1
|
Moutsoglou D, Ramakrishnan P, Vaughn BP. Microbiota transplant therapy in inflammatory bowel disease: advances and mechanistic insights. Gut Microbes 2025; 17:2477255. [PMID: 40062406 PMCID: PMC11901402 DOI: 10.1080/19490976.2025.2477255] [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: 11/06/2024] [Revised: 01/27/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Microbiota transplant therapy is an emerging therapy for inflammatory bowel disease, but factors influencing its efficacy and mechanism remain poorly understood. In this narrative review, we outline key elements affecting therapeutic outcomes, including donor factors (such as age and patient relationship), recipient factors, control selection, and elements impacting engraftment and its correlation with clinical response. We also examine potential mechanisms through inflammatory bowel disease trials, focusing on the interplay between the microbiota, host, and immune system. Finally, we briefly explore potential future directions for microbiota transplant therapy and promising emerging treatments.
Collapse
Affiliation(s)
- Daphne Moutsoglou
- Gastroenterology Section, Minneapolis VA Health Care System, Minneapolis, MN, USA
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | - Byron P. Vaughn
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
2
|
Lin A, Jiang A, Huang L, Li Y, Zhang C, Zhu L, Mou W, Liu Z, Zhang J, Cheng Q, Wei T, Luo P. From chaos to order: optimizing fecal microbiota transplantation for enhanced immune checkpoint inhibitors efficacy. Gut Microbes 2025; 17:2452277. [PMID: 39826104 DOI: 10.1080/19490976.2025.2452277] [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: 10/16/2024] [Revised: 11/22/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
The integration of fecal microbiota transplantation (FMT) with immune checkpoint inhibitors (ICIs) presents a promising approach for enhancing cancer treatment efficacy and overcoming therapeutic resistance. This review critically examines the controversial effects of FMT on ICIs outcomes and elucidates the underlying mechanisms. We investigate how FMT modulates gut microbiota composition, microbial metabolite profiles, and the tumor microenvironment, thereby influencing ICIs effectiveness. Key factors influencing FMT efficacy, including donor selection criteria, recipient characteristics, and administration protocols, are comprehensively discussed. The review delineates strategies for optimizing FMT formulations and systematically monitoring post-transplant microbiome dynamics. Through a comprehensive synthesis of evidence from clinical trials and preclinical studies, we elucidate the potential benefits and challenges of combining FMT with ICIs across diverse cancer types. While some studies report improved outcomes, others indicate no benefit or potential adverse effects, emphasizing the complexity of host-microbiome interactions in cancer immunotherapy. We outline critical research directions, encompassing the need for large-scale, multi-center randomized controlled trials, in-depth microbial ecology studies, and the integration of multi-omics approaches with artificial intelligence. Regulatory and ethical challenges are critically addressed, underscoring the imperative for standardized protocols and rigorous long-term safety assessments. This comprehensive review seeks to guide future research endeavors and clinical applications of FMT-ICIs combination therapy, with the potential to improve cancer patient outcomes while ensuring both safety and efficacy. As this rapidly evolving field advances, maintaining a judicious balance between openness to innovation and cautious scrutiny is crucial for realizing the full potential of microbiome modulation in cancer immunotherapy.
Collapse
Affiliation(s)
- Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Lihaoyun Huang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Yu Li
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Chunyanx Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Lingxuan Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Weiming Mou
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| |
Collapse
|
3
|
Dongre DS, Saha UB, Saroj SD. Exploring the role of gut microbiota in antibiotic resistance and prevention. Ann Med 2025; 57:2478317. [PMID: 40096354 PMCID: PMC11915737 DOI: 10.1080/07853890.2025.2478317] [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: 10/28/2024] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND/INTRODUCTION Antimicrobial resistance (AMR) and the evolution of multiple drug-resistant (MDR) bacteria is of grave public health concern. To combat the pandemic of AMR, it is necessary to focus on novel alternatives for drug development. Within the host, the interaction of the pathogen with the microbiome plays a pivotal role in determining the outcome of pathogenesis. Therefore, microbiome-pathogen interaction is one of the potential targets to be explored for novel antimicrobials. MAIN BODY This review focuses on how the gut microbiome has evolved as a significant component of the resistome as a source of antibiotic resistance genes (ARGs). Antibiotics alter the composition of the native microbiota of the host by favouring resistant bacteria that can manifest as opportunistic infections. Furthermore, gut dysbiosis has also been linked to low-dosage antibiotic ingestion or subtherapeutic antibiotic treatment (STAT) from food and the environment. DISCUSSION Colonization by MDR bacteria is potentially acquired and maintained in the gut microbiota. Therefore, it is pivotal to understand microbial diversity and its role in adapting pathogens to AMR. Implementing several strategies to prevent or treat dysbiosis is necessary, including faecal microbiota transplantation, probiotics and prebiotics, phage therapy, drug delivery models, and antimicrobial stewardship regulation.
Collapse
Affiliation(s)
- Devyani S Dongre
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra, India
| | - Ujjayni B Saha
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra, India
| | - Sunil D Saroj
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra, India
| |
Collapse
|
4
|
Hoops SL, Moutsoglou D, Vaughn BP, Khoruts A, Knights D. Metagenomic source tracking after microbiota transplant therapy. Gut Microbes 2025; 17:2487840. [PMID: 40229213 PMCID: PMC12005403 DOI: 10.1080/19490976.2025.2487840] [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: 12/13/2024] [Revised: 03/07/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025] Open
Abstract
Reliable engraftment assessment of donor microbial communities and individual strains is an essential component of characterizing the pharmacokinetics of microbiota transplant therapies (MTTs). Recent methods for measuring donor engraftment use whole-genome sequencing and reference databases or metagenome-assembled genomes (MAGs) to track individual bacterial strains but lack the ability to disambiguate DNA that matches both donor and patient microbiota. Here, we describe a new, cost-efficient analytic pipeline, MAGEnTa, which compares post-MTT samples to a database comprised MAGs derived directly from donor and pre-treatment metagenomic data, without relying on an external database. The pipeline uses Bayesian statistics to determine the likely sources of ambiguous reads that align with both the donor and pre-treatment samples. MAGEnTa recovers engrafted strains with minimal type II error in a simulated dataset and is robust to shallow sequencing depths in a downsampled dataset. Applying MAGEnTa to a dataset from a recent MTT clinical trial for ulcerative colitis, we found the results to be consistent with 16S rRNA gene SourceTracker analysis but with added MAG-level specificity. MAGEnTa is a powerful tool to study community and strain engraftment dynamics in the development of MTT-based treatments that can be integrated into frameworks for functional and taxonomic analysis.
Collapse
Affiliation(s)
- Susan L. Hoops
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
- Biotechnology Institute, University of Minnesota, Minneapolis, MN, USA
| | - Daphne Moutsoglou
- Gastroenterology Section, Minneapolis VA Health Care System, Minneapolis, MN, USA
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Byron P. Vaughn
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Division of Gastroenterology, University of Minnesota, Minneapolis, MN, USA
| | - Alexander Khoruts
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Division of Gastroenterology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Dan Knights
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
- Biotechnology Institute, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
5
|
An L, Li S, Chang Z, Lei M, He Z, Xu P, Zhang S, Jiang Z, Iqbal MS, Sun X, Liu H, Duan X, Wu W. Gut microbiota modulation via fecal microbiota transplantation mitigates hyperoxaluria and calcium oxalate crystal depositions induced by high oxalate diet. Gut Microbes 2025; 17:2457490. [PMID: 39873191 PMCID: PMC11776474 DOI: 10.1080/19490976.2025.2457490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 12/16/2024] [Accepted: 01/17/2025] [Indexed: 01/30/2025] Open
Abstract
Hyperoxaluria, including primary and secondary hyperoxaluria, is a disorder characterized by increased urinary oxalate excretion and could lead to recurrent calcium oxalate kidney stones, nephrocalcinosis and eventually end stage renal disease. For secondary hyperoxaluria, high dietary oxalate (HDOx) or its precursors intake is a key reason. Recently, accumulated studies highlight the important role of gut microbiota in the regulation of oxalate homeostasis. However, the underlying mechanisms involving gut microbiota and metabolite disruptions in secondary hyperoxaluria remain poorly understood. Here, we investigated the therapeutic efficacy of fecal microbiota transplantation (FMT) sourced from healthy rats fed with standard pellet diet against urinary oxalate excretion, renal damage and calcium oxalate (CaOx) crystal depositions via using hyperoxaluria rat models. We observed dose-dependent increases in urinary oxalate excretion and CaOx crystal depositions due to hyperoxaluria, accompanied by significant reductions in gut microbiota diversity characterized by shifts in Ruminococcaceae_UCG-014 and Parasutterella composition. Metabolomic analysis validated these findings, revealing substantial decreases in key metabolites associated with these microbial groups. Transplanting microbes from healthy rats effectively reduced HDOx-induced urinary oxalate excretion and CaOx crystal depositions meanwhile restoring Ruminococcaceae_UCG-014 and Parasutterella populations and their associated metabolites. Furthermore, FMT treatment could significantly decrease the urinary oxalate excretion and CaOx crystal depositions in rat kidneys via, at least in part, upregulating the expressions of intestinal barrier proteins and oxalate transporters in the intestine. In conclusion, our study emphasizes the effectiveness of FMT in countering HDOx-induced hyperoxaluria by restoring gut microbiota and related metabolites. These findings provide insights on the complex connection between secondary hyperoxaluria caused by high dietary oxalate and disruptions in gut microbiota, offering promising avenues for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Lingyue An
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Shujue Li
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhenglin Chang
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Min Lei
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhican He
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Peng Xu
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shike Zhang
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zheng Jiang
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Muhammad Sarfaraz Iqbal
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xinyuan Sun
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hongxing Liu
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaolu Duan
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenqi Wu
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
6
|
Xia J, Liu T, Wan R, Zhang J, Fu Q. Global burden and trends of the Clostridioides difficile infection-associated diseases from 1990 to 2021: an observational trend study. Ann Med 2025; 57:2451762. [PMID: 39847395 PMCID: PMC11758798 DOI: 10.1080/07853890.2025.2451762] [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: 05/17/2024] [Revised: 09/09/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND This study was aimed to explore the global burden and trends of Clostridioides difficile infections (CDI) associated diseases. METHODS Data for this study were obtained from the Global Burden of Disease Study 2021. The burden of CDI was assessed using the age-standardized rates of disability-adjusted life years (ASR-DALYs) and deaths (ASDRs). Trends in the burden of CDI were presented using average annual percentage changes (AAPCs). RESULTS The ASR-DALYs for CDI increased from 1.83 (95% UI: 1.53-2.18) per 100,000 in 1990 to 3.46 (95% UI: 3.04-3.96) per 100,000 in 2021, with an AAPC of 2.03% (95% CI: 1.67-2.4%). The ASDRs for CDI rose from 0.10 (95% UI: 0.08-0.11) per 100,000 in 1990 to 0.19 (95% UI: 0.16-0.23) per 100,000 in 2021, with an AAPC of 2.26% (95% CI: 1.74-2.79%). In 2021, higher burdens of ASR-DALYs (10.7 per 100,000) and ASDRs (0.53 per 100,000) were observed in high socio-demographic index (SDI) areas, and among age group over 70 years (31.62/100,000 for ASR-DALYs and 2.45/100,000 for ASDRs). During the COVID-19 pandemic, the global ASR-DALYs and ASDRs slightly decreased. However, in regions with low SDI, low-middle and middle SDI, those rates slightly increased. CONCLUSION The global burden of CDI has significantly increased, particularly in regions with high SDI and among individuals aged 70 years and above. During the COVID-19 pandemic period from 2020 to 2021, the burden of CDI further increased in regions with low, low-middle, and middle SDI. These findings underscore the need for increased attention and intervention, especially in specific countries and populations.
Collapse
Affiliation(s)
- Jun Xia
- Department of Neurocritical Care, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Tan Liu
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Rui Wan
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jing Zhang
- Department of Neurocritical Care, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Quanzhu Fu
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
7
|
Barrios Steed D, Koundakjian D, Harris AD, Rosato AE, Konstantinidis KT, Woodworth MH. Leveraging strain competition to address antimicrobial resistance with microbiota therapies. Gut Microbes 2025; 17:2488046. [PMID: 40195644 PMCID: PMC11988218 DOI: 10.1080/19490976.2025.2488046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 12/28/2024] [Accepted: 03/28/2025] [Indexed: 04/09/2025] Open
Abstract
The enteric microbiota is an established reservoir for multidrug-resistant organisms that present urgent clinical and public health threats. Observational data and small interventional studies suggest that microbiome interventions, such as fecal microbiota products and characterized live biotherapeutic bacterial strains, could be an effective antibiotic-sparing prevention approach to address these threats. However, bacterial colonization is a complex ecological phenomenon that remains understudied in the context of the human gut. Antibiotic resistance is one among many adaptative strategies that impact long-term colonization. Here we review and synthesize evidence of how bacterial competition and differential fitness in the context of the gut present opportunities to improve mechanistic understanding of colonization resistance, therapeutic development, patient care, and ultimately public health.
Collapse
Affiliation(s)
- Danielle Barrios Steed
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Anthony D. Harris
- Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
- Institute for Healthcare Computing, University of Maryland, Baltimore, MD, USA
| | - Adriana E Rosato
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
| | | | - Michael H Woodworth
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
8
|
Jin T, Li SY, Zheng HL, Liu XD, Huang Y, Ma G, Zhao YX, Zhao XT, Yang L, Wang QH, Wang HJ, Gu C, Pan Z, Lin F. Gut microbes-spinal connection is required for itch sensation. Gut Microbes 2025; 17:2495859. [PMID: 40289281 PMCID: PMC12036491 DOI: 10.1080/19490976.2025.2495859] [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: 08/26/2024] [Revised: 03/23/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
The gut microbiota has been linked to a number of neurological disorders. However, it is unclear whether the gut microbiota is involved in the genesis of chronic itch, a refractory condition that afflicts patients both physically and mentally. Here, we report that depletion of gut microbiota enhances tolerance to itch in mice orally administered with antibiotics (ABX) and mice free of germ. Of note, oral gavage with Bacteroides fragilis (B. fragilis), a prominent species of the genus Bacteroides with most differential change, corrected the ABX-induced itch dysfunction through its driven metabolite acetyl-l-carnitine (ALC). Mechanistically, gut microbiota or B. fragilis depletion caused a decrease in RNA N6-methyladenosine (m6A) demethylase FTO expression in the dorsal horn and a consequent increase in RNA m6A sites in Mas-related G protein-coupled receptor F (MrgprF) mRNA, leading to decreased MRGPRF protein. The downregulation of FTO was triggered by inactivation of ETS proto-oncogene 1 (ETS1), a transcription factor that binds to the Fto promoter. These findings support a gut microbe - spinal connection in modulation of itch sensation in RNA m6A epigenetic-dependent manner and highlight a critical role of ALC in linking the altered B. fragilis and itch dysfunction.
Collapse
Affiliation(s)
- Tong Jin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Department of Pain, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Si-Yuan Li
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Hong-Li Zheng
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Department of Pain, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Anesthesiology Department, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Xiao-Dan Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Department of Anesthesiology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Yue Huang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Gan Ma
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Ya-Xuan Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Xiao-Tian Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Li Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Qi-Hui Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Hong-Jun Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Chengyong Gu
- Anesthesiology Department, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Zhiqiang Pan
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Fuqing Lin
- Department of Pain, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
9
|
Steinert RE, Rehman A, Sadabad MS, Milanese A, Wittwer-Schegg J, Burton JP, Spooren A. Microbial micronutrient sharing, gut redox balance and keystone taxa as a basis for a new perspective to solutions targeting health from the gut. Gut Microbes 2025; 17:2477816. [PMID: 40090884 PMCID: PMC11913388 DOI: 10.1080/19490976.2025.2477816] [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: 09/13/2024] [Revised: 02/05/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025] Open
Abstract
In health, the gut microbiome functions as a stable ecosystem maintaining overall balance and ensuring its own survival against environmental stressors through complex microbial interaction. This balance and protection from stressors is maintained through interactions both within the bacterial ecosystem as well as with its host. As a consequence, the gut microbiome plays a critical role in various physiological processes including maintaining the structure and function of the gut barrier, educating the gut immune system, and modulating the gut motor, digestive/absorptive, as well as neuroendocrine system all of which are crucial for human health and disease pathogenesis. Pre- and probiotics, widely available and clinically established, offer various health benefits primarily by beneficially modulating the gut microbiome. However, their clinical outcomes can vary significantly due to differences in host physiology, diets, individual microbiome compositions, and other environmental factors. This perspective paper highlights emerging scientific insights into the importance of microbial micronutrient sharing, gut redox balance, keystone species, and the gut barrier in maintaining a diverse and functional microbial ecosystem, and their relevance to human health. We propose a novel approach that targets microbial ecosystems and keystone taxa performance by supplying microbial micronutrients in the form of colon-delivered vitamins, and precision prebiotics [e.g. human milk oligosaccharides (HMOs) or synthetic glycans] as components of precisely tailored ingredient combinations to optimize human health. Such a strategy may effectively support and stabilize microbial ecosystems, providing a more robust and consistent approach across various individuals and environmental conditions, thus, overcoming the limitations of current single biotic solutions.
Collapse
Affiliation(s)
- Robert E Steinert
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
- Department of Surgery and Transplantation, University Hospital Zurich (USZ) and University of Zurich (UZH), Zürich, Switzerland
| | - Ateequr Rehman
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
| | | | - Alessio Milanese
- Data Science, Science & Research, Dsm-Firmenich, Delft, Netherlands
| | | | - Jeremy P Burton
- Department of Microbiology and Immunology, The University of Western Ontario, London, Canada
| | - Anneleen Spooren
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
| |
Collapse
|
10
|
Gustafson KL, Rodriguez TR, McAdams ZL, Coghill LM, Ericsson AC, Franklin CL. Failure of colonization following gut microbiota transfer exacerbates DSS-induced colitis. Gut Microbes 2025; 17:2447815. [PMID: 39812347 PMCID: PMC11740679 DOI: 10.1080/19490976.2024.2447815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025] Open
Abstract
To study the impact of differing specific pathogen-free gut microbiomes (GMs) on a murine model of inflammatory bowel disease, selected GMs were transferred using embryo transfer (ET), cross-fostering (CF), and co-housing (CH). Prior work showed that the GM transfer method and the microbial composition of donor and recipient GMs can influence microbial colonization and disease phenotypes in dextran sodium sulfate-induced colitis. When a low richness GM was transferred to a recipient with a high richness GM via CH, the donor GM failed to successfully colonize, and a more severe disease phenotype resulted when compared to ET or CF, where colonization was successful. By comparing CH and gastric gavage for fecal material transfer, we isolated the microbial component of this effect and determined that differences in disease severity and survival were associated with microbial factors rather than the transfer method itself. Mice receiving a low richness GM via CH and gastric gavage exhibited greater disease severity and higher expression of pro-inflammatory immune mediators compared to those receiving a high richness GM. This study provides valuable insights into the role of GM composition and colonization in disease modulation.
Collapse
Affiliation(s)
- Kevin L. Gustafson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Trevor R. Rodriguez
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
| | - Zachary L. McAdams
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- Molecular Pathogenesis and Therapeutics Program, University of Missouri, Columbia, MO, USA
| | - Lyndon M. Coghill
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- University of Missouri Bioinformatics and Analytics Core, University of Missouri, Columbia, MO, USA
| | - Aaron C. Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- University of Missouri College of Veterinary Medicine, Columbia, MO, USA
- University of Missouri Metagenomics Center, Columbia, MO, USA
| | - Craig L. Franklin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| |
Collapse
|
11
|
Hoffmann DE, Javitt GH, Kelly CR, Keller JJ, Baunwall SMD, Hvas CL. Fecal microbiota transplantation: a tale of two regulatory pathways. Gut Microbes 2025; 17:2493901. [PMID: 40302307 PMCID: PMC12054926 DOI: 10.1080/19490976.2025.2493901] [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: 01/07/2025] [Revised: 04/04/2025] [Accepted: 04/10/2025] [Indexed: 05/02/2025] Open
Abstract
Fecal microbiota transplantation (FMT) is a procedure involving the transfer of intestinal microbiota from a healthy donor to a patient to restore a functional intestinal microbiome. First described in modern science in 1958, the use of FMT has been practiced for decades, but only during the past dozen years have clinical frameworks and legal regulations from competent authorities been developed. Future development of microbiota-derived medical therapies will be shaped by the regulatory frameworks of various jurisdictions. This review examines the historical development and status of FMT regulations in the United States and Europe, with particular attention to their respective approaches to ensuring the safety and quality of the therapeutic product and patient access.
Collapse
Affiliation(s)
- Diane E. Hoffmann
- University of Maryland Francis King Carey School of Law, Baltimore, MD, USA
| | | | - Colleen R. Kelly
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Josbert J. Keller
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, The Hague, Netherlands
- Department of Gastroenterology, Haaglanden Medical Center, The Hague, Netherlands
- Netherlands Donor Feces Bank, Leiden, Netherlands
| | | | - Christian Lodberg Hvas
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
12
|
Faith JJ. Assessing live microbial therapeutic transmission. Gut Microbes 2025; 17:2447836. [PMID: 39746875 DOI: 10.1080/19490976.2024.2447836] [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: 09/23/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
The development of fecal microbiota transplantation and defined live biotherapeutic products for the treatment of human disease has been an empirically driven process yielding a notable success of approved drugs for the treatment of recurrent Clostridioides difficile infection. Assessing the potential of this therapeutic modality in other indications with mixed clinical results would benefit from consistent quantitative frameworks to characterize drug potency and composition and to assess the impact of dose and composition on the frequency and duration of strain engraftment. Monitoring these drug properties and engraftment outcomes would help identify minimally sufficient sets of microbial strains to treat disease and provide insights into the intersection between microbial function and host physiology. Broad and correct usage of strain detection methods is essential to this advancement. This article describes strain detection approaches, where they are best applied, what data they require, and clinical trial designs that are best suited to their application.
Collapse
Affiliation(s)
- Jeremiah J Faith
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
13
|
Guo S, Li G, Du W, Situ F, Li Z, Lei J. The performance of ChatGPT and ERNIE Bot in surgical resident examinations. Int J Med Inform 2025; 200:105906. [PMID: 40220627 DOI: 10.1016/j.ijmedinf.2025.105906] [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: 10/30/2024] [Revised: 03/10/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025]
Abstract
STUDY PURPOSE To assess the application of these two large language models (LLMs) for surgical resident examinations and to compare the performance of these LLMs with that of human residents. STUDY DESIGN In this study, 596 questions with a total of 183,556 responses were first included from the Medical Vision World, an authoritative medical education platform across China. Both Chinese prompted and non-prompted questions were input into ChatGPT-4.0 and ERNIE Bot-4.0 to compare their performance in a Chinese question database. Additionally, we screened another 210 surgical questions with detailed response results from 43 residents to compare the performance of residents and these two LLMs. RESULTS There were no significant differences in the correctness of the responses to the 596 questions with or without prompts between the two LLMs (ChatGPT-4.0: 68.96 % [without prompt], 71.14 % [with prompts], p = 0.411; ERNIE Bot-4.0: 78.36 % [without prompt], 78.86 % [with prompts], p = 0.832), but ERNIE Bot-4.0 displayed higher correctness than ChatGPT-4.0 did (with prompts: p = 0.002; without prompts: p < 0.001). For another 210 questions with prompts, the two LLMs, especially ERNIE Bot-4.0 (ranking in the top 95 % of the 43 residents' scores), significantly outperformed the residents. CONCLUSIONS The performance of ERNIE Bot-4.0 was superior to that of ChatGPT-4.0 and that of residents on surgical resident examinations in a Chinese question database.
Collapse
Affiliation(s)
- Siyin Guo
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; The Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Genpeng Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; The Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Wei Du
- Beijing Medical Vision Times Technology Development Company Limited, Beijing, China.
| | - Fangzhi Situ
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Zhihui Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; The Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Jianyong Lei
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; The Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
14
|
Silva EN, Dos Santos TCF, Teixeira VC, Belo TCA, Augusto LV, de Almeida LA, Corsetti PP. Amoxicillin-induced bacterial gut dysbiosis: A critical influence on mice reproduction and their offspring development. Microb Pathog 2025; 204:107594. [PMID: 40246155 DOI: 10.1016/j.micpath.2025.107594] [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: 01/07/2025] [Revised: 04/08/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
The use of antibiotics such as amoxicillin can induce intestinal dysbiosis leading to rupture the essential microbiota role in regulating immune, metabolic, and reproductive functions. This study assessed the effects of amoxicillin-induced intestinal dysbiosis on the female mice reproductive function and its repercussions on their offspring. Female mice were treated with amoxicillin for 15 days (AMOX) showed an increase in Proteobacteria and a decrease in Firmicutes and Bacteroidetes in feces and estrous cycle changes, with a predominance of the metestrus and diestrus phases in the treated mice. During gestation the AMOX group presented reduced number of implantations and decreased embryonic viability, resulting in a higher rate of resorption. Differential gene expression of reproductive hormones in AMOX-treated female mice suggested that intestinal dysbiosis interferes with hormonal regulation during pregnancy. The survival, body development, and intestinal microbiota composition of offspring showed significantly altered patterns in the AMOX mice. These findings indicate that amoxicillin-induced intestinal dysbiosis affects not only the estrous cycle and reproductive hormones but also has lasting impacts on offspring development. The study highlights the need for caution in the use of antibiotics during pregnancy to avoid potential harm to maternal and offspring health.
Collapse
Affiliation(s)
- Evandro Neves Silva
- Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas (UNIFAL), Alfenas, 37130-001, Minas Gerais, Brazil; Professor Edson Antônio Velano University (UNIFENAS), Alfenas, 37132-440, Minas Gerais, Brazil
| | - Thaís Cristina Ferreira Dos Santos
- Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas (UNIFAL), Alfenas, 37130-001, Minas Gerais, Brazil; Professor Edson Antônio Velano University (UNIFENAS), Alfenas, 37132-440, Minas Gerais, Brazil; Laboratory National Biosciences, Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, 13083-100, São Paulo, Brazil
| | - Vanessa Coelho Teixeira
- Professor Edson Antônio Velano University (UNIFENAS), Alfenas, 37132-440, Minas Gerais, Brazil
| | - Thiago Caetano Andrade Belo
- Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas (UNIFAL), Alfenas, 37130-001, Minas Gerais, Brazil
| | - Lara Vilela Augusto
- Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas (UNIFAL), Alfenas, 37130-001, Minas Gerais, Brazil
| | - Leonardo Augusto de Almeida
- Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas (UNIFAL), Alfenas, 37130-001, Minas Gerais, Brazil.
| | - Patrícia Paiva Corsetti
- Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas (UNIFAL), Alfenas, 37130-001, Minas Gerais, Brazil.
| |
Collapse
|
15
|
Yu RL, Weber HC. Irritable bowel syndrome, the gut microbiome, and diet. Curr Opin Endocrinol Diabetes Obes 2025; 32:102-107. [PMID: 39968682 DOI: 10.1097/med.0000000000000905] [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] [Indexed: 02/20/2025]
Abstract
PURPOSE OF REVIEW To provide an update of recent studies exploring the role of the gut microbiota and diet in the pathogenesis and treatment of irritable bowel syndrome (IBS). RECENT FINDINGS The human gut microbiome has been recognized as an important, active source of signaling molecules that explain in part the disorder of the gut brain interaction (DGBI) in IBS. Subsequent changes in the metabolome such as the production of short-chain fatty acids (SCFA) and serotonin are associated with IBS symptoms. Dietary components are recognized as important triggers of IBS symptoms and a diet low in fermentable oligo-, di-, monosaccharides, and polyols (FODMAPs) has been shown effective and safe, even when used long-term. Fecal microbiota transplantation (FMT) in IBS has not shown sustained and effective IBS symptom reduction in controlled clinical trials. SUMMARY This update elucidates recent developments in IBS as it relates to clinical trial results targeting dietary and gut microbiota interventions. The gut microbiome is metabolically active and affects the bi-directional signaling of the gut-brain axis.
Collapse
Affiliation(s)
- Rosa Lu Yu
- Boston University Chobanian & Avedisian School of Medicine
| | - H Christian Weber
- Boston University Chobanian & Avedisian School of Medicine
- VA Boston Healthcare System, Section of Gastroenterology and Hepatology, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Lee Y, Koh HG, Kim KH, Jin YS, Sung BH, Kim J. Enhancing the persistence of engineered biotherapeutics in the gut: Adhesion, glycan metabolism, and environmental resistance. Adv Drug Deliv Rev 2025; 221:115591. [PMID: 40250567 DOI: 10.1016/j.addr.2025.115591] [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: 12/30/2024] [Revised: 03/20/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Engineered live biotherapeutic products (eLBPs) are receiving increasing attention as next-generation therapeutics to treat a variety of diseases with high specificity and effectiveness. Despite their potential, eLBPs face challenges, such as limited colonization, competition with native microbiota, nutrient depletion, and susceptibility to gastrointestinal stresses, which ultimately reduce their persistence in the gut and hinder their therapeutic efficacy. This review examines the key strategies to enhance the persistence and activity of eLBPs in the gut environment. First, methods to strengthen the adhesion capacity of eLBPs are discussed, including genetic engineering to express adhesins and chemical surface modifications to improve their binding to mucus and epithelial cells. Second, strategies to improve the ability of eLBPs to efficiently use mucin-derived sugars, which are continuously secreted by intestinal epithelial cells, were highlighted. These strategies involve the introduction and optimization of glycan-degrading enzymes and metabolic pathways for key mucin sugars, such as N-acetylglucosamine, galactose, and sialic acid, to support sustained energy production and enhance gut colonization. Third, strategies to improve the resistance of eLBPs against environmental stress are discussed, including genetic modifications to stabilize cell membranes, enhancement of ion pump activity, overexpression of stress-response proteins, and encapsulation techniques to provide protection. The implementation of these strategies can address challenges related to gut colonization by eLBPs, thereby enhancing their metabolic activity and enabling sustained and efficient secretion of therapeutic molecules. This review offers a comprehensive framework for developing and optimizing eLBPs, paving the way for their successful clinical application with enhanced effectiveness in treating gastrointestinal and systemic diseases.
Collapse
Affiliation(s)
- Yujin Lee
- Institute of Food Industrialization, Institutes of Green Bioscience and Technology, Seoul National University, Gangwon-do 25354, Republic of Korea; Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun 25354 Gangwon-do, Republic of Korea
| | - Hyun Gi Koh
- Department of Biological and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Yong-Su Jin
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Bong Hyun Sung
- Synthetic Biology and Bioengineering Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Jungyeon Kim
- Institute of Food Industrialization, Institutes of Green Bioscience and Technology, Seoul National University, Gangwon-do 25354, Republic of Korea; Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun 25354 Gangwon-do, Republic of Korea.
| |
Collapse
|
17
|
Chen Y, Tilves C, Bohn B, Doyon M, Bouchard L, Perron P, Guerin R, Masse E, Hivert MF, Mueller NT. Gut microbiota and microbial metabolites are associated with body composition in 5-year-old children: A cross-sectional study in the Gen3G cohort. Pediatr Obes 2025; 20:e70007. [PMID: 40059505 PMCID: PMC12058418 DOI: 10.1111/ijpo.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 05/08/2025]
Abstract
OBJECTIVE To examine gut microbiota diversity, composition and metabolites in relation to overall mass (OM), fat mass (FM) and lean soft tissue mass (LSTM) measured by dual x-ray absorptiometry (DXA) in 5-year-old children. METHODS Mothers of the Gen3G cohort were enrolled prenatally in 2010-2013 in Quebec, Canada; 153 children from the cohort had data on gut microbiota and DXA scans at 5-6.4 years of age, and 140 also had plasma metabolite data. We characterized gut microbiota by 16S rRNA Illumina sequencing and metabolites by untargeted multiplatform mass spectrometry. We examined associations of microbial alpha diversity, beta diversity, composition (amplicon sequence variants; ASVs) and metabolites (microbial metabolites) with DXA measures, adjusting for age, sex, diet and drinking water. RESULTS Of the 153 children, 43.1% were female, and 96.1% self-identified as white. The median BMI was the 52nd percentile. Microbial richness (alpha diversity) was positively associated with OM, FM and LSTM. Of the 542 ASVs tested, 7 were associated with OM, 5 with FM and 4 with LSTM. One Veillonella ASV and two Blautia ASVs were significantly associated with all outcomes. Among 278 microbial metabolites, no metabolites were associated with FM, while glycoursodeoxycholate was associated with OM, and glycoursodeoxycholate, 3-hydroxybutyrate and gamma-glutamylalanine were associated with LSTM. CONCLUSIONS In 5-year-old children, gut microbiota alpha diversity, richness and specific gut microbes were associated with OM, FM and LSTM. Many of the associations followed a similar pattern for FM and LSTM, suggesting they may not be specific to adiposity but rather reflect overall growth.
Collapse
Affiliation(s)
- Yingan Chen
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado, Aurora, Colorado, USA
| | - Curtis Tilves
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado, Aurora, Colorado, USA
| | - Bruno Bohn
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado, Aurora, Colorado, USA
| | - Myriam Doyon
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Quebec, Canada
| | - Luigi Bouchard
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Quebec, Canada
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Quebec, Canada
- Department of Medical Biology, CIUSSS-SLSJ, Quebec, Canada
| | - Patrice Perron
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Quebec, Canada
- Department of Medicine, Université de Sherbrooke, Quebec, Canada
| | - Renee Guerin
- Department of Medical Biology, CIUSSS-SLSJ, Quebec, Canada
| | - Eric Masse
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Quebec, Canada
| | - Marie-France Hivert
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Quebec, Canada
- Department of Medicine, Université de Sherbrooke, Quebec, Canada
- Division of Chronic Disease Across the Lifecourse (CoRAL), Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, Massachusetts, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Noel T Mueller
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado, Aurora, Colorado, USA
- Department of Pediatrics, School of Medicine, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
18
|
Muteeb G, Kazi RNA, Aatif M, Azhar A, Oirdi ME, Farhan M. Antimicrobial resistance: Linking molecular mechanisms to public health impact. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 33:100232. [PMID: 40216324 DOI: 10.1016/j.slasd.2025.100232] [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: 02/15/2025] [Revised: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Antimicrobial resistance (AMR) develops into a worldwide health emergency through genetic and biochemical adaptations which enable microorganisms to resist antimicrobial treatment. β-lactamases (blaNDM, blaKPC) and efflux pumps (MexAB-OprM) working with mobile genetic elements facilitate fast proliferation of multidrug-resistant (MDR) and exttreme drug-resistant (XDR) phenotypes thus creating major concerns for healthcare systems and community health as well as the agricultural sector. OBJECTIVES The review dissimilarly unifies molecular resistance pathways with public health implications through the study of epidemiological data and monitoring approaches and innovative therapeutic solutions. Previous studies separating their attention between molecular genetics and clinical outcomes have been combined into our approach which delivers an all-encompassing analysis of AMR. KEY INSIGHTS The report investigates the resistance mechanisms which feature enzymatic degradation and efflux pump overexpression together with target modification and horizontal gene transfer because these factors represent important contributors to present-day AMR developments. This review investigates AMR effects on hospital and community environments where it affects pathogens including MRSA, carbapenem-resistant Klebsiella pneumoniae, and drug-resistant Pseudomonas aeruginosa. This document explores modern AMR management methods that comprise WHO GLASS molecular surveillance systems and three innovative strategies such as CRISPR-modified genome editing and bacteriophage treatments along with antimicrobial peptides and artificial intelligence diagnostic tools. CONCLUSION The resolution of AMR needs complete scientific and global operational methods alongside state-of-the-art therapeutic approaches. Worldwide management of drug-resistant infection burden requires both enhanced infection prevention procedures with next-generation antimicrobial strategies to reduce cases effectively.
Collapse
Affiliation(s)
- Ghazala Muteeb
- Department of Nursing, College of Applied Medical Science, King Faisal University, Al-Ahsa, Saudi Arabia.
| | - Raisa Nazir Ahmed Kazi
- Department of Respiratory Therapy, College of Applied Medical Science, King Faisal, University, Al-Ahsa, Saudi Arabia
| | - Mohammad Aatif
- Department of Public Health, College of Applied Medical Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Asim Azhar
- NAP Life Sciences; Metropolitan Region, Maharashtra 401208, India
| | - Mohamed El Oirdi
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia; Department of Basic Sciences, Preparatory Year, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Mohd Farhan
- Department of Basic Sciences, Preparatory Year, King Faisal University, Al-Ahsa, Saudi Arabia; Department of Chemistry, College of Science, King Faisal University, Al Ahsa, Saudi Arabia.
| |
Collapse
|
19
|
Zhang YJ, Wang ML, Li Y, Lu XL, Liu XR, Hu DX, Chen A, Yin Q. Probiotic effects of Clostridium cellabutyricum YQ-FP-027 T on DSS-induced colitis mice via modulating gut microbiota and preventing inflammation. Int Immunopharmacol 2025; 155:114642. [PMID: 40228421 DOI: 10.1016/j.intimp.2025.114642] [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: 10/18/2024] [Revised: 03/08/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Butyrate-producing Clostridium spp. play fundamental roles in maintaining gut microbiota homeostasis by producing short-chain fatty acids. However, exploration of the strain resources and their probiotic effects remains limited. In previous work, a novel species of butyrate-producing Clostridium, Clostridium cellabutyricum YQ-FP-027T, was isolated from the pit mud of Chinese Baijiu brewing, which possesses antibacterial activities by modulating gut microbiota. The present study investigated the probiotic effects of YQ-FP-027T on mice with dextran sulfate sodium (DSS)-induced colitis and the underlying mechanism. The results showed that the mice treated with YQ-FP-027T exhibited lower disease activity index (DAI), recovered weight loss, and reduced colon shortening. Regarding gut microbial composition, these mice showed increased Shannon and Species richness observed (Sobs) indexes and a rising abundance of beneficial bacteria such as Akkermansia and Alistipes. Additionally, the expression levels of inflammatory factors Interleukin-1 beta (IL-1β), Interleukin-6 (IL-6), and Tumor Necrosis Factor-alpha (TNF-α) were suppressed, while those of anti-inflammatory factor Interleukin-10 (IL-10) and the cathelicidin-related antimicrobial peptide (CRAMP) were enhanced under YQ-FP-027T treatment. These results suggest that YQ-FP-027T holds potential as a promising probiotic against DSS-induced colitis via optimizing gut microbial community structure and modulating gut immune microenvironment.
Collapse
Affiliation(s)
- Yuan-Jie Zhang
- College of Public Health, Chongqing Medical University, No. 61 Daxuecheng Middle Road, Shapingba District, Chongqing 401334, PR China
| | - Meng-Lin Wang
- College of Public Health, Chongqing Medical University, No. 61 Daxuecheng Middle Road, Shapingba District, Chongqing 401334, PR China
| | - Yingli Li
- College of Public Health, Chongqing Medical University, No. 61 Daxuecheng Middle Road, Shapingba District, Chongqing 401334, PR China
| | - Xiao-Ling Lu
- College of Public Health, Chongqing Medical University, No. 61 Daxuecheng Middle Road, Shapingba District, Chongqing 401334, PR China
| | - Xiang-Ru Liu
- College of Public Health, Chongqing Medical University, No. 61 Daxuecheng Middle Road, Shapingba District, Chongqing 401334, PR China
| | - Dai-Xin Hu
- College of Public Health, Chongqing Medical University, No. 61 Daxuecheng Middle Road, Shapingba District, Chongqing 401334, PR China
| | - Anyi Chen
- College of Public Health, Chongqing Medical University, No. 61 Daxuecheng Middle Road, Shapingba District, Chongqing 401334, PR China
| | - Qi Yin
- College of Public Health, Chongqing Medical University, No. 61 Daxuecheng Middle Road, Shapingba District, Chongqing 401334, PR China.
| |
Collapse
|
20
|
Shatila M, Cruz CC, Lu L, Abdul-Baki K, Baerman E, Takigawa K, Rivera AU, Lee IJA, Ngo S, Sperling G, Aleem AS, Menon R, Sullivan A, Vemulapalli V, Natha C, Gupta T, Khan A, Mittal N, Coleman G, Salim H, Wali S, Varatharajalu K, Kim KC, Reddy SA, Grivas P, Thomas AS, Wang Y. The association between metformin use, immune mediated colitis and overall survival in patients treated with checkpoint inhibitor. Eur J Cancer 2025; 221:115405. [PMID: 40239400 DOI: 10.1016/j.ejca.2025.115405] [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: 12/10/2024] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025]
Abstract
INTRODUCTION Metformin is frequently prescribed to treat type 2 diabetes. Its primarily regulates hepatic and colonic glucose metabolism, but recent studies have suggested an anti-inflammatory effect, especially in colitis. It has been suggested that metformin may enhance immune checkpoint inhibition (ICI) efficacy for cancer treatment. Our study aims to explore the impact of metformin on ICI efficacy and the risk for colitis. METHODS This was a single center, retrospective analysis of consecutive patients at a tertiary cancer center who received ICI between 01/2010-12/2022 and developed immune-mediated colitis (IMC). Patients were screened for colitis based on stool tests, then divided into two groups depending on metformin use prior to colitis onset. We collected data on demographic and colitis clinical information including treatments, and outcomes. RESULTS A total of 953 patients were included. The incidence of IMC was higher among metformin users (7.6 %) than non-metformin users (4.9 %; p < 0.01). There were no significant differences in colitis features and outcomes, except for longer hospital stay among metformin users (8 days vs 6 for non-metformin users; p = 0.03). Metformin use was associated with shorter overall survival vs non-metformin users among patients with IMC (p = 0.03). DISCUSSION Our study is among the first to explore the impact of metformin on IMC and overall survival. We found that metformin use may be associated with higher risk of IMC. We also found an association between metformin use and shorter overall survival among patients who developed IMC. Larger studies with risk-stratified analysis are needed to validate our findings.
Collapse
Affiliation(s)
- Malek Shatila
- The University of Texas MD Anderson Cancer Center, Department of Gastroenterology, Hepatology, and Nutrition, Houston, TX, United States
| | - Carolina Colli Cruz
- The University of Texas MD Anderson Cancer Center, Department of Gastroenterology, Hepatology, and Nutrition, Houston, TX, United States
| | - Linfeng Lu
- Baylor College of Medicine, Department of Internal Medicine, Houston, TX, United States
| | - Kian Abdul-Baki
- The University of Texas Medical Branch, Department of Internal Medicine, Galveston, TX, United States
| | - Elliot Baerman
- Baylor College of Medicine, Department of Internal Medicine, Houston, TX, United States
| | - Kei Takigawa
- Baylor College of Medicine, Department of Internal Medicine, Houston, TX, United States
| | - Andres Urias Rivera
- Baylor College of Medicine, Department of Internal Medicine, Houston, TX, United States
| | - Irene Jeong-Ah Lee
- Baylor College of Medicine, Department of Internal Medicine, Houston, TX, United States
| | - Sean Ngo
- The University of Texas Health Sciences Center, Department of Internal Medicine, Houston, TX, United States
| | - Gabriel Sperling
- The University of Texas Medical Branch, Department of Internal Medicine, Galveston, TX, United States
| | - Abdullah Sagar Aleem
- The University of Texas Health Sciences Center, Department of Internal Medicine, Houston, TX, United States
| | - Raakhi Menon
- The University of Texas Medical Branch, Department of Internal Medicine, Galveston, TX, United States
| | - Andrew Sullivan
- The University of Texas Health Sciences Center, Department of Internal Medicine, Houston, TX, United States
| | - Varun Vemulapalli
- The University of Texas Health Sciences Center, Department of Internal Medicine, Houston, TX, United States
| | - Cristina Natha
- The University of Texas Health Sciences Center, Department of Internal Medicine, Houston, TX, United States
| | - Tanvi Gupta
- The University of Texas Health Sciences Center, Department of Internal Medicine, Houston, TX, United States
| | - Ayesha Khan
- The University of Texas Medical Branch, Department of Internal Medicine, Galveston, TX, United States
| | - Nitish Mittal
- The University of Texas Health Sciences Center, Department of Internal Medicine, Houston, TX, United States
| | - Garrett Coleman
- The University of Texas Medical Branch, Department of Internal Medicine, Galveston, TX, United States
| | - Hamza Salim
- Memorial Hermann Hospital, Department of Internal Medicine, Houston, TX, United States
| | - Sharada Wali
- The University of Texas MD Anderson Cancer Center, Department of Gastroenterology, Hepatology, and Nutrition, Houston, TX, United States
| | - Krishnavathana Varatharajalu
- The University of Texas MD Anderson Cancer Center, Department of Gastroenterology, Hepatology, and Nutrition, Houston, TX, United States
| | - Karen Chunguhn Kim
- Stanford University, Department of Medicine, Gastroenterology & Hepatology, San Jose, CA, United States
| | - Sunil Arani Reddy
- Stanford University, Department of Medicine, Oncology, San Jose, CA, United States
| | - Petros Grivas
- University of Washington, Department of Medicine, Division of Hematology Oncology, Fred Hutch Cancer Center, Seattle, WA, United States
| | - Anusha Shirwaikar Thomas
- The University of Texas MD Anderson Cancer Center, Department of Gastroenterology, Hepatology, and Nutrition, Houston, TX, United States
| | - Yinghong Wang
- The University of Texas MD Anderson Cancer Center, Department of Gastroenterology, Hepatology, and Nutrition, Houston, TX, United States.
| |
Collapse
|
21
|
Yang J, Qian Y, Kim C, Birhanu BT, Cal Y Mayor-Luna C, Ding D, Yu X, Schroeder VA, Mobashery S, Chang M. Targeting SleC and CspB in the Inhibition of Spore Germination in Clostridioides difficile. J Med Chem 2025; 68:9357-9370. [PMID: 40286328 DOI: 10.1021/acs.jmedchem.4c03090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
Clostridioides difficile, a Gram-positive, spore-forming anaerobic bacterium, is a major healthcare threat. Its spores colonize the gut following dysbiosis caused by broad-spectrum antibiotics, remaining dormant until host's bile acid triggers germination into vegetative cells that produce toxins, leading to diarrhea, colitis, and potentially death. Current antibiotics to treat C. difficile infection target vegetative cells but not spore germination, a pivotal step in infection development. This study unveils 1,2,4-oxadiazoles as a novel class of spore germination inhibitors and delineates the structure-activity relationship. Screening of 120 oxadiazoles revealed compound 110 (IC50 = 14 ± 1 μM or 6.3 ± 0.4 μg/mL). Compound 110 targets mature SleC (Kd = 12 ± 1.0 μM) and CspB (Kd = 8.0 ± 1.0 μM) on spores, inhibiting their enzymatic activities, thus preventing spore germination. To our knowledge, compound 110 is the first reported spore germination inhibitor targeting SleC/CspB, offering a promising avenue for C. difficile therapies.
Collapse
Affiliation(s)
- Jingdong Yang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Yuanyuan Qian
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Choon Kim
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Biruk T Birhanu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Carlos Cal Y Mayor-Luna
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Derong Ding
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Xiaotan Yu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Valerie A Schroeder
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
22
|
Noviello D, Chaparro M, Viganò C, Blesl A, Barberio B, Yanai H, Orlando A, Ferreiro-Iglesias R, Bezzio C, Zilli A, Molnár T, Gheorghe C, Conforti F, Innocenti T, Saibeni S, Bossuyt P, Oliveira R, Carvalhas Gabrielli AM, Losco A, Vieujean S, Tettoni E, Pirola L, Calderone S, Kornowski Cohen M, Dragoni G, Rath T, Barreiro-de Acosta M, Savarino EV, Gisbert JP, Vecchi M, Atreya R, Caprioli F. Fidaxomicin for Clostridioides difficile infection in patients with inflammatory bowel disease: a multicenter retrospective cohort study. J Crohns Colitis 2025; 19:jjaf056. [PMID: 40168072 PMCID: PMC12060865 DOI: 10.1093/ecco-jcc/jjaf056] [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: 01/27/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND AND AIMS Inflammatory bowel disease (IBD) patients with Clostridioides difficile infection (CDI) are at increased risk of adverse outcomes. Data on fidaxomicin use in IBD remain scarce. We assessed the effectiveness and safety of fidaxomicin for CDI and its impact on IBD outcomes in a large international cohort. METHODS Adult patients with ulcerative colitis (UC) or Crohn's disease (CD) treated with fidaxomicin for documented CDI were retrospectively included. The primary outcome was CDI recurrence rate within 8 weeks (C. difficile toxin detection and CDI-targeted therapy). Secondary outcomes included sustained response (no CDI-targeted therapy within 12 weeks), IBD therapy escalation, colectomy rate, and all-cause mortality within 30, 90, and 180 days. RESULTS Ninety-six patients (57 UC and 39 CD) from 20 IBD centers were included. Most were on advanced IBD therapy. Half had a previous CDI episode, 15% a severe episode. CDI recurrence rate was 10% at week 8, and sustained response 82% at week 12. Compared with patients with previous CDI episode, patients at first episode tended to have a lower recurrence (4.3% vs 16%; P = .06) and higher sustained response (91% vs 75%; P = .04) rate. IBD therapy escalation was required in 48% with a numerically lower need for patients achieving vs not-achieving sustained response within 30 days (12% vs 20%; P = .42). Five UC patients underwent colectomy. One death unrelated to CDI or IBD occurred. One moderate and 5 mild adverse events were reported. CONCLUSIONS Fidaxomicin was effective and safe in IBD patients with CDI, with greater effectiveness in CDI-naïve patients, potentially influencing short-term IBD outcomes.
Collapse
Affiliation(s)
- Daniele Noviello
- Department of Pathophysiology and Transplantation, University of Milan, Milano, Italy
| | - María Chaparro
- Gastrointestinal Unit of Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Chiara Viganò
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- European Reference Network on Hepatological Diseases ERN RARE-LIVER, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Andreas Blesl
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Brigida Barberio
- Division of Gastroenterology, Department of Surgery Oncology and Gastroenterology DiSCOG, University of Padova, Padova, Italy
| | - Henit Yanai
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ambrogio Orlando
- Inflammatory bowel disease Unit, “Villa Sofia-Cervello” Hospital, Palermo, Italy
| | - Rocío Ferreiro-Iglesias
- Gastroenterology Department, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, A Coruña, Spain
| | - Cristina Bezzio
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Alessandra Zilli
- Department of Gastroenterology & Endoscopy, IRCCS San Raffaele Hospital, Milan, Italy
| | - Tamás Molnár
- Department of Internal Medicine, University of Szeged, Szeged, Hungary
| | - Cristian Gheorghe
- Center of Gastroenterology and Hepatology, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Francesco Conforti
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Tommaso Innocenti
- Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences “Mario Serio,” University of Florence, Florence, Italy
- IBD Referral Center, Clinical Gastroenterology Unit, Careggi University Hospital, Florence, Italy
| | - Simone Saibeni
- IBD Centre, Gastroenterology Unit, Rho Hospital, ASST Rhodense, 20017 Rho, Italy
| | - Peter Bossuyt
- Imelda GI Clinical Research Center, Imelda General Hospital, Bonheiden, Belgium
| | - Raquel Oliveira
- Gastroenterology Department, Unidade Local de Saúde do Algarve, Portimão, Portugal
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
| | | | - Alessandra Losco
- Gastroenterology and Digestive Endoscopy Unit, ASST Santi Paolo e Carlo, Ospedale San Carlo, Milan, Italy
| | - Sophie Vieujean
- Hepato-Gastroenterology and Digestive Oncology, University Hospital CHU of Liège, Liège, Belgium
| | - Enrico Tettoni
- Gastroenterology and Digestive Endoscopy Unit, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
- Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Lorena Pirola
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- European Reference Network on Hepatological Diseases ERN RARE-LIVER, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Silvia Calderone
- Inflammatory bowel disease Unit, “Villa Sofia-Cervello” Hospital, Palermo, Italy
| | - Maya Kornowski Cohen
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gabriele Dragoni
- Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences “Mario Serio,” University of Florence, Florence, Italy
- IBD Referral Center, Clinical Gastroenterology Unit, Careggi University Hospital, Florence, Italy
| | - Timo Rath
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Manuel Barreiro-de Acosta
- Gastroenterology Department, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, A Coruña, Spain
| | - Edoardo Vincenzo Savarino
- Division of Gastroenterology, Department of Surgery Oncology and Gastroenterology DiSCOG, University of Padova, Padova, Italy
| | - Javier Pérez Gisbert
- Gastrointestinal Unit of Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Maurizio Vecchi
- Department of Pathophysiology and Transplantation, University of Milan, Milano, Italy
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Raja Atreya
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Flavio Caprioli
- Department of Pathophysiology and Transplantation, University of Milan, Milano, Italy
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
23
|
Goren I, Fallek Boldes O, Boldes T, Knyazev O, Kagramanova A, Limdi JK, Liu E, Sethi-Arora K, Holvoet T, Eder P, Bezzio C, Saibeni S, Vernero M, Alimenti E, Chaparro M, Gisbert JP, Orfanoudaki E, Koutroubakis IE, Pugliese D, Cuccia G, Calviño Suarez C, Ribaldone DG, Veisman I, Sharif K, Aratari A, Papi C, Mylonas I, Mantzaris GJ, Truyens M, Lobaton Ortega T, Nancey S, Castiglione F, Nardone OM, Calabrese G, Karmiris K, Velegraki M, Theodoropoulou A, Shitrit ABG, Lukas M, Vojtechová G, Ellul P, Bugeja L, Savarino EV, Fischler TS, Dotan I, Yanai H. Post-Discharge Outcomes of Elderly Patients Hospitalized for Inflammatory Bowel Disease Flare Complicated by Clostridioides difficile Infection. J Crohns Colitis 2025; 19:jjae161. [PMID: 39435855 DOI: 10.1093/ecco-jcc/jjae161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/12/2024] [Accepted: 10/20/2024] [Indexed: 10/23/2024]
Abstract
OBJECTIVES Elderly hospitalized patients with inflammatory bowel disease (IBD) flare and concurrent Clostridioides difficile infection (CDI) are considered at high risk of IBD-related complications. We aimed to evaluate the short-,intermediate-, and long-term post-discharge complications among these patients. METHODS A retrospective multicenter cohort study assessing outcomes of elderly individuals (≥60 years) hospitalized for an IBD flare who were tested for CDI (either positive or negative) and discharged. The primary outcome was the 3-month post-discharge IBD-related complication rates defined as steroid dependency, re-admissions (emergency department or hospitalization), IBD-related surgery, or mortality. We assessed post-discharge IBD-related complications within 6 month and mortality at 12 month among secondary outcomes. Risk factors for complication were assessed by multivariable logistic regression. RESULTS In a cohort of 654 patients hospitalized for IBD {age 68.9 (interquartile range [IQR]): 63.9-75.2 years, 60.9% ulcerative colitis (UC)}, 23.4% were CDI-positive. Post-discharge complication rates at 3 and 6 months, and 12 months mortality, did not differ significantly between CDI-positive and CDI-negative patients (32% vs 33.1%, p = 0.8; 40.5% vs 42.5%, p = 0.66; and 4.6% vs 8%, p = 0.153, respectively). The Charlson comorbidity index was the only significant risk factor for complications within 3 months (aOR 1.1), whereas mesalamine (5-aminosalicylic acid [5-ASA]) use was protective (aOR 0.6). An UC diagnosis was the sole risk factor for complication at 6 months (aOR 1.5). Clostridioides difficile infection did not significantly impact outcomes or interact with IBD type. CONCLUSIONS In elderly IBD patients hospitalized for IBD flare and subsequently discharged, a concurrent CDI infection was not associated with post-discharge IBD-related complications or mortality up to 1 year.
Collapse
Affiliation(s)
- Idan Goren
- Division of Gastroenterology, Rabin Medical Center, Affiliated with the Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Division of Gastroenterology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Ortal Fallek Boldes
- Department of Internal Medicine E, Rabin Medical Center, Affiliated with the Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tomer Boldes
- Department of Otolaryngology, Meir Medical Center, Affiliated with the Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Oleg Knyazev
- Moscow Clinical Scientific Center named after A. S. Loginov, Moscow, Russia
- National Medical Research Center of Coloproctology named after A. N. Ryzhykh, Moscow, Russia
| | - Anna Kagramanova
- Moscow Clinical Scientific Center named after A. S. Loginov, Moscow, Russia
- Research Institute of Health Organization and Medical Management, Moscow, Russia
| | - Jimmy K Limdi
- Division of Gastroenterology, Northern Care Alliance Hospitals NHS Foundation Trust, Manchester, UK
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, UK
| | - Eleanor Liu
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, UK
| | - Karishma Sethi-Arora
- Division of Gastroenterology, Northern Care Alliance Hospitals NHS Foundation Trust, Manchester, UK
| | - Tom Holvoet
- Department of Gastroenterology, VITAZ, St Niklaas, Belgium
- Department of Gastroenterology, University Hospital Ghent, Ghent, Belgium
| | - Piotr Eder
- Department of Gastroenterology, Dietetics and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Cristina Bezzio
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Simone Saibeni
- Department of Gastroenterology, ASST Rhodense, Rho, Italy
| | - Marta Vernero
- Department of Gastroenterology, ASST Rhodense, Rho, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Eleonora Alimenti
- Department of Gastroenterology, ASST Rhodense, Rho, Italy
- Department of Medical Sciences, University of Pavia, Gastroenterology Unit, Pavia, Italy
| | - María Chaparro
- Departement of Gastroenterology, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid, and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Javier P Gisbert
- Departement of Gastroenterology, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid, and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Eleni Orfanoudaki
- Department of Gastroenterology, University Hospital of Heraklion, Heraklion, Greece
| | | | - Daniela Pugliese
- CEMAD - IBD UNIT - Unità Operativa Complessa di Medicina Interna e Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Giuseppe Cuccia
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Cristina Calviño Suarez
- Department of Gastroenterology and Hepatology, University Hospital of Santiago de Compostela, La Coruña, Spain
| | | | - Ido Veisman
- Department of Gastroenterology, Sheba Medical Center, affiliated to Faculty of Medicine, Tel Aviv University, Israel, Department of Gastroenterology, Ramat Gan, Israel
| | - Kassem Sharif
- Department of Gastroenterology, Sheba Medical Center, affiliated to Faculty of Medicine, Tel Aviv University, Israel, Department of Gastroenterology, Ramat Gan, Israel
| | - Annalisa Aratari
- Department of Gastroenterology, S. Filippo Neri Hospital, Gastroenterology, Rome, Italy and Department of Gastroenterology, Evangelismos-Polycliniki General Hospital, Athens, Greece
| | - Claudio Papi
- Department of Gastroenterology, S. Filippo Neri Hospital, Gastroenterology, Rome, Italy and Department of Gastroenterology, Evangelismos-Polycliniki General Hospital, Athens, Greece
| | - Iordanis Mylonas
- Department of Gastroenterology, S. Filippo Neri Hospital, Gastroenterology, Rome, Italy and Department of Gastroenterology, Evangelismos-Polycliniki General Hospital, Athens, Greece
| | - Gerassimos J Mantzaris
- Department of Gastroenterology, S. Filippo Neri Hospital, Gastroenterology, Rome, Italy and Department of Gastroenterology, Evangelismos-Polycliniki General Hospital, Athens, Greece
| | - Marie Truyens
- Department of Gastroenterology, Evangelismos-Polycliniki General Hospital, Athens, Greece
| | | | - Stéphane Nancey
- Dept. de Gastroenterologie, South Lyon University Hospital, Hospices Civils de Lyon, Lyon, France
| | - Fabiana Castiglione
- Department of Clinical Medicine and Surgery, Federico II University, Gastroenterology, Naples, Italy
| | - Olga Maria Nardone
- Department of Public Health, University of Naples Federico II, Gastroenterolog, Naples, Italy
| | - Giulio Calabrese
- Department of Public Health, University of Naples Federico II, Gastroenterolog, Naples, Italy
| | | | - Magdalini Velegraki
- Department of Gastroenterology, Venizeleio General Hospital, Heraklion, Greece
| | | | - Ariella Bar-Gil Shitrit
- IBD MOM Unit, Digestive Diseases Institute, Shaare Zedek Medical Center, affiliated with the Hebrew University, Jerusalem, Israel
| | - Milan Lukas
- Clinical and Research Center for Inflammatory Bowel Disease ISCARE and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Gabriela Vojtechová
- Clinical and Research Center for Inflammatory Bowel Disease ISCARE and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pierre Ellul
- Division of Gastroenterology, Mater Dei Hospital, Birkirkara, Malta
| | - Luke Bugeja
- Division of Gastroenterology, Mater Dei Hospital, Birkirkara, Malta
| | - Edoardo V Savarino
- Department of Surgery, Oncology and Gastroenterology, Division of Gastroenterology, University of Padua, Padua, Italy
| | - Tali Sharar Fischler
- Division of Gastroenterology, Rabin Medical Center, Affiliated with the Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Affiliated with the Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Henit Yanai
- Division of Gastroenterology, Rabin Medical Center, Affiliated with the Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
24
|
Dans-Caballero S, Juan-Cencerrado M, Mochón-Jiménez C, Roldán-Molina R, Pérez-Guijo VC. Juvenile Dermatomyositis Triggered by Influenza B: A Case Report on Viral-Induced Autoimmunity. Pediatr Dermatol 2025. [PMID: 40326606 DOI: 10.1111/pde.15975] [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: 03/18/2025] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 05/07/2025]
Abstract
A previously healthy 13-year-old boy developed juvenile dermatomyositis (JDM) shortly after a confirmed influenza B infection, presenting with progressive proximal muscle weakness and classic cutaneous findings. Laboratory tests revealed elevated muscle enzymes and myositis-specific autoantibodies, supporting the diagnosis. The temporal association suggests a potential post-viral autoimmune trigger, highlighting influenza B as a possible environmental factor in JDM pathogenesis. This case reinforces the need for heightened clinical awareness and further research into virus-associated autoimmune mechanisms in pediatric myopathies.
Collapse
Affiliation(s)
- Santiago Dans-Caballero
- Rheumatology Department, Reina Sofia University Hospital, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | | | | | - Rosa Roldán-Molina
- Rheumatology Department, Reina Sofia University Hospital, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Verónica C Pérez-Guijo
- Rheumatology Department, Reina Sofia University Hospital, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Medicine, Cordoba, Spain
| |
Collapse
|
25
|
Li MJ, Chen HM, Chen YL, Lai YH, Lai CY, Ruan JW, Chen JW, Tsai WH, Ko WC, Tsai PJ. Lactiplantibacillus plantarum GMNL-661 Ameliorates Clostridioides difficile Infection and Reconfigures Intestinal Microbiota in a Murine Model. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10556-9. [PMID: 40327311 DOI: 10.1007/s12602-025-10556-9] [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] [Accepted: 04/21/2025] [Indexed: 05/07/2025]
Abstract
Clostridioides difficile infection (CDI) is a significant global health threat, often resulting from antibiotic-induced disruption of the gut microbiota, which leads to severe gastrointestinal issues. Current treatments, such as vancomycin, are effective but can cause subsequent relapses, further microbiota disruption, and high treatment costs. Probiotics offer a promising microbiota-based therapeutic strategy. Following an in vitro screening for novel lactic acid bacterial (LAB) strains with strong anti-C. difficile ability and good tolerance to digestive challenges, Lactiplantibacillus plantarum GMNL-661 emerged as a potential solution to combat CDI. In a CDI mice model, the appropriate dose of GMNL-661 effectively alleviated CDI, which caused weight loss, gut inflammation, and mucin depletion. GMNL-661 alleviated CDI symptoms through increased gut barrier genes and downregulated IL-1 and IL-18. 16s rDNA analysis of mice stool from CDI and CDI supplemented with GMNL-661 showed distinct microbiota ecology. GMNL-661 dramatically affected the microbiome of CDI, increasing Lactobacillus spp. and Clostridium cluster XVIII while reducing Clostridium and Enterococcus species. Genome analysis of GMNL-661 revealed minimal safety concerns in antibiotic resistance and virulence genes, confirming that it is suitable for inclusion in the food chain. Antimicrobial peptide (AMP) prediction on GMNL-661 and 299v genome suggested a strong potential candidate for anti-CD antimicrobial peptides. These findings highlighted L. plantarum GMNL-661 as an effective and highly safe therapeutic agent against CDI in clinical.
Collapse
Affiliation(s)
- Meng-Jia Li
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hong-Ming Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Yanshuei District Health Station, Tainan, Taiwan
| | - Yueh-Lin Chen
- EirGenix Inc./Research & Development/Cell Line Engineering, Taipei, Taiwan
| | - Yi-Hsin Lai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Yu Lai
- Inong Agriculture Company Limited, Tainan, Taiwan
| | - Jhen-Wei Ruan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jenn-Wei Chen
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wan-Hua Tsai
- Research and Development Department, GenMont Biotech Incorporation, Tainan, Taiwan
| | - Wen-Chien Ko
- National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Department of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Pei-Jane Tsai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
26
|
Nobels A, van Marcke C, Jordan BF, Van Hul M, Cani PD. The gut microbiome and cancer: from tumorigenesis to therapy. Nat Metab 2025:10.1038/s42255-025-01287-w. [PMID: 40329009 DOI: 10.1038/s42255-025-01287-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 03/20/2025] [Indexed: 05/08/2025]
Abstract
The gut microbiome has a crucial role in cancer development and therapy through its interactions with the immune system and tumour microenvironment. Although evidence links gut microbiota composition to cancer progression, its precise role in modulating treatment responses remains unclear. In this Review, we summarize current knowledge on the gut microbiome's involvement in cancer, covering its role in tumour initiation and progression, interactions with chemotherapy, radiotherapy and targeted therapies, and its influence on cancer immunotherapy. We discuss the impact of microbial metabolites on immune responses, the relationship between specific bacterial species and treatment outcomes, and potential microbiota-based therapeutic strategies, including dietary interventions, probiotics and faecal microbiota transplantation. Understanding these complex microbiota-immune interactions is critical for optimizing cancer therapies. Future research should focus on defining microbial signatures associated with treatment success and developing targeted microbiome modulation strategies to enhance patient outcomes.
Collapse
Affiliation(s)
- Amandine Nobels
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- UCLouvain, Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Pole of Medical Imaging, Radiotherapy and Oncology (MIRO), Brussels, Belgium
| | - Cédric van Marcke
- UCLouvain, Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Pole of Medical Imaging, Radiotherapy and Oncology (MIRO), Brussels, Belgium
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Breast Clinic, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Bénédicte F Jordan
- UCLouvain, Université catholique de Louvain, Biomedical Magnetic Resonance group (REMA), Louvain Drug Research Institute (LDRI), Brussels, Belgium
| | - Matthias Van Hul
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium.
| | - Patrice D Cani
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium.
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), Brussels, Belgium.
| |
Collapse
|
27
|
Ito R, Watanabe T, Hoshino Y, Takahashi K. Evaluation of the Efficacy of Step-down Therapy with Oral Minocycline for Complicated Pyelonephritis Caused by Extended-Spectrum β-Lactamase-Producing Enterobacterales: A Retrospective Cohort Study. J Infect Chemother 2025:102724. [PMID: 40339646 DOI: 10.1016/j.jiac.2025.102724] [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: 03/19/2025] [Revised: 04/17/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Extended-spectrum β-lactamase-producing Enterobacterales (ESBL-E) infections present significant treatment challenges owing to limited oral antibiotic options. Minocycline is active against ESBL-E; however, its efficacy remains unclear. This study evaluated the efficacy of step-down therapy using oral minocycline for complicated pyelonephritis (cPN) caused by ESBL-producing Escherichia coli. METHODS This retrospective cohort study, conducted at Tohoku Rosai Hospital (2018-2023), included hospitalized patients with cPN and ESBL-E infection. Seventy-two patients receiving intravenous antibiotics followed by oral therapy were classified into the conventional or minocycline therapy groups. Primary outcomes were recurrence within 30 and 90 days post-treatment, whereas secondary outcomes included resolution of fever within 48 h, hospital stay, and adverse events. Propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) were performed. RESULTS No significant differences were observed in recurrence or fever resolution; however, hospital stay was shorter in the minocycline group than in the conventional group (PSM: p = 0.027; IPTW: p = 0.033, respectively). No adverse effects were noted, although one Clostridioides difficile infection case occurred in the conventional group. ESBL-producing E. coli was detected more frequently in the minocycline therapy group (p = 0.005), and patients with minocycline minimum inhibitory concentration (MIC) ≥4 μg/mL had higher relapse rates (p = 0.006) than those in the conventional group. CONCLUSIONS Among ESBL-E, particularly in E. coli, step-down therapy using oral minocycline for cPN appears as effective as conventional therapy and may shorten hospital stays. However, as minocycline MICs ≥4 μg/mL may be associated with recurrence, clinicians should verify MIC values before prescribing minocycline.
Collapse
Affiliation(s)
- Ryota Ito
- Department of Pharmacy, Tohoku Rosai Hospital, Sendai 981-8563, Japan.
| | - Takashi Watanabe
- Division of Clinical Pharmaceutics and Pharmacy Practice, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan; Division of Clinical Pharmacotherapeutics, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan; Department of Pharmacy, Tohoku Medical and Pharmaceutical University Hospital, Sendai 983-8512, Japan
| | - Yuta Hoshino
- Department of Pharmacy, Tohoku Rosai Hospital, Sendai 981-8563, Japan
| | - Kenichi Takahashi
- Department of Colorectal Surgery, Tohoku Rosai Hospital, Sendai 981-8563, Japan
| |
Collapse
|
28
|
Münch A, Escudero-Hernández C. Dissecting Microscopic Colitis Immunopathophysiology: Insights From Basic Research. United European Gastroenterol J 2025. [PMID: 40317876 DOI: 10.1002/ueg2.70024] [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: 09/19/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 05/07/2025] Open
Abstract
Microscopic colitis is an inflammatory bowel disease (IBD) comprising two clinically undiscernible entities: collagenous colitis and lymphocytic colitis. Collagenous colitis associates with HLA genes and displays a Th1/Tc1-Th17/Tc17 profile with pericryptal myofibroblast activity, water malabsorption and secondary fluid loss due to altered osmoregulation. Conversely, lymphocytic colitis lacks genetic associations and displays a Th1/Th2 profile and paracellular/transcellular permeability. Lymphocytic colitis subclassifies into channelopathic lymphocytic colitis due to unique alteration of ion and organic acid transport that could result from drug exposure, and inflammatory lymphocytic colitis due to the involvement of moderate immune responses compared to collagenous colitis. As microscopic colitis mucosa remains intact and immune cells seem to stay inactive, microscopic colitis is an ideal model to explore early stages of IBD if collagenous colitis and lymphocytic colitis are studied as distinct entities. Exploiting multiomic approaches and established biobanks will ensure validation of microscopic colitis patient stratification, and deepening into pathomechanisms which could enable precision medicine.
Collapse
Affiliation(s)
- Andreas Münch
- Department of Gastroenterology and Hepatology, Linköping University, Linköping, Sweden
- Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Celia Escudero-Hernández
- Institute of Clinical Molecular Biology (IKMB), Christian-Albrechts-University and University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
29
|
Wang L, Xu T, Wu S, Zhao C, Huang H. The efficacy and underlying mechanisms of berberine in the treatment of recurrent Clostridioides difficile infection. Int J Antimicrob Agents 2025; 65:107468. [PMID: 39986400 DOI: 10.1016/j.ijantimicag.2025.107468] [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: 10/14/2024] [Revised: 01/05/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
Recurrent Clostridioides difficile infection (rCDI) is a global health threat that has received considerable attention. Berberine (BBR), a natural pentacyclic isoquinoline alkaloid, has been used as a cost-effective treatment for intestinal infections in Asia for many years. However, the effect of BBR on rCDI is not clear. The efficacy and underlying mechanisms of BBR were evaluated in a vancomycin-dependent rCDI mouse model and an intestinal organoids model. The study findings showed that BBR treatment alleviated the severity of infection and increased survival rate in rCDI mice. Mechanistically, BBR alleviated intestinal epithelial damage with higher Occludin expression, suppressed some inflammatory pathways and reduced the level of inflammatory factors in both the caecum and serum. Moreover, 16S rRNA sequencing analysis indicated that BBR reshaped the gut microbiota by increasing the abundance of Firmicutes and reducing the abundance of Proteobacteria. At genus level, BBR treatment increased levels of Blautia and Bilophila, and reduced levels of Proteus. In addition, acetic acid, one of the short-chain fatty acids (SCFAs), was also increased after BBR treatment in rCDI mice. Collectively, BBR exerted a protective effect in rCDI via multiple underlying mechanisms and is a potential drug candidate for alleviating rCDI, but further research is needed in this area.
Collapse
Affiliation(s)
- Li Wang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the PRC, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Teng Xu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the PRC, Shanghai, China
| | - Shi Wu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the PRC, Shanghai, China
| | - Chao Zhao
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Haihui Huang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the PRC, Shanghai, China.
| |
Collapse
|
30
|
Quinn-Bohmann N, Carr AV, Diener C, Gibbons SM. Moving from genome-scale to community-scale metabolic models for the human gut microbiome. Nat Microbiol 2025; 10:1055-1066. [PMID: 40217129 DOI: 10.1038/s41564-025-01972-2] [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: 07/14/2023] [Accepted: 02/26/2025] [Indexed: 05/08/2025]
Abstract
Metabolic models of individual microorganisms or small microbial consortia have become standard research tools in the bioengineering and systems biology fields. However, extending metabolic modelling to diverse microbial communities, such as those in the human gut, remains a practical challenge from both modelling and experimental validation perspectives. In complex communities, metabolic models accounting for community dynamics, or those that consider multiple objectives, may provide optimal predictions over simpler steady-state models, but require a much higher computational cost. Here we describe some of the strengths and limitations of microbial community-scale metabolic models and argue for a robust validation framework for developing personalized, mechanistic and accurate predictions of microbial community metabolic behaviours across environmental contexts. Ultimately, quantitatively accurate microbial community-scale metabolic models could aid in the design and testing of personalized prebiotic, probiotic and dietary interventions that optimize for translationally relevant outcomes.
Collapse
Affiliation(s)
- Nick Quinn-Bohmann
- Institute for Systems Biology, Seattle, WA, USA
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA, USA
| | - Alex V Carr
- Institute for Systems Biology, Seattle, WA, USA
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA, USA
| | - Christian Diener
- Institute for Systems Biology, Seattle, WA, USA.
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria.
| | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, USA.
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- eScience Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
31
|
Damianos JA. Response to Endogenous Ethanol Production in the Human Alimentary Tract: A Literature Review. J Gastroenterol Hepatol 2025; 40:1317-1318. [PMID: 40017434 DOI: 10.1111/jgh.16924] [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: 02/03/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Affiliation(s)
- John A Damianos
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
32
|
Szilcz M, Wastesson JW, Bergman D, Johnell K, Ludvigsson JF. Antibiotic Use and Risk of Microscopic Colitis in Older Adults: A Nationwide Self-Controlled Case Series Study. Aliment Pharmacol Ther 2025; 61:1500-1507. [PMID: 39935241 PMCID: PMC11981551 DOI: 10.1111/apt.70028] [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: 11/05/2024] [Revised: 12/10/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Several drugs have been linked to the risk of microscopic colitis (MC), a condition characterised by watery, non-bloody diarrhoea. Antibiotics can induce similar symptoms, but their connection to MC remains unclear. AIM To investigate the antibiotic-related risks of MC in adults aged 65 years and older. METHODS This was a nationwide, self-controlled case series study including adults aged ≥ 65 years with a new prescription for antibiotics and biopsy-confirmed, incident MC (Sweden, 2007-17). We identified cases from the nationwide histopathology cohort ESPRESSO individually linked to several registers. Using conditional Poisson regression, we estimated incidence rate ratios (IRR) for four risk periods: on treatment, and 1-14, 15-91, and 92-365 days post-treatment, compared with the non-treatment periods in the same individual. We also conducted a negative control outcome analysis to assess whether the association was specific to MC or due to diagnostic workup. E-values were used to assess robustness to unmeasured confounding. RESULTS We identified 2393 persons with incident biopsy-confirmed MC (median age at diagnosis 74; 67% women). Compared with the non-treatment periods, the risk of MC in the age-adjusted analysis increased with antibiotic treatment (IRR: 1.44 [95% CI: 1.13-1.84], E-value: 2.24), 1-14 days (IRR: 1.12 [0.83-1.49]), 15-91 days (1.12 [0.97-1.31]) and 92-365 days post-treatment (1.19 [1.07-1.32]). The negative control outcome analysis showed similar risks of biopsy-confirmed normal mucosa after antibiotic use. CONCLUSION The observed association between antibiotic use and MC may result from detection bias rather than a causal relationship.
Collapse
Affiliation(s)
- Máté Szilcz
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Jonas W. Wastesson
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Aging Research Center, Department of Neurobiology, Care Sciences and SocietyKarolinska Institutet & Stockholm UniversityStockholmSweden
| | - David Bergman
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Kristina Johnell
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Jonas F. Ludvigsson
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Department of PaediatricsÖrebro University HospitalÖrebroSweden
- Department of MedicineColumbia University College of Physicians and SurgeonsNew YorkNew YorkUSA
| |
Collapse
|
33
|
Singh A, Bhardwaj A, Midha V, Sood A. Microbiome testing in clinical practice. Lancet Gastroenterol Hepatol 2025; 10:414. [PMID: 40220778 DOI: 10.1016/s2468-1253(24)00432-1] [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: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 04/14/2025]
Affiliation(s)
- Arshdeep Singh
- Department of Gastroenterology, Dayanand Medical College, Ludhiana, Punjab 141001, India
| | - Arshia Bhardwaj
- Department of Gastroenterology, Dayanand Medical College, Ludhiana, Punjab 141001, India
| | - Vandana Midha
- Department of Internal Medicine, Dayanand Medical College, Ludhiana, Punjab 141001, India
| | - Ajit Sood
- Department of Gastroenterology, Dayanand Medical College, Ludhiana, Punjab 141001, India.
| |
Collapse
|
34
|
Karabekmez ME. Harnessing Human Holobiome and Meta-Multi-Omics Analyses for Medical Applications. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025; 29:179-182. [PMID: 40197113 DOI: 10.1089/omi.2025.0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Next-generation sequencing technology has revolutionized all fields of living systems, and its applications almost reinvented some research areas including metagenomics. The microbiotas in our body, including those of the oral, nasal, ocular, alveolar, skin regions, and particularly gut microbiota, have close linkages with our health status. Maturation of experimental techniques for metagenomics has been followed by other related omics platforms, for example, metatranscriptomics, metaproteomics, and all possible metacounterparts of multiomics studies. Now, we are on the eve of a meta-multi-omics era for the analysis of human holobiome in medical research. This era will help buttress the current efforts for systems medicine by illuminating the relationships between human holobiome and health or all human diseases including not only cancers but also infectious diseases, autoimmune diseases, obesity, aging, genetic disorders, and psychiatric conditions. Equally important, meta-multi-omics era is also poised to inform the determinants of human health and, by extension, help build individually tailored precision medicine interventions.
Collapse
Affiliation(s)
- Muhammed Erkan Karabekmez
- Department of Bioengineering, Istanbul Medeniyet University, Istanbul, Türkiye
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts
| |
Collapse
|
35
|
Tome J, Pardi DS. Editorial: Association of Antibiotic Exposure With Microscopic Colitis. Aliment Pharmacol Ther 2025; 61:1547-1548. [PMID: 39985121 DOI: 10.1111/apt.70043] [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: 02/10/2025] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 02/24/2025]
Affiliation(s)
- June Tome
- Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
36
|
Porcari S, Ianiro G. Microbiome testing in clinical practice - Authors' reply. Lancet Gastroenterol Hepatol 2025; 10:414-415. [PMID: 40220779 DOI: 10.1016/s2468-1253(25)00063-9] [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: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 04/14/2025]
Affiliation(s)
- Serena Porcari
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168-Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168-Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
37
|
Call for papers on the clinical microbiome. Nat Microbiol 2025; 10:1027. [PMID: 40328941 DOI: 10.1038/s41564-025-02019-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
|
38
|
Jansson-Knodell CL, Gardinier D, Weekley K, Yang Q, Rubio-Tapia A. Artificial Intelligence Chatbots Not Yet Ready for Celiac Disease Patient Care. Clin Gastroenterol Hepatol 2025; 23:1065-1067.e1. [PMID: 39489473 DOI: 10.1016/j.cgh.2024.10.012] [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: 09/23/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 11/05/2024]
Affiliation(s)
- Claire L Jansson-Knodell
- Celiac Disease Program, Division of Gastroenterology, Hepatology, and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio
| | - David Gardinier
- Celiac Disease Program, Division of Gastroenterology, Hepatology, and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio
| | - Kendra Weekley
- Celiac Disease Program, Division of Gastroenterology, Hepatology, and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio
| | - Qijun Yang
- Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Alberto Rubio-Tapia
- Celiac Disease Program, Division of Gastroenterology, Hepatology, and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
39
|
BharathwajChetty B, Kumar A, Deevi P, Abbas M, Alqahtani A, Liang L, Sethi G, Liu L, Kunnumakkara AB. Gut microbiota and their influence in brain cancer milieu. J Neuroinflammation 2025; 22:129. [PMID: 40312370 PMCID: PMC12046817 DOI: 10.1186/s12974-025-03434-2] [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: 11/21/2024] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
Microbial communities are not simply remnants of the past but dynamic entities that continuously evolve under the selective pressures of nature, reflecting the intricate and adaptive processes of evolution. The microbiota residing in the various regions of the human body has numerous roles in different physiological processes such as nutrition, metabolism, immune regulation, etc. In the zeal of achieving empirical insights into the ambit of the gut microbiome, the research over the years led to the revelation of reciprocal interaction between the gut microbiome and the cognitive functioning of the human body. Dysbiosis in the gut microbial composition disturbs the homeostatic cognitive functioning of the human body. This dysbiosis has been associated with various chronic diseases, including brain cancer, such as glioma, glioblastoma, etc. This review explores the mechanistic role of dysbiosis-mediated progression of brain cancers and their subtypes. Moreover, it demonstrates the regulatory role of microbial metabolites produced by the gut microbiota, such as short-chain fatty acids, amino acids, lipids, etc., in the tumour progression. Further, we also provide valuable insights into the microbiota mediating the efficiency of therapeutic regimens, thereby leveraging gut microbiota as potential biomarkers and targets for improved treatment outcomes.
Collapse
Affiliation(s)
- Bandari BharathwajChetty
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Pranav Deevi
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
- International Joint M. Tech Degree in Food Science and Technology, Department of Chemical Engineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia
| | - Athba Alqahtani
- Research Centre, King Fahad Medical City, Riyadh, 11525, Saudi Arabia
| | - Liping Liang
- Guangzhou Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin Scool of Medicine, National University of Singapore, Singapore, 117699, Singapore.
| | - Le Liu
- Integrated Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China.
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
- International Joint M. Tech Degree in Food Science and Technology, Department of Chemical Engineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
40
|
Liu M, Blattman SB, Takahashi M, Mandayam N, Jiang W, Oikonomou P, Tavazoie SF, Tavazoie S. Conserved genetic basis for microbial colonization of the gut. Cell 2025; 188:2505-2520.e22. [PMID: 40187346 PMCID: PMC12048274 DOI: 10.1016/j.cell.2025.03.010] [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: 08/26/2024] [Revised: 12/02/2024] [Accepted: 03/06/2025] [Indexed: 04/07/2025]
Abstract
Despite the fundamental importance of gut microbes, the genetic basis of their colonization remains largely unexplored. Here, by applying cross-species genotype-habitat association at the tree-of-life scale, we identify conserved microbial gene modules associated with gut colonization. Across thousands of species, we discovered 79 taxonomically diverse putative colonization factors organized into operonic and non-operonic modules. They include previously characterized colonization pathways such as autoinducer-2 biosynthesis and novel processes including tRNA modification and translation. In vivo functional validation revealed YigZ (IMPACT family) and tRNA hydroxylation protein-P (TrhP) are required for E. coli intestinal colonization. Overexpressing YigZ alone is sufficient to enhance colonization of the poorly colonizing MG1655 E. coli by >100-fold. Moreover, natural allelic variations in YigZ impact inter-strain colonization efficiency. Our findings highlight the power of large-scale comparative genomics in revealing the genetic basis of microbial adaptations. These broadly conserved colonization factors may prove critical for understanding gastrointestinal (GI) dysbiosis and developing therapeutics.
Collapse
Affiliation(s)
- Menghan Liu
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Sydney B Blattman
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Mai Takahashi
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Nandan Mandayam
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Wenyan Jiang
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Panos Oikonomou
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Sohail F Tavazoie
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Saeed Tavazoie
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Department of Systems Biology, Columbia University, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
41
|
Flores-Treviño S, Bocanegra-Ibarias P, Salas-Treviño D, Ramírez-Elizondo MT, Pérez-Alba E, Camacho-Ortiz A. Microbiota transplantation and administration of live biotherapeutic products for the treatment of dysbiosis-associated diseases. Expert Opin Biol Ther 2025; 25:1-14. [PMID: 40134274 DOI: 10.1080/14712598.2025.2484303] [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: 12/18/2024] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
INTRODUCTION The microbiota composition in humans varies according to the anatomical site and is crucial for maintaining homeostasis and an overall healthy state. Several gastrointestinal, vaginal, respiratory, and skin diseases are associated with dysbiosis. Alternative therapies such as microbiota transplantation can help restore microbiota normal composition and can be implemented to treat clinically relevant diseases. AREAS COVERED Current microbiota transplantation therapies conducted in clinical trials were included in this review (after searching on MEDLINE database from years 2017 to 2025) such as fecal microbiota transplantation (FMT) against recurrent Clostridioides difficile infection (rCDI) and vaginal microbiota transplantation (VMT) against bacterial vaginosis. Washed microbiota transplantation (WMT) and live biotherapeutic products (LBPs) were also reviewed. EXPERT OPINION In microbiota-based transplantation therapy, selecting optimal donors is a limitation. A stool or a vaginal microbiota bank should be implemented to overcome the time-consuming and expensive process of donor recruitment. Microbiota-based LBPs are also promising treatment alternatives for rCDI and other dysbiosis-associated diseases. Specific LBPs could be engineered out of donor fluids-derived strains to achieve the selection of specific beneficial microorganisms for the treatment of specific dysbiosis-associated diseases. Personalized microbiota-based treatments are promising solutions for dysbiosis-associated diseases, which remains an important necessity in clinical practice.
Collapse
Affiliation(s)
- Samantha Flores-Treviño
- Department of Infectious Diseases, University Hospital "Dr. José Eleuterio González", Autonomous University of Nuevo Leon, Monterrey, Mexico
| | - Paola Bocanegra-Ibarias
- Department of Infectious Diseases, University Hospital "Dr. José Eleuterio González", Autonomous University of Nuevo Leon, Monterrey, Mexico
| | - Daniel Salas-Treviño
- Department of Infectious Diseases, University Hospital "Dr. José Eleuterio González", Autonomous University of Nuevo Leon, Monterrey, Mexico
| | - María Teresa Ramírez-Elizondo
- Department of Infectious Diseases, University Hospital "Dr. José Eleuterio González", Autonomous University of Nuevo Leon, Monterrey, Mexico
| | - Eduardo Pérez-Alba
- Department of Infectious Diseases, University Hospital "Dr. José Eleuterio González", Autonomous University of Nuevo Leon, Monterrey, Mexico
| | - Adrián Camacho-Ortiz
- Department of Infectious Diseases, University Hospital "Dr. José Eleuterio González", Autonomous University of Nuevo Leon, Monterrey, Mexico
| |
Collapse
|
42
|
Cai Z, Zhang M, Zhou L, Xiong Y, Wang H, Chen Y, Yuan J. Kai-Xin-San polysaccharides exert therapeutic effects on D-gal and Aβ 25-35-induced AD rats by regulating gut microbiota and metabolic profile. Int J Biol Macromol 2025; 306:141850. [PMID: 40058438 DOI: 10.1016/j.ijbiomac.2025.141850] [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: 10/26/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 05/11/2025]
Abstract
Metabolic abnormalities and gut microbiota imbalance are intricately linked to the onset and progression of Alzheimer's disease (AD). Kai-Xin-San (KXS) is a traditional herbal formula known for its therapeutic effects on AD. Our previous research indicated that Kai-Xin-San polysaccharide (KXS-P) exhibits a significant therapeutic impact on AD, but the precise mechanisms remain incompletely understood. In this study, untargeted fecal metabolomics and 16S rRNA gene sequencing were used to investigate the potential mechanisms by which KXS-P acts against AD. Key metabolites and gut microbial species were identified using multivariate analysis and a comprehensive examination of intestinal microecology. Our findings revealed that KXS-P improves lipid metabolism in AD rats by modulating a series of lipid molecules and bile acid levels. Additionally, KXS-P regulated gut microbiota composition and restored the symbiotic relationships within the gut microbiome. Notably, the anti-inflammatory effect of KXS-P may be related to its regulation of specific lipotypes levels and the abundance of Romboutsia, Bifidobacterium and Alloprevotella. KXS-P demonstrates the ability to alleviate symptoms of AD rats through multiple mechanisms: ① Improving lipid metabolism and maintaining lipid homeostasis; ② Reducing neuronal and inflammatory damage; ③ Regulating the composition and symbiotic relationships of gut microbiota to preserve intestinal microecological balance.
Collapse
Affiliation(s)
- Zhinan Cai
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Min Zhang
- Nanchang Key Laboratory of Detection and Control of Food Safety, Nanchang Inspection and Testing Center, Nanchang 330012, China
| | - Lifen Zhou
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yongchang Xiong
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Huijuan Wang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Ying Chen
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jinbin Yuan
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China.
| |
Collapse
|
43
|
Setshedi M, Ianiro G. Developing microbiome research in Africa: the essential role of clinician scientists. Nat Rev Gastroenterol Hepatol 2025:10.1038/s41575-025-01069-5. [PMID: 40307565 DOI: 10.1038/s41575-025-01069-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Affiliation(s)
- Mashiko Setshedi
- Department of Medicine, Division of Gastroenterology, University of Cape Town, Cape Town, South Africa
- African Helicobacter and Microbiota Study Group
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy.
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy.
- European Helicobacter and Microbiota Study Group, Vienna, Austria.
| |
Collapse
|
44
|
Kamath S, Bryant RV, Costello SP, Day AS, Forbes B, Haifer C, Hold G, Kelly CR, Li A, Pakuwal E, Stringer A, Tucker EC, Wardill HR, Joyce P. Translational strategies for oral delivery of faecal microbiota transplantation. Gut 2025:gutjnl-2025-335077. [PMID: 40301116 DOI: 10.1136/gutjnl-2025-335077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025]
Abstract
Faecal microbiota transplantation (FMT) has emerged as a transformative therapy for Clostridioides difficile infections and shows promise for various GI and systemic diseases. However, the poor patient acceptability and accessibility of 'conventional' FMT, typically administered via colonoscopies or enemas, hinders its widespread clinical adoption, particularly for chronic conditions. Oral administration of FMT (OralFMT) overcomes these limitations, yet faces distinct challenges, including a significant capsule burden, palatability concerns and poor microbial viability during gastric transit. This review provides a comprehensive analysis of emerging strategies that aim to advance OralFMT by: (1) refining processing technologies (eg, lyophilisation) that enable manufacturing of low-volume FMT formulations for reducing capsule burden and (2) developing delivery technologies that improve organoleptic acceptability and safeguard the microbiota for targeted colonic release. These advancements present opportunities for OralFMT to expand its therapeutic scope, beyond C. difficile infections, towards chronic GI conditions requiring frequent dosing regimens. While this review primarily focuses on optimising OralFMT delivery, it is important to contextualise these advancements within the broader shift towards defined microbial consortia. Live biotherapeutic products (LBPs) offer an alternative approach, yet the interplay between OralFMT and LBPs in clinical practice remains unresolved. We postulate that continued innovation in OralFMT and LBPs via a multidisciplinary approach can further increase therapeutic efficacy and scalability by enabling disease site targeting, co-delivery of therapeutic compounds and overcoming colonisation resistance. Realising these goals positions OralFMT as a cornerstone of personalised care across a range of diseases rooted in microbiome health.
Collapse
Affiliation(s)
- Srinivas Kamath
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Robert V Bryant
- Department of Gastroenterology and Hepatology, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Faculty of Health Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Samuel P Costello
- Department of Gastroenterology and Hepatology, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- The University of Adelaide, Adelaide, South Australia, Australia
| | - Alice S Day
- Faculty of Health Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- Gastroenterology, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
| | | | - Craig Haifer
- Department of Gastroenterology, St Vincent's Hospital Sydney, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Georgina Hold
- Microbiome Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Colleen R Kelly
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Anna Li
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Evance Pakuwal
- Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Andrea Stringer
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Emily C Tucker
- Faculty of Health Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- Infectious Diseases Unit, Central Adelaide Local Health Network, Adelaide, South Australia, Australia
| | - Hannah Rose Wardill
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Paul Joyce
- University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
45
|
Clavel T, Faber F, Groussin M, Haller D, Overmann J, Pauvert C, Poyet M, Selkrig J, Stecher B, Typas A, Vehreschild MJGT, Westermann AJ, Wylensek D, Maier L. Enabling next-generation anaerobic cultivation through biotechnology to advance functional microbiome research. Nat Biotechnol 2025:10.1038/s41587-025-02660-6. [PMID: 40301656 DOI: 10.1038/s41587-025-02660-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/31/2025] [Indexed: 05/01/2025]
Abstract
Microbiomes are complex communities of microorganisms that are essential for biochemical processes on Earth and for the health of humans, animals and plants. Many environmental and host-associated microbiomes are dominated by anaerobic microbes, some of which cannot tolerate oxygen. Anaerobic microbial communities have been extensively studied over the last 20 years using molecular techniques, especially next-generation sequencing. However, there is a renewed interest in microbial cultivation because isolates provide the basis for understanding the taxonomic and functional units of biodiversity, elucidating novel biochemical pathways and the mechanisms underlying microbe-microbe and microbe-host interactions and opening new avenues for biotechnological and clinical applications. In this Perspective, we present areas of research and applications that will benefit from advancement in anaerobic microbial cultivation. We highlight key technical and infrastructural hurdles associated with the development and deployment of sophisticated cultivation workflows. Improving the performance of cultivation techniques will set new trends in functional microbiome research in the coming years.
Collapse
Affiliation(s)
- Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany.
| | - Franziska Faber
- Institute for Hygiene and Microbiology, Faculty of Medicine, University of Würzburg, Würzburg, Germany
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Mathieu Groussin
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology, School of Life Sciences, Technical University of Munich, Freising, Germany
- ZIEL - Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Jörg Overmann
- Leibniz-Institut DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- Microbiology, Technical University of Braunschweig, Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Charlie Pauvert
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Mathilde Poyet
- Institute of Experimental Medicine, Kiel University, Kiel, Germany
| | - Joel Selkrig
- Host-Microbe Interactomics Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Bärbel Stecher
- ZIEL - Institute for Food and Health, Technical University of Munich, Freising, Germany
- Chair of Intestinal Microbiome, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Athanasios Typas
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany
| | - Maria J G T Vehreschild
- Goethe University Frankfurt, University Hospital Frankfurt, Department II of Internal Medicine, Infectious Diseases, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Alexander J Westermann
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
- Department of Microbiology, Biocentre, University of Würzburg, Würzburg, Germany
| | - David Wylensek
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Lisa Maier
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, Tübingen, Germany
- M3-Research Center for Malignome, Metabolome and Microbiome, University of Tübingen, Tübingen, Germany
| |
Collapse
|
46
|
Jin J, Sun X, Wang L. Association of dietary index of gut microbiota with cardiovascular disease risk: new evidence from NHANES 2007-2018. BMC Cardiovasc Disord 2025; 25:332. [PMID: 40295908 PMCID: PMC12039086 DOI: 10.1186/s12872-025-04776-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The dietary index of gut microbiota (DI-GM) is a newly proposed index for assessing dietary quality, and studies on its association with cardiovascular disease (CVD) are limited. This study aimed to investigate the association between DI-GM and the prevalence of CVD. METHODS We utilized data from the National Health and Nutrition Examination Survey (NHANES). Logistic regression analyses were performed to examine the association between DI-GM and CVD. Smoothed curve fitting was employed to explore potential nonlinear relationships. Additionally, subgroup analyses were conducted to assess the stability of the results. RESULTS The study included 22,590 participants, of whom 20,216 had no CVD and 2,374 had CVD. After adjusting for all covariates, the DI-GM score was significantly negatively associated with CVD risk, with a 4% reduction in CVD risk for each unit increase in DI-GM score (OR = 0.96, 95% CI: 0.94-0.99, P = 0.015). Notably, the highest DI-GM score group (6-12) had a 13% lower risk of CVD compared to the lowest DI-GM score group (0-3) (OR = 0.87, 95% CI: 0.76-1.00, P = 0.048). CONCLUSION The research results indicate that a higher DI-GM score protects against CVD, providing crucial empirical support for dietary intervention strategies based on gut microbiota modulation. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Jiameng Jin
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xingang Sun
- Department of Cardiology Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, Zhejiang Province, China.
| | - Lihong Wang
- Department of Cardiology Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
47
|
Qi HX, Wang Q, Zhou GQ. Association of Clostridium difficile infection with clinical outcomes of patients with inflammatory bowel disease: A meta-analysis. World J Gastrointest Surg 2025; 17:100555. [PMID: 40291861 PMCID: PMC12019072 DOI: 10.4240/wjgs.v17.i4.100555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/07/2025] [Accepted: 02/05/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Clostridium difficile infection (CDI) is common in patients with inflammatory bowel disease (IBD). AIM To assess the association of CDI with clinical outcomes of IBD. METHODS PubMed, EMBASE, Web of Science, and the Cochrane Library databases were searched from inception to March 2024. Eligible articles included observational studies that reported on outcomes such as mortality, colectomy, hospitalization, intensive care unit (ICU) admission, complication rates, and length of hospital stay in IBD patients with and without CDI. Data were extracted, and a random-effects model was used to calculate pooled odds ratios (ORs) and mean differences (MDs). RESULTS As shown in the data from 21 studies with 1249158 participants, CDI significantly increased the risk of mortality in IBD patients [pooled OR = 4.569, 95% confidence intervals (95%CI): 2.584 to 8.079]. Although the pooled OR for colectomy was 1.409 (95%CI: 0.922 to 2.155), it was not statistically significant. Similarly, CDI did not impact hospitalization (pooled OR = 1.056, 95%CI: 0.512 to 2.179) and ICU admission outcomes (pooled OR = 1.970, 95%CI: 0.420 to 9.246) of patients with IBD. The rate of complications was comparable in the two groups (pooled OR = 0.658, 95%CI: 0.378 to 1.147). However, CDI was associated with a significantly more extended hospital stay (pooled MD = 0.349 days, 95%CI: 0.002 to 0.696). CONCLUSION CDI is linked to increased mortality and prolonged hospitalization in IBD patients. These results emphasize the need for early detection and appropriate management. Implementing routine CDI screening during IBD flare-ups and stringent infection control measures could mitigate severe complications and reduce the healthcare burden.
Collapse
Affiliation(s)
- Hai-Xin Qi
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310003, Zhejiang Province, China
| | - Qi Wang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang Province, China
| | - Gui-Qun Zhou
- Department of Gastroenterology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, Zhejiang Province, China
| |
Collapse
|
48
|
Black CJ, Olano C, Quigley EMM, Ford AC. Common misconceptions and controversies in the management of irritable bowel syndrome. Nat Rev Gastroenterol Hepatol 2025:10.1038/s41575-025-01065-9. [PMID: 40281279 DOI: 10.1038/s41575-025-01065-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/28/2025] [Indexed: 04/29/2025]
Abstract
Despite an increase in our understanding of the pathophysiology of irritable bowel syndrome (IBS), in the context of abnormal gut-brain axis communication, and advances in both pharmacological and non-pharmacological treatment of the disorder, there remain areas in which there are misconceptions and controversies in the clinical management of IBS. This Perspective aims to highlight some of the most common misconceptions and controversies in IBS management, including those that the scientific literature has resolved, but for which further education of clinicians dealing with patients with IBS might be required to implement the findings from medical research. Areas of remaining contention are also discussed, as are suggestions as to how these issues could be addressed, both by advances in clinical practice and by further research.
Collapse
Affiliation(s)
- Christopher J Black
- Leeds Gastroenterology Institute, St. James's University Hospital, Leeds, UK
- Leeds Institute of Medical Research at St. James's, University of Leeds, Leeds, UK
| | - Carolina Olano
- Gastroenterology Department, Universidad de la Republica, Montevideo, Uruguay
| | - Eamonn M M Quigley
- Division of Gastroenterology and Hepatology, Lynda K and David M Underwood Center for Digestive Disorders, Houston Methodist Hospital and Weill Cornell Medical College, Houston, TX, USA
| | - Alexander C Ford
- Leeds Gastroenterology Institute, St. James's University Hospital, Leeds, UK.
- Leeds Institute of Medical Research at St. James's, University of Leeds, Leeds, UK.
| |
Collapse
|
49
|
Smits WK, Garey KW, Riley TV, Johnson S. Clostridioides difficile is a Bacterial Priority Pathogen. Anaerobe 2025:102965. [PMID: 40288746 DOI: 10.1016/j.anaerobe.2025.102965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Affiliation(s)
- Wiep Klaas Smits
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, The Netherlands.
| | - Kevin W Garey
- University of Houston College of Pharmacy, Houston TX, USA
| | - Thomas V Riley
- School of Biomedical Sciences, The University of Western Australia and Department of Microbiology, PathWest Laboratory Medicine (EA), Queen Elizabeth II Medical Centre, Nedlands 6009, Western Australia
| | - Stuart Johnson
- Loyola University Chicago (Emeritus), Hines VA Hospital Research Service, Chicago IL, USA
| |
Collapse
|
50
|
Häsler R, Mikš MH, Bajic D, Soyyilmaz B, Bendik I, van Buul VJ, Steinert RE, Rehman A. Human milk oligosaccharides modulating inflammation in infants, adults and older individuals - from concepts to applications. Adv Nutr 2025:100433. [PMID: 40287068 DOI: 10.1016/j.advnut.2025.100433] [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: 09/16/2024] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
The increasing global prevalence of inflammatory diseases such as ulcerative colitis and irritable bowel syndrome, represents a challenging task for healthcare systems. Several approaches to disease management target the intestinal microbiome, which plays a key role in health and disease. One promising approach is modulating the microbiome using human milk oligosaccharides (HMOs). Originating from human milk, HMOs are indigestible carbohydrates which act in a host-optimized prebiotic fashion by providing an energy source for health-promoting intestinal bacteria and exhibiting systemic effects. Commercial products supporting infant health and development have been the primary fields of HMO application. Advancements in the large-scale production of HMOs through bioengineering and precision fermentation have led to evaluating their potential for managing inflammatory diseases. Several in vitro studies and observations on model systems have been clinically validated in infants, resulting in a large body of evidence supporting the safety and efficacy of HMOs in inflammatory disorders. While novel approaches seek to explore interventions in adults, the primary goal for the future is to provide cost-efficient, safe, and reliable healthcare compounds across all age groups.
Collapse
Affiliation(s)
- Robert Häsler
- Department of Dermatology and Allergology, University Kiel, Rosalind-Franklin-Straße 9, 24105 Kiel, Germany
| | - Marta Hanna Mikš
- University of Warmia and Mazury in Olsztyn, Faculty of Food Science, Plac Cieszynski 1, 10-726, Olsztyn, Poland; dsm-firmenich, Kogle Allé 4, Hørsholm, DK-2970, Denmark
| | - Danica Bajic
- dsm-firmenich, Wurmisweg 576, CH-4303 Kaiseraugst, Switzerland
| | | | - Igor Bendik
- dsm-firmenich, Wurmisweg 576, CH-4303 Kaiseraugst, Switzerland
| | | | | | - Ateequr Rehman
- dsm-firmenich, Wurmisweg 576, CH-4303 Kaiseraugst, Switzerland
| |
Collapse
|