Costello
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Matthew Jachimowicz
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Miao Guo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, People's Republic of China
| | - Shian Hu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Derek Wilson
- Department of Chemistry, York University, Toronto, ON, Canada
| | - Patrick Balaguer
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - William Bourguet
- Centre de Biologie Structurale, INSERM, CNRS, Université de Montpellier, Montpellier, France
| | - Sridhar Mani
- Department of Molecular Pharmacology; Department of Genetics; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Laura Bonati
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Milan, Italy
| | - Hui Peng
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
- School of the Environment, University of Toronto, Toronto, ON, Canada
| | - John March
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, People's Republic of China.
| | - Henry M Krause
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| | - Jiabao Liu
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
191
|
Forbes AJ, Day AS, Frampton CMA, Gearry RB. Incidence of Inflammatory Bowel Disease in Canterbury, New Zealand, 2018-2023. Dig Dis Sci 2025; 70:719-727. [PMID: 39746890 DOI: 10.1007/s10620-024-08811-0] [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: 09/09/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND New diagnoses of inflammatory bowel diseases (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), can highlight health system burden and potentially give clues to disease aetiology. This population-based study aimed to measure the annual incidence of IBD over six years (2018-2023) in the Canterbury region of New Zealand. METHODS The medical records from public and private gastroenterology clinics were examined for new patients with a confirmed diagnosis of IBD from 1 January 2018 to 31 December 2023. Demographic and disease information (including the Montreal phenotype) was gathered in a secure database. Information was collected prospectively from 2021 to 2023 and retrospectively from 2018 to 2020. RESULTS The incidence of IBD in 2023 was 30.1 per 100,000 person-years (95% CI 25.9-34.7), CD: 14.8 (95% CI 11.9-18.1), and UC: 14.5 (95% CI 11.7-17.7). The average age was 35.0 years (SD 17.2) and New Zealand European was the predominant ethnicity (83.4%), with smaller proportions of Māori (7.7%) and Pacific Islanders (1.7%). Between 2018 and 2023, the annual incidence of IBD was 32.4 per 100,000 person-years. The proportion of CD diagnosed each year decreased, and in 2023, the proportion of CD was equivalent (49%) to the proportion with UC. CONCLUSIONS The incidence of IBD in Canterbury remains high, and the rates appeared stable over the six years 2018-2023, consistent with the stabilizing incidence stage of the 4-stage epidemiological model of IBD. Māori and Pacific Island ethnicities contributed a small number of cases of IBD, but the rates of IBD in these groups were higher than in previous studies.
Collapse
Affiliation(s)
- Angela J Forbes
- Department of Medicine, University of Otago Christchurch, 2 Riccarton Avenue, PO Box 4345, Christchurch, 8140, New Zealand.
| | - Andrew S Day
- Department of Paediatrics, University of Otago Christchurch, Christchurch, New Zealand
| | - Chris M A Frampton
- Department of Medicine, University of Otago Christchurch, 2 Riccarton Avenue, PO Box 4345, Christchurch, 8140, New Zealand
| | - Richard B Gearry
- Department of Medicine, University of Otago Christchurch, 2 Riccarton Avenue, PO Box 4345, Christchurch, 8140, New Zealand
| |
Collapse
|
192
|
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
|
193
|
Piccolo P, Brunetti-Pierri N. Current and Emerging Issues in Adeno-Associated Virus Vector-Mediated Liver-Directed Gene Therapy. Hum Gene Ther 2025; 36:77-87. [PMID: 39714937 DOI: 10.1089/hum.2024.179] [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] [Indexed: 12/25/2024] Open
Abstract
Adeno-associated virus (AAV) vectors have demonstrated safety and efficacy for gene transfer to hepatocytes in preclinical models, in various clinical trials and from a clinical experience with a growing number of approved gene therapy products. Although the exact duration is unknown, the expression of therapeutic genes in hepatocytes remains stable for several years after a single administration of the vector at clinically relevant doses in adult patients with hemophilia and other inherited metabolic disorders. However, clinical applications, especially for diseases requiring high AAV vector doses by intravenous administrations, have raised several concerns. These include the high prevalence of pre-existing immunity against the vector capsid, activation of the complement and the innate immunity with serious life-threatening complications, elevation of liver transaminases, liver growth associated with loss of transgene expression, underlying conditions negatively affecting AAV vector safety and efficacy. Despite these issues, the field is rapidly advancing with a better understanding of vector-host interactions and the development of new strategies to improve liver-directed gene therapy. This review provides an overview of the current and emerging challenges for AAV-mediated liver-directed gene therapy.
Collapse
Affiliation(s)
| | - Nicola Brunetti-Pierri
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- Genomics and Experimental Medicine Program, Scuola Superiore Meridionale (SSM, School of Advanced Studies), Naples, Italy
| |
Collapse
|
194
|
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
|
195
|
Bhalla A, Shahi A, Maity M, Safa F, Srividya V, Clementina R, Anugu GR, Younas S. Inflammatory Bowel Disease in Children: Current Diagnosis and Treatment Strategies. Cureus 2025; 17:e78462. [PMID: 40051947 PMCID: PMC11883196 DOI: 10.7759/cureus.78462] [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: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Pediatric inflammatory bowel disease (PIBD), including Crohn's disease and ulcerative colitis, has emerged as a significant global health challenge with rising incidence rates. Unlike adult inflammatory bowel disease, PIBD presents complexities, including growth impairment, nutritional deficiencies, and psychosocial challenges that necessitate tailored management strategies. This article reviews current diagnostic and emerging treatment strategies to highlight the evolution from traditional therapies such as aminosalicylates, corticosteroids, and immunomodulators to advanced biologic agents like infliximab and adalimumab. Emerging biological therapies, including vedolizumab and ustekinumab, show promise, while novel small molecule therapies such as Janus kinase (JAK) inhibitors are under investigation for potential use in the pediatric population. Supportive treatments, including exclusive enteral nutrition, modified diets, and probiotics, play a critical role in comprehensive disease management. Stem cell therapy and fecal microbiota transplant represent innovative approaches still under clinical evaluation. The review underscores the significance of holistic care, incorporating mind-body interventions and psychosocial support to improve patient quality of life. Key challenges persist, such as infection risks associated with long-term biological therapy use, gaps in pediatric-specific guidelines, and the limited inclusion of children in clinical trials. Future recommendations emphasize the importance of structured transition programs bridging pediatric and adult care, regular updates to clinical guidelines, and the integration of precision medicine to personalize treatment plans. Continued research and collaboration are essential for advancing the understanding and management of PIBD, ensuring that pediatric patients benefit from the most effective, evidence-based care available.
Collapse
Affiliation(s)
- Akshita Bhalla
- Internal Medicine, Punjab Institute of Medical Sciences, Jalandhar, IND
| | - Anushka Shahi
- Internal Medicine, Sri Aurobindo Institute of Medical Sciences, Indore, IND
| | - Madhurima Maity
- Critical Care Medicine, Sir H.N Reliance Foundation Hospital, Mumbai, IND
| | - Fnu Safa
- Internal Medicine, Osmania Medical College, Hyderabad, IND
| | | | | | | | - Salma Younas
- Pharmacy, Punjab University College of Pharmacy, Lahore, PAK
| |
Collapse
|
196
|
Silverman AL, Shung D, Stidham RW, Kochhar GS, Iacucci M. How Artificial Intelligence Will Transform Clinical Care, Research, and Trials for Inflammatory Bowel Disease. Clin Gastroenterol Hepatol 2025; 23:428-439.e4. [PMID: 38992406 PMCID: PMC11719376 DOI: 10.1016/j.cgh.2024.05.048] [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: 03/08/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 07/13/2024]
Abstract
Artificial intelligence (AI) refers to computer-based methodologies that use data to teach a computer to solve pre-defined tasks; these methods can be applied to identify patterns in large multi-modal data sources. AI applications in inflammatory bowel disease (IBD) includes predicting response to therapy, disease activity scoring of endoscopy, drug discovery, and identifying bowel damage in images. As a complex disease with entangled relationships between genomics, metabolomics, microbiome, and the environment, IBD stands to benefit greatly from methodologies that can handle this complexity. We describe current applications, critical challenges, and propose future directions of AI in IBD.
Collapse
Affiliation(s)
- Anna L Silverman
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona.
| | - Dennis Shung
- Section of Digestive Diseases, Department of Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Ryan W Stidham
- Division of Gastroenterology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan; Michigan Institute for Data Science, University of Michigan, Ann Arbor, Michigan
| | - Gursimran S Kochhar
- Division of Gastroenterology, Hepatology, and Nutrition, Allegheny Health Network, Pittsburgh, Pennsylvania
| | - Marietta Iacucci
- University of Birmingham, Institute of Immunology and Immunotherapy, Birmingham, United Kingdom; College of Medicine and Health, University College Cork, and APC Microbiome Ireland, Cork, Ireland
| |
Collapse
|
197
|
Liao TJ, Xia M, Hayashi P, Pan B, Aithal GP, Lucena MI, Andrade RJ, Rule JA, Lee WM, Rakela J, Huang R, Chen M. Genetic Variants of GBP4: Reduced Risks for Drug-Induced Acute Liver Failure in Non-Finnish European Population. Liver Int 2025; 45:e70011. [PMID: 39868816 PMCID: PMC11850085 DOI: 10.1111/liv.70011] [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: 10/02/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND AND AIMS Acute liver failure (ALF) is a serious condition, typically in individuals without prior liver disease. Drug-induced ALF (DIALF) constitutes a major portion of ALF cases. Our research aimed to identify potential genetic predispositions to DIALF. METHODS We analysed the potential genetic variants associated with DIALF using the whole exome sequencing data from 75 cases, including 40 non-Finnish European cases in the pilot study. Chi-square tests were performed for case-control analysis against the 1000 genomes project as the control. A replication study of 44 DIALF cases that included 24 non-Finnish Europeans was conducted to validate candidate variants. The association between clinical phenotype and genotypes was analysed using one-way analysis of variance. RESULTS Eight variants (rs561037, rs561042, rs608339, rs655260, rs1142886, rs1142888, rs1142889 and rs1142890) in the guanylate binding protein 4 (GBP4) were significantly associated with DIALF in non-Finnish Europeans in the pilot study and confirmed in the replication study. Rs561037 and rs561042 were highly significant with the lowest allele frequencies in both pilot and replication studies. An association was also found between these variants and milder clinical outcomes, indicated by lower peak levels of ALT, AST and higher Karnofsky performance scores. CONCLUSION Our study identified eight GBP4 missense variants linked to a lower risk of DIALF in the non-Finnish European population. The GBP4 protein, activated by interferon-gamma, plays a critical role in innate immunity. These findings suggest that GBP4 variants might influence immune and inflammatory responses in DIALF, though further studies are needed to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Tsung-Jen Liao
- Division of Bioinformatics and Statistics, the FDA’s National Center for Toxicological Research, Jefferson, AR, USA
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Menghang Xia
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Paul Hayashi
- Division of Hepatology and Nutrition, Office of New Drugs, FDA Center for Drug Evaluation and Research, Silver Spring, MD, USA
| | - Bohun Pan
- Division of Bioinformatics and Statistics, the FDA’s National Center for Toxicological Research, Jefferson, AR, USA
| | - Guruprasad P. Aithal
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Center at the Nottingham University Hospital NHS Trust and University of Nottingham, Nottingham, UK
| | - M. Isabel Lucena
- Servicios de Aparato Digestivo y de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga IBIMA_Plataforma Bionand, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Spain
- Centro de Investigacion Biomedica en Red Enfermedades Hepaticas y Digestivas (CIBERehd), Madrid, Spain
| | - Raúl J. Andrade
- Servicios de Aparato Digestivo y de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga IBIMA_Plataforma Bionand, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Spain
- Centro de Investigacion Biomedica en Red Enfermedades Hepaticas y Digestivas (CIBERehd), Madrid, Spain
| | - Jody A. Rule
- Division of Gastroenterology and Hepatology, University of Texas Southwestern, Dallas, TX, USA
| | - William M. Lee
- Division of Gastroenterology and Hepatology, University of Texas Southwestern, Dallas, TX, USA
| | - Jorge Rakela
- Division of Gastroenterology and Hepatology, Mayo Clinic, Phoenix, AZ, USA
| | - Ruili Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Minjun Chen
- Division of Bioinformatics and Statistics, the FDA’s National Center for Toxicological Research, Jefferson, AR, USA
| |
Collapse
|
198
|
Wen Z, Zhu L, He W, Liang T, Zhong Q, Long J, Su L. Exploring the causal inference of inflammatory bowel disease and ischemic stroke: a bidirectional two-sample Mendelian randomization study. J Thromb Thrombolysis 2025; 58:340-348. [PMID: 39720960 DOI: 10.1007/s11239-024-03065-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/05/2024] [Indexed: 12/26/2024]
Abstract
Growing evidence suggests inflammatory bowel disease (IBD) is linked to ischemic stroke (IS); however, the results are inconclusive. Therefore, it remains uncertain whether the association between IBD and IS is causal. Herein, we performed a bidirectional Mendelian randomization (MR) study to examine the causal association of IBD with IS. We obtained summary-level data for IBD and IS from several publicly released genome-wide association studies to conduct a two-sample bidirectional Mendelian randomization (MR) analysis. Herein, the inverse-variance weighted method was utilized as the primary approach. Then, we applied the weighted median and MR-Egger estimators for the follow-up sensitivity analyses. In addition, the MR-Egger intercept test was performed to detect the potential directional pleiotropy. Genetically predicted IBD was not causally associated with IS and IS subtypes (IS: OR = 0.99, 95% CI 0.98-1.01, p = 0.49; large artery atherosclerosis stroke: OR = 1.00, 95% CI: 0.96-1.05, p = 0.88; cardioembolic stroke: OR = 0.99, 95% CI 0.96-1.03, p = 0.75; small-vessel occlusion stroke: OR = 1.02, 95% CI 0.99-1.05, p = 0.16). Moreover, we did not find a significant causal effect of UC or CD on IS and IS subtypes. Furthermore, there was no significant association observed between IS and IBD in the reverse MR analysis. The estimates were consistent across sensitivity analyses. Our MR analysis does not support a bidirectional causal association between IBD and IS, despite observational studies reporting an association of IBD with IS.
Collapse
Affiliation(s)
- Zheng Wen
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Lulu Zhu
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, 530021, China
| | - Wanting He
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, 530021, China
| | - Tian Liang
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, 530021, China
| | - Qingqing Zhong
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, 530021, China
| | - Jianxiong Long
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, 530021, China.
| | - Li Su
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, 530021, China.
| |
Collapse
|
199
|
Samuels A, Irie K, Mizuno T, Reifenberg J, Punt N, Vinks AA, Minar P. Integrating early response biomarkers in pharmacokinetic models: A novel method to individualize the initial infliximab dose in patients with Crohn's disease. Clin Transl Sci 2025; 18:e70086. [PMID: 39985779 PMCID: PMC11846607 DOI: 10.1111/cts.70086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/10/2024] [Accepted: 11/05/2024] [Indexed: 02/24/2025] Open
Abstract
The use of model-informed precision dosing to personalize infliximab has been shown to improve both the acquisition of concentration targets and clinical outcomes during maintenance. Current iterations of infliximab pharmacokinetic models include time-varying covariates of drug clearance, however, not accounting for the expected improvements in the covariates can lead to indiscriminate use of higher infliximab doses and imprecise drug exposure. The aim was to identify changes in the four biomarkers associated with infliximab clearance (Xiong et al. model) and determine if integration of these dynamic changes would improve model performance during induction and early maintenance. We analyzed two cohorts of children receiving infliximab for Crohn's Disease. The Emax method was used to assess time-varying changes in covariates. Model performance (observed vs. predicted infliximab concentrations) was evaluated using median percentage error (bias) and median absolute percentage error (precision). The combined cohorts included 239 Crohn's disease patients. We found from baseline to dose 4, the maximum changes in weight, albumin, erythrocyte sedimentation rate, and neutrophil CD64 were 4.7%, +11.7%, -62.4%, and -26.5%, respectively. We also found the use of baseline covariates alone to forecast future trough concentration was inferior to the Emax time-varying method with a significant improvement observed in bias (doses 2, 3, and 4) and precision (doses 2 and 4). The integration of the four time-varying biomarkers of drug clearance with pharmacokinetic modeling improved the accuracy and precision of the predictions. This novel strategy may be key to improving drug exposure, minimizing indiscriminate dosing strategies, and reducing healthcare costs.
Collapse
Affiliation(s)
- Abigail Samuels
- Department of Internal Medicine, Department of Veterans AffairsUniversity of Cincinnati School of MedicineCincinnatiOhioUSA
| | - Kei Irie
- Division of Translational and Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Tomoyuki Mizuno
- Division of Translational and Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Jack Reifenberg
- University of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Nieko Punt
- MedimaticsMaastrichtThe Netherlands
- Department of Clinical Pharmacy and PharmacologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Alexander A. Vinks
- Division of Translational and Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Phillip Minar
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Division of Gastroenterology, Hepatology and NutritionCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| |
Collapse
|
200
|
Saberiyan M, Ghasemi N, Jafari N, Sadeghi M, Ghaderi A, Mousavi P. Investigate of LOC101928988 Regulatory Effect on the DAPK2 Transcription in Breast Tumors. Cancer Rep (Hoboken) 2025; 8:e70115. [PMID: 39971705 PMCID: PMC11839281 DOI: 10.1002/cnr2.70115] [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: 08/25/2024] [Revised: 11/24/2024] [Accepted: 12/24/2024] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND The World Health Organization has mentioned breast cancer holds the highest incidence rate among all types of cancer globally. Death-associated protein kinase 2 (DAPK2) is a serine/threonine kinase linked to various forms of malignancy, such as breast cancer. This protein assumes a pivotal function in a multitude of cellular mechanisms, including apoptosis, autophagy, and cell migration. AIMS This study aimed to study the LOC101928988 regulatory effect on the DAPK2 expression in breast cancer. METHODS In this study, 38 paired tumoral and normal tissues were selected from patients. Quantitative real-time PCR was used to analyze the expression of DAPK2 and LOC101928988. The interactions of DAPK2 and its intermediate elements with LOC101928988 were predicted by docking analysis. RESULTS The expression of DAPK2 and LOC101928988 was downregulated in tumor tissues compared to the control group. Further analysis revealed a significant positive correlation between DAPK2 and LOC101928988 levels in tumoral and adjacent normal tissues. A comparison of gene expression between different grades, stages, and HER2 statuses showed significant findings. ROC curve analysis of DAPK2 and LOC101928988 expression revealed 77% and 72% AUC for BC tissue, respectively. CONCLUSIONS Overall, our results suggest that alterations in the levels of DAPK2 and LOC101928988 may be involved in tumor initiation and progression in breast cancer. It has also been reported that LOC101928988 probably has a role in regulating DAPK2 expression through interaction with transcription factors.
Collapse
Affiliation(s)
- Mohammadreza Saberiyan
- Student Research CommitteeHormozgan University of Medical SciencesBandar AbbasIran
- Department of Medical Genetics, Faculty of MedicineHormozgan University of Medical SciencesBandar AbbasIran
| | - Nazila Ghasemi
- Department of Biology, Jahrom BranchIslamic Azad UniversityJahromIran
| | - Negar Jafari
- Department of Cardiology, School of MedicineUrmia University of Medical SciencesUrmiaIran
| | - Mahboubeh Sadeghi
- Student Research CommitteeHormozgan University of Medical SciencesBandar AbbasIran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of MedicineShiraz University of Medical SciencesShirazIran
- Department of Immunology, School of MedicineShiraz University of Medical SciencesShirazIran
| | - Pegah Mousavi
- Molecular Medicine Research CenterHormozgan Health Institute, Hormozgan University of Medical SciencesBandar AbbasIran
| |
Collapse
|