1
|
Shin S, Chen S, Xie K, Duhun SA, Ortiz-Cerda T. Evaluating the anti-inflammatory and antioxidant efficacy of complementary and alternative medicines (CAM) used for management of inflammatory bowel disease: a comprehensive review. Redox Rep 2025; 30:2471737. [PMID: 40056427 PMCID: PMC11892051 DOI: 10.1080/13510002.2025.2471737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic autoimmune condition whose pathogenesis has not been fully elucidated, and current treatments are not definitive and often carry several side effects. The Complementary and Alternative Medicine (CAM) offers a new approach to conventional medicine. However, their clinical application and mechanisms remain limited.Objective: The aim of this review is to evaluate the anti-inflammatory, impact on microbiota and antioxidant efficacy of currently available CAM for IBD.Methods: The literature collection was obtained from Google Scholar, MEDLINE, PubMed and Web of Science (WOS). Studies in both human and animal models, published in English language between 2018 and 2024, were selected. Sixty-seven studies were included in the current review after inclusion and exclusion screening processes.Results: Mostly, studies showed significant anti-inflammatory, gut microbiota restoring, antioxidant effects of polyphenols, polysaccharides, emodin, short-chain fatty acids (SCFA; including butyrate, propionate and acetate), and probiotics although some contrasting results were noted. Current evidence shows that polyphenols exhibit the most consistent result in alleviating IBD pathophysiology, primarily due to their significant SCFA-elevating effect.Discussion: Future studies may focus on human studies, narrowing down on individual factors which may change natural product's metabolism. Further research studies are also essential to obtain therapeutic recommendations.
Collapse
Affiliation(s)
- Sia Shin
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Siqi Chen
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Kangzhe Xie
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Suehad Abou Duhun
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Tamara Ortiz-Cerda
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Departamento de Citología e Histología Normal y Patológica, Facultad de medicina, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
2
|
Liu Z, Yuan J, Din MA, Tian Y, Mao F. HucMSC-Ex alleviates inflammatory bowel disease by regulating O-GlcNAcylation modification of RACK1 in intestinal epithelial cells. Colloids Surf B Biointerfaces 2025; 251:114606. [PMID: 40068238 DOI: 10.1016/j.colsurfb.2025.114606] [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: 12/28/2024] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 04/15/2025]
Abstract
Inflammatory Bowel Disease (IBD) is a chronic autoimmune disorder that severely affects the gastrointestinal tract and is difficult to cure. This study explored the mechanism by which human umbilical cord mesenchymal stem cell-derived exosomes (HucMSC-Ex) alleviate IBD through O-GlcNAc glycosylation modification and the expression of related proteins. The study analyzed the effects of HucMSC-Ex on the inhibition of pro-inflammatory factors and promotion of intestinal epithelial cells regeneration in vitro and in vivo, with a focus on the role of the O-GlcNAc glycosylation of the RACK1 protein. The findings indicated that HucMSC-Ex reverses epithelial-mesenchymal transition (EMT) by upregulating O-GlcNAc glycosylation levels and effectively alleviates IBD symptoms and inflammatory responses in mouse intestinal epithelial cells. By modulating O-GlcNAc glycosylation, HucMSC-Ex exhibits significant therapeutic potential in immune regulation and gut microbiota remodeling, offering new perspectives for IBD treatment.
Collapse
Affiliation(s)
- Ziyue Liu
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu 212002, PR China
| | - Jintao Yuan
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu 212300, PR China
| | - Muhammad AzharUd Din
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu 212002, PR China
| | - Yiqing Tian
- Department of Clinical Laboratory, Xuzhou Central Hospital, Xuzhou Institute of Medical Sciences, Xuzhou, Jiangsu 221000, PR China.
| | - Fei Mao
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu 212002, PR China.
| |
Collapse
|
3
|
Song JY, Hui X, Feng R, Zhao Y, Hu JC, Jin JY, Lu JY, Xu H, Wang JY, Zuo HT, Ye ML, Wang Y. Analysis of intestinal bacterial carboxylesterase-mediated metabolites and the potential antitumour molecular mechanism of angoroside C. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025; 27:892-909. [PMID: 39792153 DOI: 10.1080/10286020.2024.2441823] [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: 11/04/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025]
Abstract
Angoroside C (AgrC) is a compound with many pharmacological properties. However, its antitumour potential has not been well studied. The low bioavailability of AgrC suggests a strong link to gut bacteria. Therefore, we identified and quantified four AgrC metabolites in gut microbiota. Molecular docking and inhibitor-based experiments demonstrated that carboxylesterase played a key role in AgrC metabolism. Both AgrC and its metabolites inhibited the viability of CT-26 cells, and potential antitumour targets were further explored. Additionally, AgrC significantly increased the levels of propionic, butyric, valeric and isovaleric acids. This provides a new insight for the antitumour effects of AgrC.
Collapse
Affiliation(s)
- Jian-Ye Song
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100050, China
| | - Xiang Hui
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100050, China
| | - Ru Feng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100050, China
| | - Yi Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100050, China
| | - Jia-Chun Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100050, China
| | - Jing-Yu Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100050, China
| | - Jin-Yue Lu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100050, China
| | - Hui Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100050, China
| | - Jing-Yue Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100050, China
| | - Heng-Tong Zuo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100050, China
| | - Meng-Liang Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100050, China
| | - Yan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
4
|
Zheng Z, Gao J, Ma Y, Hou X. Cellular and Molecular Mechanisms of Phytochemicals Against Inflammation-Associated Diseases and Viral Infection. Cell Biol Int 2025; 49:606-633. [PMID: 40091269 DOI: 10.1002/cbin.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025]
Abstract
Inflammation-associated diseases have become widespread and pose a significant threat to human health, and the therapeutic methods for diverse diseases are inadequate due to the undesirable effects of synthetic ingredients. Recently, more and more evidence indicated that phytochemicals, plant secondary metabolites, have numerous therapeutic functions against human diseases via affecting a variety of mechanisms with their distinct advantages of high efficiency and low toxicity. Here, we highlight the mechanisms of phytochemicals to hinder inflammation-associated diseases (including Inflammatory diseases, cardiovascular diseases, metabolic syndrome, neurological disorders, skin diseases, respiratory diseases, kidney diseases, gastrointestinal diseases, retinal diseases, viral infections) by regulating the crosstalk among various signal cascades (including MicroRNAs, SIRT1, DNMTs, NF-κB, NLRP3, TGF-β, the Gasdermin-mediated pyroptosis pathway), which can be considered as a novel and potential therapeutic strategy. Furthermore, phytochemicals could prevent virus infection by disturbing different targets in the virus replication cycle. However, natural plants have shown limited bioavailability due to their low water solubility, the use of adjuvants such as liposomal phytochemicals, phytochemical nanoparticles and phytochemicals-phospholipid complex promote their bioavailability to exhibit beneficial effects against various diseases. The purpose of this review is to explore the molecular mechanisms and promising applications of phytochemicals in the fields of inflammation-associated diseases and virus infection to provide some direction.
Collapse
Affiliation(s)
- Zhaodi Zheng
- College of Medical Imaging and Laboratory, Jining Medical University, Jining, China
| | - Junying Gao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Yubing Ma
- College of Medical Imaging and Laboratory, Jining Medical University, Jining, China
| | - Xitan Hou
- College of Medical Imaging and Laboratory, Jining Medical University, Jining, China
| |
Collapse
|
5
|
Wang H, Yang Y, Li T, Chang S, Zhu Y, Liu C. Drinking Water Temperature Impacts the Pathogenesis of DSS-Induced Ulcerative Colitis by Regulating Intestinal Barrier Function and Remodeling the Gut Microbiota Composition. FASEB J 2025; 39:e70645. [PMID: 40377203 DOI: 10.1096/fj.202500062r] [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: 01/07/2025] [Revised: 04/18/2025] [Accepted: 05/07/2025] [Indexed: 05/18/2025]
Abstract
Environmental factors, including poor dietary habits and unhealthy drinking patterns, contribute to ulcerative colitis (UC). While the relationship between diet-related malnutrition and UC has been extensively explored, the impact of drinking water temperature remains largely overlooked, prompting us to investigate its influence on UC pathogenesis and explore the underlying mechanisms. In the present study, we observed that, unlike external thermal and cold therapy, varying drinking water temperatures transiently altered the internal body temperature of the digestive tract. Specifically, chronic drinking of 0°C water had significant anti-inflammatory effects and preserved the integrity of the mucosal barrier in a colitis mouse model. Mechanistically, this temperature spectrum changed the composition of the gut microbiota from inflammation-prone (25°C drinking water) to a resting pattern similar to that of the negative control. Specifically, the abundances of Blautia and Parasutterella, two beneficial genera, were strongly increased in response to 0°C water, accompanied by elevated levels of short-chain fatty acids. In contrast, drinking 40°C water had opposite effects on all the examined parameters and generally aggravated the development of colitis. This study is the first to demonstrate how modifying the temperature of habitual drinking water can modulate colitis progression, providing a novel and noninvasive approach to UC management. Specifically, chronic consumption of 0°C water alleviated the severity of colitis, whereas 40°C water aggravated the disease. Therefore, by focusing on commonly consumed drinking water temperatures, our findings suggest that this simple intervention could be a safe, convenient, and effective therapeutic strategy.
Collapse
Affiliation(s)
- Huiting Wang
- Department of Endocrinology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Provincial University Key Laboratory of Drug Discovery for Metabolic Inflammatory Diseases, China Pharmaceutical University, Nanjing, China
| | - Yiheng Yang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianyu Li
- Department of Endocrinology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Provincial University Key Laboratory of Drug Discovery for Metabolic Inflammatory Diseases, China Pharmaceutical University, Nanjing, China
| | - Shengyu Chang
- Department of Endocrinology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Provincial University Key Laboratory of Drug Discovery for Metabolic Inflammatory Diseases, China Pharmaceutical University, Nanjing, China
| | - Yao Zhu
- Department of Endocrinology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Provincial University Key Laboratory of Drug Discovery for Metabolic Inflammatory Diseases, China Pharmaceutical University, Nanjing, China
| | - Chang Liu
- Department of Endocrinology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Provincial University Key Laboratory of Drug Discovery for Metabolic Inflammatory Diseases, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
6
|
Ming S, Ou J, Huang Y, Tang Z, Qin Y, Ma H, Gan S, Li Z. Liupao tea aqueous extract alleviates dextran sulfate sodium-induced ulcerative colitis in rats by modulating the gut microbiota. Open Life Sci 2025; 20:20251106. [PMID: 40417008 PMCID: PMC12103186 DOI: 10.1515/biol-2025-1106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/13/2025] [Accepted: 03/26/2025] [Indexed: 05/27/2025] Open
Abstract
Liupao tea is known for its anti-inflammatory antioxidant and regulation of gut microecological balance properties. This study aims to investigate the therapeutic effects of Liupao tea aqueous extract (LPTAE) on ulcerative colitis (UC) induced by dextran sulfate sodium (DSS) in rats. The rats were randomly divided into five groups: the Normal group, the DSS group, the LPTL group, the LPTM group, and the LPTH group. Throughout the experiment, the rats' activity levels, stool consistency, and body weights were observed and recorded daily. After the experiment, colon length was measured, and colon tissues were collected for pathological analysis. Additionally, the colon contents were analyzed for gut microbiota composition and short-chain fatty acid (SCFA) level, while serum samples were collected to determine inflammatory and oxidative factors. The results indicated that treatment with low, medium, and high doses of LPTAE significantly inhibited weight loss, alleviated rectal bleeding, and reduced colon shortening compared to the DSS group. It also decreased the disease activity index and histopathological activity index scores in the rats. Furthermore, LPTAE reduced the levels of inflammatory cytokines such as IL-1β, IL-6, TNF-α, and malondialdehyde, while simultaneously increasing the levels of superoxide dismutase and SCFAs, including acetic acid, propionic acid, and butyric acid. 16S rDNA gene sequencing of the gut microbiota revealed that all doses of LPTAE reversed the decrease in both α and β diversities caused by UC, increased the relative abundance of beneficial bacteria such as Lactobacillus, Muribaculaceae, Alloprevotella, and Blautia, and decreased the levels of harmful bacteria such as Prevotella, Romboutsia, and Bacteroides. In summary, within the tested doses (100, 150, 250 mg/kg), LPTAE alleviated DSS-induced colitis by modulating the gut microbiota and correcting the metabolic imbalance of SCFAs.
Collapse
Affiliation(s)
- Shengjin Ming
- Department of Clinical Laboratory, Wuzhou Gongren Hospital (The Seventh Affiliated Hospital of Guangxi Medical University), 1 Gaodi Road, Wuzhou, 543001, China
| | - Jinxi Ou
- Department of Gastroenterology, Wuzhou Gongren Hospital (The Seventh Affiliated Hospital of Guangxi Medical University), 1 Gaodi Road, Wuzhou, 543001, China
| | - Ying Huang
- Department of Clinical Laboratory, Wuzhou Gongren Hospital (The Seventh Affiliated Hospital of Guangxi Medical University), 1 Gaodi Road, Wuzhou, 543001, China
| | - Zhongqing Tang
- Department of Pathology, Wuzhou Gongren Hospital (The Seventh Affiliated Hospital of Guangxi Medical University), 1 Gaodi Road, Wuzhou, 543001, China
| | - Yuechao Qin
- Department of Clinical Laboratory, Wuzhou Gongren Hospital (The Seventh Affiliated Hospital of Guangxi Medical University), 1 Gaodi Road, Wuzhou, 543001, China
| | - Hongxi Ma
- Department of Clinical Laboratory, Wuzhou Gongren Hospital (The Seventh Affiliated Hospital of Guangxi Medical University), 1 Gaodi Road, Wuzhou, 543001, China
| | - Siling Gan
- Department of Gastroenterology, Wuzhou Gongren Hospital (The Seventh Affiliated Hospital of Guangxi Medical University), 1 Gaodi Road, Wuzhou, 543001, China
| | - Zhongxia Li
- Department of Pediatrics, Wuzhou Gongren Hospital (The Seventh Affiliated Hospital of Guangxi Medical University), 1 Gaodi Road, Wuzhou, Guangxi, 543001, China
| |
Collapse
|
7
|
Xu Z, Zhao L, Wu M, Cui A, Chen W, Zheng G, Zhou J, Gao D, Shi R. Prophylactic administration of overproducing-abscisic acid Bacillus licheniformis attenuated DSS-induced colitis in mice by regulating the gut microbiota and immune activity. BMC Microbiol 2025; 25:306. [PMID: 40389822 PMCID: PMC12087060 DOI: 10.1186/s12866-025-03988-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 04/23/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) involves the complex interplay among the mucosal barrier, microbiota, immunity and genetic factors. There are currently no satisfactory treatments for IBD. Administration of the probiotic Bacillus licheniformis (Bl) can improves colitis by regulating the gut microbiota. The phytohormone abscisic acid (ABA) treatment has favorable effects on immunity, as well as on inflammatory diseases like colitis. We hypothesized that the expression of an additional cyp gene by the Bl to increase its ABA production would enhance its effects. RESULTS In this study, we found that a Bl-cyp strain overexpressing the cyp gene secreted more ABA into its supernatant than either the parental Bl stain or a Bl-pET82a strain expressing only a vector pET82a when these bacteria were grown in Nfb medium for 48 h. The prophylactic administration of the Bl-cyp strain culture more effectively attenuated dextran sodium sulfate (DSS)-induced colitis in mice compared to the Bl and Bl-pET28a strains. These findings were associated with significantly reduced epithelial barrier damage, as well as increased number of goblet cells and expression levels of occludin gene in the colonic epithelial layer, and decreased serum LPS levels in the Bl-cyp group. In addition, the administration of Bl-cyp strain effectively regulated the disordered gut microbiota by improving their diversity, richness and compositions more than the Bl or Bl-pET82a strain, including the ratio of Bacteroidota: Bacillota. It also inhibited the excessive growth of opportunistic pathogen Escherichia just like the Bl or Bl-pET82a strain. Moreover, the preventive administration of the Bl-cyp strain to mice following DSS-induced colitis enhanced the proportion of Treg cells and suppressed the proportion of Th17 cells in mesenteric lymph nodes (MLNs), decreased the levels of pro-inflammatory cytokines TNF-α, IL-6, and IL-22, and increased the level of anti-inflammatory IL-10 in colon tissues, similar to treatment with a high concentration of the ABA standard (ABA-H). Notably, the treatment with the Bl-cyp strain more effectively regulated the disordered microbiota than the ABA-H. CONCLUSIONS The administration of the Bl-cyp strain may provide a novel preventive approach for IBD, and may exert its effects by modulating the gut microbiota and host's immune status.
Collapse
Affiliation(s)
- Zeyan Xu
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Lijiang Zhao
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Mengting Wu
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Anqi Cui
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Wei Chen
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Guohao Zheng
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Jingyi Zhou
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Daqing Gao
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China.
| | - Ruihua Shi
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China.
- Department of Gastroenterology, Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|
8
|
Han J, Meng X, Kong H, Li X, Chen P, Zhang XA. Links between short-chain fatty acids and osteoarthritis from pathology to clinic via gut-joint axis. Stem Cell Res Ther 2025; 16:251. [PMID: 40390010 PMCID: PMC12090658 DOI: 10.1186/s13287-025-04386-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 05/09/2025] [Indexed: 05/21/2025] Open
Abstract
Short-chain fatty acids (SCFAs), the primary metabolites produced by the microbial fermentation of dietary fibers in the gut, have a key role in protecting gut health. Increasing evidence indicates SCFAs can exert effects on distant tissues and organs beyond the gut via blood circulation. Osteoarthritis (OA) is a chronic inflammatory joint disease that severely diminishes the physical function and quality of life. However, effective clinical treatments for OA remain elusive. Recent studies have shown that SCFAs can exert beneficial effects on damaged joints in OA. SCFAs can mitigate OA progression by preserving intestinal barrier function and maintaining the integrity of cartilage and subchondral bone, suggesting that they have substantial potential to be the adjunctive treatment strategy for OA. This review described the SCFAs in the human body and their cellular signaling mechanism, and summarized the multiple effects of SCFAs (especially butyrate, propionate, and acetate) on the prevention and treatment of OA by regulating the gut-joint axis, providing novel insights into their promising clinical applications.
Collapse
Affiliation(s)
- Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Xin Meng
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Hui Kong
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Xinran Li
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Peijie Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Xin-An Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China.
| |
Collapse
|
9
|
Ma Y, Jing J, Gao Y, Yu Y, Mao J, Zhang Y, Li T. MLIF inhibits inflammation and maintains intestinal flora homeostasis in a dextran sulfate sodium (DSS)-induced colitis mouse model. Food Chem Toxicol 2025; 202:115545. [PMID: 40354872 DOI: 10.1016/j.fct.2025.115545] [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: 04/14/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease primarily affecting the colon, characterized by mucosal inflammation and ulceration. Monocyte locomotion inhibitory factor (MLIF), a heat-stable pentapeptide derived from Entamoeba histolytica, has demonstrated the anti-inflammatory capacity. The aim of the current work was to test the protective effects of MLIF in a dextran sulfate sodium (DSS)-induced colitis mouse model. Our findings indicated that MLIF significantly inhibition of colitis development, including body weight, DAI score, colon length, and spleen index. MLIF slowing the progression of inflammation in the colon of mice exposed to DSS, evidenced by HE staining and mRNA expression levels of Il1b, Il6, Il18 and Il10. MLIF significantly alleviated intestinal barrier dysfunction in mice exposed to DSS, evidenced by AB-PAS staining and mRNA expression levels of Tjp1, Ocln and Muc2. Importantly, the administration of MLIF in colitis mice exerted beneficial effects on the gut microbiota, enhancing microbial diversity and abundance, and promoting the restoration of gut microbiota homeostasis. Non-targeted metabolomics results suggest that the benefits of MLIF may arise from its modulation of tryptophan metabolism pathways. In conclusion, MLIF prevention inflammation induction and preserves intestinal homeostasis against colitis induced by DSS.
Collapse
Affiliation(s)
- Yulin Ma
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, 200125, China; School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Jing Jing
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, 200125, China
| | - Yuan Gao
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yongsheng Yu
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Junqin Mao
- Department of Clinical Pharmacy, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201803, China
| | - Yuefan Zhang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Tiejun Li
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, 200125, China.
| |
Collapse
|
10
|
Hu Y, Bai F, Guo W, Chen G, Cai H. Bioavailability and gut microbiota modulation of polyphenol from Camellia oleifera shells during digestion and fermentation in vitro. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025. [PMID: 40347027 DOI: 10.1002/jsfa.14344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/24/2025] [Accepted: 04/09/2025] [Indexed: 05/12/2025]
Abstract
BACKGROUND Camellia oleifera shells (COS) are a by-product of camellia oil processing and are rich in polyphenols. To date, the polyphenols extracted from COS have not been fully developed. In this study, two polyphenol fractions, referred to as 20P and 40P, were obtained by sequentially eluting the COS crude extract from an AB-8 macroporous resin column using ethanol at concentrations of 20% and 40% with different polarities. In addition, the biotransformation processes of these two polyphenols and their effects on the gut microbiota were studied. RESULTS The results of in vitro digestion showed that at the end of the intestinal digestion stage the total phenolic content (TPC) content (0.34 ± 0.050 mg gallic acid equivalent (GAE) mg-1 dry weight (DW)) and antioxidant activity (0.53 ± 0.0076 mg vitamin C (Vc) mg-1 DW) of the 20P component were significantly higher than those of the 40P component (0.19 ± 0.020 mg GAE mg-1 DW; 0.39 ± 0.016 mg Vc mg-1 DW). In addition, the experiment also found that the phenolic composition and transformation process of 20P and 40P components were significantly different, and the TPC content (0.30 ± 0.058 mg GAE mg-1 DW) and antioxidant capacity (0.73 ± 0.034 mg Vc mg-1 DW) of the 40P component were significantly higher than that of the 20P (0.13 ± 0.035 mg GAE mg-1 DW); (0.19 ± 0.013 mg Vc mg-1 DW) component during in vitro fermentation. This may be related to the types of compounds extracted by solvents with different polarities. In addition, both 20P and 40P components significantly regulated the gut microbiota and promoted the production of short-chain fatty acids. CONCLUSION These results indicate that polyphenols from different polar sources of COS can serve as valuable bioactive compounds with potential health-regulating properties. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yanan Hu
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Anhui Agricultural University, Hefei, China
- College of Food and Nutrition, Anhui Agricultural University, Hefei, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, China
- Anhui Provincial Joint Construction Key Laboratory of Industrial New-Style Tea Beverage Green Manufacturing, Anhui Agricultural University, Hefei, China
- Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, Hefei, China
| | - Fuqing Bai
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Anhui Agricultural University, Hefei, China
- College of Food and Nutrition, Anhui Agricultural University, Hefei, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, China
- Anhui Provincial Joint Construction Key Laboratory of Industrial New-Style Tea Beverage Green Manufacturing, Anhui Agricultural University, Hefei, China
- Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, Hefei, China
| | - Wei Guo
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Anhui Agricultural University, Hefei, China
- College of Food and Nutrition, Anhui Agricultural University, Hefei, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, China
- Anhui Provincial Joint Construction Key Laboratory of Industrial New-Style Tea Beverage Green Manufacturing, Anhui Agricultural University, Hefei, China
- Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, Hefei, China
| | - Guijie Chen
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Anhui Agricultural University, Hefei, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, China
- Anhui Provincial Joint Construction Key Laboratory of Industrial New-Style Tea Beverage Green Manufacturing, Anhui Agricultural University, Hefei, China
- Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, Hefei, China
| | - Huimei Cai
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Anhui Agricultural University, Hefei, China
- College of Food and Nutrition, Anhui Agricultural University, Hefei, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, China
- Anhui Provincial Joint Construction Key Laboratory of Industrial New-Style Tea Beverage Green Manufacturing, Anhui Agricultural University, Hefei, China
- Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, Hefei, China
| |
Collapse
|
11
|
Jia Y, Liu X, Gao X, Yin S, Wu K, Meng X, Ren H, Liu J, Liu Z, Li H, Jiang Y. Plantamajoside alleviates DSS-induced ulcerative colitis by modulating gut microbiota, upregulating CBS, and inhibiting NF-κB. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156827. [PMID: 40381501 DOI: 10.1016/j.phymed.2025.156827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 04/18/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Plantamajoside (PMS) is a natural bioactive compound derived from medicinal, food homologous plants of the genus Plantago. PURPOSE AND METHODS This study aimed to investigate the protective effects of PMS on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in mice and explore the associated mechanisms. RESULTS We found that PMS treatment significantly alleviated UC symptoms in mice by preventing body weight loss, increasing colon length, and reducing disease activity index scores. Moreover, PMS alleviated colonic lesions, increased the number of goblet cells, upregulated the expression of intestinal barrier proteins (ZO-1, occludin, and claudin-3), and decreased the levels of pro-inflammatory factors. PMS treatment modulated the gut microbiota by increasing the relative abundance of Bacteroidota and Verrucomicrobiota and decreasing that of Firmicutes and Proteobacteria at the phylum level. At the genus level, PMS suppressed the abundance of pathogenic bacteria, such as Turicibacter and upregulated the abundance of [Eubacterium]_xylanophilum_group. Fecal microbiota transplantation experiments further confirmed that PMS treatment alleviated UC by modulating the gut microbiota. Transcriptomic analysis of colon tissues, coupled with reverse transcription-quantitative polymerase chain reaction and western blotting, showed that PMS treatment upregulated cystathionine beta-synthase (CBS) expression and inhibited NF-κB pathway activation. In a lipopolysaccharide-induced inflammation model in RAW264.7 cells, PMS treatment inhibited the secretion of pro-inflammatory cytokines, upregulated CBS expression, and prevented NF-κB pathway activation. CONCLUSION PMS protects against UC in mice via multiple mechanisms, including modulating the gut microbiota, increasing the expression levels of CBS, and inhibiting the NF-κB pathway.
Collapse
Affiliation(s)
- Yongheng Jia
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Xianjun Liu
- College of Biological and Food Engineering, Jilin Engineering Normal University, No. 3050 Kaixuan Street, Changchun 130000, China; Postdoctoral Research Workstation, Changchun Gangheng Electronics Company Limited, Changchun 130000, China
| | - Xinyi Gao
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Siyuan Yin
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Kun Wu
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Xianglong Meng
- Department of Gastroenterology, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Hui Ren
- Department of General surgery, the China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Jiawei Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Zijing Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Hao Li
- College of Biological and Food Engineering, Jilin Engineering Normal University, No. 3050 Kaixuan Street, Changchun 130000, China
| | - Yang Jiang
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China.
| |
Collapse
|
12
|
Yang H, Kuang Y, Wang L, Ma X, Gálvez JAV, Lu J, Dai Y, Liu S, Yao J, Chen X, Cao Y. Pterostilbene attenuates intestinal barrier damage and secondary liver oxidative stress in a murine model of Clostridium difficile infection by regulating the gut microbiota. Food Funct 2025; 16:3325-3343. [PMID: 40190207 DOI: 10.1039/d4fo06413e] [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: 05/07/2025]
Abstract
Clostridium difficile infection (CDI) is a significant infectious disease with limited treatment options. Pterostilbene, an active compound found in blueberries, is known for its antioxidant and anti-inflammatory properties. This study investigated the effects of pterostilbene on intestinal barrier damage and secondary liver oxidative stress induced by CDI in mice. Pathological changes in the colon and liver, the levels of anti-inflammatory cytokines and antioxidants, and the expression of related genes were evaluated. Additionally, 16S rRNA sequencing and targeted metabolomics analyses of the gut microbiota and bile acids were conducted. Pterostilbene reduced the abundance of harmful bacteria such as Enterococcus, while increasing beneficial bacteria like Lactobacillus, thereby reshaping the gut microbiota and bile acid profile and reducing the accumulation of T-βMCA. This process activated intestinal FXR signaling, which alleviated colonic inflammation and reduced intestinal permeability. The reduction in intestinal permeability prevented the translocation of bacteria and bacterial toxins into the liver via the portal vein, thereby reducing liver inflammation and oxidative stress. Pterostilbene presented a promising strategy for maintaining intestinal health through the regulation of dysbiosis and bile acid disturbances caused by CDI. When integrated into the food system, pterostilbene has the potential to improve intestinal health, mitigate the risk of CDI associated with contaminated agricultural products, and enhance public health and food safety. Additionally, we identified that regulating the intestinal bile acid profile and the FXR receptor could serve as potential therapeutic targets for CDI, thereby facilitating the development of novel treatment options and dietary strategies.
Collapse
Affiliation(s)
- Hao Yang
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, China.
| | - Yanling Kuang
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, China.
| | - Lamei Wang
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, China.
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts 02215, USA.
| | - Xinru Ma
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, China.
| | - Javier A Villafuerte Gálvez
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts 02215, USA.
| | - Jing Lu
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, China.
| | - Yanfei Dai
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, China.
| | - Shimin Liu
- Institute of Agriculture, The University of Western Australia, Perth, Australia
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, China.
| | - Xinhua Chen
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts 02215, USA.
| | - Yangchun Cao
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, China.
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts 02215, USA.
| |
Collapse
|
13
|
Jiménez-González C, Alonso-Peña M, Argos Vélez P, Crespo J, Iruzubieta P. Unraveling MASLD: The Role of Gut Microbiota, Dietary Modulation, and AI-Driven Lifestyle Interventions. Nutrients 2025; 17:1580. [PMID: 40362889 PMCID: PMC12073168 DOI: 10.3390/nu17091580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 05/01/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Gut microbiota has a crucial role in the pathophysiology of metabolic-associated steatotic liver disease (MASLD), influencing various metabolic mechanisms and contributing to the development of the disease. Dietary interventions targeting gut microbiota have shown potential in modulating microbial composition and mitigating MASLD progression. In this context, the integration of multi-omics analysis and artificial intelligence (AI) in personalized nutrition offers new opportunities for tailoring dietary strategies based on individual microbiome profiles and metabolic responses. The use of chatbots and other AI-based health solutions offers a unique opportunity to democratize access to health interventions due to their low cost, accessibility, and scalability. Future research should focus on the clinical validation of AI-powered dietary strategies, integrating microbiome-based therapies and precision nutrition approaches. Establishing standardized protocols and ethical guidelines will be crucial for implementing AI in MASLD management, paving the way for a more personalized, data-driven approach to disease prevention and treatment.
Collapse
Affiliation(s)
- Carolina Jiménez-González
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases, Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, 39011 Santander, Spain; (C.J.-G.); (M.A.-P.); (P.A.V.); (P.I.)
| | - Marta Alonso-Peña
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases, Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, 39011 Santander, Spain; (C.J.-G.); (M.A.-P.); (P.A.V.); (P.I.)
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria, 39011 Santander, Spain
| | - Paula Argos Vélez
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases, Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, 39011 Santander, Spain; (C.J.-G.); (M.A.-P.); (P.A.V.); (P.I.)
| | - Javier Crespo
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases, Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, 39011 Santander, Spain; (C.J.-G.); (M.A.-P.); (P.A.V.); (P.I.)
| | - Paula Iruzubieta
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases, Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, 39011 Santander, Spain; (C.J.-G.); (M.A.-P.); (P.A.V.); (P.I.)
| |
Collapse
|
14
|
Yang J, Ren H, Cao J, Fu J, Wang J, Su Z, Lu S, Sheng K, Wang Y. Gut commensal Lachnospiraceae bacteria contribute to anti-colitis effects of Lactiplantibacillus plantarum exopolysaccharides. Int J Biol Macromol 2025; 309:142815. [PMID: 40187461 DOI: 10.1016/j.ijbiomac.2025.142815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 03/20/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
The probiotic Lactiplantibacillus plantarum (L. plantarum) could ameliorate colitis. Alterations in the composition of gut microbiota (GM) have been proved in cases of colitis. The exopolysaccharides from L. plantarum HMPM2111 (LPE) could be effective in colitis through altering the composition of the GM. These effects were linked to inhibiting intestinal inflammation, regulating the TXNIP/NLRP3 inflammasome axis, and attenuating colonic barrier dysfunction. The combination of fecal microbiota transplantation (FMT) and antibiotic inducement showed that gut bacteria susceptible to vancomycin were inversely associated with colitis features and were necessary for the anti-inflammatory effects of LPE. The elevated abundances of gut commensal Lachnospiraceae bacteria were associated with the restoration of colitis treated by LPE. Metabolomics analysis showed that colitis mice treated with LPE had higher levels of propionate and tryptophan metabolites generated from gut bacteria. The administration of these metabolites protected colitis and resulted in a reduction in inflammatory responses. The outcomes of our investigation emerge the significance of the GM in controlling the protective implications of LPE against colitis. Lachnospiraceae bacteria, together with downstream metabolites, contribute substantially to protection. This work elucidates the essential function of the GM-metabolite axis in producing comprehensive protection versus colitis and identifies prospective treatment targets.
Collapse
Affiliation(s)
- Jian Yang
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China
| | - Huijuan Ren
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China
| | - Jialing Cao
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China
| | - Jingjing Fu
- Department of Pharmacy, Anhui No.2 Provincial People's Hospital, Hefei 230041, Anhui, China; Anhui No.2 Provincial People's Hospital Clinical College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Junhui Wang
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China
| | - Ziwei Su
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China
| | - Shiqi Lu
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China
| | - Kangliang Sheng
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China.
| | - Yongzhong Wang
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China.
| |
Collapse
|
15
|
Yong H, Yun D, Xu F, Tang C, Chen D, Kan J, Huang J, Yu H, Liu J. Dialdehyde starch-epicatechin gallate conjugate alleviates inflammation in lipopolysaccharide-stimulated RAW264.7 cells and dextran sulfate sodium-induced colitis mice. Int J Biol Macromol 2025; 306:141343. [PMID: 39988158 DOI: 10.1016/j.ijbiomac.2025.141343] [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: 11/24/2024] [Revised: 02/09/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
In this study, epicatechin gallate (ECG), a natural anti-inflammatory agent, was conjugated onto dialdehyde starch (DAS) to achieve high physiological stability. The anti-inflammatory effect of DAS-ECG conjugate was evaluated by lipopolysaccharide (LPS)-stimulated RAW264.7 cells and dextran sulfate sodium (DSS)-induced colitis mice models. Results showed that 25-800 μg/mL of DAS-ECG conjugate was non-cytotoxic to RAW264.7 cells. DAS-ECG conjugate effectively inhibited the abnormal morphology, the production of nitric oxide, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1β (IL-1β) and reactive oxygen species, and the apoptosis of LPS-stimulated RAW264.7 cells in a dose-dependent manner. DAS-ECG conjugate significantly reduced the disease activity index, thymus atrophy, spleen enlargement, colon shortening and colon damage of DSS-induced colitis mice. Meanwhile, DAS-ECG conjugate remarkably reduced the levels of TNF-α, IL-6, IL-1β and malondialdehyde but increased the levels of superoxide dismutase and glutathione in the colon tissue of DSS-induced colitis mice. Moreover, DAS-ECG conjugate increased the relative abundance of beneficial bacteria (Akkermansia, Candidatus_Saccharimonas, unclassified_f_Muribaculaceae, Alistipes and Parabacteroides), promoted the production of short-chain fatty acids, and decreased the relative abundance of harmful bacterium (norank_f_Ruminococcaceae) in DSS-induced colitis mice. Therefore, DAS-ECG conjugate could be considered as a promising anti-inflammatory agent.
Collapse
Affiliation(s)
- Huimin Yong
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Dawei Yun
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Fengfeng Xu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Chao Tang
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Dan Chen
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Juan Kan
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Jinbao Huang
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, China
| | - Hai Yu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Jun Liu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China.
| |
Collapse
|
16
|
Yang JZ, Li JH, Liu JL, Zhou AD, Wang H, Xie XL, Zhang KK, Wang Q. Multiomics analysis revealed the effects of polystyrene nanoplastics at different environmentally relevant concentrations on intestinal homeostasis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 372:126050. [PMID: 40086783 DOI: 10.1016/j.envpol.2025.126050] [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: 11/25/2024] [Revised: 02/16/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Nanoplastics pollution is a global issue, with the digestive tract being one of the first affected organs, requiring further research on its impact on intestinal health. This study involved orally exposing mice to polystyrene nanoplastics (PS-NPs) at doses of 0.1, 0.5, or 2.5 mg/d for 42 days. The effects on intestinal health were thoroughly assessed via microbiomics, metabolomics, transcriptomics, and molecular biology. Our study demonstrated that the administration of all three doses of PS-NPs resulted in increased colonic permeability, heightened colonic and peripheral inflammation, reduced levels of antimicrobial peptides, and shortened colonic length. These effects may be attributed to a reduction in the abundance of probiotic bacteria, such as Clostridia_UCG-014, Roseburia, and Akkermansia, alongside an increase in the abundance of the pathogenic bacterium Desulfovibrionaceae induced by PS-NPs. Furthermore, we underscored the crucial role of histidine metabolism in PS-NPs-induced colonic injury, characterized by a significant reduction of L-histidine, which is closely related to microbial ecological dysregulation. Corresponding to microbiota deterioration and metabolic dysregulation, transcriptome analysis revealed that PS-NPs may disrupt colonic immune homeostasis by activating the TLR4/MyD88/NF-κB/NLRP3 signaling pathway. In conclusion, this study provided novel insights into the mechanisms by which PS-NPs disrupt intestinal homeostasis through integrated multiomics analysis, revealing critical molecular pathway and providing a scientific basis for future risk assessment of nanoplastics exposure.
Collapse
Affiliation(s)
- Jian-Zheng Yang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Ji-Hui Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jia-Li Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - An-Ding Zhou
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Hui Wang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, National Children's Medical Center for South Central Region, Guangzhou, 510623, China
| | - Xiao-Li Xie
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), Guangzhou, Guangdong, 510515, China
| | - Kai-Kai Zhang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Qi Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
17
|
Sun C, Liu Z, Feng M, Wang J, Jiang Y, Zhao C. Mixtures of EGCG, bamboo leaf flavonoids, and broccoli seed water extracts exhibit anti-glycation and skin-protective effects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156592. [PMID: 40081290 DOI: 10.1016/j.phymed.2025.156592] [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: 11/05/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Skin aging is a multifaceted process. Glycation significantly contributes to skin aging and the development of complications. Researchers are currently investigating various substances, particularly those from natural sources, to combat skin glycation. PURPOSE This study aimed to comprehensively evaluate the anti-glycation effect of a new natural combination, EBB, which includes (-)-epigallocatechin-3 gallate (EGCG), bamboo leaf flavonoids, and broccoli seed water extracts, using cell and animal models and to explore its potential anti-glycation mechanism. METHODS The components of EBB were identified using HPLC and UHPLC-MS/MS. Additionally, a glycation cell model induced by glyceraldehyde, advanced glycation end products (AGEs), and methylglyoxal was established in HaCaT cells to evaluate the efficacy of EBB in alleviating glycation. Differential genes, signalling pathways, and biological processes were analysed through RNA sequencing to explore the mechanisms of the anti-glycation effects of EBB, which were further validated using qRT-PCR and Western blotting. Finally, the protective effects of EBB against glycation and skin damage were assessed in zebrafish and mouse in vivo models through histological studies and the measurement of various skin physiological parameters. RESULTS Glucoraphanin, Sinapine and orientin were identified in EBB, which effectively reduced the formation of AGEs and decreased the expression level of the RAGE protein in HaCaT cells. Transcriptomic analyses revealed that EBB regulated the expression of 576 differentially expressed genes. These genes were enriched in various biological processes, such as chronic inflammation and immune responses, and participated in the regulation of multiple signalling pathways, including TNF. Glycation upregulated the expression of the ROS1 gene and protein, while EBB reversed this effect. Furthermore, EBB attenuated the glycation response by downregulating the expression levels of proteins such as p-p38, p-ERK1/2, p-p65, and TNF-α. Additionally, the reduction of AGE accumulation by EBB was confirmed in a zebrafish model. Similarly, histological analyses of mouse skin tissue and various physiological parameters demonstrated that EBB significantly mitigated damage induced by glycation. CONCLUSIONS Our results show that EBB effectively inhibited glycation reactions. The mechanism of action may involve the reduction of inflammation by downregulating the expression levels of RAGE and ROS1, thereby decreasing the accumulation of AGEs in keratinocytes and alleviating skin damage. This paves the way for the potential application of EBB as a valuable anti-glycation functional ingredient in the food and cosmetic industries.
Collapse
Affiliation(s)
- Chang Sun
- Beijing Qingyan Boshi Health Management Co., Ltd, No. 36, Chuangyuan Road, Chaoyang District, Beijing, China.
| | - Zibin Liu
- Beijing Qingyan Boshi Health Management Co., Ltd, No. 36, Chuangyuan Road, Chaoyang District, Beijing, China.
| | - Mengmeng Feng
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
| | - Junbo Wang
- School of Public Health, Peking University, Beijing, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, China; Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China.
| | - Yanfei Jiang
- Beijing Qingyan Boshi Health Management Co., Ltd, No. 36, Chuangyuan Road, Chaoyang District, Beijing, China.
| | - Chunyue Zhao
- Beijing Qingyan Boshi Health Management Co., Ltd, No. 36, Chuangyuan Road, Chaoyang District, Beijing, China.
| |
Collapse
|
18
|
Yang H, Zhao Y, Zhang R, Zhao L, Yang H, Liao X. CiLi (Rosa roxburghii Tratt.) polyphenols improve colitis via gut microbiota-lipid mediator-immunity axis. Food Res Int 2025; 209:116257. [PMID: 40253185 DOI: 10.1016/j.foodres.2025.116257] [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: 12/28/2024] [Revised: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
Dysbiosis of gut microbiome is one of the most important factors leading to inflammatory bowel disease (IBD). Intake of phytochemicals from fruits and vegetables is an effective way to improve IBD, but how these bioactivators regulate gut microbiota to exert healthy effects remains unclear. Here, we found that pretreatment with CiLi juice, particularly its polyphenol component, alleviated dextran sulfate sodium (DSS)-induced colitis while preserving intestinal barrier integrity. CiLi polyphenols (CL_PP) reduced inflammation and oxidative stress in colon tissue and enriched fecal short-chain fatty acids. Importantly, CL_PP significantly regulated the gut microbiome diversity, increasing beneficial bacteria (e.g., Clostridia_UCG-014, f_Muribaculaceae and Ileibacterium_valens) while decreasing harmful bacteria (Escherichia-Shigella and Romboutsia). Multiomics analysis revealed that CL_PP upregulated bioactive lipid metabolites, particularly those derived from polyunsaturated fatty acids (e.g., resolvin D2, prostaglandin A1, and glycerophosphocholine) related gene expressions (Pltp, Alox15 and Pld4). Additionally, CL-PP downregulated the oxidative stress markers (oxidized glutathione and glutathione peroxidase 3), and immune cell markers (CD8 and CD68). Fecal microbiota transplantation confirmed that the fecal microbiota from CL_PP-treated mice exhibited anti-colitis effects. These effects were diminished in antibiotic-treated mice, underscoring the importance of the gut microbiota in mediating the CL_PP's anti-inflammatory benefits. This study suggests that CL_PP is a potential modulator of gut microbiome dysbiosis for the prevention and treatment of colitis.
Collapse
Affiliation(s)
- Huanzhi Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yang Zhao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ruiqi Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Liang Zhao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Haixia Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Xiaojun Liao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
19
|
Hu J, Xu F, Zhu L, Cui Y, Au R, Li Y, Tong Y, Shen H. Angelica dahurica Polysaccharides Ameliorate Colitis by Reducing the Restriction of Gut Microbiota-Derived Imidazole Propionate on PPAR-γ Signaling Activation. Phytother Res 2025; 39:2072-2090. [PMID: 40045660 PMCID: PMC12087948 DOI: 10.1002/ptr.8466] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 05/21/2025]
Abstract
Angelica dahurica radix (ADR), the root of the botanical family Apiaceae (genus Angelica, species Angelica dahurica (Hoffm.)), has been used to treat colitis in clinical practice. The immunomodulatory effects of ADR are attributed to its polysaccharides (RP). However, its mechanism of action has not been elucidated. In this study, RP's structure was determined through nuclear magnetic resonance analysis. Dextran sulfate sodium-induced colitis in mice was utilized to assess the therapeutic efficacy of RP, while experiments involving fecal microbiota transplantation (FMT) and antibiotic treatment were performed to investigate the contribution of gut microbiota to RP's protective function. Non-targeted metabolomics was utilized to identify potential targets for elucidating the underlying mechanisms. RP is likely composed of (→4)-α-D-Glcp-(1→ and →4)-α-D-Galp-(1→). It effectively alleviated DSS-induced colitis by restoring the balance of the gut microbial community, a finding validated through FMT and antibiotic intervention experiments. Imidazole propionate (ImP) emerged as a potential target for RP's efficacy in treating colitis, which inhibits the activation of peroxisome proliferator-activated receptor gamma (PPAR-γ). Our findings suggest that RP may confer protection against colitis by activating the PPAR-γ signaling pathway through alleviating the constraint imposed by ImP.
Collapse
Affiliation(s)
- Jingyi Hu
- Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine)NanjingChina
| | - Feng Xu
- First Clinical Medical CollegeNanjing University of Chinese MedicineNanjingChina
| | - Lei Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine)NanjingChina
| | - Yuan Cui
- First Clinical Medical CollegeNanjing University of Chinese MedicineNanjingChina
| | - Ryan Au
- International Education CollegeNanjing University of Chinese MedicineNanjingChina
| | - Yanan Li
- First Clinical Medical CollegeNanjing University of Chinese MedicineNanjingChina
| | - Yiheng Tong
- First Clinical Medical CollegeNanjing University of Chinese MedicineNanjingChina
| | - Hong Shen
- Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine)NanjingChina
| |
Collapse
|
20
|
Cao Y, Xiao S, He B, Shi X, Xiao N, Liu X, Liu D, Zhou Z, Wang P. Chronic Exposure to Fluxapyroxad Exacerbated Susceptibility to Colitis in Mice via a Gut Microbiota-Indole Derivatives-Th17/Treg Cell Balance Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:10172-10185. [PMID: 40244699 DOI: 10.1021/acs.jafc.5c02749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Fluxapyroxad is the most commonly used succinate dehydrogenase inhibitor fungicide. This work investigated its adverse effects on colitis susceptibility and explored the underlying mechanisms based on a mouse model. After 13 weeks of exposure at the acceptable daily intake (ADI) level, fluxapyroxad exacerbated the susceptibility to colitis, impaired the intestinal barrier, and elevated proinflammatory cytokines and chemokines of the colon in mice. It was found that this toxic effect was caused by the disruption of the gut microbiome. Specifically, the abundance of Lachnospiraceae and Muribaculaceae decreased, while Desulfovibrionaceae and Eggerthellaceae increased. Altered microbiota reduced fecal indole derivatives, including indole-3-lactic acid (ILA), indole-3-acetic acid (IAA), and indole-3-acrylic acid (IArA), inhibiting aryl hydrocarbon receptor (AHR) activation, disrupting immune homeostasis by overactivating Th17 cells and insufficient Treg cell differentiation, and causing mild colonic inflammation. Oral antibiotic-treated mice and fecal transfer experiments validated the pathway. Susceptibility to colitis induced by fluxapyroxad was not detected in the oral antibiotic-treated mice. Fecal transfer of the disordered gut microbiota caused by fluxapyroxad could aggravate the severity of colitis in recipient oral antibiotic-treated mice that did not receive fluxapyroxad exposure. In conclusion, chronic fluxapyroxad exposure at the ADI level exacerbated colitis via a gut microbiota-indole derivatives-Treg/Th17 cell balance axis, offering a new risk assessment perspective of fluxapyroxad.
Collapse
Affiliation(s)
- Yue Cao
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Shouchun Xiao
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Bingying He
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Xinlei Shi
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Nan Xiao
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Xueke Liu
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Donghui Liu
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Zhiqiang Zhou
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Peng Wang
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| |
Collapse
|
21
|
Liu M, Ge Y, Xu E, Zhu T, Ruan H, Zheng J. The regulatory effects of epigallocatechin gallate on growth performance, systemic antioxidant status, immune response, and gut microbiota structure in geese. Poult Sci 2025; 104:105178. [PMID: 40267569 DOI: 10.1016/j.psj.2025.105178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025] Open
Abstract
The effects of dietary supplementation with epigallocatechin-3-gallate (EGCG) on growth performance, antioxidant capacity, immune function, and gut microbiota in geese were investigated. Seventy-two healthy 35-day-old male geese were randomly divided into a control group (basal diet) or an EGCG group (basal diet + 200 mg/kg EGCG), with 36 geese per group, which was further subdivided into 6 replicates (6 geese per replicate). The experiment lasted 21 days. Geese in the EGCG group exhibited significantly higher final body weight (2.93 kg vs. 2.28 kg, P < 0.01) and average daily gain (72.38 g/d vs. 41.4 g/d, P < 0.01) along with a 42.8 % reduction in the feed conversion ratio (3.95 vs. 6.91, P < 0.01) versus the control. Liver weights in the EGCG group were significantly elevated compared to the CON group on days 14 and 21 (P < 0.05) and a strong correlation between liver weight and body weight. EGCG significantly increased catalase, glutathione peroxidase, and superoxide dismutase activities, and the total antioxidant capacity in serum and jejunum while decreasing malondialdehyde (MDA) levels (P < 0.05). Immunological analyses revealed elevated serum immunoglobulin (Ig)A, IgG, and IgM and lysozyme in the EGCG group (P < 0.05), accompanied by a decrease in the pro-inflammatory cytokines interleukin-6 and interferon-γ. Intestinal morphology demonstrated increased villus height-to-crypt depth ratios in the duodenum and ileum (P < 0.05). 16S rRNA sequencing indicated that EGCG increased the relative abundance of Akkermansia and Verrucomicrobiota (P < 0.05) in the cecal content and enriched microbial functions related to inorganic ion transport and metabolism. Correlation analysis revealed positive associations between Akkermansia and IgA, and between Firmicutes and oxidative damage markers (MDA). Overall, dietary supplementation with 200 mg/kg EGCG could improve growth performance, antioxidant capacity, immune response, and gut microbiota structure in geese, supporting its potential as a plant-based feed additive.
Collapse
Affiliation(s)
- Mengyu Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University,163319, Daqing, Heilongjiang, PR China
| | - Yansong Ge
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University,163319, Daqing, Heilongjiang, PR China
| | - Enshuang Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University,163319, Daqing, Heilongjiang, PR China
| | - Tingting Zhu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University,163319, Daqing, Heilongjiang, PR China
| | - Hongri Ruan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University,163319, Daqing, Heilongjiang, PR China
| | - Jiasan Zheng
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University,163319, Daqing, Heilongjiang, PR China.
| |
Collapse
|
22
|
Du J, Guan Y, Zhang E. Regulatory role of gut microbiota in immunotherapy of hepatocellular carcinoma. Hepatol Int 2025:10.1007/s12072-025-10822-6. [PMID: 40229514 DOI: 10.1007/s12072-025-10822-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/07/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND The gut microbiota plays a role in triggering innate immunity and regulating the immune microenvironment (IME) of hepatocellular carcinoma (HCC) by acting on various signaling receptors and transcription factors through its metabolites and related molecules. Furthermore, there is an increasing recognition of the gut microbiota as a potential therapeutic target for HCC, given its ability to modulate the efficacy of immune checkpoint inhibitors (ICIs). OBJECTIVE This review will discuss the mechanisms of gut microbiota in modulating immunotherapy of HCC, the predictive value of efficacy, and the therapeutic strategies for modulating the gut microbiota in detail. METHODS We conducted a systematic literature search in PubMed, Embase, Scopus, Cochrane Library, China National Knowledge Infrastructure, and Wanfang Chinese databases for articles involving the influence of gut microbiota on HCC immunotherapy. RESULTS The mechanisms underlying the effect of gut microbiota on HCC immunotherapy include gut-liver axis, tumor immune microenvironment (TIME), and antibodies. Patients who benefit from ICIs exhibit a higher abundance of gut microbiota. Antibiotics, fecal microbiota transplantation (FMT), probiotics, and prebiotics are effective methods to regulate gut microbiota. CONCLUSION The strong connection between the liver and gut will provide numerous opportunities for the development of microbiome-based diagnostics, treatments, or prevention strategies for HCC patients.
Collapse
Affiliation(s)
- Jiajia Du
- Hepatic Surgery Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Yan Guan
- Hepatic Surgery Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Erlei Zhang
- Hepatic Surgery Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China.
| |
Collapse
|
23
|
Wang Y, Li Q, Hua J, Que H, Xu H, Xu X, Feng N. Causal relationship between gut microbiota and dental caries: a two-sample mendelian randomization study. BDJ Open 2025; 11:35. [PMID: 40210870 PMCID: PMC11986140 DOI: 10.1038/s41405-025-00328-6] [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: 09/19/2024] [Revised: 01/26/2025] [Accepted: 02/07/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND In recent years, an increasing number of studies have revealed a close relationship between the gut microbiota and a variety of human diseases. At the same time, it has also been shown that dysregulation of the oral microbiota may lead to changes in the gut microbiota. However, it remains unclear whether the gut microbiota affects the occurrence and development of oral diseases. Therefore, the aim of this study was to explore the potential effects of gut microbiota on dental caries and to reveal possible mechanisms of the gut-oral microbiota axis. METHODS First, gut microbiota and dental caries data from genome-wide association studies (GWAS) were analyzed using Mendelian randomization analysis. Inverse variance weighted (IVW) was used as the main criterion (P value < 0.05). Then, MR-Egger regression, IVW regression and leave-one-out tests were used to test the reliability and stability of the mendelian randomization results. Finally, the potential mechanisms and significance of the relationship between gut microbiota and dental caries were explored. RESULTS The analysis showed that Eubacteriumbrachygroup [odds ratio (OR) = 1.001, 95% confidence interval (CI): 1.000-1.002, P = 0.046] and Terrisporobacter (OR = 1.002, 95% CI: 1.0001-1.0041, P = 0.035) were positively correlated with dental caries. Escherichia.Shigella (OR = 0.997, 95% CI: 0.995-0.999, P = 0.047), Oscillibacter (OR = 0.998, 95% CI: 0.997-0.999, P = 0.038), RuminococcaceaeUCG014 (OR = 0.998, 95% CI: 0.996-0.999, P = 0.044) and Oscillospira (OR = 0.997, 95% CI: 0.995-0.999, P = 0.038) were negatively correlated with dental caries. CONCLUSION The present study demonstrated a significant causal relationship between the gut microbiota and the development of dental caries, providing new insights into influencing the development of dental caries by affecting the composition of the gut microbiota.
Collapse
Affiliation(s)
- Yang Wang
- Department of urology, Jiangnan University Medical Center (Wuxi No. 2 People's Hospital), Wuxi, 214000, China
| | - Quan Li
- Department of urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jinqi Hua
- Department of urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Hongliang Que
- Department of urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Haoxiang Xu
- Department of urology, Jiangnan University Medical Center (Wuxi No. 2 People's Hospital), Wuxi, 214000, China
| | - Xinyu Xu
- Department of urology, Jiangnan University Medical Center (Wuxi No. 2 People's Hospital), Wuxi, 214000, China
| | - Ninghan Feng
- Department of urology, Jiangnan University Medical Center (Wuxi No. 2 People's Hospital), Wuxi, 214000, China.
| |
Collapse
|
24
|
Eslami M, Adampour Z, Fadaee Dowlat B, Yaghmayee S, Motallebi Tabaei F, Oksenych V, Naderian R. A Novel Frontier in Gut-Brain Axis Research: The Transplantation of Fecal Microbiota in Neurodegenerative Disorders. Biomedicines 2025; 13:915. [PMID: 40299512 PMCID: PMC12025253 DOI: 10.3390/biomedicines13040915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
The gut-brain axis (GBA) represents a sophisticated bidirectional communication system connecting the central nervous system (CNS) and the gastrointestinal (GI) tract. This interplay occurs primarily through neuronal, immune, and metabolic pathways. Dysbiosis in gut microbiota has been associated with multiple neurodegenerative diseases, such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS). In recent years, fecal microbiota transplantation (FMT) has gained attention as an innovative therapeutic approach, aiming to restore microbial balance in the gut while influencing neuroinflammatory and neurodegenerative pathways. This review explores the mechanisms by which FMT impacts the gut-brain axis. Key areas of focus include its ability to reduce neuroinflammation, strengthen gut barrier integrity, regulate neurotransmitter production, and reinstate microbial diversity. Both preclinical and clinical studies indicate that FMT can alleviate motor and cognitive deficits in PD and AD, lower neuroinflammatory markers in MS, and enhance respiratory and neuromuscular functions in ALS. Despite these findings, several challenges remain, including donor selection complexities, uncertainties about long-term safety, and inconsistencies in clinical outcomes. Innovations such as synthetic microbial communities, engineered probiotics, and AI-driven analysis of the microbiome hold the potential to improve the precision and effectiveness of FMT in managing neurodegenerative conditions. Although FMT presents considerable promise as a therapeutic development, its widespread application for neurodegenerative diseases requires thorough validation through well-designed, large-scale clinical trials. It is essential to establish standardized protocols, refine donor selection processes, and deepen our understanding of the molecular mechanisms behind its efficacy.
Collapse
Affiliation(s)
- Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran;
- Department of Bacteriology and Virology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Zarifeh Adampour
- Institute of Science, Biotechnology and Biosafety Department, Eskishehir Osmangazi University, Eskishehir 26040, Türkiye;
| | - Bahram Fadaee Dowlat
- School of Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Shayan Yaghmayee
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Faezeh Motallebi Tabaei
- Department of Medical Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan 49189-36316, Iran
| | | | - Ramtin Naderian
- Clinical Research Development Unit, Kowsar Educational, Research and Therapeutic Hospital, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| |
Collapse
|
25
|
Cheng X, He J, Yang Y, He Y, Chen G, Ling B, Wang A. Targeted metabolomics unravels the mechanism by phenylpropanoid-rich of the peel of Zea mays L. ameliorates metabolic disorders in diabetic mice through gut microbiota modulation. Front Pharmacol 2025; 16:1551713. [PMID: 40271058 PMCID: PMC12014729 DOI: 10.3389/fphar.2025.1551713] [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: 12/26/2024] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Background Diabetes is one common clinical symptoms of metabolic disorders. The peel of Zea mays L. is a folk remedy for diabetes that has not been thoroughly studied. The effects and mechanisms on diabetes complicated glucose and lipid metabolism disorders are still unknown now. Purpose The research is intended to elucidate the constituent of phenylpropanoid enriched of Zea mays L. (YMP), and investigate the treatment and mechanism on amending glucose and lipid metabolism disorders. Methods The constituents of YMP were systematacially identified by HPLC-Q-TOF-MS/MS and NMR. To assess the effects of varying YMP doses, diabetic mice induced by streptozotocin and a high-fat diet were divided into groups. Targeted serum metabolomics investigations were conducted using UHPLC-LTQ-Orbitrap MS. Moreover, 16S rRNA analysis was employed to elucidate the intricate mechanisms through the gut microbiota modulates lipid and glucose metabolism. Results It demonstrated that the primary component of YMP was luteolin. At a high dosage of 160 mg/kg/day, YMP considerably reduced the values of the oral glucose tolerance test, insulin, and blood glucose (p < 0.001). After administration, insulin resistance indexes decreased. YMP reversed the accumulation of glycogen in the liver and reduced hepatic lipid deposition. Compared to MOD group, the concentration of luteolin is higher and its metabolite, indicating that luteolin may be adequately absorbed and have an influence on the circulatory system. The results of 16S rRNA sequencing demonstrated that YMP and gut microbiota interacted to positively regulate beneficial bacteria such as Bifidobacterium, Ligilactobacillus, and Lactobacillus. Conclusion This work investigated the regulating effect of YMP on the liver glycolipid metabolism for the first time, and it also showed the underlying mechanism through gut microbiota. According to these studies, YMP has a lot of potential to be used as a supplemental treatment for complex metabolic illnesses like diabetes. It offered empirical support for the use of alternative medicine in the area to treat complex problems of glucose and lipid metabolism in diabetes.
Collapse
Affiliation(s)
- Xiaotian Cheng
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
- Department of Pharmacy, The Yancheng Clinical College of Xuzhou Medical University & The First people’s Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Jinyan He
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yuru Yang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yaonan He
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Guangtong Chen
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Bai Ling
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
- Department of Pharmacy, The Yancheng Clinical College of Xuzhou Medical University & The First people’s Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Andong Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
26
|
Zhang Y, Si L, Shu X, Qiu C, Wan X, Li H, Ma S, Jin X, Wei Z, Hu H. Gut microbiota contributes to protection against porcine deltacoronavirus infection in piglets by modulating intestinal barrier and microbiome. MICROBIOME 2025; 13:93. [PMID: 40189556 PMCID: PMC11974153 DOI: 10.1186/s40168-025-02092-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 03/14/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND Gut microbiota plays a critical role in counteracting enteric viral infection. Our previous study demonstrated that infection of porcine deltacoronavirus (PDCoV) disturbs gut microbiota and causes intestinal damage and inflammation in piglets. However, the influence of gut microbiota on PDCoV infection remains unclear. RESULTS Firstly, the relationship between gut microbiota and disease severity of PDCoV infection was evaluated using 8-day-old and 90-day-old pigs. The composition of gut microbiota was significantly altered in 8-day-old piglets after PDCoV infection, leading to severe diarrhea and intestinal damage. In contrast, PDCoV infection barely affected the 90-day-old pigs. Moreover, the diversity (richness and evenness) of microbiota in 90-day-old pigs was much higher compared to the 8-day-old piglets, suggesting the gut microbiota is possibly associated with the severity of PDCoV infection. Subsequently, transplanting the fecal microbiota from the 90-day-old pigs to the 3-day-old piglets alleviated clinical signs of PDCoV infection, modulated the diversity and composition of gut microbiota, and maintained the physical and chemical barrier of intestines. Additionally, metabolomic analysis revealed that the fecal microbiota transplantation (FMT) treatment upregulated the swine intestinal arginine biosynthesis, FMT significantly inhibited the inflammatory response in piglet intestine by modulating the TLR4/MyD88/NF-κB signaling pathway. CONCLUSIONS PDCoV infection altered the structure and composition of the gut microbiota in neonatal pigs. FMT treatment mitigated the clinical signs of PDCoV infection in the piglets by modulating the gut microbiota composition and intestinal barrier, downregulating the inflammatory response. The preventive effect of FMT provides novel targets for the development of therapeutics against enteropathogenic coronaviruses. Video Abstract.
Collapse
Affiliation(s)
- Yunfei Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Lulu Si
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Xiangli Shu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Congrui Qiu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Xianhua Wan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Haiyan Li
- College of Sport, Yan'an University, Yanan, 716000, People's Republic of China
| | - Shijie Ma
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
- Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou, 450046, People's Republic of China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, People's Republic of China
| | - Xiaohui Jin
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
- Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou, 450046, People's Republic of China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, People's Republic of China
| | - Zhanyong Wei
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China.
- Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou, 450046, People's Republic of China.
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, People's Republic of China.
- Longhu Laboratory of Henan Province, Zhengzhou, 450046, People's Republic of China.
| | - Hui Hu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China.
- Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou, 450046, People's Republic of China.
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, People's Republic of China.
- Longhu Laboratory of Henan Province, Zhengzhou, 450046, People's Republic of China.
| |
Collapse
|
27
|
Liu S, Cai P, You W, Yang M, Tu Y, Zhou Y, Valencak TG, Xiao Y, Wang Y, Shan T. Enhancement of gut barrier integrity by a Bacillus subtilis secreted metabolite through the GADD45A-Wnt/β-catenin pathway. IMETA 2025; 4:e70005. [PMID: 40236773 PMCID: PMC11995189 DOI: 10.1002/imt2.70005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 04/17/2025]
Abstract
Inflammatory bowel disease (IBD) represents a significant challenge to global health, characterized by intestinal inflammation, impaired barrier function, and dysbiosis, with limited therapeutic options. In this study, we isolated a novel strain of Bacillus subtilis (B. subtilis) and observed promising effects in protecting against disruption of the gut barrier. Our findings indicate that the enhancement of intestinal barrier function is primarily attributed to its metabolites. We identified a novel metabolite, 2-hydroxy-4-methylpentanoic acid (HMP), derived from B. subtilis, that significantly improved intestinal barrier function. We also show that growth arrest and DNA damage 45A (GADD45A) is a key regulator of mucosal barrier integrity, which is activated by HMP and subsequently activates the downstream Wnt/β-catenin pathway. Our findings potentially contribute to the development of probiotics-derived metabolites or targeted "postbiotics" as novel therapeutics for the treatment or prevention of IBD and other diseases associated with intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Shiqi Liu
- College of Animal SciencesZhejiang UniversityHangzhouChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐quality Animal ProductsHangzhouChina
| | - Peiran Cai
- College of Animal SciencesZhejiang UniversityHangzhouChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐quality Animal ProductsHangzhouChina
| | - Wenjing You
- College of Animal SciencesZhejiang UniversityHangzhouChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐quality Animal ProductsHangzhouChina
| | - Mingshun Yang
- College of Animal SciencesZhejiang UniversityHangzhouChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐quality Animal ProductsHangzhouChina
| | - Yuang Tu
- College of Animal SciencesZhejiang UniversityHangzhouChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐quality Animal ProductsHangzhouChina
| | - Yanbing Zhou
- College of Animal SciencesZhejiang UniversityHangzhouChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐quality Animal ProductsHangzhouChina
| | | | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐products, Institute of Agro‐product Safety and NutritionZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Yizhen Wang
- College of Animal SciencesZhejiang UniversityHangzhouChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐quality Animal ProductsHangzhouChina
| | - Tizhong Shan
- College of Animal SciencesZhejiang UniversityHangzhouChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐quality Animal ProductsHangzhouChina
| |
Collapse
|
28
|
Yao Q, Zhu H, Wang L, Zuo Z, Li X, Gao D. Preparation and intestinal gastrointestinal delivery performance of Malus baccata (Linn.) polyphenols loaded nanoparticles based on yeast membranes. Food Res Int 2025; 207:116129. [PMID: 40086951 DOI: 10.1016/j.foodres.2025.116129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/16/2025]
Abstract
The currently available polyphenols delivery systems require the complicated preparation process and participation of multiple food-graded materials. Yeast membranes (YMS), as unitary encapsulation material, not only can load natural products by their porous structure but also can be specifically degraded by β-glucanase in intestine. Therefore, this study fabricated Malus baccata (Linn.) polyphenols loaded nanoparticles based on yeast membranes (YMS@MBP) by hydrogen bonding and hydrophobic interaction. YMS@MBP with the lamellar aggregated morphology possessed the non-crystalline feature and excellent thermal stability, and their average particle size was 997.2 ± 22.1 nm. Through establishing the model of gastrointestinal digestion in vitro, YMS@MBP presented the sustained release and intestinal targeting release characteristics, and the maximum release rates in gastric and small intestine were 16.04 % and 79.39 %, respectively. HPLC-MS/MS analysis showed that MBP were mainly composed by quercetin and its derivatives, phloretin, catechins, anthocyanins and phenolic acids. After digestion, the phenolic composition of MBP was perfectly protected by encapsulation of YMS, which was much closer to that of undigested MBP. This study provides a new strategy for construction of polyphenols delivery system applied in functional food field.
Collapse
Affiliation(s)
- Qiuying Yao
- Nano-biotechnology Key Laboratory of Hebei Province, State Key Laboratory of Metastable Materials Science and Technology, School of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, PR China
| | - Huipeng Zhu
- Nano-biotechnology Key Laboratory of Hebei Province, State Key Laboratory of Metastable Materials Science and Technology, School of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, PR China
| | - Lu Wang
- Nano-biotechnology Key Laboratory of Hebei Province, State Key Laboratory of Metastable Materials Science and Technology, School of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, PR China.
| | - Zhigang Zuo
- Intensive care department, Qinhuangdao First Hospital, Qinhuangdao 066000, PR China
| | - Xiaoyu Li
- Nano-biotechnology Key Laboratory of Hebei Province, State Key Laboratory of Metastable Materials Science and Technology, School of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, PR China.
| | - Dawei Gao
- Nano-biotechnology Key Laboratory of Hebei Province, State Key Laboratory of Metastable Materials Science and Technology, School of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, PR China
| |
Collapse
|
29
|
Tang L, Li J, Luan M, Qin M, Zhong C, Zhang Y, Xie Y, Shi M, Qiu L, Yu J. Edgeworthia gardneri (Wall.) Meisn protects against HFD-induced murine atherosclerosis through improving gut microbiota-mediated intestinal barrier integrity. Atherosclerosis 2025; 403:119132. [PMID: 40015156 DOI: 10.1016/j.atherosclerosis.2025.119132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/18/2025] [Accepted: 02/07/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Gut microbiota plays a crucial role in the development and progression of atherosclerosis. Edgeworthia gardneri (Wall.) Meisn, a member of the Thymelaeaceae family and the Edgeworthia genus, has been previously shown in our studies to attenuate atherogenesis when administered orally as an ethanolic extract (EEEG). However, the interaction between EEEG and gut microbiota, and the mechanism by which gut microbiota exerts anti-atherosclerotic effects, remains unclear. AIMS This study aims to determine whether the anti-atherosclerotic properties of EEEG are associated with gut microbiota remodeling. METHOD Atherosclerosis was induced in ApoE-/- mice using a high-fat diet (HFD). The mice were treated with EEEG or Lactobacillus plantarum for 16 weeks. The composition of gut microbiota was analyzed through 16S rDNA sequencing. To assess whether the anti-atherosclerotic effects of EEEG depend on the gut microbiota, HFD-fed mice were treated with a cocktail of antibiotics or underwent fecal microbiota transplantation (FMT). Simultaneously, plaque areas in the aortic roots and whole aortas of apolipoprotein E deficient (ApoE-/-) mice were evaluated using oil red O staining and hematoxylin-eosin staining. Serum levels of LPS, fluorescein isothiocyanate-dextran, and expression levels of tight junction proteins were measured to identify the effects of EEEG on gut barrier dysfunction in HFD-fed ApoE-/- mice. RESULTS The results revealed that EEEG treatment significantly reduced atherosclerotic lesions by ameliorating lipid accumulation and preserving gut barrier integrity. The protective effects were abrogated by antibiotics administration, concomitant with an increase in gut barrier permeability by decreasing expression of tight junction proteins. The microbial analysis indicated an augmented abundance of Lactobacillus, Turicibacter, Faecalibacterium, Akkermansia, and Desulfovibrio following EEEG treatment. Meanwhile, transplantation of fecal microbiota from EEEG-treated mice exerted the anti-atherosclerotic effect in the high-fat diet (HFD)-fed ApoE-/- recipient mice, accompanied by improvement of gut barrier integrity through upregulation of tight junction protein expression. Furthermore, exogenous supplementation of Lactobacillus plantarum mitigated AS in ApoE-/- mice and improved the gut epithelial barrier function by increasing the expression level of Zo-1. CONCLUSION These results suggest that the anti-atherosclerotic efficacy of EEEG is attributed to the preservation of gut barrier integrity mediated by gut microbiota. EEEG and its enriched Lactobacillus plantarum may be promising adjuncts for AS management. IMPORTANCE Atherosclerosis (AS) is the primary pathological basis of cardiovascular disease (CVD). The gut microbiota is known to play an important role in the development and progression of atherosclerosis. In the clinical management of AS, pharmacological classes such as antioxidants, lipid-lowering drugs, and antiplatelet agents are commonly utilized. Despite their ability to decelerate the progression of AS, complications and adverse reactions still limit their application. Edgeworthia gardneri (Wall.) Meisn, a member of the Thymelaeaceae family and Edgeworthia Meisn genus, has been shown in previous studies to attenuate atherogenesis when orally administered as an ethanolic extract (EEEG). However, the interaction between EEEG and the gut microbiota, as well as the mechanism by which the gut microbiota exerts its anti-atherosclerotic effects, remain unclear. The significance of our research lies in identifying the mechanism behind the anti-atherosclerotic effect of Edgeworthia gardneri. The expected results will provide an important scientific basis for the clinical development and application of Edgeworthia gardneri in the prevention and treatment of AS.
Collapse
Affiliation(s)
- Le Tang
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Jiangsheng Li
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Mingxuan Luan
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Manman Qin
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Chao Zhong
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Yifeng Zhang
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Yanfei Xie
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Min Shi
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Liang Qiu
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| | - Jun Yu
- Department of Cardiovascular Sciences and Centre for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
30
|
Hou S, Yu J, Li Y, Zhao D, Zhang Z. Advances in Fecal Microbiota Transplantation for Gut Dysbiosis-Related Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413197. [PMID: 40013938 PMCID: PMC11967859 DOI: 10.1002/advs.202413197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/22/2025] [Indexed: 02/28/2025]
Abstract
This article provides an overview of the advancements in the application of fecal microbiota transplantation (FMT) in treating diseases related to intestinal dysbiosis. FMT involves the transfer of healthy donor fecal microbiota into the patient's body, aiming to restore the balance of intestinal microbiota and thereby treat a variety of intestinal diseases such as recurrent Clostridioides difficile infection (rCDI), inflammatory bowel disease (IBD), constipation, short bowel syndrome (SBS), and irritable bowel syndrome (IBS). While FMT has shown high efficacy in the treatment of rCDI, further research is needed for its application in other chronic conditions. This article elaborates on the application of FMT in intestinal diseases and the mechanisms of intestinal dysbiosis, as well as discusses key factors influencing the effectiveness of FMT, including donor selection, recipient characteristics, treatment protocols, and methods for assessing microbiota. Additionally, it emphasizes the key to successful FMT. Future research should focus on optimizing the FMT process to ensure long-term safety and explore the potential application of FMT in a broader range of medical conditions.
Collapse
Affiliation(s)
- Shuna Hou
- Department of OrthopedicsThe Fourth Affiliated Hospital of China Medical UniversityChina Medical UniversityLiao NingShen Yang110032P. R. China
- Department of general surgeryThe Fourth Affiliated Hospital of China Medical UniversityChina Medical UniversityLiao NingShen Yang110032P. R. China
| | - Jiachen Yu
- Department of OrthopedicsThe Fourth Affiliated Hospital of China Medical UniversityChina Medical UniversityLiao NingShen Yang110032P. R. China
| | - Yongshuang Li
- Department of general surgeryThe Fourth Affiliated Hospital of China Medical UniversityChina Medical UniversityLiao NingShen Yang110032P. R. China
| | - Duoyi Zhao
- Department of OrthopedicsThe Fourth Affiliated Hospital of China Medical UniversityChina Medical UniversityLiao NingShen Yang110032P. R. China
| | - Zhiyu Zhang
- Department of OrthopedicsThe Fourth Affiliated Hospital of China Medical UniversityChina Medical UniversityLiao NingShen Yang110032P. R. China
| |
Collapse
|
31
|
Fu W, Huang Z, Li W, Xu L, Yang M, Ma Y, Liu H, Qian H, Wang W. Copper-luteolin nanocomplexes for Mediating multifaceted regulation of oxidative stress, intestinal barrier, and gut microbiota in inflammatory bowel disease. Bioact Mater 2025; 46:118-133. [PMID: 39760067 PMCID: PMC11697280 DOI: 10.1016/j.bioactmat.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Oxidative stress, dysbiosis, and immune dysregulation have been confirmed to play pivotal roles in the complex pathogenesis of inflammatory bowel disease (IBD). Herein, we design copper ion-luteolin nanocomplexes (CuL NCs) through a metal-polyphenol coordination strategy, which plays a multifaceted role in the amelioration of IBD. The fabricated CuL NCs function as therapeutic agents with exceptional antioxidant and anti-inflammatory capabilities because of their great stability and capacity to scavenge reactive oxygen species (ROS). It can effectively modulate the inflammatory microenvironment including facilitating the efficient reduction of pro-inflammatory cytokine levels, protecting intestinal epithelial cells, promoting mucosal barrier repair and regulating intestinal microbiota. In addition, CuL NCs have been found to enhance cellular antioxidant and anti-inflammatory capacities by regulating the nuclear factor erythroid 2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) oxidative stress pathway and nuclear factor kappa B (NF-κB) signaling pathway, respectively. Notably, CuL NCs demonstrate significant prophylactic and therapeutic efficacy in mouse models with typical IBD, including ulcerative colitis (UC) and Crohn's disease (CD). This study provides a new approach for building multifaceted therapeutic platforms for natural products to treat IBD.
Collapse
Affiliation(s)
- Wanyue Fu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Engineering Research Center for Medical Micro-Nano Devices, Anhui Medical University, Hefei, 230011, PR China
| | - Zhongshi Huang
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang, 443002, PR China
| | - Weiqi Li
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Engineering Research Center for Medical Micro-Nano Devices, Anhui Medical University, Hefei, 230011, PR China
| | - Lingling Xu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Engineering Research Center for Medical Micro-Nano Devices, Anhui Medical University, Hefei, 230011, PR China
| | - Miaomiao Yang
- The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, 230012, PR China
| | - Yan Ma
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Engineering Research Center for Medical Micro-Nano Devices, Anhui Medical University, Hefei, 230011, PR China
| | - Hanghang Liu
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang, 443002, PR China
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Engineering Research Center for Medical Micro-Nano Devices, Anhui Medical University, Hefei, 230011, PR China
| | - Wanni Wang
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Engineering Research Center for Medical Micro-Nano Devices, Anhui Medical University, Hefei, 230011, PR China
| |
Collapse
|
32
|
Ma Z, Wen X, Zhang Y, Ai Z, Zhao X, Dong N, Dou X, Shan A. Thymol Alleviates Colitis by Modulating Intestinal Barrier Damage, Gut Microbiota, and Amino Acid Metabolic Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7211-7227. [PMID: 40077957 DOI: 10.1021/acs.jafc.4c10406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Thymol (THY) is a phenolic monoterpene compound that has garnered attention due to its various biological properties, including antioxidant, anti-inflammatory, and immune-regulatory effects. The purpose of this study was to determine the therapeutic and protective effects of THY in colitic mice, with a particular focus on the mechanisms involving gut microbiota. The results showed that early intervention with THY (40 and 80 mg/kg) not only alleviated the clinical symptoms and colonic damage in mice with dextran sodium sulfate (DSS)-induced colitis but also suppressed the colonic production of inflammatory cytokines (IL-1β, IL-6, and IL-18) and enhanced the expression of mucins (MUC1 and MUC2) and trefoil factor family 3 (TFF3), thereby improving the integrity of the intestinal epithelial barrier. In addition, THY altered the composition of the gut microbiota in colitis mice by increasing the abundance of Bacteroides and reducing the abundance of Proteobacteria. Fecal microbial transplantation (FMT) results demonstrated that FM from THY donor mice significantly improved symptoms of inflammatory bowel disease (IBD), confirming the crucial role of the gut microbiota. Metagenomic and untargeted metabolomic studies found that the characteristic microbiota of THY is Prevotellaceae, and THY significantly upregulated the amino acid metabolic pathways related to arginine and proline metabolism, arginine biosynthesis, and glycerophospholipid metabolism. In summary, THY holds significant potential as a functional additive to enhance host intestinal activity.
Collapse
Affiliation(s)
- Ziwen Ma
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Xin Wen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Yahan Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Zichun Ai
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Xinyi Zhao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Na Dong
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Xiujing Dou
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| |
Collapse
|
33
|
Zhang Y, Lin H, Xiong Y, Zhang Z, Zeng L, Liu Z. Fu Brick Tea Protects the Intestinal Barrier and Ameliorates Colitis in Mice by Regulating Gut Microbiota. Foods 2025; 14:1122. [PMID: 40238292 PMCID: PMC11989102 DOI: 10.3390/foods14071122] [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: 02/09/2025] [Revised: 03/16/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
Ulcerative colitis (UC) pathogenesis is strongly linked to gut microbiota dysbiosis and compromised intestinal barrier integrity. Emerging evidence suggests that targeted dietary interventions may restore microbial homeostasis and ameliorate colitis progression. In this study, we evaluated the therapeutic potential of Fu Brick tea (FBT) using a dextran sulfate sodium (DSS)-induced murine colitis model. The results indicated that oral administration of FBT extract significantly improved the disease index, reduced inflammatory response, protected intestinal barrier protein (e.g., ZO-1), and maintained intestinal structure integrity. Furthermore, FBT intake increased the diversity of gut microbiota, promoted the growth of beneficial bacteria (e.g., Akkermansia), inhibited the proliferation of harmful bacteria (e.g., Desulfovibrioceae, Escherichia, and Helicobacter), restored intestinal homeostasis, and alleviated colitis symptoms including diarrhea. These findings position FBT as a promising nutraceutical candidate for UC management via multi-target modulation of mucosal immunity and microbial ecology.
Collapse
Affiliation(s)
- Yangbo Zhang
- School of Pharmacy, Shaoyang University, Shaoyang 422000, China;
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; (H.L.); (Y.X.)
| | - Haiyan Lin
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; (H.L.); (Y.X.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Yifan Xiong
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; (H.L.); (Y.X.)
| | - Zhixu Zhang
- Yuelushan Laboratory, Changsha 410128, China
| | - Li Zeng
- School of Pharmacy, Shaoyang University, Shaoyang 422000, China;
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; (H.L.); (Y.X.)
- Yuelushan Laboratory, Changsha 410128, China
| |
Collapse
|
34
|
Xu Q, Xue L, Wu Z, Kang S, Li J, Wu Y, Wu Y, Zhao J, Wu R, Lv H, Wang J, Han D. Dietary Qiwenghuangbo powder-enriched Limosilactobacillus reuteri protects the intestinal epithelium and alleviates inflammation via a strain-specific mechanism. Animal Model Exp Med 2025. [PMID: 40109036 DOI: 10.1002/ame2.70016] [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: 01/11/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Qiwenghuangbo powder (QP), composed of Astragalus, Phellodendron chinensis, and Radix pulsatilla, is a traditional Chinese herbal formula, but its effects on weaned piglets remained unclear. METHODS Weaned piglets fed with 0.5 kg/t QP (QP1), 1 kg/t QP (QP2), low-zinc oxide (ZnO; negative control), and high-ZnO (positive control) diets in two phases, respectively, and the growth performance, intestinal morphology, cytokines, and microbial communities were profiled. The mouse models of colitis induced by Citrobacter rodentium and dextran sulfate sodium (DSS) were employed to elucidate the potential role of QP-fed enriched key species. RESULTS Dietary 1.0 kg/t QP alleviated diarrhea and inflammation and improved intestinal development and growth performance of weaned piglets. Moreover, this dietary intervention notably altered microbiota composition, characterized by the enrichment of Limosilactobacillus reuteri. Furthermore, out of three isolated L. reuteri, two strains could alleviate pathogen infection and intestinal inflammation, respectively. Specifically, the anti-inflammatory effect of one strain was achieved by promoting the colonization resistance of C. rodentium as significantly reduced pathogen loads. The other strain mitigated DSS-induced colitis by enhancing the goblet cell function and inhibiting the secretion of pro-inflammatory cytokines, particularly interleukin-1β (IL-1ß) and tumor necrosis factor-α (TNF-α). CONCLUSIONS Dietary QP improved the growth performance and intestinal health of weaned piglets by promoting the colonization of L. reuteri. The isolated commensal L. reuteri control colitis in a strain-specific mechanism, highlighting the potential of QP and L. reuteri in providing evidence for gut health promotion.
Collapse
Affiliation(s)
- Qian Xu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lei Xue
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhenhua Wu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shuaishuai Kang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jia Li
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yifan Wu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yujun Wu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Rujuan Wu
- Peking Centre Technology Co., Ltd., Beijing, China
| | - Huiyuan Lv
- Peking Centre Technology Co., Ltd., Beijing, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
35
|
Luo T, Hou L, Cao Y, Li M, Sheng X, Cheng W, Yan L, Zheng L. Tea Extracellular Vesicle-Derived MicroRNAs Contribute to Alleviate Intestinal Inflammation by Reprogramming Macrophages. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6745-6757. [PMID: 40047388 DOI: 10.1021/acs.jafc.5c01990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
The clinical use of conventional medications for inflammatory bowel disease (IBD) is often limited by significant side effects. The extracellular vesicles derived from plant-based diets have shown promise in mitigating disease. Here, we discovered that natural extracellular vesicles from tea (TEVs) can achieve an appropriate transition from proinflammatory (M1) to anti-inflammatory (M2) macrophages and inhibit inflammation response both in vitro and in vivo. More importantly, the therapeutic effects of TEVs were at least partially attributed to RNA in a DSS-induced colitis model. Small RNA sequencing revealed a distinct enrichment of miRNAs in TEVs, with target genes primarily linked to IBD. TEVs were absorbed by macrophages in a time-dependent manner, carrying miRNAs that modulate gene expression within host cells. Notably, TEV-derived osa-miR166d-5p and gma-miR396a-3p were shown to enhance M2 macrophage polarization and reduce inflammation in vitro. Mechanistically, the osa-miR166d-5p- and gma-miR396a-3p-mediated targeting of the 3'-UTRs of AKT1 and IKBKB decreased NF-κB levels. Overall, we demonstrated that TEVs can ameliorate mouse colitis by reprogramming macrophage polarization and contain a unique miRNA repertoire, including osa-miR166d-5p and gma-miR396a-3p, with a novel function of alleviating intestinal inflammation.
Collapse
Affiliation(s)
- Tianyu Luo
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Linhai Hou
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Yaqi Cao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Meiqi Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Xinyue Sheng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Wenqi Cheng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Ling Yan
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
- Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230009, China
- Anhui Province Key Laboratory of Agricultural Products Modern Processing, Hefei 230009, China
| | - Lei Zheng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
- Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230009, China
- Research Laboratory of Agricultural Environment and Food Safety, Anhui Modern Agricultural Industry Technology System, Hefei 230009, China
| |
Collapse
|
36
|
Zhang Z, Ding Y, Yuan H, Rui C, Fan P, Ji Y, Xiao Y, Dai J, Li L. A multiple-crosslinked injectable hydrogel for modulating tissue microenvironment and accelerating infected diabetic wound repair. J Nanobiotechnology 2025; 23:218. [PMID: 40102884 PMCID: PMC11917161 DOI: 10.1186/s12951-025-03285-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 03/02/2025] [Indexed: 03/20/2025] Open
Abstract
Elevated oxidative stress and inflammation, bacterial infections, and vascular impairment undoubtedly impede the normal diabetic wound healing process, which has encouraged the development of high-performance dressings for wound management. Herein, a new type of multiple-crosslinked injectable hydrogel, GCP, was developed via the radical polymerization of propenyl groups and the formation of copper‒polyphenol coordination bonds and Schiff base bonds. The copper‒polyphenol coordination and Schiff base bonds in the GCP hydrogel were disrupted in the acidic microenvironment of diabetic wound, resulting in the release of copper ions and protocatechualdehyde (PA) to scavenge reactive oxygen species (ROS), promote angiogenesis and cell migration, and exert antibacterial and anti-inflammatory activities via the CuPA complexes. Consequently, markedly accelerated infected diabetic wounds healing was achieved through this tissue microenvironment remodeling strategy. Moreover, the underlying mechanism of the antibacterial properties was investigated by 16S rRNA sequencing. The results indicated that the CuPA complexes can clearly inhibit the growth and reproduction of S. aureus by downregulating specific genes associated with ABC transporters, hindering bacterial protein synthesis, and enhancing oxidoreductase activity. This innovative hydrogel platform for wound management may inspire new methods for the preparation of high-performance biomedical materials and the treatment of other clinical diseases.
Collapse
Affiliation(s)
- Zhengduo Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Yuanyuan Ding
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Huipu Yuan
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Chen Rui
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Pengfei Fan
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Yinwen Ji
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Ying Xiao
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China.
| | - Jiayong Dai
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China.
| | - Lei Li
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China.
| |
Collapse
|
37
|
Fan J, Wu Y, Wang X, Ullah H, Ling Z, Liu P, Wang Y, Feng P, Ji J, Li X. The probiotic enhances donor microbiota stability and improves the efficacy of fecal microbiota transplantation for treating colitis. J Adv Res 2025:S2090-1232(25)00177-8. [PMID: 40089059 DOI: 10.1016/j.jare.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025] Open
Abstract
INTRODUCTION The stability and metabolic functionality of donor microbiota are critical determinants of fecal microbiota transplantation (FMT) efficacy in inflammatory bowel disease (IBD). While probiotics show potential to enhance microbiota resilience, their role in optimizing donor microbiota for FMT remains underexplored. OBJECTIVES This study investigated whether pretreatment of donor microbiota with L. plantarum GR-4 could improve FMT outcomes in a DSS-induced colitis model by modulating microbial stability, metabolic activity, and host-microbiome interactions. METHODS Donor mice received L. plantarum GR-4 for 3 weeks to generate modified FMT (MFMT). DSS-colitis mice were treated with MFMT, conventional FMT, or 5-aminosalicylic acid (5-ASA). Multi-omics analyses and functional assays (stress resistance, engraftment efficiency) were used to evaluate therapeutic mechanisms. RESULTS GR-4 pretreatment conferred three key advantages to donor microbiota: Ecological stabilization: 1. GR-4-driven acidification (pH 3.97 vs. 4.59 for LGG, p < 0.0001) enriched butyrogenic Butyricicoccus (73 % butyrate increase, p < 0.05) and improved stress resistance to bile acids/gastric conditions (1.25 × survival vs. FMT). 2. Metabolic reprogramming: GR-4 metabolized 25.3 % of tryptophan (vs. 10.3 % for LGG) to generate immunomodulatory indoles (ILA, IAA), activating aryl hydrocarbon receptor (AHR) signaling and upregulating anti-inflammatory IL-10/IL-22. 3. Bile acid remodeling: MFMT restored sulfolithocholic acid and β-MCA levels, outperforming FMT in resolving DSS-induced dysregulation. MFMT achieved an 83 % remission rate (vs. 50 % for FMT), enhanced gut barrier integrity, and reversed colitis-associated metabolic dysregulation (e.g., elevated spermidine, 7-sulfocholic acid). Probiotic preconditioning improved donor engraftment by 1.25 × and enriched success-associated taxa (Sporobacter, Butyricimonas), while suppressing pathogens (Clostridium papyrosolvens). CONCLUSIONS L. plantarum GR-4 optimizes donor microbiota via pH-driven niche engineering, immunometabolic reprogramming, and bile acid modulation, addressing key limitations of conventional FMT. The multi-targeted efficacy of MFMT, evidenced by superior remission rates and metabolic restoration, establishes this approach as a translatable strategy for IBD therapy. This study establishes probiotic-enhanced FMT as a paradigm for precision microbiome interventions.
Collapse
Affiliation(s)
- Jingjing Fan
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Ying Wu
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Xing Wang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Habib Ullah
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Zhenmin Ling
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Pu Liu
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Yu Wang
- Nutrition and Health Research Center, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Pengya Feng
- Department of Children Rehabilitation Medicine, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Jing Ji
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China.
| | - Xiangkai Li
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China.
| |
Collapse
|
38
|
Ye L, Yao Z, Xuan Q, Liu Q, Bo T. The impact of sleeve gastrectomy on MASH development by regulating the composition of gut microbiota and metabolic homeostasis. Biochem Biophys Res Commun 2025; 752:151466. [PMID: 39938449 DOI: 10.1016/j.bbrc.2025.151466] [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: 11/22/2024] [Revised: 01/21/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
The prevalence of metabolic dysfunction-associated steatohepatitis (MASH) is increasing annually, which is a global public health issue. Although clinical trials are lacking, observational studies indicate that bariatric surgery can alleviate the progression of MASH. Here, we performed sleeve gastrectomy (SG) and Sham surgery on 8-week-old mice, and then fed a AMLN diet for 24 weeks to construct a diet-inducted MASH mice model after 4-week post-surgery recovery. Applying a multi-omics approach combining metagenomics, metabolomics, and transcriptomics, we found that SG prevents the development of hepatic steatosis, inflammation, and fibrosis in MASH mice not only by significantly altering the structure of gut microbiota including s_Akkermansia muciniphila, s_Alistiples dispar, g_Helicobacter and s_uc_Oscillospiraceae, but also by modulating the levels of serum metabolites including l-arginine and taurocholic acid (TCA). These results suggest that SG and the alteration of gut microbiota and its related serum metabolites can be served as the effective therapeutic strategies for MASH.
Collapse
Affiliation(s)
- Lingxi Ye
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Zhenyu Yao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Qiuhui Xuan
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Qiaoran Liu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250021, China; Key Laboratory of Metabolism and Gastrointestinal Tumor, the First Affiliated Hospital of Shandong First Medical University, China; Key Laboratory of Laparoscopic Technology, the First Affiliated Hospital of Shandong First Medical University, China.
| | - Tao Bo
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| |
Collapse
|
39
|
Li J, Liu T, Xian M, Zhou K, Wei J. The Power of Exercise: Unlocking the Biological Mysteries of Peripheral-Central Crosstalk in Parkinson's Disease. J Adv Res 2025:S2090-1232(25)00143-2. [PMID: 40049515 DOI: 10.1016/j.jare.2025.03.001] [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: 07/23/2024] [Revised: 01/06/2025] [Accepted: 03/01/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Exercise is a widely recognized non-pharmacological treatment for Parkinson's Disease (PD). The bidirectional regulation between the brain and peripheral organs has emerged as a promising area of research, with the mechanisms by which exercise impacts PD closely linked to the interplay between peripheral signals and the central nervous system. AIM OF REVIEW This review aims to summarize the mechanisms by which exercise influences peripheral-central crosstalk to improve PD, discuss the molecular processes mediating these interactions, elucidate the pathways through which exercise may modulate PD pathophysiology, and identify directions for future research. KEY SCIENTIFIC CONCEPTS OF REVIEW This review examines how exercise-induced cytokine release promotes neuroprotection in PD. It discusses how exercise can stimulate cytokine secretion through various pathways, including the gut-brain, muscle-brain, liver-brain, adipose-brain, and bone-brain axes, thereby alleviating PD symptoms. Additionally, the potential contributions of the heart-brain, lung-brain, and spleen-brain axes, as well as multi-axis crosstalk-such as the brain-gut-muscle and brain-gut-bone axes-are explored in the context of exercise therapy. The study highlights the need for further research into peripheral-central crosstalk and outlines future directions to address challenges in clinical PD therapy.
Collapse
Affiliation(s)
- Jingwen Li
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Ke Zhou
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China.
| | - Jianshe Wei
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China; Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
40
|
Xu X, Li K, Liu Q, Zhang H, Li L. Epigallocatechin Gallate Alleviates Lipopolysaccharide-Induced Intestinal Inflammation in Wenchang Chicken by Inhibiting the TLR4/MyD88/NF-κB Signaling Pathway. Vet Sci 2025; 12:225. [PMID: 40266904 PMCID: PMC11945909 DOI: 10.3390/vetsci12030225] [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: 01/13/2025] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 04/25/2025] Open
Abstract
Intestinal inflammation significantly compromises broiler health and adversely affects growth performance. Epigallocatechin gallate (EGCG) was found to maintain the gut health of animals. However, the role and mechanism of EGCG in preventing lipopolysaccharide (LPS)-induced intestinal inflammation in chicks have not yet been fully elucidated. In the 35-day study, 140 one-day-old Wenchang chickens were randomly assigned to four treatments: CON (basal diet), LPS (basal diet + 1 mg/kg body weight (BW) LPS), L-EGCG (basal diet + 40 mg/kg BW EGCG + 1 mg/kg BW LPS), and H-EGCG (basal diet + 60 mg/kg BW EGCG + 1 mg/kg BW LPS). On days 31, 33, and 35 of age, broilers in the LPS, L-EGCG, and H-EGCG treatments received intraperitoneal injections of LPS. The LPS reduced jejunal villus height, villus height/crypt depth ratio, Claudin1 mRNA, catalase (CAT) activity, and interleukin-10 (IL-10) levels compared to CON while elevating diamine oxidase (DAO), interleukin-1β (IL-1β), and tumor necrosis factor α (TNF-α). EGCG improved growth performance in LPS-challenged broilers, elevating jejunal villus height and Claudin1/ZO-1 mRNA with reduced serum DAO. It enhanced antioxidant capacity via increased serum total antioxidant capacity (T-AOC), total superoxide dismutase (T-SOD), CAT, glutathione peroxidase (GSH-Px) activities, and a decreased malondialdehyde (MDA) concentration. Concurrently, EGCG lowered IL-1β/TNF-α and raised IL-10 in serum/jejunum. Crucially, EGCG suppressed jejunal TLR4/MyD88/NF-κB mRNA and protein expression under LPS. These findings demonstrate EGCG's protective role against LPS-induced intestinal inflammation in Wenchang chickens through TLR4/MyD88/NF-κB pathway inhibition.
Collapse
Affiliation(s)
- Xin Xu
- Key Laboratory of Tropical Animal Breeding and Epidemic Disease Research of Hainan Province, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (X.X.); (K.L.); (Q.L.); (H.Z.)
| | - Kunpeng Li
- Key Laboratory of Tropical Animal Breeding and Epidemic Disease Research of Hainan Province, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (X.X.); (K.L.); (Q.L.); (H.Z.)
- School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Qian Liu
- Key Laboratory of Tropical Animal Breeding and Epidemic Disease Research of Hainan Province, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (X.X.); (K.L.); (Q.L.); (H.Z.)
| | - Haiwen Zhang
- Key Laboratory of Tropical Animal Breeding and Epidemic Disease Research of Hainan Province, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (X.X.); (K.L.); (Q.L.); (H.Z.)
| | - Lianbin Li
- Key Laboratory of Tropical Animal Breeding and Epidemic Disease Research of Hainan Province, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (X.X.); (K.L.); (Q.L.); (H.Z.)
| |
Collapse
|
41
|
Wang Z, Liu T, Liu L, Xie J, Tang F, Pi Y, Zhong Y, He Z, Zhang W, Zheng C. Lactobacillus vaginalis alleviates DSS induced colitis by regulating the gut microbiota and increasing the production of 3-indoleacrylic acid. Pharmacol Res 2025; 213:107663. [PMID: 39961405 DOI: 10.1016/j.phrs.2025.107663] [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: 12/23/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disorder, and its incidence is experiencing an upward trend worldwide. UC can result in gut microbiota dysbiosis, impaired intestinal epithelial barrier, and systemic inflammation, for all of which there is presently no definitive treatment available. Lactobacillus is known to regulate gut microbiota and related metabolites to intervene in the development of UC. The objective of this study was to explore the underlying mechanism through which a novel probiotic, Lactobacillus vaginalis, alleviates DSS-induced colitis. Specifically, L. vaginalis were found to ameliorate the DSS-induced UC phenotype, restore intestinal microbiota balance and intestinal barrier function, and elevate the levels of 3-indoleacrylic acid (IAA) in mouse feces. Furthermore, fecal microbiota transplantation and fecal filtrate transplantation provide additional evidence that L. vaginalis alleviate DSS-induced colitis through metabolic products. Additionally, IAA has been shown to alleviate DSS-induced colitis symptoms, decrease inflammatory responses, and enhance intestinal barrier function. Finally, our findings confirm that L. vaginal and metabolites possess the capability to regulate the immune microenvironment in mice with colitis. And the RNA-seq analysis suggests that L. vaginal may play a pivotal role in alleviating colitis by modulating the PPAR signaling pathway. In conclusion, our findings suggest that oral administration of L. vaginalis alleviates DSS induced colonic inflammation by increasing the levels of IAA. L. vaginalis, as an emerging probiotic, provides a potential therapeutic strategy for clinical UC.
Collapse
Affiliation(s)
- Zhuoya Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Tian Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Li Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Graduate School of Jiangxi University of Chinese Medicine, Nanchang 330004, PR China
| | - Jian Xie
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Furui Tang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Yimin Pi
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Yuchun Zhong
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Zhidong He
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Wenming Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Province Key Laboratory of Precision Cell Therapy, The Institute of Translational Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China.
| | - Cihua Zheng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
42
|
Li Y, Yue X, Ren X, Pang Y, Wang T, Huangfu B, Mikhailovich ZA, Vasilievich KV, Zhang M, Luan Y, Wang Q, He X. Mare milk and fermented mare milk alleviate dextran sulfate sodium salt-induced ulcerative colitis in mice by reducing inflammation and modulating intestinal flora. J Dairy Sci 2025; 108:2182-2198. [PMID: 39647629 DOI: 10.3168/jds.2024-25181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/10/2024] [Indexed: 12/10/2024]
Abstract
Mare milk (MM) and fermented mare milk (FM) are specialized animal milks with high nutritional value, containing a variety of functionally active substances that are capable of resisting inflammatory responses and oxidative stress. However, little relevant research on the maintenance of intestinal homeostasis has been performed. This study aimed to investigate the effects of MM and FM on the prevention of dextran sulfate sodium salt (DSS)-induced ulcerative colitis in a mouse model and to preliminarily elucidate the underlying mechanisms. The results showed that MM and FM had different degrees of protective effects against the damage caused by DSS and alleviated ulcerative colitis by inhibiting weight loss, reducing colon length shortening, and restoring intestinal structure. Additionally, MM and FM maintained intestinal tight junction protein levels to repair barrier function, downregulated inflammatory cytokines (e.g., IL-1β, TNF-α, IL-6, and iNOS) and bolstered the body's antioxidant defense system. Moreover, MM and FM regulated dysregulation of the intestinal microenvironment by improving the diversity of the gut microbiota and reshaping its structure, including increasing the proportion of Firmicutes and Bacteroidetes and the relative abundance of beneficial bacterial genera (e.g., Akkermansia). In summary, MM and FMM can serve as dietary resources for preventing ulcerative colitis and maintaining intestinal homeostasis.
Collapse
Affiliation(s)
- Yi Li
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China 100083
| | - Xiaoyu Yue
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China 100193
| | - Xinxin Ren
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China 100083
| | - Yang Pang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China 100083
| | - Teng Wang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China 100083
| | - Bingxin Huangfu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China 100083
| | | | | | - Mu Zhang
- Shenyang Agricultural University, Shenyang, China 110161
| | - Yue Luan
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China 100193
| | - Qin Wang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China 100193.
| | - Xiaoyun He
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China 100083.
| |
Collapse
|
43
|
Li S, Zhuge A, Chen H, Han S, Shen J, Wang K, Xia J, Xia H, Jiang S, Wu Y, Li L. Sedanolide alleviates DSS-induced colitis by modulating the intestinal FXR-SMPD3 pathway in mice. J Adv Res 2025; 69:413-426. [PMID: 38582300 PMCID: PMC11954817 DOI: 10.1016/j.jare.2024.03.026] [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: 11/29/2023] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/08/2024] Open
Abstract
INTRODUCTION Inflammatory bowel disease (IBD) is a global disease with limited therapy. It is reported that sedanolide exerts anti-oxidative and anti-inflammatory effects as a natural phthalide, but its effects on IBD remain unclear. OBJECTIVES In this study, we investigated the impacts of sedanolide on dextran sodium sulfate (DSS)-induced colitis in mice. METHODS The mice were administered sedanolide or vehicle followed by DSS administration, after which colitis symptoms, inflammation levels, and intestinal barrier function were evaluated. Transcriptome analysis, 16S rRNA sequencing, and targeted metabolomics analysis of bile acids and lipids were performed. RESULTS Sedanolide protected mice from DSS-induced colitis, suppressed the inflammation, restored the weakened epithelial barrier, and modified the gut microbiota by decreasing bile salt hydrolase (BSH)-expressing bacteria. The downregulation of BSH activity by sedanolide increased the ratio of conjugated/unconjugated bile acids (BAs), thereby inhibiting the intestinal farnesoid X receptor (FXR) pathway. The roles of the FXR pathway and gut microbiota were verified using an intestinal FXR-specific agonist (fexaramine) and germ-free mice, respectively. Furthermore, we identified the key effector ceramide, which is regulated by sphingomyelin phosphodiesterase 3 (SMPD3). The protective effects of ceramide (d18:1/16:0) against inflammation and the gut barrier were demonstrated in vitro using the human cell line Caco-2. CONCLUSION Sedanolide could reshape the intestinal flora and influence BA composition, thus inhibiting the FXR-SMPD3 pathway to stimulate the synthesis of ceramide, which ultimately alleviated DSS-induced colitis in mice. Overall, our research revealed the protective effects of sedanolide against DSS-induced colitis in mice, which indicated that sedanolide may be a clinical treatment for colitis. Additionally, the key lipid ceramide (d18:1/16:0) was shown to mediate the protective effects of sedanolide, providing new insight into the associations between colitis and lipid metabolites.
Collapse
Affiliation(s)
- Shengjie Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Aoxiang Zhuge
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hui Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shengyi Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jian Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Kaicen Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiafeng Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - He Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shiman Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Youhe Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250000, China.
| |
Collapse
|
44
|
Wu X, Wei J, Ran W, Liu D, Yi Y, Gong M, Liu X, Gong Q, Li H, Gao J. The Gut Microbiota-Xanthurenic Acid-Aromatic Hydrocarbon Receptor Axis Mediates the Anticolitic Effects of Trilobatin. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412234. [PMID: 39836604 PMCID: PMC11904984 DOI: 10.1002/advs.202412234] [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: 10/01/2024] [Revised: 12/16/2024] [Indexed: 01/23/2025]
Abstract
Current treatments for ulcerative colitis (UC) remain limited, highlighting the need for novel therapeutic strategies. Trilobatin (TLB), a naturally derived food additive, exhibits potential anti-inflammatory properties. In this study, a dextran sulfate sodium (DSS)-induced animal model is used to investigate the effects of TLB on UC. It is found TLB significantly alleviates DSS-induced UC in mice, as evidenced by a reduction in the disease activity index, an increase in colon length, improvement in histopathological lesions. Furthermore, TLB treatment results in a decrease in proinflammatory cytokines and an increase in anti-inflammatory cytokines. TLB mitigates UC by modulating the intestinal microbiota, particularly Akkermansia, which enhances tryptophan metabolism and upregulates the production of xanthurenic acid (XANA). To confirm the role of TLB-induced microbiota changes, experiments are performed with pseudogerm-free mice and fecal transplantation. It is also identified XANA as a key metabolite that mediates TLB's protective effects. Both TLB and XANA markedly activate the aromatic hydrocarbon receptor (AhR). Administration of an AhR antagonist abrogates their protective effects, thereby confirming the involvement of AhR in the underlying mechanism. In conclusion, the study reveals a novel mechanism through which TLB alleviates UC by correcting microbiota imbalances, regulating tryptophan metabolism, enhancing XANA production, and activating AhR.
Collapse
Affiliation(s)
- Xiaoyu Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
- Faculty of Functional Food and WineShenyang Pharmaceutical UniversityShenyang110016China
| | - Jiajia Wei
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| | - Wang Ran
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| | - Dongjing Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| | - Yang Yi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| | - Miaoxian Gong
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| | - Xin Liu
- School of Traditional Chinese MedicineLiaoning University of Traditional Chinese MedicineShenyang110016China
| | - Qihai Gong
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| | - Haibo Li
- School of Traditional Chinese MedicineLiaoning University of Traditional Chinese MedicineShenyang110016China
| | - Jianmei Gao
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| |
Collapse
|
45
|
Tao S, Fan J, Li J, Wu Z, Yao Y, Wang Z, Wu Y, Liu X, Xiao Y, Wei H. Extracellular vesicles derived from Lactobacillus johnsonii promote gut barrier homeostasis by enhancing M2 macrophage polarization. J Adv Res 2025; 69:545-563. [PMID: 38508446 PMCID: PMC11954842 DOI: 10.1016/j.jare.2024.03.011] [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: 10/05/2023] [Revised: 02/19/2024] [Accepted: 03/16/2024] [Indexed: 03/22/2024] Open
Abstract
INTRODUCTION Diarrheic disease is a common intestinal health problem worldwide, causing great suffering to humans and animals. Precise manipulation strategies based on probiotics to combat diarrheic diseases have not been fully developed. OBJECTIVES The aim of this study was to investigate the molecular mechanisms by which probiotics manipulate macrophage against diarrheic disease. METHODS Metagenome reveals gut microbiome profiles of healthy and diarrheic piglets. Fecal microbial transplantation (FMT) was employed to explore the causal relationship between gut microbes and diarrhea. The protective role of probiotics and their derived extracellular vesicles (EVs) was investigated in ETEC K88-infected mice. Macrophage depletion was performed to assess the role of macrophages in EVs against diarrhea. Execution of in vitro cell co-culture and transcriptome analyses elucidated the molecular mechanisms by which EVs modulate the macrophage and intestinal epithelial barrier. RESULTS Escherichia coli was enriched in weaned diarrheic piglets, while Lactobacillus johnsonii (L. john) showed a negative correlation with Escherichia coli. The transmission of diarrheic illness symptoms was achieved by transferring fecal microbiota, but not metabolites, from diarrheic pigs to germ-free (GF) mice. L. john's intervention prevented the transmission of disease phenotypes from diarrheic piglets to GF mice. L. john also reduces the gut inflammation induced by ETEC K88. The EVs secreted by L. john demonstrated enhanced efficacy in mitigating the adverse impacts induced by ETEC K88 through the modulation of macrophage phenotype. In vitro experiments have revealed that EVs activate M2 macrophages in a manner that shuts down ERK, thereby inhibiting NLRP3 activation in intestinal epithelial cells. CONCLUSION Our results reveal that intestinal microbiota drives the onset of diarrheic disease and that probiotic-derived EVs ameliorate diarrheic disease symptoms by modulating macrophage phenotypes. These findings can enhance the advancement of innovative therapeutic approaches for diarrheic conditions based on probiotic-derived EVs.
Collapse
Affiliation(s)
- Shiyu Tao
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jinping Fan
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jingjing Li
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhifeng Wu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yong Yao
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhenyu Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Yujun Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Xiangdong Liu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China.
| | - Hong Wei
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
46
|
Bai X, Liu B, Fan D, Lu Y, Zhao X. Modulating the gut microbiota: A novel perspective in colorectal cancer treatment. Cancer Lett 2025; 612:217459. [PMID: 39805389 DOI: 10.1016/j.canlet.2025.217459] [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: 10/08/2024] [Revised: 01/07/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Colorectal cancer (CRC), the second leading cause of cancer-related deaths worldwide, is intricately linked to the dysregulation of the gut microbiota. Manipulating the gut microbiota has emerged as a novel strategy for the prevention and treatment of CRC. Natural products, a pivotal source in new drug discovery, have shown promise in recent research as regulators of the gut microbiota, offering potential applications in the prevention and treatment of CRC. In this work, commencing with a focus on the gut microbiota, we first elucidate the latest research on the intricate relationship between the gut microbiota and CRC. Additionally, we explore the impact of the gut microbiota on immunotherapy and chemotherapy treatments for CRC. Subsequently, we review the latest research findings on the regulation of the gut microbiota for CRC prevention through various mechanisms by natural products. These mechanisms include promoting the growth of beneficial bacteria, eradicating harmful bacteria, and enhancing the synthesis of beneficial metabolites. Furthermore, we summarize the advancements in research on natural products that alleviate chemotherapy toxicity and enhance the efficacy of immunotherapy by modulating the gut microbiota. Ultimately, we aspire to leverage advancements in nanomedicine and multiomics technologies to gain a deeper understanding of the mechanisms by which natural products regulate the gut microbiota. This work leverages gut microbiota as a focal point, aiming to offer new perspectives for developing novel natural products for colorectal cancer prevention and treatment.
Collapse
Affiliation(s)
- Xue Bai
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Boyang Liu
- Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710000, China
| | - Daiming Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yuanyuan Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Xiaodi Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
47
|
Zhao C, Bao L, Shan R, Zhao Y, Wu K, Shang S, Li H, Liu Y, Chen K, Zhang N, Ye C, Hu X, Fu Y. Maternal Gut Inflammation Aggravates Acute Liver Failure Through Facilitating Ferroptosis via Altering Gut Microbial Metabolism in Offspring. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411985. [PMID: 39808540 PMCID: PMC11884527 DOI: 10.1002/advs.202411985] [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: 09/27/2024] [Revised: 01/01/2025] [Indexed: 01/16/2025]
Abstract
Microbial transmission from mother to infant is important for offspring microbiome formation and health. However, it is unclear whether maternal gut inflammation (MGI) during lactation influences mother-to-infant microbial transmission and offspring microbiota and disease susceptibility. In this study, it is found that MGI during lactation altered the gut microbiota of suckling pups by shaping the maternal microbiota in the gut and mammary glands. MGI-induced changes in the gut microbiota of suckling pups lasted into adulthood, resulting in the exacerbation of acute liver failure (ALF) caused by acetaminophen (APAP) in offspring. Specifically, MGI reduced the abundance of Lactobacillus reuteri (L. reuteri) and its metabolite indole-3-acetic acid (IAA) level in adult offspring. L. reuteri and IAA alleviated ALF in mice by promoting intestinal IL-22 production. Mechanistically, IL-22 limits APAP-induced excessive oxidative stress and ferroptosis by activating STAT3. The intestinal abundances of L. reuteri and IAA are inversely associated with the progression of patients with ALF. Overall, the study reveals the role of MGI in mother-to-infant microbial transmission and disease development in offspring, highlighting potential strategies for intervention in ALF based on the IAA-IL-22-STAT3 axis.
Collapse
Affiliation(s)
- Caijun Zhao
- Department of GynecologyChina‐Japan Union Hospital of Jilin UniversityChangchun130033China
- Department of Clinical Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchun130062China
| | - Lijuan Bao
- Department of Clinical Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchun130062China
| | - Ruping Shan
- Department of Clinical Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchun130062China
| | - Yihong Zhao
- Department of Clinical Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchun130062China
| | - Keyi Wu
- Department of Clinical Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchun130062China
| | - Shan Shang
- Department of Clinical Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchun130062China
| | - Haiqi Li
- Department of NeurologyChina‐Japan Union Hospital of Jilin UniversityChangchun130033China
| | - Yi Liu
- Department of Orthopedic CenterThe First Hospital of Jilin UniversityChangchun130012China
| | - Ke Chen
- Department of GynecologyChina‐Japan Union Hospital of Jilin UniversityChangchun130033China
| | - Naisheng Zhang
- Department of Clinical Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchun130062China
| | - Cong Ye
- Department of GynecologyChina‐Japan Union Hospital of Jilin UniversityChangchun130033China
| | - Xiaoyu Hu
- Department of Clinical Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchun130062China
| | - Yunhe Fu
- Department of Clinical Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchun130062China
| |
Collapse
|
48
|
Hua D, Yang Q, Li X, Zhou X, Kang Y, Zhao Y, Wu D, Zhang Z, Li B, Wang X, Qi X, Chen Z, Cui G, Hong W. The combination of Clostridium butyricum and Akkermansia muciniphila mitigates DSS-induced colitis and attenuates colitis-associated tumorigenesis by modulating gut microbiota and reducing CD8 + T cells in mice. mSystems 2025; 10:e0156724. [PMID: 39840995 PMCID: PMC11834468 DOI: 10.1128/msystems.01567-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
The gut microbiota is closely associated with inflammatory bowel disease (IBD) and colorectal cancer (CRC). Probiotics such as Clostridium butyricum (CB) or Akkermansia muciniphila (AKK) have the potential to treat inflammatory bowel disease (IBD) or colorectal cancer (CRC). However, research on the combined therapeutic effects and immunomodulatory mechanisms of CB and AKK in treating IBD or CRC has never been studied. This study evaluates the potential of co-administration of CB and AKK in treating DSS/AOM-induced IBD and colitis-associated CRC. Our results indicate that compared to mono-administration, the co-administration of CB and AKK not only significantly alleviates symptoms such as weight loss, colon shortening, and increased Disease Activity Index in IBD mice but also regulates the gut microbiota composition and effectively suppresses colonic inflammatory responses. In the colitis-associated CRC mice model, a combination of CB and AKK significantly alleviates weight loss and markedly reduces inflammatory infiltration of macrophages and cytotoxic T lymphocytes (CTLs) in the colon, thereby regulating anti-tumor immunity and inhibiting the occurrence of inflammation-induced CRC. In addition, we found that the combined probiotic therapy of CB and AKK can enhance the sensitivity of colitis-associated CRC mice to the immune checkpoint inhibitor anti-mouse PD-L1 (aPD-L1), significantly improving the anti-tumor efficacy of immunotherapy and the survival rate of colitis-associated CRC mice. Furthermore, fecal microbiota transplantation therapy showed that transplanting feces from CRC mice treated with the co-administration of CB and AKK into other CRC mice alleviated the tumor loads in the colon and significantly extended their survival rate. Our study suggests that the combined use of two probiotics, CB and AKK, can not only alleviate chronic intestinal inflammation but also inhibit the progression to CRC. This may be a natural and relatively safe method to support the gut microbiota and enhance the host's immunity against cancer. IMPORTANCE Our study suggests that the combined administration of CB and AKK probiotics, as opposed to a single probiotic strain, holds considerable promise in preventing the advancement of IBD to CRC. This synergistic effect is attributed to the ability of this probiotic combination to more effectively modulate the gut microbiota, curb inflammatory reactions, bolster the efficacy of immunotherapeutic approaches, and optimize treatment results via fecal microbiota transplantation.
Collapse
Affiliation(s)
- Dengxiong Hua
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Qin Yang
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Xiaowei Li
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Xuexue Zhou
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Yingqian Kang
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Yan Zhao
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
| | - Daoyan Wu
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Zhengrong Zhang
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Boyan Li
- School of Public Health, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xinxin Wang
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou, China
| | - Zhenghong Chen
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou, China
| | - Guzhen Cui
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou, China
| | - Wei Hong
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou, China
| |
Collapse
|
49
|
Dobani S, Kirsty Pourshahidi L, Ternan NG, McDougall GJ, Pereira-Caro G, Bresciani L, Mena P, Almutairi TM, Crozier A, Tuohy KM, Del Rio D, Gill CIR. A review on the effects of flavan-3-ols, their metabolites, and their dietary sources on gut barrier integrity. Food Funct 2025; 16:815-830. [PMID: 39807528 DOI: 10.1039/d4fo04721d] [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: 01/16/2025]
Abstract
Impairment of gut barrier integrity is associated with the pathogenesis of gastrointestinal diseases, including inflammatory bowel disease, colorectal cancer, and coeliac disease. While many aspects of diet have been linked to improved barrier function, (poly)phenols, a broad group of bioactive phytochemicals, are of potential interest. The (poly)phenolic sub-class, flavan-3-ols, have been investigated in some detail owing to their abundance in commonly consumed foods, including grapes, tea, apples, cocoa, berries, and nuts. This review summarises studies on the effects of flavan-3-ols, their microbiome-mediated metabolites, and food sources of these compounds, on gut barrier structure. Extensive evidence demonstrates that flavan-3-ol rich foods, individual flavan-3-ols (e.g., (epi)catechin, epi(gallo)catechin-3-O-gallate, and pro(antho)cyanidins), and their related microbiota-mediated metabolites, could be effective in protecting and restoring the integrity of the gut barrier. In this context, various endpoints are assessed, including transepithelial electrical resistance of the epithelial layer and expression of tight junction proteins and mucins, in ex vivo, in vitro, and animal models. The differences in bioactivity reported for barrier integrity are structure-function dependent, related to the (poly)phenolic source or the tested compound, as well as their dose, exposure time, and presence or absence of a stressor in the experimental system. Overall, these results suggest that flavan-3-ols and related compounds could help to maintain, protect, and restore gut barrier integrity, indicating that they might contribute to the beneficial properties associated with the intake of their dietary sources. However, rigorous and robustly designed human intervention studies are needed to confirm these experimental observations.
Collapse
Affiliation(s)
- Sara Dobani
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK.
| | - L Kirsty Pourshahidi
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK.
| | - Nigel G Ternan
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK.
| | - Gordon J McDougall
- Environmental and Biochemical Sciences Department, The James Hutton Institute, Invergowrie, Dundee, UK
| | - Gema Pereira-Caro
- Department of Agroindustry and Food Quality, IFAPA-Alameda Del Obispo, Córdoba, Spain
| | - Letizia Bresciani
- Human Nutrition Unit, Department of Food and Drug, University of Parma, Parma, Italy
| | - Pedro Mena
- Human Nutrition Unit, Department of Food and Drug, University of Parma, Parma, Italy
- Microbiome Research Hub, Department of Food and Drug, University of Parma, Parma, Italy
| | | | - Alan Crozier
- Department of Chemistry, King Saud University Riyadh, Saudi Arabia
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Kieran M Tuohy
- School of Food Science & Nutrition, University of Leeds, Leeds, UK
| | - Daniele Del Rio
- Human Nutrition Unit, Department of Food and Drug, University of Parma, Parma, Italy
- Microbiome Research Hub, Department of Food and Drug, University of Parma, Parma, Italy
| | - Chris I R Gill
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK.
| |
Collapse
|
50
|
Xu E, Sun Y, Yu Z, Zheng J. Epigallocatechin Gallate Alleviates Cisplatin Induced Intestinal Injury in Rats via Inhibiting NRF2/Keap1 Signaling Pathway and Regulating Gut Microbiota and Metabolites. Mol Nutr Food Res 2025; 69:e202400784. [PMID: 39757492 DOI: 10.1002/mnfr.202400784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025]
Abstract
Cisplatin (CIS) is a broad-spectrum anticancer drug widely used in the clinic; however, one of its side effects is that it can cause intestinal damage such as loss of appetite, vomiting, and diarrhea in patients. Epigallocatechin gallate (EGCG) is one of the main active substances in green tea, which has the effects of antitumor multiple drug resistance, antioxidation, and antiinflammatory properties. The aim of this study was to explore the protective effect of EGCG on CIS-induced intestinal injury in rats. First, physiological indices and HE staining indicated that compared with the control group, the physiological state of rats in the CIS group was worse, and the intestinal tissue was damaged, especially the ileum. In contrast, pretreatment with EGCG (20, 40, and 80 mg/kg) effectively alleviated the intestinal damage induced by CIS, with the 40 mg/kg dose demonstrating the most substantial protective effect. Additionally, 40 mg/kg EGCG pretreatment mitigated CIS-induced morphological and ultrastructural damage to intestinal tissues, reduced bacterial translocation, and preserved the integrity of the intestinal barrier. This treatment also altered the abundance of 19 bacterial species, including Lactobacillus and Shigella, and influenced amino acid metabolism and 15 metabolic pathways, including vitamin B6 metabolism by 16S RNA and metabolome sequencing. Furthermore, the expression of proteins associated with autophagy and the NRF2/Keap1 signaling pathway was inhibited. Lastly, ML385 (NRF2 signaling pathway inhibitor) reversed the protective effects of EGCG. Taken together, our findings indicate that EGCG ameliorates CIS induced hepatoenteric toxicity in rats by regulating the intestinal flora and targeting the Nrf2/Keap1 signal axis.
Collapse
Affiliation(s)
- Enshuang Xu
- Department of Veterinary Surgery, College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yue Sun
- Department of Veterinary Surgery, College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Zhiying Yu
- Department of Veterinary Surgery, College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jiasan Zheng
- Department of Veterinary Surgery, College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|