Pub Date : 2026-01-02DOI: 10.1136/gutjnl-2025-337298
Chhagan L Birda, Anuraag Jena, Joana Torres, Siew C Ng, Paulo Gustavo Kotze, Shaji Sebastian, Vishal Sharma
IBD is rising worldwide and is now a global disease. With the expanding armamentarium of medical therapies, including biologics and small molecules, there is a decline in hospitalisation rates and IBD-related surgeries. However, high costs, injectable therapy, risk of opportunistic infections and the lifelong nature of the disease pose significant challenges in the management of IBD. Developing countries are also constrained by a lack of trained manpower, as well as economic and infrastructural limitations. Strategies aimed at the prevention of IBD may alleviate the suffering and cost of this disease. Suggested approaches include implementation of prevention and interception trials using dietary, pharmacological and precision medicine approaches. However, these would necessitate massive funding and equitable infrastructural support for identifying the population at risk (for prevention trials) and those with preclinical disease (for interception trials). Hence, these strategies are unlikely to be globally practicable or economically viable, particularly in the Global South. It is believed that IBD, like certain non-communicable diseases (NCDs) such as metabolic syndrome and cardiovascular disorders, may be preventable by modifying the risk factors. Therefore, in this review, we advocate for an alternative approach of combining evidence-based IBD prevention strategies with the time-tested strategies of NCD prevention approaches already being implemented. We suggest a sieving strategy for selecting preventive measures through a series of sieves-interventions that have evidence to support prevention, align with NCD prevention and are economically viable.
{"title":"Road to IBD prevention in the Global South: a conceptual framework modelling from non-communicable diseases.","authors":"Chhagan L Birda, Anuraag Jena, Joana Torres, Siew C Ng, Paulo Gustavo Kotze, Shaji Sebastian, Vishal Sharma","doi":"10.1136/gutjnl-2025-337298","DOIUrl":"https://doi.org/10.1136/gutjnl-2025-337298","url":null,"abstract":"<p><p>IBD is rising worldwide and is now a global disease. With the expanding armamentarium of medical therapies, including biologics and small molecules, there is a decline in hospitalisation rates and IBD-related surgeries. However, high costs, injectable therapy, risk of opportunistic infections and the lifelong nature of the disease pose significant challenges in the management of IBD. Developing countries are also constrained by a lack of trained manpower, as well as economic and infrastructural limitations. Strategies aimed at the prevention of IBD may alleviate the suffering and cost of this disease. Suggested approaches include implementation of prevention and interception trials using dietary, pharmacological and precision medicine approaches. However, these would necessitate massive funding and equitable infrastructural support for identifying the population at risk (for prevention trials) and those with preclinical disease (for interception trials). Hence, these strategies are unlikely to be globally practicable or economically viable, particularly in the Global South. It is believed that IBD, like certain non-communicable diseases (NCDs) such as metabolic syndrome and cardiovascular disorders, may be preventable by modifying the risk factors. Therefore, in this review, we advocate for an alternative approach of combining evidence-based IBD prevention strategies with the time-tested strategies of NCD prevention approaches already being implemented. We suggest a sieving strategy for selecting preventive measures through a series of sieves-interventions that have evidence to support prevention, align with NCD prevention and are economically viable.</p>","PeriodicalId":12825,"journal":{"name":"Gut","volume":" ","pages":""},"PeriodicalIF":25.8,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145892335","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-31DOI: 10.1136/gutjnl-2025-337873
Jonas F Ludvigsson
{"title":"My first 1000 manuscript rejections.","authors":"Jonas F Ludvigsson","doi":"10.1136/gutjnl-2025-337873","DOIUrl":"https://doi.org/10.1136/gutjnl-2025-337873","url":null,"abstract":"","PeriodicalId":12825,"journal":{"name":"Gut","volume":" ","pages":""},"PeriodicalIF":25.8,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145877872","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-31DOI: 10.1136/gutjnl-2025-337431
Xiao-Dong Zhou, Qiong-Yue Fan, Christopher D Byrne, Giovanni Targher, Mark D Muthiah, Daniel Q Huang, Qin-Fen Chen, Mazen Noureddin, Wenhao Li, Vlad Ratziu, Rohit Loomba, Sven M Francque, Arun J Sanyal, Ming-Hua Zheng
Metabolic dysfunction-associated steatohepatitis (MASH) is a multifactorial metabolic liver disease that occurs in the context of obesity, insulin resistance and cardiometabolic comorbidities. Monotherapy targeting single pathways often provides only partial improvements in histological liver fibrosis and systemic metabolic parameters, prompting growing interest in multitargeted combination therapies. Multitargeted and combination strategies for MASH can modulate multiple and complementary pathogenic processes, potentially improving efficacy, promoting liver fibrosis regression, optimising systemic metabolic outcomes and reducing treatment-related adverse effects. Liver-directed combination therapies, such as thyroid hormone receptor-beta (THR-β) agonists along with acetyl-CoA carboxylase (ACC) inhibitors or peroxisome proliferator-activated receptor agonists, aim to improve histological features of MASH. Regimens combining systemic metabolic and liver-specific agents, such as glucagon-like peptide-1 (GLP-1) receptor agonists with fibroblast growth factor-21 analogues or THR-β agonists, aim to optimise metabolic outcomes, including body weight, insulin resistance and plasma lipids. Thoughtfully designed drug pairings, such as diacylglycerol O-acyltransferase 2 inhibitors combined with ACC inhibitors or GLP-1 and glucagon receptor dual agonists, could improve safety and tolerability by leveraging complementary mechanisms that may mitigate adverse effects. These therapeutic strategies aim to achieve more comprehensive and durable improvements in both liver pathology and systemic health than single-agent therapy alone. This review integrates current knowledge on multitargeted and combination therapies for MASH, examines mechanistic rationales and emerging clinical evidence and addresses practical considerations for patient-centred implementation. Therefore, we aim to provide clinicians and researchers with a comprehensive framework to optimise individualised management and improve both hepatic and systemic outcomes in individuals living with MASH.
{"title":"Combination therapies for metabolic dysfunction-associated steatohepatitis: challenges and opportunities.","authors":"Xiao-Dong Zhou, Qiong-Yue Fan, Christopher D Byrne, Giovanni Targher, Mark D Muthiah, Daniel Q Huang, Qin-Fen Chen, Mazen Noureddin, Wenhao Li, Vlad Ratziu, Rohit Loomba, Sven M Francque, Arun J Sanyal, Ming-Hua Zheng","doi":"10.1136/gutjnl-2025-337431","DOIUrl":"https://doi.org/10.1136/gutjnl-2025-337431","url":null,"abstract":"<p><p>Metabolic dysfunction-associated steatohepatitis (MASH) is a multifactorial metabolic liver disease that occurs in the context of obesity, insulin resistance and cardiometabolic comorbidities. Monotherapy targeting single pathways often provides only partial improvements in histological liver fibrosis and systemic metabolic parameters, prompting growing interest in multitargeted combination therapies. Multitargeted and combination strategies for MASH can modulate multiple and complementary pathogenic processes, potentially improving efficacy, promoting liver fibrosis regression, optimising systemic metabolic outcomes and reducing treatment-related adverse effects. Liver-directed combination therapies, such as thyroid hormone receptor-beta (THR-β) agonists along with acetyl-CoA carboxylase (ACC) inhibitors or peroxisome proliferator-activated receptor agonists, aim to improve histological features of MASH. Regimens combining systemic metabolic and liver-specific agents, such as glucagon-like peptide-1 (GLP-1) receptor agonists with fibroblast growth factor-21 analogues or THR-β agonists, aim to optimise metabolic outcomes, including body weight, insulin resistance and plasma lipids. Thoughtfully designed drug pairings, such as diacylglycerol O-acyltransferase 2 inhibitors combined with ACC inhibitors or GLP-1 and glucagon receptor dual agonists, could improve safety and tolerability by leveraging complementary mechanisms that may mitigate adverse effects. These therapeutic strategies aim to achieve more comprehensive and durable improvements in both liver pathology and systemic health than single-agent therapy alone. This review integrates current knowledge on multitargeted and combination therapies for MASH, examines mechanistic rationales and emerging clinical evidence and addresses practical considerations for patient-centred implementation. Therefore, we aim to provide clinicians and researchers with a comprehensive framework to optimise individualised management and improve both hepatic and systemic outcomes in individuals living with MASH.</p>","PeriodicalId":12825,"journal":{"name":"Gut","volume":" ","pages":""},"PeriodicalIF":25.8,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145877828","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-31DOI: 10.1136/gutjnl-2025-336451
Bingqi Han, Yutong Jiang, Haihua Liu, Fangling Zhang, Yi Xu, Zhuang Li, Songyuan Zheng, Fopei Ma, Hao Shi, Xinlei Jia, Jinhao Chen, Leyi Tan, Huijie Fan, Shuaijun Lv, Lei Gao, Xiaoshan Zhao, Hong-Wei Zhou, Juan Li, Mukeng Hong
Background: Gut microbiota has been widely recognised as playing a critical role in maintaining immune imbalance and the development of rheumatoid arthritis (RA). As key roles mediating interkingdom crosstalk among plants, microbiomes and mammals, plant-derived exosome-like nanoparticles (ELNs) could use lipids and microRNA components to precisely modulate gene expression of gut microbiota, showing potential as a dietary intervention for RA treatment.
Objective: We aimed to investigate gut microbiota-immune interactions inducing immune dysregulation in RA and explore potential applications of edible plant-derived ELNs for RA treatment through gut microbiota manipulation.
Design: Combinations of microbial analysis of clinical cohort, metabolomics, in vivo and in vitro examination were performed to establish potential gut-immune mechanisms for interventions. Several representative edible plants ELNs were chosen to compare the modulation effects based on the above mechanism. Small RNA sequencing and lipidomic analysis were performed to identify key components and reveal the related mechanisms mediating therapeutic effects.
Results: Ruminococcus gnavus was significantly enriched in RA and aggravated arthritis through secreting phenylethylamine (PEA) to induce excessive neutrophil extracellular traps (NETs) formation. Among several plants ELNs, Pueraria lobata-derived ELNs (Pu-ELNs) were preferentially taken up by R. gnavus and decreased PEA production. Mechanistically, the lipid components of Pu-ELNs induced intestinal accumulation of ELN-derived gma-miR4412, which reduces phenylalanine decarboxylase (PDC) expression, relieving the arthritis aggravation caused by R. gnavus through acting on the PEA-Bruton's tyrosine kinase (BTK)-NETs axis.
Conclusions: Our findings suggest the crucial role of R. gnavus in aggravating RA and underscore the application of plant-derived ELNs for microbiota manipulation.
{"title":"Pueraria lobata-derived exosome-like nanovesicles alleviate rheumatoid arthritis via targeting <i>Ruminococcus gnavus</i> phenylethylamine production.","authors":"Bingqi Han, Yutong Jiang, Haihua Liu, Fangling Zhang, Yi Xu, Zhuang Li, Songyuan Zheng, Fopei Ma, Hao Shi, Xinlei Jia, Jinhao Chen, Leyi Tan, Huijie Fan, Shuaijun Lv, Lei Gao, Xiaoshan Zhao, Hong-Wei Zhou, Juan Li, Mukeng Hong","doi":"10.1136/gutjnl-2025-336451","DOIUrl":"https://doi.org/10.1136/gutjnl-2025-336451","url":null,"abstract":"<p><strong>Background: </strong>Gut microbiota has been widely recognised as playing a critical role in maintaining immune imbalance and the development of rheumatoid arthritis (RA). As key roles mediating interkingdom crosstalk among plants, microbiomes and mammals, plant-derived exosome-like nanoparticles (ELNs) could use lipids and microRNA components to precisely modulate gene expression of gut microbiota, showing potential as a dietary intervention for RA treatment.</p><p><strong>Objective: </strong>We aimed to investigate gut microbiota-immune interactions inducing immune dysregulation in RA and explore potential applications of edible plant-derived ELNs for RA treatment through gut microbiota manipulation.</p><p><strong>Design: </strong>Combinations of microbial analysis of clinical cohort, metabolomics, in vivo and in vitro examination were performed to establish potential gut-immune mechanisms for interventions. Several representative edible plants ELNs were chosen to compare the modulation effects based on the above mechanism. Small RNA sequencing and lipidomic analysis were performed to identify key components and reveal the related mechanisms mediating therapeutic effects.</p><p><strong>Results: </strong><i>Ruminococcus gnavu</i>s was significantly enriched in RA and aggravated arthritis through secreting phenylethylamine (PEA) to induce excessive neutrophil extracellular traps (NETs) formation. Among several plants ELNs, Pueraria lobata-derived ELNs (Pu-ELNs) were preferentially taken up by <i>R. gnavus</i> and decreased PEA production. Mechanistically, the lipid components of Pu-ELNs induced intestinal accumulation of ELN-derived gma-miR4412, which reduces phenylalanine decarboxylase (PDC) expression, relieving the arthritis aggravation caused by <i>R. gnavus</i> through acting on the PEA-Bruton's tyrosine kinase (BTK)-NETs axis.</p><p><strong>Conclusions: </strong>Our findings suggest the crucial role of <i>R. gnavus</i> in aggravating RA and underscore the application of plant-derived ELNs for microbiota manipulation.</p>","PeriodicalId":12825,"journal":{"name":"Gut","volume":" ","pages":""},"PeriodicalIF":25.8,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145878101","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-31DOI: 10.1136/gutjnl-2025-337345
Yunha Noh, Ahhyung Choi, Hyesung Lee, Dong Keon Yon, Hyun-Soo Kim, Suyeon Kim, Ju-Young Shin, Laurent Azoulay
{"title":"Proton pump inhibitors use and risk of inflammatory bowel disease in children.","authors":"Yunha Noh, Ahhyung Choi, Hyesung Lee, Dong Keon Yon, Hyun-Soo Kim, Suyeon Kim, Ju-Young Shin, Laurent Azoulay","doi":"10.1136/gutjnl-2025-337345","DOIUrl":"https://doi.org/10.1136/gutjnl-2025-337345","url":null,"abstract":"","PeriodicalId":12825,"journal":{"name":"Gut","volume":" ","pages":""},"PeriodicalIF":25.8,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145877862","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-25DOI: 10.1136/gutjnl-2025-336527
Yasi Pan,Xiang Zhang,Chi Chun Wong,Huarong Chen,Stephanie Ma,Terence Kin-Wah Lee,Kai Yuan,Cong Liang,Xingyu Zhou,Harry Cheuk Hay Lau,Pingmei Huang,Danyu Chen,Lina Wang,Yanqiang Ding,Qinyao Wei,Alvin Ho Kwan Cheung,Ka Fai To,Jun Yu
OBJECTIVEMetabolic dysfunction-associated steatotic liver disease-related hepatocellular carcinoma (MASLD-HCC) is an emerging malignancy with limited therapeutic options. The identity and function of cancer stem cells (CSCs) in MASLD-HCC remain poorly understood. In this study, we characterised CSCs in MASLD-HCC and investigated their contribution to MASLD-HCC tumourigenesis and therapy response.DESIGNWe performed expression profiling in human MASLD-HCC samples (n=29 pairs of tumour and adjacent normal tissues). Advanced in vivo genetic lineage tracing coupled with single-cell RNA sequencing was used to characterise CD133+ CSCs in preclinical models. To establish causality, we developed a hepatocyte-specific CD133-overexpressing mouse model of MASLD-HCC. We identified CD133 protein interactors by mass spectrometry. A novel strategy combining CD133-targeted small interfering RNA (siRNA) nanoparticles with first-line therapy was assessed in clinically relevant MASLD-HCC models.RESULTSCD133+ CSCs were significantly enriched in human MASLD-HCC tumours and positively correlated with established markers of malignancy. In vivo genetic lineage tracing in mice revealed that CD133+ cells exhibit hallmark CSC properties, including self-renewal, tumour-initiating capacity and multipotent differentiation, as compared with CD133- counterparts. Hepatocyte-specific CD133 overexpression in mice accelerated MASLD-HCC tumourigenesis. Mechanistically, CD133 interacts with myosin heavy chain 9 (MYH9) to stabilise active β-catenin, thereby propagating Wnt/β-catenin signalling that drives CSC phenotypes and tumourigenic potential. Therapeutically, genetic ablation of CD133+ cells or systemic delivery of CD133-siRNA nanoparticles potently sensitised MASLD-HCC to sorafenib and lenvatinib, significantly improving outcomes in MASLD-HCC.CONCLUSIONThis study established CD133+ CSCs as critical mediators through the CD133-MYH9/β-catenin axis in MASLD-HCC. Targeting CD133 enhances multikinase inhibitor efficacy, offering a promising therapeutic strategy for MASLD-HCC.
{"title":"Targeting cancer stem cells enhances multikinase inhibitor therapy in metabolic dysfunction-associated steatotic liver disease-related hepatocellular carcinoma.","authors":"Yasi Pan,Xiang Zhang,Chi Chun Wong,Huarong Chen,Stephanie Ma,Terence Kin-Wah Lee,Kai Yuan,Cong Liang,Xingyu Zhou,Harry Cheuk Hay Lau,Pingmei Huang,Danyu Chen,Lina Wang,Yanqiang Ding,Qinyao Wei,Alvin Ho Kwan Cheung,Ka Fai To,Jun Yu","doi":"10.1136/gutjnl-2025-336527","DOIUrl":"https://doi.org/10.1136/gutjnl-2025-336527","url":null,"abstract":"OBJECTIVEMetabolic dysfunction-associated steatotic liver disease-related hepatocellular carcinoma (MASLD-HCC) is an emerging malignancy with limited therapeutic options. The identity and function of cancer stem cells (CSCs) in MASLD-HCC remain poorly understood. In this study, we characterised CSCs in MASLD-HCC and investigated their contribution to MASLD-HCC tumourigenesis and therapy response.DESIGNWe performed expression profiling in human MASLD-HCC samples (n=29 pairs of tumour and adjacent normal tissues). Advanced in vivo genetic lineage tracing coupled with single-cell RNA sequencing was used to characterise CD133+ CSCs in preclinical models. To establish causality, we developed a hepatocyte-specific CD133-overexpressing mouse model of MASLD-HCC. We identified CD133 protein interactors by mass spectrometry. A novel strategy combining CD133-targeted small interfering RNA (siRNA) nanoparticles with first-line therapy was assessed in clinically relevant MASLD-HCC models.RESULTSCD133+ CSCs were significantly enriched in human MASLD-HCC tumours and positively correlated with established markers of malignancy. In vivo genetic lineage tracing in mice revealed that CD133+ cells exhibit hallmark CSC properties, including self-renewal, tumour-initiating capacity and multipotent differentiation, as compared with CD133- counterparts. Hepatocyte-specific CD133 overexpression in mice accelerated MASLD-HCC tumourigenesis. Mechanistically, CD133 interacts with myosin heavy chain 9 (MYH9) to stabilise active β-catenin, thereby propagating Wnt/β-catenin signalling that drives CSC phenotypes and tumourigenic potential. Therapeutically, genetic ablation of CD133+ cells or systemic delivery of CD133-siRNA nanoparticles potently sensitised MASLD-HCC to sorafenib and lenvatinib, significantly improving outcomes in MASLD-HCC.CONCLUSIONThis study established CD133+ CSCs as critical mediators through the CD133-MYH9/β-catenin axis in MASLD-HCC. Targeting CD133 enhances multikinase inhibitor efficacy, offering a promising therapeutic strategy for MASLD-HCC.","PeriodicalId":12825,"journal":{"name":"Gut","volume":"8 1","pages":""},"PeriodicalIF":24.5,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145830453","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
BACKGROUNDPerianal fistulising disease (PFD) is a complication that affects about 20% of patients with Crohn's disease (CD) whose aetiology remains unknown.OBJECTIVESTo identify predisposing events driving fistula formation.DESIGNRectal biopsies from patients with CD with or without PFD (CD+PFD and CD, respectively; n=31) were collected and subjected to single-cell RNA sequencing. Functional analyses were conducted using peripheral CD3+ T cells, intestinal tissue explants, primary fibroblasts and two-dimensional epithelial monolayer cell cultures.RESULTSThe rectal mucosa of patients with CD+PFD is imprinted with cellular and transcriptomic alterations specific to PFD and independent of luminal inflammation, potentially driven by tumour necrosis factor-like ligand 1A (TL1A) activation in CD4+ T cells. We identified lymphotoxin beta (LTB or its functional heterotrimer LTα1β2) as a novel mediator downstream of TL1A that, along with interleukin (IL)-22, induces a PFD-associated signature in rectal fibroblast and epithelial cells, respectively. This signature includes an increased abundance of fibroblasts, an induction of matrix-degrading enzymes, transcriptomic rewiring of the lamina propria S1 fibroblasts and an anti-bacterial and immune responses in epithelial cells. Notably, the induction of LTα1β2 and IL-22 occurs independently of tumour necrosis factor (TNF) signalling, revealing a new TL1A-LTα1β2/IL-22 axis that remains active under anti-TNF therapy.CONCLUSIONOur findings revealed unique cellular alterations in the rectum of patients with CD+PFD, highlighting the previously unrecognised involvement of TL1A in mediating this signature and supporting the need for exploring the role of TL1A inhibition as a therapeutic approach for PFD.
{"title":"TL1A-activated T cells remodel the rectal mucosa in patients with Crohn's disease with perianal fistulising disease.","authors":"Victoria Gudiño,Jae Won Cho,Berta Caballol,Ángela Sanzo-Machuca,Ana Corraliza,Marisol Veny,Isabella Dotti,Livia Moreira Genaro,Elisa Melón-Ardanaz,Maria Carme Masamunt,Miriam Esteller,Iris Teubel,Lisseth Robbins,Àngel Giner,Cristina Prieto,Elena Ferrer,Raquel Franco Leal,Albert Martin-Cardona,Carme Loras,Maria Esteve,Jordi Rimola,Agnès Fernández-Clotet,Ingrid Ordás,Elena Ricart,Julian Panés,Martin Hemberg,Azucena Salas","doi":"10.1136/gutjnl-2025-336246","DOIUrl":"https://doi.org/10.1136/gutjnl-2025-336246","url":null,"abstract":"BACKGROUNDPerianal fistulising disease (PFD) is a complication that affects about 20% of patients with Crohn's disease (CD) whose aetiology remains unknown.OBJECTIVESTo identify predisposing events driving fistula formation.DESIGNRectal biopsies from patients with CD with or without PFD (CD+PFD and CD, respectively; n=31) were collected and subjected to single-cell RNA sequencing. Functional analyses were conducted using peripheral CD3+ T cells, intestinal tissue explants, primary fibroblasts and two-dimensional epithelial monolayer cell cultures.RESULTSThe rectal mucosa of patients with CD+PFD is imprinted with cellular and transcriptomic alterations specific to PFD and independent of luminal inflammation, potentially driven by tumour necrosis factor-like ligand 1A (TL1A) activation in CD4+ T cells. We identified lymphotoxin beta (LTB or its functional heterotrimer LTα1β2) as a novel mediator downstream of TL1A that, along with interleukin (IL)-22, induces a PFD-associated signature in rectal fibroblast and epithelial cells, respectively. This signature includes an increased abundance of fibroblasts, an induction of matrix-degrading enzymes, transcriptomic rewiring of the lamina propria S1 fibroblasts and an anti-bacterial and immune responses in epithelial cells. Notably, the induction of LTα1β2 and IL-22 occurs independently of tumour necrosis factor (TNF) signalling, revealing a new TL1A-LTα1β2/IL-22 axis that remains active under anti-TNF therapy.CONCLUSIONOur findings revealed unique cellular alterations in the rectum of patients with CD+PFD, highlighting the previously unrecognised involvement of TL1A in mediating this signature and supporting the need for exploring the role of TL1A inhibition as a therapeutic approach for PFD.","PeriodicalId":12825,"journal":{"name":"Gut","volume":"25 1","pages":""},"PeriodicalIF":24.5,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145830317","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
BACKGROUNDParasutterella excrementihominis (P. excrementihominis), a Betaproteobacteria species enriched in ulcerative colitis (UC) patients, is implicated in chronic inflammation. However, its mechanistic role in UC progression and colitis-associated colorectal cancer (CAC) remains unclear.OBJECTIVEThis study investigates the pathogenic role of P. excrementihominis in UC and CAC, focusing on its induction of neutrophil extracellular traps (NETs) and underlying mechanisms.DESIGNClinical stool samples from UC patients and healthy controls were analysed for P. excrementihominis abundance. Murine models of dextran sulphate sodium (DSS)-induced colitis and azoxymethane/DSS-induced CAC were used to evaluate bacterial pathogenicity. RNA sequencing and metabolomic analyses were conducted on germ-free mice with monocolonisation, and in vitro cell experiments were carried out to elucidate the role of bacterial metabolites in NETosis.RESULTSP. excrementihominis was significantly enriched in UC patients and exacerbated colitis and CAC in mice by expanding colonic neutrophils and NETs formation. Metabolomic profiling revealed that P. excrementihominis enhances the host's carbohydrate metabolic capacity, leading to increased production of succinic acid (Suc) and 6-hydroxyhexanoic acid (6-HHA). These metabolites activated gasdermin D (GSDMD)-dependent NETosis in lipopolysaccharide-primed neutrophils through the succinate receptor 1/G protein-coupled receptor 84 signalling pathway. Conversely, neutrophil-specific GSDMD deletion attenuated metabolite-driven tumourigenesis.CONCLUSIONOur findings identify P. excrementihominis as a critical microbial driver of UC and CAC pathogenesis. This bacterium significantly accelerates disease progression by producing specific metabolites (Suc and 6-HHA) that induce pathogenic NETosis. Targeting this bacterium or its metabolic axis offers novel therapeutic strategies for inflammation-driven colorectal carcinogenesis.
{"title":"Parasutterella excrementihominis exacerbates experimental colitis and colitis-associated colorectal cancer via pathogenic NETosis activation.","authors":"Huishi Tan,Linwen Huang,Jun Wang,Hongli Huang,Zelong Lin,Siqi Yang,Yanqiang Shi,Jierui Li,Haiyan Zhang,Yongjian Zhou,Chongyang Huang","doi":"10.1136/gutjnl-2025-335887","DOIUrl":"https://doi.org/10.1136/gutjnl-2025-335887","url":null,"abstract":"BACKGROUNDParasutterella excrementihominis (P. excrementihominis), a Betaproteobacteria species enriched in ulcerative colitis (UC) patients, is implicated in chronic inflammation. However, its mechanistic role in UC progression and colitis-associated colorectal cancer (CAC) remains unclear.OBJECTIVEThis study investigates the pathogenic role of P. excrementihominis in UC and CAC, focusing on its induction of neutrophil extracellular traps (NETs) and underlying mechanisms.DESIGNClinical stool samples from UC patients and healthy controls were analysed for P. excrementihominis abundance. Murine models of dextran sulphate sodium (DSS)-induced colitis and azoxymethane/DSS-induced CAC were used to evaluate bacterial pathogenicity. RNA sequencing and metabolomic analyses were conducted on germ-free mice with monocolonisation, and in vitro cell experiments were carried out to elucidate the role of bacterial metabolites in NETosis.RESULTSP. excrementihominis was significantly enriched in UC patients and exacerbated colitis and CAC in mice by expanding colonic neutrophils and NETs formation. Metabolomic profiling revealed that P. excrementihominis enhances the host's carbohydrate metabolic capacity, leading to increased production of succinic acid (Suc) and 6-hydroxyhexanoic acid (6-HHA). These metabolites activated gasdermin D (GSDMD)-dependent NETosis in lipopolysaccharide-primed neutrophils through the succinate receptor 1/G protein-coupled receptor 84 signalling pathway. Conversely, neutrophil-specific GSDMD deletion attenuated metabolite-driven tumourigenesis.CONCLUSIONOur findings identify P. excrementihominis as a critical microbial driver of UC and CAC pathogenesis. This bacterium significantly accelerates disease progression by producing specific metabolites (Suc and 6-HHA) that induce pathogenic NETosis. Targeting this bacterium or its metabolic axis offers novel therapeutic strategies for inflammation-driven colorectal carcinogenesis.","PeriodicalId":12825,"journal":{"name":"Gut","volume":"8 1","pages":""},"PeriodicalIF":24.5,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145830329","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-24DOI: 10.1136/gutjnl-2025-335618
Mengxi Du, Xinyu Wang, Dong Hang, Fenglei Wang, Yujia Lu, Kai Wang, Alaina M Bever, Ana Nogal, Danielle Haslam, Shuji Ogino, Jeffrey A Meyerhardt, Liming Liang, Qi Sun, Curtis Huttenhower, Andrew T Chan, Frank B Hu, Mingyang Song
Background: High ultra-processed food (UPF) intake has been linked to colorectal cancer (CRC), but underlying mechanisms remain unclear.
Objective: To evaluate a metabolomic pattern of UPF intake and its association with CRC risk.
Design: Integrating food frequency questionnaire data and high-throughput metabolomic profiling in 1740 participants (mean age at blood draw: 59.9 years; >95% non-Hispanic white participants) from nested case-control studies within the Nurses' Health Study and Health Professionals Follow-up Study, we derived and validated a UPF-related metabolomic pattern as a weighted sum of metabolites selected via elastic net regression with 10-fold cross-validation. We evaluated prospective associations of this pattern and individual metabolites with CRC risk using multivariable conditional logistic regression in 686 pairs of incident CRC cases and matched controls.
Results: Among 222 metabolites, we constructed a UPF metabolomic pattern comprising 50 metabolites, primarily lipids and amino acids, with 22 positively and 28 inversely associated with total UPF intake (pattern vs intake: Spearman rho=0.35). The pattern was associated with higher CRC risk (highest vs lowest quintile: OR (95% CI) 1.71 (1.15 to 2.53), p value trend=0.002). Correlations of individual metabolites with UPF intake were moderately aligned with their associations with CRC risk (rho=0.50). N2, N2-dimethylguanosine, a marker of meat/poultry intake, was positively associated with CRC risk (1.96 (1.27 to 3.03)), while 21-deoxycortisol, related to cortisol biosynthesis, was inversely associated (0.59 (0.41 to 0.86)).
Conclusion: We developed a UPF metabolomic pattern. The pattern and several metabolites were associated with CRC risk, providing biological insights into potential pathways underlying the UPF-CRC relationship.
{"title":"Metabolomic pattern of ultraprocessed food intake and its association with colorectal cancer risk.","authors":"Mengxi Du, Xinyu Wang, Dong Hang, Fenglei Wang, Yujia Lu, Kai Wang, Alaina M Bever, Ana Nogal, Danielle Haslam, Shuji Ogino, Jeffrey A Meyerhardt, Liming Liang, Qi Sun, Curtis Huttenhower, Andrew T Chan, Frank B Hu, Mingyang Song","doi":"10.1136/gutjnl-2025-335618","DOIUrl":"https://doi.org/10.1136/gutjnl-2025-335618","url":null,"abstract":"<p><strong>Background: </strong>High ultra-processed food (UPF) intake has been linked to colorectal cancer (CRC), but underlying mechanisms remain unclear.</p><p><strong>Objective: </strong>To evaluate a metabolomic pattern of UPF intake and its association with CRC risk.</p><p><strong>Design: </strong>Integrating food frequency questionnaire data and high-throughput metabolomic profiling in 1740 participants (mean age at blood draw: 59.9 years; >95% non-Hispanic white participants) from nested case-control studies within the Nurses' Health Study and Health Professionals Follow-up Study, we derived and validated a UPF-related metabolomic pattern as a weighted sum of metabolites selected via elastic net regression with 10-fold cross-validation. We evaluated prospective associations of this pattern and individual metabolites with CRC risk using multivariable conditional logistic regression in 686 pairs of incident CRC cases and matched controls.</p><p><strong>Results: </strong>Among 222 metabolites, we constructed a UPF metabolomic pattern comprising 50 metabolites, primarily lipids and amino acids, with 22 positively and 28 inversely associated with total UPF intake (pattern vs intake: Spearman rho=0.35). The pattern was associated with higher CRC risk (highest vs lowest quintile: OR (95% CI) 1.71 (1.15 to 2.53), p value trend=0.002). Correlations of individual metabolites with UPF intake were moderately aligned with their associations with CRC risk (rho=0.50). N2, N2-dimethylguanosine, a marker of meat/poultry intake, was positively associated with CRC risk (1.96 (1.27 to 3.03)), while 21-deoxycortisol, related to cortisol biosynthesis, was inversely associated (0.59 (0.41 to 0.86)).</p><p><strong>Conclusion: </strong>We developed a UPF metabolomic pattern. The pattern and several metabolites were associated with CRC risk, providing biological insights into potential pathways underlying the UPF-CRC relationship.</p>","PeriodicalId":12825,"journal":{"name":"Gut","volume":" ","pages":""},"PeriodicalIF":25.8,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145827583","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
BACKGROUNDGermline RNF43 mutations cause a dominantly inherited syndrome of colorectal cancer (CRC) and serrated polyps. However, these data originate from highly selected families.OBJECTIVEWe assessed germline RNF43 variants in patients more representative of the general population and compared these with somatic RNF43mutations in CRCs.DESIGNWe studied 49 823 CRC and/or polyp cases from the CORGI study, 100 000 Genomes (100kGP) and UK Biobank (UKB), alongside 165 250 controls. Somatic mutations were analysed in 2722 CRCs.RESULTSConsistent with the literature, a germline loss-of-function RNF43 variant (p.Thr158ProfsTer6) was found in a multigenerational CORGI family with early-onset CRC and serrated and/or filiform polyps. However, while 23 CRC/polyp cases and 47 controls from 100kGP or UKB had germline RNF43 mutations, cases often lacked multiple polyps or a notable family history. Sometimes, CRCs developed independently of the germline RNF43 mutation. In case-control analyses, germline RNF43 variants were associated with CRC risk (OR=2.696, p=0.010), but penetrance was much greater for germline mutations in the N-terminal half of the gene. Germline C-terminal mutations conferred no increased CRC risk. However, somatic C-terminal mutations were pathogenic, perhaps because their relatively weak effects are supplemented by accompanying mutations in Wnt genes, including ZNRF3 and a new driver, SFRP4.CONCLUSIONRNF43 is a CRC predisposition gene, but risks are moderate, the reported polyposis phenotype is often absent and molecular phenocopies can occur. N-terminal germline RNF43 variants confer higher risk, although weak effects of C-terminal variants cannot be excluded. Genetic testing and patient management should incorporate these factors.
{"title":"Comparison between germline and somatic loss-of-function RNF43 mutations reveals different genotype-phenotype associations and provides insights into the genetic mechanisms of colorectal tumourigenesis.","authors":"Claire Palles,Luke Freeman-Mills,Edward Arbe-Barnes,Nathalie Feeley,Laura Chegwidden,Helen Curley,Sara Galavotti,Connor Woolley,Jeremy Cheadle,Dmitri Mouradov,Oliver Sieber,Silvia Salatino,Steve Thorn,Anshita Goel,Juan Fernandez-Tajes,Sulochana Omwenga,Sujata Biswas,Timothy Maughan,Simon J Leedham, , , , ,Viktor Hendrik Koelzer,Lai Mun Wang,Roland Arnold,James Edward East,Ian Tomlinson","doi":"10.1136/gutjnl-2025-337030","DOIUrl":"https://doi.org/10.1136/gutjnl-2025-337030","url":null,"abstract":"BACKGROUNDGermline RNF43 mutations cause a dominantly inherited syndrome of colorectal cancer (CRC) and serrated polyps. However, these data originate from highly selected families.OBJECTIVEWe assessed germline RNF43 variants in patients more representative of the general population and compared these with somatic RNF43mutations in CRCs.DESIGNWe studied 49 823 CRC and/or polyp cases from the CORGI study, 100 000 Genomes (100kGP) and UK Biobank (UKB), alongside 165 250 controls. Somatic mutations were analysed in 2722 CRCs.RESULTSConsistent with the literature, a germline loss-of-function RNF43 variant (p.Thr158ProfsTer6) was found in a multigenerational CORGI family with early-onset CRC and serrated and/or filiform polyps. However, while 23 CRC/polyp cases and 47 controls from 100kGP or UKB had germline RNF43 mutations, cases often lacked multiple polyps or a notable family history. Sometimes, CRCs developed independently of the germline RNF43 mutation. In case-control analyses, germline RNF43 variants were associated with CRC risk (OR=2.696, p=0.010), but penetrance was much greater for germline mutations in the N-terminal half of the gene. Germline C-terminal mutations conferred no increased CRC risk. However, somatic C-terminal mutations were pathogenic, perhaps because their relatively weak effects are supplemented by accompanying mutations in Wnt genes, including ZNRF3 and a new driver, SFRP4.CONCLUSIONRNF43 is a CRC predisposition gene, but risks are moderate, the reported polyposis phenotype is often absent and molecular phenocopies can occur. N-terminal germline RNF43 variants confer higher risk, although weak effects of C-terminal variants cannot be excluded. Genetic testing and patient management should incorporate these factors.","PeriodicalId":12825,"journal":{"name":"Gut","volume":"9 1","pages":""},"PeriodicalIF":24.5,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145823987","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}