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
|
Sall I, Foxall R, Felth L, Maret S, Rosa Z, Gaur A, Calawa J, Pavlik N, Whistler JL, Whistler CA. Gut dysbiosis was inevitable, but tolerance was not: temporal responses of the murine microbiota that maintain its capacity for butyrate production correlate with sustained antinociception to chronic morphine. Gut Microbes 2025; 17:2446423. [PMID: 39800714 PMCID: PMC11730370 DOI: 10.1080/19490976.2024.2446423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 11/24/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
The therapeutic benefits of opioids are compromised by the development of analgesic tolerance, which necessitates higher dosing for pain management thereby increasing the liability for drug dependence and addiction. Rodent models indicate opposing roles of the gut microbiota in tolerance: morphine-induced gut dysbiosis exacerbates tolerance, whereas probiotics ameliorate tolerance. Not all individuals develop tolerance, which could be influenced by differences in microbiota, and yet no study design has capitalized upon this natural variation. We leveraged natural behavioral variation in a murine model of voluntary oral morphine self-administration to elucidate the mechanisms by which microbiota influences tolerance. Although all mice shared similar morphine-driven microbiota changes that largely masked informative associations with variability in tolerance, our high-resolution temporal analyses revealed a divergence in the progression of dysbiosis that best explained sustained antinociception. Mice that did not develop tolerance maintained a higher capacity for production of the short-chain fatty acid (SCFA) butyrate known to bolster intestinal barriers and promote neuronal homeostasis. Both fecal microbial transplantation (FMT) from donor mice that did not develop tolerance and dietary butyrate supplementation significantly reduced the development of tolerance independently of suppression of systemic inflammation. These findings could inform immediate therapies to extend the analgesic efficacy of opioids.
Collapse
Affiliation(s)
- Izabella Sall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Graduate program in Molecular and Evolutionary Systems Biology, University of New Hampshire, Durham, NH, USA
| | - Randi Foxall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Lindsey Felth
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Soren Maret
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Zachary Rosa
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Anirudh Gaur
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Jennifer Calawa
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Microbiology Graduate Program, University of New Hampshire, Durham, NH, USA
| | - Nadia Pavlik
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Jennifer L. Whistler
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
- Department of Physiology and Membrane Biology, UC Davis School of Medicine, Davis, CA, USA
| | - Cheryl A. Whistler
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
3
|
Loison L, Huré M, Lefranc B, Leprince J, Bôle-Feysot C, Coëffier M, Ribet D. Staphylococcus warneri dampens SUMOylation and promotes intestinal inflammation. Gut Microbes 2025; 17:2446392. [PMID: 39819277 DOI: 10.1080/19490976.2024.2446392] [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: 04/28/2024] [Revised: 11/28/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025] Open
Abstract
Gut bacteria play key roles in intestinal physiology, via the secretion of diversified bacterial effectors. Many of these effectors remodel the host proteome, either by altering transcription or by regulating protein post-translational modifications. SUMOylation, a ubiquitin-like post-translational modification playing key roles in intestinal physiology, is a target of gut bacteria. Mutualistic gut bacteria can promote SUMOylation, via the production of short- or branched-chain fatty acids (SCFA/BCFA). In contrast, several pathogenic bacteria were shown to dampen SUMOylation in order to promote infection. Here, we demonstrate that Staphylococcus warneri, a natural member of the human gut microbiota, decreases SUMOylation in intestinal cells. We identify that Warnericin RK, a hemolytic toxin secreted by S. warneri, targets key components of the host SUMOylation machinery, leading to the loss of SUMO-conjugated proteins. We further demonstrate that Warnericin RK promotes inflammation in intestinal and immune cells using both SUMO-dependent and SUMO-independent mechanisms. We finally show that Warnericin RK regulates the expression of genes involved in intestinal tight junctions. Together, these results highlight the diversity of mechanisms used by bacteria from the gut microbiota to manipulate host SUMOylation. They further highlight that changes in gut microbiota composition may impact intestinal inflammation, by altering the equilibrium between bacterial effectors promoting or dampening SUMOylation.
Collapse
Affiliation(s)
- Léa Loison
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, Rouen, France
| | - Marion Huré
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, Rouen, France
| | - Benjamin Lefranc
- Univ Rouen Normandie, INSERM, Normandie Univ, NorDiC, UMR 1239, PRIMACEN, Rouen, France
| | - Jérôme Leprince
- Univ Rouen Normandie, INSERM, Normandie Univ, NorDiC, UMR 1239, PRIMACEN, Rouen, France
| | - Christine Bôle-Feysot
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, Rouen, France
| | - Moïse Coëffier
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, CHU Rouen, Department of Nutrition, CIC-CRB1404, Rouen, France
| | - David Ribet
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, Rouen, France
| |
Collapse
|
4
|
Park SJ, Kim KW, Lee EJ. Gut-brain axis and environmental factors in Parkinson's disease: bidirectional link between disease onset and progression. Neural Regen Res 2025; 20:3416-3429. [PMID: 39688568 PMCID: PMC11974660 DOI: 10.4103/nrr.nrr-d-24-00994] [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: 08/28/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Parkinson's disease has long been considered a disorder that primarily affects the brain, as it is defined by the dopaminergic neurodegeneration in the substantia nigra and the brain accumulation of Lewy bodies containing α-synuclein protein. In recent decades, however, accumulating research has revealed that Parkinson's disease also involves the gut and uncovered an intimate and important bidirectional link between the brain and the gut, called the "gut-brain axis." Numerous clinical studies demonstrate that gut dysfunction frequently precedes motor symptoms in Parkinson's disease patients, with findings including impaired intestinal permeability, heightened inflammation, and distinct gut microbiome profiles and metabolites. Furthermore, α-synuclein deposition has been consistently observed in the gut of Parkinson's disease patients, suggesting a potential role in disease initiation. Importantly, individuals with vagotomy have a reduced Parkinson's disease risk. From these observations, researchers have hypothesized that α-synuclein accumulation may initiate in the gut and subsequently propagate to the central dopaminergic neurons through the gut-brain axis, leading to Parkinson's disease. This review comprehensively examines the gut's involvement in Parkinson's disease, focusing on the concept of a gut-origin for the disease. We also examine the interplay between altered gut-related factors and the accumulation of pathological α-synuclein in the gut of Parkinson's disease patients. Given the accessibility of the gut to both dietary and pharmacological interventions, targeting gut-localized α-synuclein represents a promising avenue for developing effective Parkinson's disease therapies.
Collapse
Affiliation(s)
- Soo Jung Park
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Kyung Won Kim
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon, South Korea
| | - Eun Jeong Lee
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
5
|
Su F, Su M, Wei W, Wu J, Chen L, Sun X, Liu M, Sun S, Mao R, Bourgonje AR, Hu S. Integrating multi-omics data to reveal the host-microbiota interactome in inflammatory bowel disease. Gut Microbes 2025; 17:2476570. [PMID: 40063366 PMCID: PMC11901428 DOI: 10.1080/19490976.2025.2476570] [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: 12/17/2024] [Revised: 02/14/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Numerous studies have accelerated the knowledge expansion on the role of gut microbiota in inflammatory bowel disease (IBD). However, the precise mechanisms behind host-microbe cross-talk remain largely undefined, due to the complexity of the human intestinal ecosystem and multiple external factors. In this review, we introduce the interactome concept to systematically summarize how intestinal dysbiosis is involved in IBD pathogenesis in terms of microbial composition, functionality, genomic structure, transcriptional activity, and downstream proteins and metabolites. Meanwhile, this review also aims to present an updated overview of the relevant mechanisms, high-throughput multi-omics methodologies, different types of multi-omics cohort resources, and computational methods used to understand host-microbiota interactions in the context of IBD. Finally, we discuss the challenges pertaining to the integration of multi-omics data in order to reveal host-microbiota cross-talk and offer insights into relevant future research directions.
Collapse
Affiliation(s)
- Fengyuan Su
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Meng Su
- The First Clinical Medical School, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Wenting Wei
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jiayun Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Leyan Chen
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xiqiao Sun
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Moyan Liu
- Amsterdam UMC location Academic Medical Center, Department of Experimental Vascular Medicine, Amsterdam, The Netherlands
| | - Shiqiang Sun
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Arno R. Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shixian Hu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
6
|
Mukhopadhya I, Martin JC, Shaw S, Gutierrez-Torrejon M, Boteva N, McKinley AJ, Gratz SW, Scott KP. Novel insights into carbohydrate utilisation, antimicrobial resistance, and sporulation potential in Roseburia intestinalis isolates across diverse geographical locations. Gut Microbes 2025; 17:2473516. [PMID: 40089923 PMCID: PMC11913394 DOI: 10.1080/19490976.2025.2473516] [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: 07/28/2024] [Revised: 02/11/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
Roseburia intestinalis is one of the most abundant and important butyrate-producing human gut anaerobic bacteria that plays an important role in maintaining health and is a potential next-generation probiotic. We investigated the pangenome of 16 distinct strains, isolated over several decades, identifying local and time-specific adaptations. More than 50% of the genes in each individual strain were assigned to the core genome, and 77% of the cloud genes were unique to individual strains, revealing the high level of genome conservation. Co-carriage of the same enzymes involved in carbohydrate binding and degradation in all strains highlighted major pathways in carbohydrate utilization and reveal the importance of xylan, starch and mannose as key growth substrates. A single strain had adapted to use rhamnose as a sole growth substrate, the first time this has been reported. The ubiquitous presence of motility and sporulation gene clusters demonstrates the importance of these phenotypes for gut survival and acquisition of this bacterium. More than half the strains contained functional, potentially transferable, tetracycline resistance genes. This study advances our understanding of the importance of R. intestinalis within the gut ecosystem by elucidating conserved metabolic characteristics among different strains, isolated from different locations. This information will help to devise dietary strategies to increase the abundance of this species providing health benefits.
Collapse
Affiliation(s)
- Indrani Mukhopadhya
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
- Microbiology and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Jennifer C Martin
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Sophie Shaw
- Centre for Genome Enabled Biology and Medicine, University of Aberdeen, Aberdeen, UK
- All Wales Medical Genomics Service, Institute of Medical Genetics, University Hospital of Wales, Heath Park, Cardiff, UK
| | | | - Nikoleta Boteva
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Aileen J McKinley
- Department of Surgery, Aberdeen Royal Infirmary Foresterhill, Aberdeen, UK
| | - Silvia W Gratz
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Karen P Scott
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| |
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
|
Shen Y, Fan N, Ma S, Cheng X, Yang X, Wang G. Gut Microbiota Dysbiosis: Pathogenesis, Diseases, Prevention, and Therapy. MedComm (Beijing) 2025; 6:e70168. [PMID: 40255918 PMCID: PMC12006732 DOI: 10.1002/mco2.70168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/22/2025] Open
Abstract
Dysbiosis refers to the disruption of the gut microbiota balance and is the pathological basis of various diseases. The main pathogenic mechanisms include impaired intestinal mucosal barrier function, inflammation activation, immune dysregulation, and metabolic abnormalities. These mechanisms involve dysfunctions in the gut-brain axis, gut-liver axis, and others to cause broader effects. Although the association between diseases caused by dysbiosis has been extensively studied, many questions remain regarding the specific pathogenic mechanisms and treatment strategies. This review begins by examining the causes of gut microbiota dysbiosis and summarizes the potential mechanisms of representative diseases caused by microbiota imbalance. It integrates clinical evidence to explore preventive and therapeutic strategies targeting gut microbiota dysregulation, emphasizing the importance of understanding gut microbiota dysbiosis. Finally, we summarized the development of artificial intelligence (AI) in the gut microbiota research and suggested that it will play a critical role in future studies on gut dysbiosis. The research combining multiomics technologies and AI will further uncover the complex mechanisms of gut microbiota dysbiosis. It will drive the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Yao Shen
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
| | - Nairui Fan
- Basic Medical College of Jiamusi UniversityHeilongjiangChina
| | - Shu‐xia Ma
- Basic Medical College of Jiamusi UniversityHeilongjiangChina
| | - Xin Cheng
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
- International SchoolGuangzhou Huali College, ZengchengGuangzhouChina
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
- Guangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryGuangdong Second Provincial General HospitalSchool of MedicineJinan UniversityGuangzhouChina
| |
Collapse
|
9
|
Zuo G, Li M, Guo X, Wang L, Yao Y, Huang JA, Liu Z, Lin Y. Fu brick tea supplementation ameliorates non-alcoholic fatty liver disease and associated endotoxemia via maintaining intestinal homeostasis and remodeling hepatic immune microenvironment. Food Res Int 2025; 209:116207. [PMID: 40253128 DOI: 10.1016/j.foodres.2025.116207] [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: 11/06/2024] [Revised: 01/27/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent disorder of excessive fat accumulation and inflammation in the liver that currently lacks effective therapeutic interventions. Fu brick tea (FBT) has been shown to ameliorate liver damage and modulate gut microbiota dysbiosis in NAFLD, but the potential mechanisms have not been comprehensively elucidated, especailly whether its hepatoprotective effects are determined to depend on the homeostasis of gut microbiota, intestinal barrier function and hepatic immune microenvironment. In this study, our results further demonstrated that FBT not only alleviated NAFLD symptoms and related endotoxemia in high-fat diet (HFD)-fed rats, but also attenuated intestinal barrier dysfunction and associated inflammation, also confirmed in Caco-2 cell experiment. Meanwhile, FBT intervention significantly relieved HFD-induced gut microbiota dysbiosis, characterized by increased diversity and composition, particularly facilitating beneficial microbes, including short chain fatty acids (SCFAs) and bile acids producers, such as Blautia and Fusicatenibacter, and inhibiting Gram-negative bacteria, such as Prevotella_9 and Phascolarctobacterium. Also, the gut microbiota-dependent hepatoprotective effects of FBT were verified by fecal microbiota transplantation (FMT) experiment. Thus, the beneficial moulation of gut microbiota altered by FBT in levels of SCFAs, bile acids and lipopolysaccharides, intestinal barrier function and TLR4/NF-κB pathway contributed to alleviate liver steatosis and inflammation. Additionally, the hepatoprotective effects of FBT was further demonstrated by suppressing Kupffer cell activation and regulating lipid metabolism using an ex vivo model of liver organoid. Therefore, FBT supplementation can maintain intenstinal homeostasis and remodel hepatic immune microenvironment to prevent NAFLD and associated endotoxemia.
Collapse
Affiliation(s)
- Gaolong Zuo
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China
| | - Menghua Li
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China
| | - Xiaoli Guo
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China
| | - Ling Wang
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China
| | - Yanyan Yao
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China
| | - Jian-An Huang
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, PR China.
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, PR China.
| | - Yong Lin
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, PR China.
| |
Collapse
|
10
|
Qu Y, An K, Wang D, Yu H, Li J, Min Z, Xiong Y, Xue Z, Mao Z. Short-Chain Fatty Acid Aggregates Alpha-Synuclein Accumulation and Neuroinflammation via GPR43-NLRP3 Signaling Pathway in a Model Parkinson's Disease. Mol Neurobiol 2025; 62:6612-6625. [PMID: 39904963 DOI: 10.1007/s12035-025-04726-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
Parkinson's disease (PD) is characterized by the aggregation of α-synuclein (α-syn) and the loss of dopaminergic (DA) neurons, with growing evidence suggesting a significant role of gut microbiota and their metabolites in the disease's pathogenesis. This study explores the effects of short-chain fatty acids (SCFAs) on PD progression, focusing on the G protein-coupled receptor 43 (GPR43) and the NLRP3 signaling pathway in both in vitro and in vivo models. Employing the1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model and SH-SY5Y cells with SCFAs-treated, this study investigated the impact of SCFAs on α-syn deposition, DA loss, and neuroinflammation. In vitro, supernatant from STC-1 cells was administered to SH-SY5Y cells, and the effects were assessed following the knockdown of NLRP3 or GPR43. In vivo, mice were treated with NLRP3 or GPR43 inhibitors after feeding with SCFAs, and the motor deficits, α-syn pathology, DA neuronal loss, and inflammatory responses were observed. SCFAs were found to exacerbate motor and gastrointestinal dysfunctions in PD models, intensifying α-syn pathology and neuroinflammation. The activation of the NLRP3 inflammasome through GPR43 emerged as a key pathological mechanism, with inhibition of these molecules mitigating the observed effects. Such interventions reduced α-syn accumulation, DA loss, and inflammatory responses, highlighting the pivotal role of the SCFA/GPR43-NLRP3 pathway in PD. The findings from this study elucidate a critical link between gut-derived metabolic changes and neuroinflammatory processes in PD via the SCFA/GPR43-NLRP3 pathway. Targeting this pathway offers a promising therapeutic strategy and enriches our understanding of the gut-brain axis' role in PD progression.
Collapse
Affiliation(s)
- Yi Qu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ke An
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Danlei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haoheng Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingyi Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhe Min
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yongjie Xiong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zheng Xue
- Department of General Practice, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhijuan Mao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
11
|
Murugesan R, Kumar J, Leela KV, Meenakshi S, Srivijayan A, Thiruselvam S, Satheesan A, Chaithanya V. The role of gut microbiota and bacterial translocation in the pathogenesis and management of type 2 diabetes mellitus: Mechanisms, impacts, and dietary therapeutic strategies. Physiol Behav 2025; 293:114838. [PMID: 39922411 DOI: 10.1016/j.physbeh.2025.114838] [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: 12/06/2024] [Revised: 01/17/2025] [Accepted: 01/31/2025] [Indexed: 02/10/2025]
Abstract
PURPOSE OF REVIEW The influence of gut microbiota on Type 2 Diabetes Mellitus (T2DM) is an emerging area of research. This review investigates the relationship between gut microbiota dysbiosis, bacterial translocation, and T2DM. It aims to elucidate how microbial imbalances contribute to the progression of T2DM through bacterial translocation and to evaluate dietary and therapeutic strategies to manage these effects. RECENT FINDINGS Recent studies highlight that dysbiosis in T2DM patients often leads to increased systemic inflammation, impaired glucose metabolism, and disrupted gut barrier integrity. These disruptions promote elevated levels of harmful bacterial components, such as lipopolysaccharides, in the bloodstream. This, in turn, is linked to worsening insulin resistance and metabolic dysfunction. Advances in molecular methods and biomarkers have provided deeper insights into bacterial translocation and its impact on diabetes. Dietary interventions, including nutraceutical agents, high-fiber and low-glycemic index diets, as well as the use of probiotics and prebiotics, have shown promise in restoring gut health and mitigating bacterial translocation. CONCLUSION Maintaining a balanced gut microbiota and intestinal barrier integrity is crucial for managing T2DM. Therapeutic strategies, including dietary modifications and nutraceuticals, have demonstrated potential in reducing bacterial translocation and systemic inflammation. Continued research is needed to refine these approaches and explore novel treatment modalities for improving metabolic health in T2DM patients.
Collapse
Affiliation(s)
- Ria Murugesan
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India.
| | - Janardanan Kumar
- Department of General Medicine, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India.
| | - Kakithakara Vajravelu Leela
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India
| | - Sachdev Meenakshi
- Department of Dietary, Tamil Nadu Government Multi Super Speciality Hospital, Chennai 600002, Tamil Nadu, India
| | - Appandraj Srivijayan
- Department of Internal Medicine, Melmaruvathur Adhiparasakthi Institute of Medical Sciences and Research, Melmaruvathur 603319, Tamil Nadu, India
| | - Shubhashree Thiruselvam
- Department of Obstetrics and Gynaecology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India
| | - Abhishek Satheesan
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India
| | - Venkata Chaithanya
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India
| |
Collapse
|
12
|
Wang M, Ma X, He J, Sun J, Cai F, Liu C, Duan J. Characterizing the impact of podophyllotoxin on pulmonary toxicity and gut-lung microbiota interactions in SD rats based on TEC concept. Microbiol Spectr 2025:e0165324. [PMID: 40277406 DOI: 10.1128/spectrum.01653-24] [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: 07/08/2024] [Accepted: 02/19/2025] [Indexed: 04/26/2025] Open
Abstract
Podophyllotoxin (PPT), an extract from the traditional medicinal plant Dysosma, offers anti-viral and anti-cancer benefits, though its use is limited by toxicity. The mechanisms of PPT's inherent pulmonary toxicity remain elusive. This study leverages the novel "Toxicological Evidence Chain" theory to explore the potential involvement of the "gut-lung axis" in PPT-induced pulmonary toxicity. In this study, we examined injury phenotypes in rats, evaluated pulmonary pathological changes, measured pro-inflammatory factors, and conducted comprehensive analyses of both pulmonary and gut microbiomes and metabolomics. Our findings indicate that exposure to PPT leads to significant pulmonary damage in these animals. The PPT group exhibited significantly elevated levels of total protein, albumin, alkaline phosphatase, and lactate dehydrogenase in bronchoalveolar lavage fluid, accompanied by marked upregulation of interleukin (IL)-18, tumor necrosis factor-alpha, IL-6, and IL-1β expression in lung tissue. Furthermore, 16S rRNA gene sequencing analysis revealed significant increases of Akkermansia, Escherichia-Shigella, and Bacteroides in both intestinal contents and lung tissue of PPT-treated animals, concomitant with notable elevations in short-chain fatty acids (SCFAs) such as isobutyric acid and isovaleric acid, and reductions in acetic acid, propionic acid, and butyric acid. The increased abundance of Akkermansia and Escherichia-Shigella may enhance pulmonary inflammatory factors through effects on intestinal barrier integrity and direct immune stimulation, while elevated Bacteroides may alter SCFA production, exacerbating pulmonary inflammation under PPT treatment, suggesting a potential role in the manifestation of PPT-induced pulmonary toxicity. This study offers new insights into the mechanisms of PPT-induced pulmonary toxicity, highlights the role of the gut-lung axis, and provides avenues for therapeutic intervention. IMPORTANCE PPT, derived from the medicinal plant Dysosma, is known for its anti-cancer and anti-viral properties but limited by severe pulmonary toxicity. This study illuminates the gut-lung microbiota axis's role in mediating this toxicity, revealing how specific microbial and metabolic alterations contribute to lung damage. By uncovering these mechanisms, our research opens avenues for interventions that could mitigate PPT's side effects, potentially enhancing its safety and widening its therapeutic use.
Collapse
Affiliation(s)
- Min Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, , Luoyang, China
| | - Xiao Ma
- Department of Clinical Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, , Luoyang, China
| | - Junjie He
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiaxing Sun
- Department of Clinical Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, , Luoyang, China
| | - Feng Cai
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Chuanxin Liu
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, , Luoyang, China
| | - Jiajia Duan
- Department of Clinical Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, , Luoyang, China
| |
Collapse
|
13
|
Ren Q, Wang Y, Han X, Wang Q, Liang G. The relationship of cardiometabolic index with bowel movement frequency: an NHANES-based cross-sectional analysis. Lipids Health Dis 2025; 24:154. [PMID: 40275266 DOI: 10.1186/s12944-025-02567-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Prior studies have indicated a notable link between gut health and metabolic syndrome (MetS). The cardiometabolic index (CMI), an innovative indicator of metabolic health, effectively predicts MetS. Bowel movement frequency (BMF) closely reflects gastrointestinal function and is a key sign of gut health. Nonetheless, the relationship between CMI and BMF is still unclear. Our research explores the possible association between these two variables. METHODS This study employed 2005 to 2010 National Health and Nutrition Examination Survey data. The CMI for each participant was determined by triglycerides, high-density lipoprotein cholesterol, and the waist-to-hip ratio. Multiple regression, smooth curve fitting, and threshold effect analyses were employed to investigate the association between CMI and BMF. The association's stability across populations was assessed through subgroup analyses and interaction tests. RESULTS The study included 9,678 participants in total. After controlling for potential confounding variables, those in the uppermost CMI quartile had a 0.69 more increase in BMF than the bottom quartile (β = 0.69, 95% CI: 0.34, 1.03). The trend analyses showed that BMF increased steadily with the advancement of the CMI quartiles (P for trend < 0.0001). Associations between CMI and BMF were shown to be nonlinear through smooth curve fitting and threshold effect analyses. Specifically, when CMI ranged from 4.97 to 11.75, a negative connection was observed (β = -0.78, 95% CI: -1.33, -0.23), while positive associations were identified in other ranges. Subgroup analyses and interaction tests indicated significant CMI and BMF association variations when stratified by depression and age categories (P for interaction < 0.05). CONCLUSIONS This research indicates that CMI is generally associated with an increase in BMF. However, when CMI ranges from 4.97 to 11.75, it is associated with a BMF decrease. Notably, the association of CMI and BMF is more potent in young, middle-aged, and depressed people.
Collapse
Affiliation(s)
- Qianyi Ren
- School of First Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Yanan Wang
- Department of Intensive Care Rehabilitation 1, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150001, China
| | - Xinhui Han
- School of First Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Qingyi Wang
- School of First Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Guoying Liang
- Department of Gastroenterology 1, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
14
|
Ye X, An X, Zhang T, Kong Y, Jia S, Wu J. CGA protects against experimental colitis by modulating host purine metabolism through the gut microbiota. Int Immunopharmacol 2025; 153:114547. [PMID: 40147263 DOI: 10.1016/j.intimp.2025.114547] [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: 11/26/2024] [Revised: 02/24/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Alterations in the gut microbiota may contribute to the development of inflammatory bowel disease (IBD). Chlorogenic acid (CGA), a product of the esterification of caffeic acid and quinic acid, is one of the most abundant polyphenols in the human diet and has potential beneficial effects on gut function. However, the underlying mechanisms remain unclear. In this study, the pharmacological effects of CGA on colitis and the potential underlying mechanisms were investigated. METHODS A mouse model of colitis was induced via the use of 4 % dextran sulfate sodium (DSS), and the mice were treated with 200 mg/kg CGA. Body weight, colon length, colon tissue pathology, and plasma and colon inflammatory cytokine levels were assessed. RNA sequencing was used to detect changes in gene expression in mouse colon tissues, and 16S rRNA sequencing was used to analyze the composition and structure of the gut microbiota. Fecal metabolomic analysis was performed, and fecal microbiota transplantation (FMT) was used to evaluate the contribution of the gut microbiota. RESULTS CGA significantly alleviated DSS-induced colitis, alleviating intestinal mucosal barrier damage and gut microbiota dysbiosis. It significantly enriched bacteria that produce short-chain fatty acids (SCFAs). CGA inhibited the accumulation of purine metabolites derived from the microbiota and suppressed immune-related signaling cascades, exerting immunomodulatory effects. Furthermore, the gut microbiota of CGA-treated mice alleviated DSS-induced colitis through FMT. CONCLUSION CGA alleviates colitis in a gut microbiota-dependent manner, potentially providing a new strategy for the treatment of IBD.
Collapse
Affiliation(s)
- Xiaolin Ye
- Department of Gastroenterology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Xueying An
- Department of Gastroenterology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Tianzhuo Zhang
- Department of Gastroenterology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Yan Kong
- Department of Gastroenterology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Shuangzhen Jia
- Department of Gastroenterology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Jie Wu
- Department of Gastroenterology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| |
Collapse
|
15
|
Zaher A, Moura Nascimento Santos MJ, Elsaygh H, Peterson SJ, Colli Cruz C, Thomas AS, Wang Y. Management of refractory checkpoint inhibitor-induced colitis. Expert Opin Drug Saf 2025:1-10. [PMID: 40251944 DOI: 10.1080/14740338.2025.2496431] [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: 01/21/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
INTRODUCTION This review discusses the epidemiology, pathophysiology, and factors associated with refractory immune-mediated diarrhea and colitis (r-IMDC), emphasizing tailored treatment strategies. AREAS COVERED The current literature on r-IMDC was reviewed using PubMed (2015-2025), focusing on clinical trials, meta-analyses, and case reports relevant to its management. EXPERT OPINION Effectively managing r-IMDC is crucial for balancing toxicities and antitumor response. Available second and third-line management options for r-IMDC cases must be carefully evaluated. Future perspectives include development of standardized protocols beyond second-line therapies and predictive biomarkers to enable personalized treatment.
Collapse
Affiliation(s)
- Anas Zaher
- Department of Internal Medicine, New York Presbyterian - Brooklyn Methodist/Weill Cornell Medicine, Brooklyn, NY, USA
| | | | - Hassan Elsaygh
- Department of Internal Medicine, University of Debrecen, Debrecen, Hungary
| | - Stephen J Peterson
- Department of Internal Medicine, New York Presbyterian - Brooklyn Methodist/Weill Cornell Medicine, Brooklyn, NY, USA
| | - Carolina Colli Cruz
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anusha Shirwaikar Thomas
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yinghong Wang
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
16
|
Cuisiniere T, Hajjar R, Oliero M, Calvé A, Fragoso G, Rendos HV, Gerkins C, Taleb N, Gagnon-Konamna M, Dagbert F, Loungnarath R, Sebajang H, Schwenter F, Wassef R, Ratelle R, De Broux É, Richard C, Santos MM. Initial gut microbiota composition is a determining factor in the promotion of colorectal cancer by oral iron supplementation: evidence from a murine model. MICROBIOME 2025; 13:100. [PMID: 40259408 PMCID: PMC12013013 DOI: 10.1186/s40168-025-02101-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/26/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) development is influenced by both iron and gut microbiota composition. While iron supplementation is routinely used to manage anemia in CRC patients, it may also impact gut microbiota and promote tumorigenesis. In this study, we investigated the impact of initial gut microbiota composition on iron-promoted tumorigenesis. We performed fecal microbiota transplantation (FMT) in ApcMin/+ mice using samples from healthy controls, CRC patients, and mice, followed by exposure to iron sufficient or iron excess diets. RESULTS We found that iron supplementation promoted CRC and resulted in distinct gut microbiota changes in ApcMin/+ mice receiving FMT from CRC patients (FMT-CRC), but not from healthy controls or mice. Oral treatment with identified bacterial strains, namely Faecalibaculum rodentium, Holdemanella biformis, Bifidobacterium pseudolongum, and Alistipes inops, protected FMT-CRC mice against iron-promoted tumorigenesis. CONCLUSIONS Our findings suggest that microbiota-targeted interventions may mitigate tumorigenic effects of iron supplementation in anemic patients with CRC.
Collapse
Affiliation(s)
- Thibault Cuisiniere
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Roy Hajjar
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Manon Oliero
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Annie Calvé
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Gabriela Fragoso
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Hervé Vennin Rendos
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Claire Gerkins
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Nassima Taleb
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Marianne Gagnon-Konamna
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - François Dagbert
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Rasmy Loungnarath
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Herawaty Sebajang
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Frank Schwenter
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Ramses Wassef
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Richard Ratelle
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Éric De Broux
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Carole Richard
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Manuela M Santos
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada.
- Institut du Cancer de Montréal, Montréal, Québec, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
17
|
Li X, Su Q, Xue J, Wei S. Mechanisms, structure-activity relationships, and skin applications of natural polysaccharides in anti-aging: A review. Int J Biol Macromol 2025; 310:143320. [PMID: 40258559 DOI: 10.1016/j.ijbiomac.2025.143320] [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: 02/19/2025] [Revised: 04/03/2025] [Accepted: 04/16/2025] [Indexed: 04/23/2025]
Abstract
Natural polysaccharides, a class of biological macromolecules found in nature, have recently attracted considerable interest owing to their notable anti-aging capabilities. This article provides a comprehensive review of the intricate mechanisms through which natural polysaccharides combat aging, as well as their applications in addressing skin aging. Primarily, these polysaccharides manifest their anti-aging effects via diverse pathways, such as antioxidation, gut microbiota regulation, metabolic modulation, and immune system regulation. The anti-aging efficacy of natural polysaccharides is intrinsically linked to their structure-activity relationships, with critical determinants including molecular weight, monosaccharide composition, and chemical architecture. Polysaccharides with lower molecular weights typically demonstrate enhanced biological activity, whereas specific monosaccharide configurations and chemical modifications can markedly augment their anti-aging potential. The utilization of natural polysaccharides in skin aging holds significant promise, offering benefits such as anti-aging, wrinkle reduction, anti-glycation, and the facilitation of skin regeneration. In conclusion, this article synthesizes the advancements in research on natural polysaccharides within the anti-aging sector and forecasts future trajectories, to establish a robust foundation for the innovation of new polysaccharide-derived anti-aging formulations.
Collapse
Affiliation(s)
- Xiujuan Li
- Pharmaceutical Department, The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Qingqi Su
- Skills Training Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Jingwei Xue
- Tumor Precise Intervention and Translational Medicine Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China.
| | - Song Wei
- Tumor Precise Intervention and Translational Medicine Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China.
| |
Collapse
|
18
|
Min M, Afzal N, Maloh J, Dulai AS, Ahmad N, Pinzauti D, Sivamani RK. Prospective Comparative Study of an Oral Synbiotic and a Myoinositol-Based Herbal Supplement in Modifying Hormone Levels and the Gut Microbiome in Non-cystic Acne. Dermatol Ther (Heidelb) 2025:10.1007/s13555-025-01411-4. [PMID: 40246799 DOI: 10.1007/s13555-025-01411-4] [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: 02/11/2025] [Accepted: 04/06/2025] [Indexed: 04/19/2025] Open
Abstract
INTRODUCTION Acne pathogenesis is multifactorial, involving systemic factors including gut dysbiosis, hormones, and chronic inflammation. Probiotics, myoinositol, and plant-derived molecules may modulate acne by targeting these factors. The objective is to compare a synbiotic containing herbs against a myoinositol-based herbal supplement on how they influence acne, the gut microbiome, short chain fatty acids (SCFAs), and hormonal profiles. METHODS This was an 8-week, randomized study involving 36 male and female patients aged 12 to 45 years with non-cystic acne. Subjects received either a synbiotic or a myoinositol-based herbal supplement (MBHS). Acne lesions were counted, stool samples were collected for gut microbiome and SCFA analyses, and hormone collections were performed at baseline, 4, and 8 weeks. RESULTS Several gut bacteria increased by at least threefold at both week 4 and 8 in the synbiotic (Erysipelatoclostridium merdavium, Blautia argi, Faecalibacterium prausnitzii, Prevotella copri, Streptococcus sp001556435, Blautia sp900541955) and MBHS group (Megamonas funiformis, Ligilactobacillus ruminis, Prevotella ssp015074785, Prevotella copri, Gca-900199835 sp900176495). Acne lesion counts decreased significantly in both groups at week 4 (p < 0.0001) and week 8 (synbiotic, p < 0.0001; MBHS, p < 0.0001). There were significant and trending increases in stool and plasma SCFAs in both cohorts at week 4 and 8. After 8 weeks of MBHS, 17-OHP and androstenedione significantly decreased from 27.3 to 11.3 pg/ml (p = 0.001) and 94.9 to 68.0 pg/ml (p = 0.04), respectively. CONCLUSION Both the synbiotic and MBHS improved gut health, augmented SCFAs, and reduced lesion counts in those with non-cystic acne. The MBHS may act by reducing hormone levels of 17-OHP and androstenedione. CLINICAL TRIAL REGISTRATION www. CLINICALTRIALS gov (NCT05919810).
Collapse
Affiliation(s)
- Mildred Min
- Integrative Skin Science and Research, Sacramento, CA, USA
- Integrative Research Institute, Sacramento, CA, USA
- College of Medicine, California Northstate University, Elk Grove, CA, USA
| | - Nasima Afzal
- Integrative Skin Science and Research, Sacramento, CA, USA
- Integrative Research Institute, Sacramento, CA, USA
| | | | - Ajay S Dulai
- Integrative Skin Science and Research, Sacramento, CA, USA
- Integrative Research Institute, Sacramento, CA, USA
| | - Nabeel Ahmad
- Integrative Skin Science and Research, Sacramento, CA, USA
- Integrative Research Institute, Sacramento, CA, USA
- College of Medicine, University of Houston, Houston, TX, USA
| | - David Pinzauti
- The Bio Arte Limited, Laboratories at Life Science Park, Triq San Giljan, San Gwann, Malta
| | - Raja K Sivamani
- Integrative Skin Science and Research, Sacramento, CA, USA.
- Integrative Research Institute, Sacramento, CA, USA.
- College of Medicine, California Northstate University, Elk Grove, CA, USA.
- Department of Dermatology, University of California-Davis, Sacramento, CA, USA.
- Pacific Skin Institute, Sacramento, CA, USA.
| |
Collapse
|
19
|
Xie X, Gu Y, Liu Y, Shen M, Ji J, Gao J, Li J. An inulin-type fructan from Codonopsis pilosula ameliorates cyclophosphamide-induced immunosuppression and intestinal barrier injury in mice. Int J Biol Macromol 2025; 310:143312. [PMID: 40250123 DOI: 10.1016/j.ijbiomac.2025.143312] [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: 01/09/2025] [Revised: 04/08/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
In the present study, an inulin-type fructan (ITF) with the degree polymerization (DP) of 21 was isolated from Codonopsis pilosula roots and its structure was characterized by FT-IR, MALDI-TOF-MS and NMR. The immunomodulatory and intestinal protective effects of ITF were investigated on immunosuppressive mice. Male BALB/c mice were pretreated with cyclophosphamide (Cy) for 3 days to establish an immunosuppressive model followed by ITF treatment. The results demonstrated that compared with the model group, ITF administration significantly increased immune organ index (P<0.05), alleviated intestinal villus damage, stimulated serum cytokine secretion including Ig G, IL-4, IL-6, IL-2, TNF-α, and INF-γ (P<0.05), upregulated the expression of Occludin and Claudin-1 (P<0.05), and increased CD4+ and CD8+ T cells of ileum in Cy-induced mice (P<0.05). Furthermore, ITF restored the intestinal microbiota dysbiosis caused by Cy by increasing the abundance of Muribaculaceae, Blautia, Odoribacter, Lactobacillus and decreasing the abundance of Lachnospiraceae_NK4A136_group (P<0.05). Meanwhile, ITF increased the production of short-chain fatty acids (SCFAs) including acetic acid, propionic acid and butyric acid (P<0.05). These results indicated that ITF can ameliorate cyclophosphamide-induced immunosuppression and intestinal barrier injury, and restore gut microbiota dysbiosis. This study provided important evidences for the immunomodulatory and intestinal protective effects of the ITF from C. pilosula.
Collapse
Affiliation(s)
- Xingfang Xie
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Yao Gu
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Yi Liu
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Mingyue Shen
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Jiaojiao Ji
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Jianping Gao
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Jiankuan Li
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China.
| |
Collapse
|
20
|
Palanivelu L, Chang CW, Li SJ, Liang YW, Lo YC, Chen YY. Interplay of Neuroinflammation and Gut Microbiota Dysbiosis in Alzheimer's Disease Using Diffusion Kurtosis Imaging Biomarker in 3 × Tg-AD Mouse Models. ACS Chem Neurosci 2025; 16:1511-1528. [PMID: 40195658 PMCID: PMC12006996 DOI: 10.1021/acschemneuro.5c00063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/09/2025] Open
Abstract
The relationship between alterations in brain microstructure and dysbiosis of gut microbiota in Alzheimer's disease (AD) has garnered increasing attention, although the functional implications of these changes are not yet fully elucidated. This research examines how neuroinflammation, systemic inflammation, and gut microbiota interact in male 3 × Tg-AD and B6129SF1/J wild-type (WT) mice at 6 months-old (6-MO) and 12 months-old (12-MO). Employing a combination of behavioral assessments, diffusion kurtosis imaging (DKI), microbiota profiling, cytokine analysis, short-chain fatty acids (SCFAs), and immunohistochemistry, we explored the progression of AD-related pathology. Significant memory impairments in AD mice at both assessed ages were correlated with altered DKI parameters that suggest neuroinflammation and microstructural damage. We observed elevated levels of pro-inflammatory cytokines, such as IL-1β, IL-6, TNFα, and IFN-γ, in the serum, which were associated with increased activity of microglia and astrocytes in brain regions critical for memory. Although gut microbiota analysis did not reveal significant changes in alpha diversity, it did show notable differences in beta diversity and a diminished Firmicutes/Bacteroidetes (F/B) ratio in AD mice at 12-MO. Furthermore, a reduction in six kinds of SCFAs were identified at two time points of 6-MO and 12-MO, indicating widespread disruption in gut microbial metabolism. These findings underscore a complex bidirectional relationship between systemic inflammation and gut dysbiosis in AD, highlighting the gut-brain axis as a crucial factor in disease progression. This study emphasizes the potential of integrating DKI metrics, microbiota profiling, and SCFA analysis to enhance our understanding of AD pathology and to identify new therapeutic targets.
Collapse
Affiliation(s)
- Lalitha Palanivelu
- International
Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, 7F., No. 250, Wuxing Street, Xinyi District, Taipei 11031, Taiwan
| | - Ching-Wen Chang
- Department
of Biomedical Engineering, National Yang
Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei 112304, Taiwan
| | - Ssu-Ju Li
- Department
of Biomedical Engineering, National Yang
Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei 112304, Taiwan
| | - Yao-Wen Liang
- Department
of Biomedical Engineering, National Yang
Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei 112304, Taiwan
| | - Yu-Chun Lo
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, 12F., Education and Research Building, Shuang-Ho
Campus, No. 301, Yuantong Road, New Taipei
City 23564, Taiwan
| | - You-Yin Chen
- Department
of Biomedical Engineering, National Yang
Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei 112304, Taiwan
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, 12F., Education and Research Building, Shuang-Ho
Campus, No. 301, Yuantong Road, New Taipei
City 23564, Taiwan
| |
Collapse
|
21
|
Carrera Silva EA, Puyssegur J, Errasti AE. Coevolutionary interplay: Helminths-trained immunity and its impact on the rise of inflammatory diseases. eLife 2025; 14:e105393. [PMID: 40231720 PMCID: PMC12002795 DOI: 10.7554/elife.105393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
The gut biome, a complex ecosystem of micro- and macro-organisms, plays a crucial role in human health. A disruption in this evolutive balance, particularly during early life, can lead to immune dysregulation and inflammatory disorders. 'Biome repletion' has emerged as a potential therapeutic approach, introducing live microbes or helminth-derived products to restore immune balance. While helminth therapy has shown some promise, significant challenges remain in optimizing clinical trials. Factors such as patient genetics, disease status, helminth species, and the optimal timing and dosage of their products or metabolites must be carefully considered to train the immune system effectively. We aim to discuss how helminths and their products induce trained immunity as prospective to treat inflammatory and autoimmune diseases. The molecular repertoire of helminth excretory/secretory products (ESPs), which includes proteins, peptides, lipids, and RNA-carrying extracellular vesicles (EVs), underscores their potential to modulate innate immune cells and hematopoietic stem cell precursors. Mimicking natural delivery mechanisms like synthetic exosomes could revolutionize EV-based therapies and optimizing production and delivery of ESP will be crucial for their translation into clinical applications. By deciphering and harnessing helminth-derived products' diverse modes of action, we can unleash their full therapeutic potential and pave the way for innovative treatments.
Collapse
Affiliation(s)
- Eugenio Antonio Carrera Silva
- EACS and JP Institute of Experimental Medicine, National Scientific and Technical Research Council, National Academy of Medicine (IMEX-CONICET-ANM)Buenos AiresArgentina
| | - Juliana Puyssegur
- EACS and JP Institute of Experimental Medicine, National Scientific and Technical Research Council, National Academy of Medicine (IMEX-CONICET-ANM)Buenos AiresArgentina
| | - Andrea Emilse Errasti
- AEE Institute of Pharmacology, School of Medicine, University of Buenos AiresBuenos AiresArgentina
- National Scientific and Technical Research Council (CONICET)Buenos AiresArgentina
| |
Collapse
|
22
|
Spencer NR, Gunabalasingam M, Dial K, Di X, Malcolm T, Magarvey NA. An integrated AI knowledge graph framework of bacterial enzymology and metabolism. Proc Natl Acad Sci U S A 2025; 122:e2425048122. [PMID: 40193601 PMCID: PMC12012490 DOI: 10.1073/pnas.2425048122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
The study of bacterial metabolism holds immense significance for improving human health and advancing agricultural practices. The prospective applications of genomically encoded bacterial metabolism present a compelling opportunity, particularly in the light of the rapid expansion of genomic sequencing data. Current metabolic inference tools face challenges in scaling with large datasets, leading to increased computational demands, and often exhibit limited inter-relatability and interoperability. Here, we introduce the Integrated Biosynthetic Inference Suite (IBIS), which employs deep learning models and a knowledge graph to facilitate rapid, scalable bacterial metabolic inference. This system leverages a series of Transformer based models to generate high quality, meaningful embeddings for individual enzymes, biosynthetic domains, and metabolic pathways. These embedded representations enable rapid, large-scale comparisons of metabolic proteins and pathways, surpassing the capabilities of conventional methodologies. The examination of evolutionary and functionally conserved metabolites across diverse bacterial species is facilitated by integrating the predictive capabilities of IBIS into a graph database enriched with comprehensive metadata. The consideration of both primary and specialized metabolism, combined with an embedding logic for enzyme discovery, uniquely positions IBIS to identify potential novel metabolic pathways. With the expansion of genomic data necessitating transformative approaches to advance molecular metabolism research, IBIS delivers an AI-driven holistic investigation of bacterial metabolism.
Collapse
Affiliation(s)
- Norman R. Spencer
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ONL8S 4L8, Canada
| | - Mathusan Gunabalasingam
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ONL8S 4L8, Canada
| | - Keshav Dial
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ONL8S 4L8, Canada
| | - Xiaxia Di
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ONL8S 4L8, Canada
| | - Tonya Malcolm
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ONL8S 4L8, Canada
| | - Nathan A. Magarvey
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ONL8S 4L8, Canada
| |
Collapse
|
23
|
Lu B, Pan S, He J, Li B, Cao N, Fu X, Liu W, Huang Y, Tian Y, Xu D, Li W. Protective effects of polysaccharide of Atractylodes macrocephala Koidz and Jiawei Si-jun-zi Decoction on gut health and immune function in cyclophosphamide-treated chicks. Poult Sci 2025; 104:105160. [PMID: 40267565 DOI: 10.1016/j.psj.2025.105160] [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: 01/14/2025] [Revised: 04/08/2025] [Accepted: 04/13/2025] [Indexed: 04/25/2025] Open
Abstract
The gut serves not only as digestive but also as critical immune organ, playing a vital role in maintaining the growth performance and immune function of poultry. Atractylodes macrocephala Koidz (AMK) is known for its antioxidant, anti-inflammatory and immunomodulatory properties. This study utilized a Cyclophosphamide (CTX)-induced gut injury model to explore the effects of Polysaccharide of Atractylodes macrocephala Koidz (PAMK) and the Jiawei Si-jun-zi Decoction (JSD) on alleviating gut injury and modulating immune function. The experimental results demonstrated that CTX significantly reduced the average daily gain (ADG) and antioxidant capacity of broiler chicks, disrupted intestinal barrier function, and induced gut microbiota dysbiosis. However, supplementation with PAMK and JSD significantly improved ADG, enhanced antioxidant enzyme activity, alleviated oxidative stress, and upregulated the expression of barrier-related genes such as ZO-1 and Occludin. Additionally, PAMK and JSD significantly increased anti-inflammatory cytokines, including IL-10 and TGF-β, improved gut microbiota diversity, enriched beneficial microbial populations, and restored the microbiota balance disrupted by CTX. These findings suggest that PAMK and JSD effectively mitigate CTX-induced intestinal injury by regulating the antioxidant system, strengthening intestinal barrier function, and restoring gut microbiota structure. This study provides a scientific basis for the development of safe and effective feed additives and proposes a novel strategy to reduce antibiotic use in poultry farming.
Collapse
Affiliation(s)
- Baili Lu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Shirou Pan
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Jiayu He
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Bingxin Li
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Nan Cao
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Xinliang Fu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Wenjun Liu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Yunmao Huang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Yunbo Tian
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Danning Xu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Wanyan Li
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China.
| |
Collapse
|
24
|
Zhang M, Feng C, Zhang B, Yin Y, Chen J, Liu H, Farag MA, Mamadalieva NZ, Li N, Sun J, Sun S, Liu C. In vitro and in vivo immune-enhancing effects of punicic acid and the action mechanisms as revealed via microbiome and lipid profiling. Food Funct 2025; 16:3120-3133. [PMID: 40159912 DOI: 10.1039/d4fo05023a] [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: 04/02/2025]
Abstract
Punicic acid (PA) is a chief component of pomegranate seed oil with several health benefits. In this study, the in vitro immunomodulatory activity of PA was assessed using RAW264.7 cells, revealing that PA activated the macrophages, facilitated the concentration of immune-related cytokines and enzymes, and regulated the immune-related NF-κB and MAPK signaling pathways. Further, the in vivo immune-enhancing effect of PA was evaluated with the cyclophosphamide (CTX)-induced immune-compromised mouse model with 16S rDNA amplicon sequencing and relative quantification of lipidome. Results indicated that high doses of PA (200 mg kg-1) remarkably restored CTX-induced immune injury by enhancing the innate and adaptive immunity to stimulate the secretion of immune-related factors. In addition, PA improved gut microbiota dysbiosis and ameliorated lipid metabolism disorders. Our research provides a theoretical basis for the exploitation of PA as a functional component with immune-enhancing effects and adds to the potential health uses of pomegranate seed oil.
Collapse
Affiliation(s)
- Mengqi Zhang
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
| | - Caiyun Feng
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
- College of Life Sciences, Shandong Normal University, Jinan, 250014, PR China
| | - Bo Zhang
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China
| | - Yanlei Yin
- Shandong Institute of Pomology, Tai'an, 271000, China
| | - Jinlong Chen
- Work Station of Forest Fruit Industry in Kashi, Kashi, 844000, PR China
| | - Haoran Liu
- JiMei One Health Industry (Shandong) Co., Ltd, Zaozhuang, 277300, PR China
| | - Mohamed A Farag
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Nilufar Z Mamadalieva
- Institute of the Chemistry of Plant Substances of the Academy Sciences of Uzbekistan, Tashkent, 100170, Uzbekistan
| | - Ningyang Li
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, PR China
| | - Jinyue Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
| | - Shutao Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
| | - Chao Liu
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
- Shandong Aojing Biotechnology Co., Ltd, Jining, 273500, PR China
| |
Collapse
|
25
|
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] [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
|
26
|
Jena PK, Wakita D, Gomez AC, Carvalho TT, Atici AE, Aubuchon E, Narayanan M, Lee Y, Fishbein MC, Takasato Y, Kurashima Y, Kiyono H, Cani PD, de Vos WM, Underhill DM, Devkota S, Chen S, Shimada K, Crother TR, Arditi M, Rivas MN. Intestinal Microbiota Contributes to the Development of Cardiovascular Inflammation and Vasculitis in Mice. Circ Res 2025; 136:e53-e72. [PMID: 40026151 PMCID: PMC11985309 DOI: 10.1161/circresaha.124.325079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Alterations in the intestinal microbiota contribute to the pathogenesis of various cardiovascular disorders, but how they affect the development of Kawasaki disease (KD) an acute pediatric vasculitis, remains unclear. METHODS We used the Lactobacillus casei cell wall extract (LCWE) murine model of KD vasculitis to assess the contribution of the intestinal microbiota to the development of vascular inflammation. We evaluated the severity of vasculitis in microbiota-depleted mice. 16S rRNA gene sequencing was used to characterize the fecal microbiome composition of LCWE-injected mice. Some groups of mice were orally treated with selected live or pasteurized bacteria, short-chain fatty acids, or Amuc_1100, the Toll-like receptor 2 signaling outer membrane protein from Akkermansia muciniphila, and their impact on vasculitis development was assessed. RESULTS We report that depleting the gut microbiota reduces the development of cardiovascular inflammation in a murine model mimicking KD vasculitis. The development of cardiovascular lesions was associated with alterations in the intestinal microbiota composition and, notably, a decreased abundance of Akkermansia muciniphila and Faecalibacterium prausnitzii. Oral supplementation with either of these live or pasteurized individual bacteria or with short-chain fatty acids produced by them attenuated cardiovascular inflammation, as reflected by decreased local immune cell infiltrations. Treatment with Amuc_1100 also reduced the severity of vascular inflammation. CONCLUSIONS This study reveals an underappreciated gut microbiota-cardiovascular inflammation axis in KD vasculitis pathogenesis and identifies specific intestinal commensals that regulate vasculitis in mice by producing metabolites or via extracellular proteins capable of enhancing and supporting gut barrier function.
Collapse
Affiliation(s)
- Prasant K. Jena
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daiko Wakita
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Angela C. Gomez
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Thacyana T. Carvalho
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Asli E. Atici
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Emily Aubuchon
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Meena Narayanan
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Youngho Lee
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael C. Fishbein
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, CA, USA
| | - Yoshihiro Takasato
- Department of Allergy, Allergy and Immunology Center, Aichi Children’s Health and Medical Center, Obu, Japan
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yosuke Kurashima
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Japan
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Japan
| | - Patrice D. Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, WELBIO department, WEL Research Institute, Wavre, Belgium
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - David M. Underhill
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- F. Widjaja Inflammatory Bowel Diseases Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Suzanne Devkota
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- F. Widjaja Inflammatory Bowel Diseases Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Human Microbiome Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shuang Chen
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kenichi Shimada
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Timothy R. Crother
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
27
|
Ye Z, Tan Q, Woltemate S, Tan X, Römermann D, Grassl GA, Vital M, Seidler U, Kini A. Escherichia coli Nissle Improves Short-Chain Fatty Acid Absorption and Barrier Function in a Mouse Model for Chronic Inflammatory Diarrhea. Inflamm Bowel Dis 2025; 31:1109-1120. [PMID: 39708301 PMCID: PMC11985405 DOI: 10.1093/ibd/izae294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Defects in SLC26A3, the major colonic Cl-/HCO3- exchanger, result in chloride-rich diarrhea, a reduction in short-chain fatty acid (SCFA)-producing bacteria, and a high incidence of inflammatory bowel disease in humans and in mice. Slc26a3-/- mice are, therefore, an interesting animal model for spontaneous but mild colonic inflammation and for testing strategies to reverse or prevent the inflammation. This study investigates the effect of Escherichia coli Nissle (EcN) application on the microbiome, SCFA production, barrier integrity, and mucosal inflammation in slc26a3-/- mice. METHODS In vivo fluid absorption and bicarbonate secretion were assessed in the gut of slc26a3+/+ and slc26a3-/- mice before and during luminal perfusion with 100 mM sodium acetate. Age-matched slc26a3+/+ and slc26a3-/- mice were intragastrically gavaged twice daily with 2 × 108 CFU/100 µL of EcN for 21 days. Body weight and stool water content were assessed daily, and stool and tissues were collected for further analysis. RESULTS Addition of sodium acetate to the lumen of the proximal colon significantly increased fluid absorption and luminal alkalinization in the slc26a3-/- mice. Gavage with EcN resulted in a significant increase in SCFA levels and the expression of SCFA transporters in the slc26a3-/- cecum, the predominant habitat of EcN in mice. This was accompanied by an increase in mucus-producing goblet cells and a decrease in the expression of inflammatory markers as well as host defense antimicrobial peptides. EcN did not improve the overall diversity of the luminal microbiome but resulted in a significant increase in SCFA producers Lachnospiraceae and Ruminococcaceae in the slc26a3-/- feces. CONCLUSIONS These findings suggest that EcN is able to proliferate in the inflamed cecum, resulting in increased microbial SCFA production, decreased inflammation, and improved gut barrier properties. In sufficient dosage, probiotics may thus be an effective anti-inflammatory strategy in the diseased gut.
Collapse
Affiliation(s)
- Zhenghao Ye
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Qinghai Tan
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Department of Gastroenterology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Sabrina Woltemate
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Xinjie Tan
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dorothee Römermann
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Guntram A Grassl
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
- German Centre for Infection Research DZIF, Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Marius Vital
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Ursula Seidler
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Archana Kini
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
28
|
Wang M, Liu K, Bao W, Hang B, Chen X, Zhu X, Li G, Liu L, Xiang H, Hu H, Lu Y, Song Z, Chen J, Wang Y. Gut microbiota protect against colorectal tumorigenesis through lncRNA Snhg9. Dev Cell 2025; 60:1008-1017.e7. [PMID: 39755115 DOI: 10.1016/j.devcel.2024.12.013] [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: 11/09/2023] [Revised: 09/30/2024] [Accepted: 12/05/2024] [Indexed: 01/06/2025]
Abstract
The intestinal microbiota is a key environmental factor in the development of colorectal cancer (CRC). Here, we report that, in the context of mild colonic inflammation, the microbiota protects against colorectal tumorigenesis in mice. This protection is achieved by microbial suppression of the long non-coding RNA (lncRNA) Snhg9. Snhg9 promotes tumor growth through inhibition of the tumor suppressor p53. Snhg9 suppresses p53 activity by dissociating the p53 deacetylase sirtuin 1 (SIRT1) from the cell cycle and apoptosis regulator 2 (CCAR2). Consequently, the depletion of the microbiota by antibiotics causes upregulation of Snhg9 and accelerates CRC progression. Moreover, Snhg9 is functionally conserved. Human SNHG9 promotes tumor growth via the same mechanism as mouse Snhg9, despite their low sequence similarity.
Collapse
Affiliation(s)
- Meng Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Kailin Liu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Wu Bao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Bingqing Hang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Xianjiong Chen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Xinyi Zhu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Guifang Li
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Lihong Liu
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
| | - Haoyi Xiang
- Department of Colorectal Surgery and Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Hai Hu
- Breast Cancer Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Yanhui Lu
- School of Nursing, Peking University, Beijing 100191, China
| | - Zhangfa Song
- Department of Colorectal Surgery and Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China.
| | - Jiaxin Chen
- Department of Breast Surgery and Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China.
| | - Yuhao Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310029, Zhejiang, China; Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou 310029, Zhejiang, China.
| |
Collapse
|
29
|
Zhang C, Liu B, Cui Z, Wu K, Huang H, Wang Y, Ma X, Tan B. Effects of Magnolia officinalis extract on the growth performance and immune function of weaned piglets. Porcine Health Manag 2025; 11:16. [PMID: 40181480 PMCID: PMC11969803 DOI: 10.1186/s40813-025-00430-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Magnolia officinalis is a medicinal herb known for its pharmacological properties and as a potential natural feed additive. We aimed to assess the effects of dietary Magnolia officinalis extract (MOE) on the growth performance and immune function of piglets, and explored the potential of MOE as a natural alternative to antibiotics for piglet nutrition during weaning. RESULTS Compared with the basal diet group (CK), the MOE diet significantly increased average daily feed intake and reduced diarrhea incidence and serum interleukin-6 (IL-6) levels. Compared with 0.1% MOE group, the 0.05% MOE group had lower diarrhea rates, eosinophils (EOS) count, EOS' percentage, and serum interleukin-4 levels. Compared with CK, 0.05% MOE supplementation in the diet could reduce the diarrhea incidence and the thymus index by elevating the levels of transforming growth factor-β (TGF-β) and interleukin-10 (IL-10) in the serum, jejunum, and ileum. Compared with the basal diet group, 0.05% MOE supplementation upregulated the mRNA expressions of IL-10 and TGF-β1 in the jejunum and ileum (P < 0.05) and those of IL-10, interleukin-1β (IL-1β), and interferon-γ (IFN-γ) in the thymus (P < 0.05). Moreover, 0.05% MOE increased the levels of butyric, isobutyric, isovaleric, and valeric acids in the colon. CONCLUSIONS MOE supplementation could modulate the immune status of animals, lower production costs, and contribute to more sustainable and ethical pig farming practices by promoting healthier growth and reducing disease susceptibility. Our findings offer a sustainable solution to antibiotic use in animal farming, addressing concerns about antibiotic resistance and food safety.
Collapse
Affiliation(s)
- Chen Zhang
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, 230031, China.
- Yuelushan Laboratory, Changsha, 410128, China.
- Institute of Yunnan Circular Agricultural Industry, Pu'er, 665000, China.
| | - Bifan Liu
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
- Yuelushan Laboratory, Changsha, 410128, China
- Institute of Yunnan Circular Agricultural Industry, Pu'er, 665000, China
| | - Zhijuan Cui
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
| | - Kunfu Wu
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
- Yuelushan Laboratory, Changsha, 410128, China
| | - Haibo Huang
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, 230031, China
- Yuelushan Laboratory, Changsha, 410128, China
- Institute of Yunnan Circular Agricultural Industry, Pu'er, 665000, China
| | - Yongliang Wang
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
- Yuelushan Laboratory, Changsha, 410128, China
- Institute of Yunnan Circular Agricultural Industry, Pu'er, 665000, China
| | - Xiaokang Ma
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
- Yuelushan Laboratory, Changsha, 410128, China
| | - Bi'e Tan
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
- Yuelushan Laboratory, Changsha, 410128, China.
- Institute of Yunnan Circular Agricultural Industry, Pu'er, 665000, China.
| |
Collapse
|
30
|
Takagi K, Tamura Y, Narita N, Komatsu S, Yamazaki S, Matsumura A, Kubota K, Matsumiya T, Sawada K, Nakaji S, Mikami T, Kobayashi W. Involvement of Megasphaera in the oral microbiome and dyslipidemia onset: evidence from a community-based study in Japan. Folia Microbiol (Praha) 2025:10.1007/s12223-025-01258-4. [PMID: 40175821 DOI: 10.1007/s12223-025-01258-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/22/2025] [Indexed: 04/04/2025]
Abstract
Dyslipidemia is a major risk factor for cardiovascular diseases and is influenced by genetic and environmental factors, including diet. Emerging research suggests a link between the gut microbiome and metabolic disorders. While the connection between the gut microbiota and dyslipidemia is well documented, the specific relationship between oral bacteria and dyslipidemia has not been thoroughly investigated. This study aimed to identify oral bacterial species associated with dyslipidemia in a community-based Japanese population. We conducted a metagenomic analysis on tongue coating samples from 763 participants in the Iwaki Health Promotion Project, which were collected during health checkups in 2017 and 2019. Dyslipidemia was diagnosed using standard lipid level criteria. The oral microbiome was analyzed via 16S rDNA amplicon sequencing. Statistical analyses included multiple regression and β diversity assessments. Our analysis revealed that the abundances of several bacterial genera, including Veillonella, Atopobium, Stomatobaculum, Tanneralla, and Megasphaera, are significantly associated with dyslipidemia. A higher relative abundance of Megasphaera was specifically observed in individuals with dyslipidemia. Moreover, Megasphaera abundance was closely associated with the onset of dyslipidemia (P = 0.038, odds ratio: 1.005, 95% confidence interval: 1.000-1.009), suggesting its role in metabolic regulation. This study revealed a significant association between the abundance of specific oral bacteria and dyslipidemia, suggesting the potential of using the oral microbiota as a biomarker for the early detection and management of dyslipidemia. Future research should explore the mechanisms through which oral bacteria influence lipid metabolism and the potential for microbioma-based therapies.
Collapse
Affiliation(s)
- Koki Takagi
- Department of Oral and Maxillofacial Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yoshihiro Tamura
- Department of Oral and Maxillofacial Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Norihiko Narita
- Department of Oral and Maxillofacial Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shotaro Komatsu
- Department of Oral and Maxillofacial Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shunya Yamazaki
- Department of Oral and Maxillofacial Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Akihiro Matsumura
- Department of Oral and Maxillofacial Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kosei Kubota
- Department of Oral and Maxillofacial Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Tomoh Matsumiya
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, Hirosaki, Japan.
| | - Kaori Sawada
- Department of Preemptive Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shigeyuki Nakaji
- Department of Preemptive Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Tatsuya Mikami
- Department of Preemptive Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Wataru Kobayashi
- Department of Oral and Maxillofacial Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
31
|
Xu P, Mageswaran UM, Nisaa AA, Balasubramaniam SD, Rajendran D, Ismail EHBE, Kadir MN, Oon CE, Tan CS, Sany SB, Liong MT. Roles of probiotics against HPV through the gut-vaginal axis. Int J Gynaecol Obstet 2025; 169:1-8. [PMID: 39520180 DOI: 10.1002/ijgo.16005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Human papillomavirus (HPV) is a sexually transmitted virus, whose persistent infection is the main reason for invasive cervical cancer (ICC), which is the fourth most common type of cancer in women, with more than 500 000 new cases every year. After infection, various alterations occur in the host, facilitating the virus's evasion of immune system clearance and promoting its proliferation. Oral probiotic consumption can influence the whole body's immunity, inflammatory reflection, neural, endocrine humoral, metabolic pathways and other organs by adjusting the components of gut microbiota (GM). Some evidence shows there is a tight connection between GM and vaginal microbiota (VM), which is referred to as the gut-vaginal axis. This review investigates the potential role of probiotics in clearing HPV via the gut-vagina axis, emphasizing the effectiveness of Lactobacillus in preventing vaginal diseases and suggesting its potential for HPV clearance. Understanding the role of probiotics in the gut-vagina axis could pave the way for new strategies to reduce and eliminate HPV and related diseases.
Collapse
Affiliation(s)
- Pei Xu
- Bioprocess Technology, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, Penang, Malaysia
- Faculty of Cuisine, Sichuan Tourism University, Chengdu, China
| | - Uma Mageswary Mageswaran
- Bioprocess Technology, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, Penang, Malaysia
| | - Azka Ainun Nisaa
- Bioprocess Technology, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, Penang, Malaysia
| | | | - Deepa Rajendran
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor, Penang, Malaysia
| | - Engku Husna Binti Engku Ismail
- Department of Obstetrics and Gynecology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Muhammad Nashriq Kadir
- Department of Obstetrics and Gynecology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Chern-Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor, Penang, Malaysia
| | - Cheng-Siang Tan
- Faculty of Medicine and Health Sciences (FMHS), Universiti Malaysia Sarawak (UNIMAS), Kota Samarahan, Sarawak, Malaysia
| | - Salina Binti Sany
- Obstetrics and Gynecology, Sunway Medical Centre Penang, Gelugor, Penang, Malaysia
| | - Min Tze Liong
- Bioprocess Technology, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, Penang, Malaysia
- Renewable Biomass Transformation Cluster, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, Penang, Malaysia
| |
Collapse
|
32
|
Chen Z, Ge X, Wang Y, Zhang J, Sui Y, Yin X, Wu N, Yang L, Xu J, Zhou H, Wu Q, Zeng F, Liu L, Shao R, Xu W. Ruditapes philippinarum Polysaccharide Alleviates Hyperglycemia by Modulating Gut Microbiota in a Mouse Model of Type 2 Diabetes Mellitus. Mol Nutr Food Res 2025; 69:e202400996. [PMID: 39981981 DOI: 10.1002/mnfr.202400996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/22/2025]
Abstract
Type 2 diabetes mellitus (T2DM), a widespread chronic metabolic disorder, presents a global challenge. Metformin hydrochloride, although widely prescribed, is associated with notable side effects. This study aims to explore safer, more effective alternatives by assessing the impact of Ruditapes philippinarum polysaccharides (RPPs) on glycemic control and modulation of microbiota in T2DM mice. A T2DM mouse model was established through a high-sucrose/high-fat diet and intraperitioneal streptozotocin injection. Glycometabolism indicators, histopathological features, and gut microbiota composition in caecum samples were assessed. Following 4 weeks of RPPs intervention, fasting blood glucose (FBG), glycated serum protein (GSP), area under the curve (AUC) of oral glucose tolerance test (OGTT), total cholesterol (TC), triglyceride (TG), and low-density lipoprotein cholesterol (LDL-c) levels were reduced in T2DM mice, while AKT-1 and GLUT-2 transcription levels were significant increased. Short-chain fatty acids (SCFAs) concentrations notably increased in the RPP-L group compared to the Model group, with improvements also observed in medium-chain fatty acids (MCFAs) and secondary bile acids (SBAs). Regarding gut microbiota, the Firmicutes-to-Bacteroidetes (F/B) ratio in RPP-L was substantially lower than in the Model group. At the genus level, beneficial bacteria, such as Akkermansia, Alloprevotella, Tidjanibacter, and Faecalibaculum demonstrated increased abundance. Correlation analysis identified Muribaculum, Paramuribaculum, Lacrimispora, and Turicibacter as microbial taxa associated with T2DM progression. RPPs significantly alleviated hyperglycemic symptoms in T2DM mice while enhancing the presence of beneficial gut bacteria, leading to a marked improvement in intestinal microbiota composition. This research offers foundational insights for the potential use of R. philippinarum in biomedical and nutraceutical applications.
Collapse
Affiliation(s)
- Zhuo Chen
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Xiaodong Ge
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Yaolin Wang
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Jiawei Zhang
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Yinzi Sui
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Xuemei Yin
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Na Wu
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Lei Yang
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Jianda Xu
- Department of Orthopaedics, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, China
| | - Hongcheng Zhou
- School of Medicine, Jiangsu Medicine College, Yancheng, China
| | - Qin Wu
- School of Medicine, Jiangsu Medicine College, Yancheng, China
| | - Feng Zeng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Liang Liu
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Rong Shao
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Wei Xu
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| |
Collapse
|
33
|
Zhou Y, Yang Y, Zhu W, Kourkoumelis N, Wang Y, Chen Y, Hong L, Wang J, Zhu J, Zhu C, Zhang X. Microbial Influences on Calcium-Phosphorus Homeostasis and Metabolic Bone Diseases: A Bidirectional Mendelian Randomisation Study on the Gut-Bone Axis. J Cell Mol Med 2025; 29:e70491. [PMID: 40167025 PMCID: PMC11959414 DOI: 10.1111/jcmm.70491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 10/12/2024] [Accepted: 03/05/2025] [Indexed: 04/02/2025] Open
Abstract
Observational studies have shown that the gut microbiota (GM) is associated with bone diseases, particularly calcium-phosphorus metabolic bone diseases, demonstrating the existence of a gut-bone axis. However, whether these associations are causal effects remains to be determined. This study employed bidirectional two-sample Mendelian randomisation (MR) using summary data from Genome-Wide Association Studies (GWAS) of 211 gut microbial taxa and six metabolic bone diseases (osteoporosis, Osteopenia, osteonecrosis, osteomyelitis, hypoparathyroidism and hyperparathyroidism) to explore causal relationships and their directionality. Comprehensive sensitivity analyses were conducted to ensure the robustness of the results, and a false discovery rate-corrected pFDR of < 0.05 was used as a threshold to support strong associations. Additionally, co-localisation analysis was conducted to consolidate the findings. We identified 35 causal relationships between GM and metabolic bone diseases, with 17 exhibiting positive and 18 negative correlations. Furthermore, reverse MR analysis indicated that osteomyelitis was associated with elevated abundance of two GMs (pFDR < 0.05, PP.H4 < 75%). No evidence of horizontal pleiotropy or heterogeneity was observed, and co-localisation analysis further strengthened the evidence for these causal relationships. The study underscores the critical role of GM in influencing bone health through the gut-bone axis, paving the way for future therapeutic interventions targeting the gut-bone axis and offering new directions for research in bone metabolism and diseases.
Collapse
Affiliation(s)
- Yanling Zhou
- Department of OrthopedicsThe First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiAnhuiChina
- Department of OrthopedicsThe Second Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
| | - Yao Yang
- Department of OrthopedicsThe Second Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
| | - Wanbo Zhu
- Department of OrthopedicsShanghai Sixth People's Hospital Affiliated to Shanghai, Jiao Tong University School of Medicine, Shanghai Jiao Tong UniversityShanghaiP. R. China
| | - Nikolaos Kourkoumelis
- Department of Medical PhysicsSchool of Health Sciences, University of IoanninaIoanninaGreece
| | - Yingjie Wang
- Department of OrthopedicsThe Second Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
| | - Yuan Chen
- Department of OrthopedicsThe Second Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
| | - Lingxiang Hong
- Department of OrthopedicsThe Second Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
| | - Junjie Wang
- Department of OrthopedicsThe Second Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
| | - Junchen Zhu
- Department of OrthopedicsThe Second Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
| | - Chen Zhu
- Department of OrthopedicsThe First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiAnhuiChina
| | - Xianzuo Zhang
- Department of OrthopedicsThe First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiAnhuiChina
| |
Collapse
|
34
|
Li H, Wu Y, Zhou Q, Wu P, Xing Y, Zhuang Z, Zhao X, Zhang X. Polysaccharide from steamed Polygonatum sibiricum ameliorates ulcerative colitis by protecting the intestinal mucosal barrier and regulating gut microbiota. Int J Biol Macromol 2025; 301:140343. [PMID: 39880230 DOI: 10.1016/j.ijbiomac.2025.140343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 12/02/2024] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Steamed Polygonatum sibiricum is widely applied in clinical practice for its tonic effect on gastrointestinal tract. A novel polysaccharide named PSSP-EF was extracted from the steamed roots of Polygonatum sibiricum using hot water extraction, ethanol precipitation, and chromatographic purification. PSSP-EF, with a molecular weight of 2.24 × 104 Da, was consisted of mannose, glucosamine hydrochloride, glucose, galactose, xylose, and arabinose in a molar ratio of 10.25: 0.26: 3.56: 80.55: 1.72: 3.66, and its main chain was constituted by→4)-β-D-Galp-(1→4)-β-D-Galp-(1→ and →4)-β-D-Manp-(1→4)-β-D-2ace-Manp-(1→ residues, with branching from β-D-Galp-(1→ residues. PSSP-EF could dramatically relieve clinical symptoms of ulcerative colitis (UC) in mice. Treatment with PSSP-EF significantly alleviated colon inflammation (TNF-α, IL-1β, and IL-6), repaired intestinal mucosal barrier (Occludin, ZO-1, and Claudin-1) and regulated the balance of gut microbiota by increased the levels of Muribaculaeae, while decreasing the levels of Bacteroides, Erysipelatoclostridium, and Romboutsia. Notably, PSSP-EF remarkably increased the levels of acetic, propionic, isobutyric, butyric, valeric, and isovaleric acid in the cecal contents of UC mice. In conclusion, PSSP-EF has a significant therapeutic effect on UC by balancing gut microbiota, protecting intestinal mucosal barrier, and regulating short-chain fatty acid production, and can be developed as a functional food.
Collapse
Affiliation(s)
- Haoran Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yi Wu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qiao Zhou
- Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Peng Wu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yue Xing
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ziming Zhuang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xin Zhao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Xuelan Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
35
|
Dai S, Long J, Han W, Zhang L, Chen B. Alleviative effect of probiotics and prebiotics on dry eye in type 2 diabetic mice through the gut-eye axis. Ocul Surf 2025; 36:244-260. [PMID: 39922458 DOI: 10.1016/j.jtos.2025.02.004] [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: 10/11/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Diabetes Mellitus (DM) is a metabolic disease that manifests as a state of "chronic low-grade inflammation". Patients with DM have a disorder of intestinal flora. There is a discernible correlation between this disorder of intestinal flora and the onset and progression of eye diseases, which offers novel insights into treating eye diseases through the modulation of intestinal flora. Here, we demonstrated that a high-fat diet and streptozotocin injection-induced intestinal microbiota dysbiosis can lead to dry eye-like manifestations in T2DM mice. Probiotic and prebiotic treatments not only alleviated intestinal inflammation and barrier disruption, but also mitigated damage to the lacrimal barrier and suppressed immune cell infiltration and inflammatory responses. Additional mechanism investigation found that probiotics and prebiotics inhibited the TLR4/NF-κB signaling pathway and its downstream pro-inflammatory products both in the lacrimal gland and colon. 16S RNA sequencing identified a reduction in the bacterial genera Akkermansia and Lactobacillus in the fecal samples of DM mice. By contrast, treatment with probiotics and prebiotics led to a reshaping of the intestinal microbial community and a reduction in bile acid metabolites, such as taurocholic acid and deoxycholic acid. Our current study demonstrates that probiotic and prebiotic treatments can ameliorate dry eye-like symptoms and associated pathological changes in T2DM mice. Moreover, we proved that a high-fat diet and STZ-induced microbiota dysbiosis were involved in diabetic dry eye through the gut-eye axis.
Collapse
Affiliation(s)
- Shirui Dai
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| | - Jianfeng Long
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| | - Wentao Han
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| | - Liwei Zhang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| | - Baihua Chen
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| |
Collapse
|
36
|
Sangfuang N, McCoubrey LE, Awad A, Marzorati M, Ghyselinck J, Verstrepen L, Munck JD, Medts JD, Gaisford S, Basit AW. Effects of senotherapeutics on gut microbiome dysbiosis and intestinal inflammation in Crohn's disease: A pilot study. Transl Res 2025; 278:36-47. [PMID: 39986536 DOI: 10.1016/j.trsl.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Inflammatory Bowel Disease (IBD) is characterized by chronic inflammation in the gastrointestinal tract, and is usually accompanied by dysbiosis in the gut microbiome, a factor that contributes to disease progression. Excessive production of reactive oxygen species (ROS) because of gut microbiome dysbiosis-one of the hallmark features of IBD-promotes chronic inflammation and facilitates the transformation of normal cells into senescent cells. Cellular senescence is associated with the development of various chronic and age-related diseases. We hypothesise that senolytic agents, specifically dasatinib (D) and quercetin (Q), could have a beneficial effect on both the gut microbiome and intestinal cells in IBD. The modulatory effects of a combination of D + Q was assessed in the M-SHIME model with faecal microbiota sourced from Crohn's disease patients. D + Q significantly modulated butyrate and lactate levels in the samples from specific patients. In addition, metabolomic analysis showed that D + Q positively impacted the abundance of anti-inflammatory bacteria while also significantly reducing the several species of pathogenic bacteria. Findings from a Caco-2 cell/THP1 co-culture model of IBD demonstrated that D + Q exerted strong immunomodulatory effects on the gut epithelium, evidenced by reduced NF-kB activity, and lower levels of the pro-inflammatory markers TNF-α, CXCL-10, and MCP-1. Furthermore, D + Q induced the secretion of anti-inflammatory cytokines, including IL-6 and IL-10. However, it should be noted that D + Q also led to the secretion of the pro-inflammatory cytokines IL-8. These findings suggest that D + Q could offer a novel therapeutic approach for advanced IBD management by modulating both the gut microbiome and inflammatory pathways. The results support the potential repurposing of senotherapeutic agents as a strategy for addressing the chronic inflammation central to IBD pathogenesis.
Collapse
Affiliation(s)
| | - Laura E McCoubrey
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK; Now at Drug Product Development, GSK R&D, Ware SG12 0GX, UK
| | - Atheer Awad
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK; Department of Clinical, Pharmaceutical and Biological Sciences, University of Hertfordshire, College Lane, Hatfield AL10 9AB, UK
| | | | | | | | | | | | - Simon Gaisford
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Abdul W Basit
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK.
| |
Collapse
|
37
|
Kaden T, Alonso‐Román R, Stallhofer J, Gresnigt MS, Hube B, Mosig AS. Leveraging Organ-on-Chip Models to Investigate Host-Microbiota Dynamics and Targeted Therapies for Inflammatory Bowel Disease. Adv Healthc Mater 2025; 14:e2402756. [PMID: 39491534 PMCID: PMC12004439 DOI: 10.1002/adhm.202402756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/29/2024] [Indexed: 11/05/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic gastrointestinal disease with drastically increasing incidence rates. Due to its multifactorial etiology, a precise investigation of the pathogenesis is extremely difficult. Although reductionist cell culture models and more complex disease models in animals have clarified the understanding of individual disease mechanisms and contributing factors of IBD in the past, it remains challenging to bridge research and clinical practice. Conventional 2D cell culture models cannot replicate complex host-microbiota interactions and stable long-term microbial culture. Further, extrapolating data from animal models to patients remains challenging due to genetic and environmental diversity leading to differences in immune responses. Human intestine organ-on-chip (OoC) models have emerged as an alternative in vitro model approach to investigate IBD. OoC models not only recapitulate the human intestinal microenvironment more accurately than 2D cultures yet may also be advantageous for the identification of important disease-driving factors and pharmacological interventions targets due to the possibility of emulating different complexities. The predispositions and biological hallmarks of IBD focusing on host-microbiota interactions at the intestinal mucosal barrier are elucidated here. Additionally, the potential of OoCs to explore microbiota-related therapies and personalized medicine for IBD treatment is discussed.
Collapse
Affiliation(s)
- Tim Kaden
- Dynamic42 GmbH07745JenaGermany
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
| | - Raquel Alonso‐Román
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | | | - Mark S. Gresnigt
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | - Bernhard Hube
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Institute of MicrobiologyFaculty of Biological SciencesFriedrich Schiller University07743JenaGermany
| | - Alexander S. Mosig
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
| |
Collapse
|
38
|
Xiang M, Wu S, Liu M, Zhang B, Xia X, Tan W, Xiang S. Iota-carrageenan oligosaccharide ameliorates DSS-induced colitis in mice by mediating gut microbiota dysbiosis and modulating SCFAs-PI3K-AKT pathway. Inflammopharmacology 2025:10.1007/s10787-025-01718-w. [PMID: 40167852 DOI: 10.1007/s10787-025-01718-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/04/2025] [Indexed: 04/02/2025]
Abstract
Iota-carrageenan oligosaccharides (iCOs), derived from marine red algae, are traditionally used as antithrombotic and anti-inflammatory agents in folk medicinal practice. Despite the prevailing emphasis on these aspects in their applications, the potential of iCOs as a prebiotic agent for gut health and its subsequent impact on intestinal disorders such as colitis remains largely unexplored. A DSS-induced colitis model was employed in C57BL/6 male mice to analyze the gut microbiota via 16S rRNA sequencing. Fecal microbiota transplantation (FMT) was used to assess the therapeutic effects of iCOs on colitis. RNA sequencing (RNA-Seq) identified pathways and genes affected by iCOs. ELISA measured inflammatory cytokines, while western blot and RT-qPCR evaluated protein and gene expressions, respectively. The iCOs increased beneficial bacteria, such as Lactobacillus, Bifidobacterium, and Akkermansia. They enhanced short-chain fatty acid production and upregulated GPR41, GPR43, and GPR109A mRNA, influencing cytokine secretion. The iCOs reduced mRNA of SPHK1, BDKRB1, LCN2, and so on, potentially through PI3K-Akt pathway inhibition, and promoted tight junction protein expression. Our findings highlight the novel therapeutic potential of iCOs in colitis, indicating a multifaceted approach to treatment that includes gut microbiota modulation, intestinal barrier restoration, and the suppression of inflammatory responses.
Collapse
Affiliation(s)
- Meixian Xiang
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, Hubei, People's Republic of China.
| | - Songtao Wu
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Hongshan District, No. 16, Huangjiahu West Road, Wuhan, 430065, People's Republic of China
| | - Minxin Liu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, Hubei, People's Republic of China
| | - Bin Zhang
- Department of Pharmacy, Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, People's Republic of China
| | - Xiankun Xia
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, Hubei, People's Republic of China
| | - Wenjing Tan
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, Hubei, People's Republic of China
| | - Shijian Xiang
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People's Republic of China.
| |
Collapse
|
39
|
Sun J, Geng L, Zhou D, Teng X, Chen M. Gut microbiota participates in polystyrene microplastics-induced defective implantation through impairing uterine receptivity. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 380:124997. [PMID: 40101486 DOI: 10.1016/j.jenvman.2025.124997] [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: 11/13/2024] [Revised: 02/22/2025] [Accepted: 03/13/2025] [Indexed: 03/20/2025]
Abstract
Microplastics (MPs) are widespread in global ecosystems and could pose risks to human health. However, crucial information on the impact of MP exposure on female reproductive health remains insufficient. In this study, we constructed an MP-exposure mice model through oral administration of polystyrene microplastics (PS-MPs) and found that it resulted in impaired uterine receptivity and defective implantation. An accumulation of plastic particles was detected in MP mice intestines. Metagenomic sequencing of feces samples indicated a structural and functional alteration of gut microbiota. Alistipes played a prominent role in MP biodegradation, while among the biodegradable functional genes, ACSL made the greatest contribution. Both had a significant increase in MP group, suggesting a potential occurrence of ferroptosis. Ferroptosis, a form of programmed cell death, is closely associated with uterine receptivity impairment and defective implantation. We detected MDA contents and ferroptosis-related proteins, and the results indicated the activation of ferroptosis in the process. Our research is the first to elucidate that exposure to MPs impairs uterine receptivity and results in deficient implantation, while also providing initial evidence that gut microbiota plays a critical role in this process.
Collapse
Affiliation(s)
- Jiani Sun
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Lulu Geng
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Dan Zhou
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiaoming Teng
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Miaoxin Chen
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
40
|
Zahedi E, Naseri FM, Zamani E, Nikbakhtzadeh M, Rastegar T, Sanaeirad A, Sadr SS. Ginger Extract Improves Cognitive Dysfunction via Modulation of Gut Microbiota-Derived Short-Chain Fatty Acids in D-Galactose/Ovariectomy-Induced Alzheimer-Like Disease. Mol Neurobiol 2025; 62:5095-5108. [PMID: 39505806 DOI: 10.1007/s12035-024-04583-w] [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: 03/29/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia with complex causes and limited treatment options. Recent research has suggested a connection between the progression of AD and the activity of gut microbiota. Ginger, a plant known for its anti-inflammatory, antioxidant, and neuroprotective properties, has gained attention as a potential treatment for alleviating AD symptoms. In this study, we induced an AD model in female rats through ovariectomy and D-galactose injection and then investigated the protective effects of oral administration of ginger ethanolic extract. We assessed changes in short-chain fatty acids (SCFAs), learning and memory abilities, neuroinflammatory markers in plasma, and the hippocampus, as well as histological changes in the intestine and hippocampus in sham-operated, diseased, and treatment groups. Oral administration of ginger ethanolic extract improved gut microbiota activity, increased SCFA levels, and enhanced the expression of tight junction proteins. Additionally, ginger extract reduced the concentrations of TNF-α and IL-1β in both plasma and the hippocampus. Furthermore, it significantly reduced cell death and amyloid plaque deposition in the hippocampal tissue. These physiological changes resulted in improved performance in learning and memory tasks in rats treated with ginger compared with the disease group. These findings provide compelling evidence for the beneficial effects of ginger on the gut-brain axis, leading to improvements in learning and memory through the reduction of neuroinflammation.
Collapse
Affiliation(s)
- Elham Zahedi
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Mokhtari Naseri
- Physiology Department and Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Zamani
- Department of Psychology, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Marjan Nikbakhtzadeh
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Rastegar
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashkan Sanaeirad
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Shahabeddin Sadr
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
41
|
Jia Y, Huang Q, Song R, Tang Y, Feng M, Lu J. Effects of fermented bamboo fiber on intestinal health and fecal pollutants in weaned piglets. Front Nutr 2025; 12:1538560. [PMID: 40236635 PMCID: PMC11998670 DOI: 10.3389/fnut.2025.1538560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/07/2025] [Indexed: 04/17/2025] Open
Abstract
Introduction Weaning stress adversely affects piglet growth and development, thereby reducing the economic efficiency of pig farming operations. Furthermore, pig feces are a major source of environmental pollution, underscoring the need for effective strategies to mitigate fecal output at its source. Methods This study investigated the effects of dietary supplementation with fermented bamboo fiber (FBF) on growth performance, intestinal barrier integrity, gut microbiota composition, and fecal pollutant levels in weaned piglets. A total of 144 Duroc × Landrace × Yorkshire piglets, weaned at 21 days of age, were randomly assigned to 4 groups, with six replicates per group and 6 piglets per replicate. The control group (CON) received a basal diet, while the three treatment groups were fed the basal diet supplemented with 1, 1.5, and 2% FBF, respectively. The trial lasted 30 days. Results The findings revealed that FBF supplementation fortified the intestinal barrier, modulated colonic microbial communities, and decreased fecal pollutant levels. Among the treatment groups, supplementation with 1.5% FBF produced the most significant improvements in piglets' growth performance and intestinal barrier function, as well as the strongest microbial interactions and the greatest reduction in fecal pollutants. Discussion These results suggest that FBF supplementation can alleviate weaning stress and mitigate the environmental impact of pig feces, with 1.5% identified as the optimal supplementation level.
Collapse
Affiliation(s)
- Yubiao Jia
- Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Qiuming Huang
- Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Rui Song
- Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Yanling Tang
- Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Mengxin Feng
- Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Jianjun Lu
- Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
42
|
Lun J, Liu M, Zhang W, Huang G, Ma M, Jin W, Zhu Y, Qu Q, Lv W, Guo S. Influence of purslane extract on immuno-antioxidant status, intestinal barrier, and microbiota of chicks after experimental infection with Escherichia coli O78. Poult Sci 2025; 104:105106. [PMID: 40245541 DOI: 10.1016/j.psj.2025.105106] [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: 01/27/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
This study focused on examining how Portulaca oleracea L. extract (POLE) influences production performance, antioxidant capacity, immunity, anti-apoptotic ability, and intestinal health in chicks exposed to Escherichia coli O78(E. coli). A 2 × 2 factorial design was used to randomly divide male chicks into 4 groups: (1) basal diet (CON group), (2) basal diet supplemented with 0.5 g/kg POL (CPOL group), (3) basal diet with E. coli challenge (ECON group), and (4) basal diet supplemented with 0.5 g/kg POL as well as E. coli challenge (EPOL group).The experiment lasted for 19 days. The POL supplementation was found to decrease the feed conversion ratio and liver and spleen indexes and increase the average daily gain and bursa indexes (P<0.05). Moreover, in chicks infected with E. coli, POL reduced the levels of serum DAO and caspase 8 (P<0.05). POL notably elevated the serum levels of immunoglobulins and IL-10, an anti-inflammatory cytokine, which also decreased the levels of proinflammatory cytokines TNF-α and IFN-γ. At the same time, dietary POL enhanced the mRNA expression of genes related to antioxidation and reduced the expression of genes associated with proinflammatory cytokines and apoptosis in the liver, spleen, jejunum, and ileum. In addition, dietary POL repaired the intestinal barrier and injury. More importantly, by boosting the abundance of beneficial bacteria such as Butyricicoccus and Blautia, and reducing harmful bacteria like Enterococcus and Escherichia, POL induced changes in the gut microbiota composition. Spearman's correlation study revealed that Dorea and Butyricoccus had a strong positive link with intestinal barrier function and antioxidant capacity, while Escherichia had a substantial positive correlation with pro-inflammatory cytokines. In summary, this study suggests adding POL to the diet as a beneficial supplement to enhance chick performance.
Collapse
Affiliation(s)
- Jianchi Lun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, PRC
| | - Mengjie Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, PRC; School of Animal Science and Technology, Foshan University, Foshan, PRC
| | - Wenbo Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, PRC
| | - Gengxiong Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, PRC
| | - Ming Ma
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, PRC
| | - Wenxin Jin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, PRC
| | - Yongqi Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, PRC
| | - Qian Qu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, PRC
| | - Weijie Lv
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, PRC; Guangdong Technology Research Center for Traditional Chinese Veterinary Medicine and Natural Medicine, Guangzhou, PRC
| | - Shining Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, PRC; Guangdong Technology Research Center for Traditional Chinese Veterinary Medicine and Natural Medicine, Guangzhou, PRC.
| |
Collapse
|
43
|
Huang F, Yang N, Zhang Q, Luo C, Wang J, Yang Y, Yue B, Chen P, Zhang X. Marine-Derived Enterococcus faecalis HY0110 as a Next-Generation Functional Food Probiotic: Comprehensive In Vitro and In Vivo Bioactivity Evaluation and Synergistic Fermentation of Periplaneta americana Extract Powder. Foods 2025; 14:1181. [PMID: 40238337 PMCID: PMC11988638 DOI: 10.3390/foods14071181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Addressing the escalating global burdens of inflammatory bowel disease and antimicrobial resistance demanded innovative food-based approaches to fortify gut health and suppress pathogens. We introduced a novel edible probiotic, Enterococcus faecalis HY0110, isolated from marine Thunnus thynnus. Through comprehensive in vitro, in vivo, and metabolomic analyses, we demonstrated its superior antibacterial effects compared to Lactobacillus rhamnosus GG, along with significantly enhanced antioxidant and free-radical scavenging capacities. Notably, elevated acetic acid production strongly correlated with its antimicrobial efficacy (R ≥ 0.999). HY0110 also exerted antiproliferative effects on HT-29 colorectal cancer cells by attenuating β-catenin and BCL-2 expression while upregulating pro-apoptotic markers P62 and c-PARP. In a DSS-induced colitis model, HY0110 alleviated inflammation, restored gut microbial homeostasis, and enhanced deterministic processes in community assembly dynamics. Furthermore, fermenting Periplaneta americana powder with HY0110 triggered extensive metabolic remodeling, notably a 668.73-fold rise in astragaloside A, plus increases in L-Leucyl-L-Alanine, S-lactoylglutathione, and 16,16-dimethyl prostaglandin A1. These shifts diminished harmful components and amplified essential amino acids and peptides to bolster immune modulation, redox balance, and anti-inflammatory responses. This work established a transformative paradigm for utilizing marine probiotics and novel entomological substrates in functional foods, presenting strategic pathways for precision nutrition and inflammatory disease management.
Collapse
Affiliation(s)
- Feiyun Huang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Nan Yang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610041, China;
| | - Qingqing Zhang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Cuiling Luo
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Jingheng Wang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Yu Yang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Bisong Yue
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Peng Chen
- MOE Key Laboratory of Deep Earth Science and Engineering, College of Architecture and Environment, Sichuan University, Chengdu 610065, China
| | - Xiuyue Zhang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu 610065, China
| |
Collapse
|
44
|
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 2025:1-14. [PMID: 40134274 DOI: 10.1080/14712598.2025.2484303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
45
|
Jayasinghe T, Jenkins J, Medara N, Choowong P, Dharmarathne G, Kong F, Cho H, Kim SH, Zhang Y, Franco-Duarte R, Eberhard J, Spahr A. Dietary Fibre Modulates Body Composition, Blood Glucose, Inflammation, Microbiome, and Metabolome in a Murine Model of Periodontitis. Nutrients 2025; 17:1146. [PMID: 40218904 PMCID: PMC11990244 DOI: 10.3390/nu17071146] [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: 02/24/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Dietary fibre plays a crucial role in metabolic regulation, inflammation, and microbiome composition. However, its impact on systemic and oral health, particularly in periodontitis, remains unclear. This study investigated the effects of high- and low-fibre diets on body composition, glycaemic control, inflammation, microbiome, and metabolome in a murine model of experimental periodontitis. Methods: Thirty-six male C57BL/6 mice were randomised to a high-fibre (40% fibre) or low-fibre (5% fibre) diet for eight weeks. Body weight, fat mass, lean mass, fasting blood glucose, serum inflammatory markers, alveolar bone loss, and root length were assessed. Oral and faecal microbiome composition was analysed using 16S rRNA sequencing. Metabolomic and short-chain fatty acid (SCFA) profiling was conducted using liquid chromatography-mass spectrometry (LC-MS). Results: Mice on the high-fibre diet exhibited significantly lower body weight (p < 0.0001), fat mass (p = 0.0007), and lean mass (p < 0.0001) compared to the low-fibre group. Fasting blood glucose levels were significantly lower in the high-fibre group (p = 0.0013). TNF-α and IFN-γ levels were significantly elevated in the low-fibre group (p < 0.0001), suggesting a heightened pro-inflammatory state. While alveolar bone loss and root length did not differ significantly, microbiome analysis revealed distinct bacterial compositions (PERMANOVA, p < 0.05), with fibre-fermenting taxa enriched in high-fibre-fed mice. Metabolomic analysis identified 19 significantly altered metabolites, indicating dietary adaptations. Conclusions: A high-fibre diet improves glycaemic control, reduces systemic inflammation, and alters microbial and metabolic profiles in experimental periodontitis. These findings highlight dietary fibre's role in modulating metabolic and inflammatory pathways relevant to periodontal and systemic diseases.
Collapse
Affiliation(s)
- Thilini Jayasinghe
- The Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (J.J.); (P.C.); (J.E.)
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Josie Jenkins
- The Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (J.J.); (P.C.); (J.E.)
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Nidhi Medara
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Phannaphat Choowong
- The Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (J.J.); (P.C.); (J.E.)
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Gangani Dharmarathne
- Australian Laboratory Services Global, Water and Hydrographic, Hume, ACT 2620, Australia;
| | - Fay Kong
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Hanna Cho
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Se Hun Kim
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Yuchen Zhang
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Ricardo Franco-Duarte
- Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, 4710-057 Braga, Portugal;
| | - Joerg Eberhard
- The Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (J.J.); (P.C.); (J.E.)
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Axel Spahr
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| |
Collapse
|
46
|
Lv J, Kong X, Liu W, Su Z, Luo F, Suo F, Wang Z, Cao L, Liu Z, Li M, Xiao W. Rhodiola crenulata polysaccharide alleviates dextran sulfate sodium-induced ulcerative colitis in mice by repairing the intestinal barrier and regulating the intestinal microecology. Front Pharmacol 2025; 16:1519038. [PMID: 40206066 PMCID: PMC11979201 DOI: 10.3389/fphar.2025.1519038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Polysaccharides, vital biological macromolecules ubiquitous in organisms, have garnered attention as potential therapeutic candidates for ulcerative colitis (UC). However, the therapeutic potential of Rhodiola crenulata polysaccharides (RCP) in UC remains largely unexplored. The RCP was prepared by boiling water extraction, 80% alcohol precipitation, membrane separation, and D101 macroporous resin purification. The monosaccharide composition of RCP (Mw = 67.848 kDa) includes mannose, rhamnose, glucuronic acid, galacturonic acid, glucose, galactose, and arabinose, with a molar ratio of 0.22:1:0.07:7.03:2.88:0.64:4.12. In vivo experiments have shown that RCP can improve DSS induced weight loss in UC mice, decrease disease activity index (DAI), alleviate histopathological changes in colon tissue, and suppress the levels of pro-inflammatory cytokine IL-6 and MPO activity. Immunohistochemical results showed that essential tight junction proteins such as occludin, claudin1, and ZO-1 were upregulated, improving the integrity of the intestinal barrier. Importantly, RCP regulated the abundance of the intestinal microbiota by reducing the Firmicutes-to-Bacteroidetes ratio (F/B), increasing beneficial bacteria such as Muribaculaceae and Bifidobacterium, decreasing harmful bacteria including Erysipelotrichaceae, Faecalibaculum, Lachnospiraceae_unclassified, Parabacteroides, and Ruminiclostridium_9. Additionally, it enhanced the restoration of acetic acid, propionic acid, isovaleric acid, and valeric acid to maintain intestinal SCFA levels, thereby restoring the intestinal microecology. Therefore, RCP has excellent therapeutic effects on UC and is worthy of further drug development and clinical treatment.
Collapse
Affiliation(s)
- Jia Lv
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyu Kong
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Wenjun Liu
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Zhenzhen Su
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Fengshou Luo
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Fengtai Suo
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Zhenzhong Wang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Liang Cao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mengxuan Li
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Wei Xiao
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| |
Collapse
|
47
|
Zhra M, Elahi MA, Tariq A, Abu-Zaid A, Yaqinuddin A. Sirtuins and Gut Microbiota: Dynamics in Health and a Journey from Metabolic Dysfunction to Hepatocellular Carcinoma. Cells 2025; 14:466. [PMID: 40136715 PMCID: PMC11941559 DOI: 10.3390/cells14060466] [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: 01/27/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction leading to non-alcoholic fatty liver disease (NAFLD) exhibits distinct molecular and immune signatures that are influenced by factors like gut microbiota. The gut microbiome interacts with the liver via a bidirectional relationship with the gut-liver axis. Microbial metabolites, sirtuins, and immune responses are pivotal in different metabolic diseases. This extensive review explores the complex and multifaceted interrelationship between sirtuins and gut microbiota, highlighting their importance in health and disease, particularly metabolic dysfunction and hepatocellular carcinoma (HCC). Sirtuins (SIRTs), classified as a group of NAD+-dependent deacetylases, serve as crucial modulators of a wide spectrum of cellular functions, including metabolic pathways, the inflammatory response, and the process of senescence. Their subcellular localization and diverse functions link them to various health conditions, including NAFLD and cancer. Concurrently, the gut microbiota, comprising diverse microorganisms, significantly influences host metabolism and immune responses. Recent findings indicate that sirtuins modulate gut microbiota composition and function, while the microbiota can affect sirtuin activity. This bidirectional relationship is particularly relevant in metabolic disorders, where dysbiosis contributes to disease progression. The review highlights recent findings on the roles of specific sirtuins in maintaining gut health and their implications in metabolic dysfunction and HCC development. Understanding these interactions offers potential therapeutic avenues for managing diseases linked to metabolic dysregulation and liver pathology.
Collapse
Affiliation(s)
- Mahmoud Zhra
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Muhammad Affan Elahi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.A.E.); (A.A.-Z.)
| | - Aamira Tariq
- Department of Biosciences, COMSATS University Islamabad, Islamabad Campus, Islamabad 45550, Pakistan
| | - Ahmed Abu-Zaid
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.A.E.); (A.A.-Z.)
| | - Ahmed Yaqinuddin
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| |
Collapse
|
48
|
Wu Y, Huang X, Li Q, Yang C, Huang X, Du H, Situ B, Zheng L, Ou Z. Reducing severity of inflammatory bowel disease through colonization of Lactiplantibacillus plantarum and its extracellular vesicles release. J Nanobiotechnology 2025; 23:227. [PMID: 40114208 PMCID: PMC11924789 DOI: 10.1186/s12951-025-03280-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by compromised intestinal barrier function and a lack of effective treatments. Probiotics have shown promise in managing IBD due to their ability to modulate the gut microbiota, enhance intestinal barrier function, and exert anti-inflammatory effects. However, the specific mechanisms through which probiotics exert these therapeutic effects in IBD treatment remain poorly understood. Our research revealed a significant reduction of Lactiplantibacillus plantarum (L. plantarum) in the gut microbiota of IBD patients. L. plantarum is a well-known probiotic strain in the list of edible probiotics, recognized for its beneficial effects on gut health, including its ability to strengthen the intestinal barrier and reduce inflammation. We demonstrated that supplementation with L. plantarum could alleviate IBD symptoms in mice, primarily by inhibiting apoptosis in intestinal epithelial cells through L. plantarum's bacterial extracellular vesicles (L. plant-EVs). This protective effect is dependent on the efficient uptake of L. plant-EVs by intestinal cells. Intriguingly, watermelon enhances L. plantarum colonization and L. plant-EVs release, further promoting intestinal barrier repair. Our findings contribute to the understanding of L. plant-EVs in the probiotic-based therapeutic approach for IBD, as they are promising candidates for nanoparticle-based therapeutic methods that are enhanced by natural diets such as watermelon. This study thereby offers a potential breakthrough in the management and treatment of IBD.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinyue Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qianbei Li
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chaoqun Yang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xixin Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hualongyue Du
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bo Situ
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Zihao Ou
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
49
|
Ge Y, Yang H, Fu Y, Zhou J, Cheng Z, Fan X, Yu Y. A Mendelian randomization study to reveal gut-disc axis: causal associations between gut microbiota with intervertebral disc diseases. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2025:10.1007/s00586-025-08795-z. [PMID: 40105993 DOI: 10.1007/s00586-025-08795-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/15/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE Emerging evidence suggests a link between gut microbiota and intervertebral disc diseases (IDDs); however, the causal relationships remain unclear. This study aimed to evaluate the causal effects of gut microbiota on the risk of cervical disc disorders (CDD), other intervertebral disc disorders (OIDD), pyogenic intervertebral disc infections, and discitis, shedding light on the potential "gut-disc axis". METHODS Genetic variation data for 202 gut microbiota taxa were obtained from the Dutch Microbiome Project, and disease outcome data were sourced from the FinnGen consortium. A Mendelian Randomization (MR) approach was employed to assess causal relationships, using genetic variants as instrumental variables. Sensitivity analyses, including tests for pleiotropy, heterogeneity, and reverse causation, ensured robust findings. RESULTS The study identified 20 gut microbial taxa with significant associations to IDDs. Notably, taxa within the Erysipelotrichaceae family showed consistent protective effects against OIDD after Bonferroni correction (P < 0.05). Associations between several species and specific diseases, such as Alistipes senegalensis with CDD and Ruminococcus lactaris with discitis, were also observed. Sensitivity analyses confirmed no evidence of confounding or reverse causation. CONCLUSION This study provides evidence of causal relationships between specific gut microbiota and IDDs, supporting the existence of a "gut-disc axis." The findings suggest that microbial dysbiosis may influence spinal health through systemic inflammation and immune regulation. These insights open new possibilities for microbiota-targeted interventions, such as probiotics or dietary modifications, to prevent or manage IDDs. However, further research is required to validate these therapeutic strategies.
Collapse
Affiliation(s)
- Yuanxin Ge
- Sichuan Provincial Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Huifang Yang
- Sichuan Provincial Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Yang Fu
- Department of Rehabilitation, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Jie Zhou
- Department of Rehabilitation, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Zilin Cheng
- Sichuan Provincial Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohong Fan
- Sichuan Provincial Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Yang Yu
- Sichuan Provincial Hospital of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
50
|
Zhu D, Li S, Xu Z, Kulyar MF, Bai X, Wang Y, Wang B, Khateeb E, Deng D, Wang L, Chen Y, Guo A, Shen Y. Comparative analysis of gut microbiota in healthy and diarrheic foals. Microbiol Spectr 2025:e0087124. [PMID: 40105330 DOI: 10.1128/spectrum.00871-24] [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: 04/05/2024] [Accepted: 02/01/2025] [Indexed: 03/20/2025] Open
Abstract
Diarrhea presents a substantial risk of high morbidity and mortality among foals. Although studies have shown connections between gut microbiota and several gastrointestinal diseases, there is still inadequate information on gut microbial alterations in foals during diarrhea. In this study, we conducted 16S rRNA and ITS gene amplicon sequencing to investigate gut bacterial and fungal differences between healthy and diarrheic foals. The results unveiled significant reductions in gut bacterial and fungal diversities among foals experiencing diarrhea, accompanied by notable shifts in the composition of gut microbial communities. A considerable decrease was observed in the relative abundance of 30 bacterial and 34 fungal genera. Moreover, two bacterial and eight fungal genera were utterly undetectable in the gut microbiota of diarrheic foals. Some decreased genera, such as Bifidobacterium and Saccharomyces, were deemed beneficial and recognized as probiotics. The study revealed significant alterations in foals' gut bacterial and fungal communities during diarrhea, which enriched our comprehension of gut microbial dynamics in foals across varying health statuses. These findings offer valuable insights for managing diarrhea through gut microbiota modulation, suggesting that probiotics may be superior to antibiotics in preventing and controlling foal diarrhea.IMPORTANCEThis research advances the understanding of gut bacterial and fungal dynamics in foals, highlighting gut microbiota dysbiosis as a potential contributor to foal diarrhea. Additionally, we observed that many altered bacteria and fungi were downregulated during diarrhea, including some probiotic strains. Consequently, our findings provide evidence that probiotics may offer superior efficacy compared with antibiotics as potential candidates for preventing and treating foal diarrhea.
Collapse
Affiliation(s)
- Di Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Siyu Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhixiang Xu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Md F Kulyar
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xu Bai
- China Horse Industry Association, Beijing, China
| | - Yu Wang
- China Horse Industry Association, Beijing, China
| | - Boya Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Emaan Khateeb
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Dandan Deng
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lidan Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yuji Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Aizhen Guo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yaoqin Shen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|