1
|
Qi C, Li Z, Tu H, Sun F, Guo W, Di C, He R, Ze X, Zhang L, Gao R, Hu P, Yang W, Li K, Liu J, Pan X, Jin Z, Sun J. 2'-FL and cross-feeding bifidobacteria reshaped the gut microbiota of infants with atopic dermatitis ex vivo and prevented dermatitis in mice post-microbiota transplantation through retinol metabolism activation. Gut Microbes 2025; 17:2474148. [PMID: 40025650 PMCID: PMC11881859 DOI: 10.1080/19490976.2025.2474148] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
2'-Fucosyllactose (2'-FL), a predominant human milk oligosaccharide, plays a crucial role in the development of the infant gut microbiota and immune system. However, the microbiota of infants with atopic dermatitis (AD) often has difficulty utilizing 2'-FL. Here, we found that strains from human milk, Bifidobacterium bifidum FN120 and Bifidobacterium longum subsp. longum FN103, utilized 2'-FL for growth by cross-feeding. Through an ex vivo continuous fermentation system, we found that 2'-FL and cross-feeding bifidobacteria synergistically enhanced the production of short-chain fatty acids (SCFAs), particularly acetate and propionate, while reshaping the gut microbiota in infants with AD. The reshaped microbiota was then transplanted into oxazolone-induced mice. We observed that AD symptoms in mice were effectively prevented, with significant changes in the ileum microbiota and increased intestinal SCFA levels. RNA sequencing analysis of Peyer's patches in the small intestine revealed activation of the retinol metabolic pathway. Nontargeted metabolomics analysis revealed a significant increase in plasma retinoate levels, which correlated markedly with AD-related markers. Collectively, our study demonstrated that supplementation with cross-feeding bifidobacteria and 2'-FL reshaped the gut microbiota, activated retinol metabolic pathways, promoted immune tolerance, and thereby prevented AD. Our findings provide novel insights into the therapeutic potential of combining prebiotics and probiotics to modulate the gut - skin axis and support immune tolerance in early life, offering a promising strategy for infantile AD management and prevention.
Collapse
Affiliation(s)
- Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Zhongxia Li
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Huayu Tu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Fang Sun
- Pediatrics, Jiaozhou Maternal and Child Health and Family Planning Service Center, Qingdao, China
| | - Wenbo Guo
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Can Di
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Ruikun He
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Xiaolei Ze
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Lintao Zhang
- Pediatrics, Jiaozhou Maternal and Child Health and Family Planning Service Center, Qingdao, China
| | - Ruijuan Gao
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Pengyue Hu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Wenjing Yang
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Kexin Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Jiayi Liu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Xiaonan Pan
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Zilu Jin
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Sall I, Foxall R, Felth L, Maret S, Rosa Z, Gaur A, Calawa J, Pavlik N, Whistler JL, Whistler CA. Gut dysbiosis was inevitable, but tolerance was not: temporal responses of the murine microbiota that maintain its capacity for butyrate production correlate with sustained antinociception to chronic morphine. Gut Microbes 2025; 17:2446423. [PMID: 39800714 PMCID: PMC11730370 DOI: 10.1080/19490976.2024.2446423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 11/24/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
The therapeutic benefits of opioids are compromised by the development of analgesic tolerance, which necessitates higher dosing for pain management thereby increasing the liability for drug dependence and addiction. Rodent models indicate opposing roles of the gut microbiota in tolerance: morphine-induced gut dysbiosis exacerbates tolerance, whereas probiotics ameliorate tolerance. Not all individuals develop tolerance, which could be influenced by differences in microbiota, and yet no study design has capitalized upon this natural variation. We leveraged natural behavioral variation in a murine model of voluntary oral morphine self-administration to elucidate the mechanisms by which microbiota influences tolerance. Although all mice shared similar morphine-driven microbiota changes that largely masked informative associations with variability in tolerance, our high-resolution temporal analyses revealed a divergence in the progression of dysbiosis that best explained sustained antinociception. Mice that did not develop tolerance maintained a higher capacity for production of the short-chain fatty acid (SCFA) butyrate known to bolster intestinal barriers and promote neuronal homeostasis. Both fecal microbial transplantation (FMT) from donor mice that did not develop tolerance and dietary butyrate supplementation significantly reduced the development of tolerance independently of suppression of systemic inflammation. These findings could inform immediate therapies to extend the analgesic efficacy of opioids.
Collapse
Affiliation(s)
- Izabella Sall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Graduate program in Molecular and Evolutionary Systems Biology, University of New Hampshire, Durham, NH, USA
| | - Randi Foxall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Lindsey Felth
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Soren Maret
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Zachary Rosa
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Anirudh Gaur
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Jennifer Calawa
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Microbiology Graduate Program, University of New Hampshire, Durham, NH, USA
| | - Nadia Pavlik
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Jennifer L. Whistler
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
- Department of Physiology and Membrane Biology, UC Davis School of Medicine, Davis, CA, USA
| | - Cheryl A. Whistler
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
3
|
Gao H, Sun M, Li A, Gu Q, Kang D, Feng Z, Li X, Wang X, Chen L, Yang H, Cong Y, Liu Z. Microbiota-derived IPA alleviates intestinal mucosal inflammation through upregulating Th1/Th17 cell apoptosis in inflammatory bowel disease. Gut Microbes 2025; 17:2467235. [PMID: 39956891 PMCID: PMC11834480 DOI: 10.1080/19490976.2025.2467235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/09/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025] Open
Abstract
The gut microbiota-derived metabolite indole-3-propionic acid (IPA) plays an important role in maintaining intestinal mucosal homeostasis, while the molecular mechanisms underlying IPA regulation on mucosal CD4+ T cell functions in inflammatory bowel disease (IBD) remain elusive. Here we investigated the roles of IPA in modulating mucosal CD4+ T cells and its therapeutic potential in treatment of human IBD. Leveraging metabolomics and microbial community analyses, we observed that the levels of IPA-producing microbiota (e.g. Peptostreptococcus, Clostridium, and Fournierella) and IPA were decreased, while the IPA-consuming microbiota (e.g. Parabacteroides, Erysipelatoclostridium, and Lachnoclostridium) were increased in the feces of IBD patients than those in healthy donors. Dextran sulfate sodium (DSS)-induced acute colitis and CD45RBhighCD4+ T cell transfer-induced chronic colitis models were then established in mice and treated orally with IPA to study its role in intestinal mucosal inflammation in vivo. We found that oral administration of IPA attenuated mucosal inflammation in both acute and chronic colitis models in mice, as characterized by increased body weight, and reduced levels of pro-inflammatory cytokines (e.g. TNF-α, IFN-γ, and IL-17A) and histological scores in the colon. We further utilized RNA sequencing, molecular docking simulations, and surface plasmon resonance analyses and identified that IPA exerts its biological effects by interacting with heat shock protein 70 (HSP70), leading to inducing Th1/Th17 cell apoptosis. Consistently, ectopic expression of HSP70 in CD4+ T cells conferred resistance to IPA-induced Th1/Th17 cell apoptosis. Therefore, these findings identify a previously unrecognized pathway by which IPA modulates intestinal inflammation and provide a promising avenue for the treatment of IBD.
Collapse
Affiliation(s)
- Han Gao
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Mingming Sun
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Ai Li
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Qiaoyan Gu
- Department of Gastroenterology, Yanan University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dengfeng Kang
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Zhongsheng Feng
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Xiaoyu Li
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Xuehong Wang
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liang Chen
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Hong Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yingzi Cong
- Division of Gastroenterology and Hepatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Human Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Zhanju Liu
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| |
Collapse
|
4
|
Zünd JN, Mujezinovic D, Reichlin M, Plüss S, Caflisch M, Robinson S, Lacroix C, Pugin B. Novel cross-feeding human gut microbes metabolizing tryptophan to indole-3-propionate. Gut Microbes 2025; 17:2501195. [PMID: 40336187 PMCID: PMC12064059 DOI: 10.1080/19490976.2025.2501195] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 04/26/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Tryptophan-derived indoles produced by the gut microbiota, particularly indole-3-propionate (IPA), are key compounds associated with gastrointestinal balance and overall health. Reduced levels of IPA have been associated with inflammatory bowel disease, type 2 diabetes, and colorectal cancer. Since fiber-rich diets have been shown to promote IPA, we aimed to decipher fiber-specific effects and identify associated IPA-producing taxa in a range of healthy individuals. We cultured fecal microbiota from 16 adults with tryptophan and eight different dietary fibers and monitored community shifts by 16S rRNA gene amplicon sequencing and tryptophan-derived indoles using targeted liquid chromatography with diode array detection. The concentrations and types of indoles produced were donor-specific, with pectin strongly promoting IPA production in certain donors. IPA production was not associated with any known IPA producer but with the pectin-utilizing species Lachnospira eligens, which produced indole-3-lactate (ILA) in vitro, the IPA precursor. Supplementation of ILA in additional fecal microbiota cultures (n = 6) revealed its effective use as a substrate for IPA production. We identified a novel IPA producer, Enterocloster aldenensis, which produced IPA exclusively from ILA but not from tryptophan. Co-culture of L. eligens and E. aldenensis resulted in IPA production, providing new evidence for an ILA cross-feeding mechanism that may contribute to the IPA-promoting effects observed with pectin. Overall, we highlight the potential for targeted dietary interventions to promote beneficial gut taxa and metabolites.
Collapse
Affiliation(s)
- Janina N. Zünd
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Denisa Mujezinovic
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Markus Reichlin
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Serafina Plüss
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Marina Caflisch
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Serina Robinson
- Department of Environmental Microbiology, Eawag, Dübendorf, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Benoit Pugin
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
5
|
Wang H, Xu F, Wang C. Metabolic reprogramming of tumor microenviroment by engineered bacteria. Semin Cancer Biol 2025; 112:58-70. [PMID: 40157514 DOI: 10.1016/j.semcancer.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/16/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
The tumor microenvironment (TME) is a complex ecosystem that plays a crucial role in tumor progression and response to therapy. The metabolic characteristics of the TME are fundamental to its function, influencing not only cancer cell proliferation and survival but also the behavior of immune cells within the tumor. Metabolic reprogramming-where cancer cells adapt their metabolic pathways to support rapid growth and immune evasion-has emerged as a key factor in cancer immunotherapy. Recently, the potential of engineered bacteria in cancer immunotherapy has gained increasing recognition, offering a novel strategy to modulate TME metabolism and enhance antitumor immunity. This review summarizes the metabolic properties and adaptations of tumor and immune cells within the TME and summarizes the strategies by which engineered bacteria regulate tumor metabolism. We discuss how engineered bacteria can overcome the immunosuppressive TME by reprogramming its metabolism to improve antitumor therapy. Furthermore, we examine the advantages, potential challenges, and future clinical translation of engineered bacteria in reshaping TME metabolism.
Collapse
Affiliation(s)
- Heng Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Fang Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Chao Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
6
|
Guo Z, Yang S, Qi L, Ma X, Wang Y, Li B, He J. Lactobacillus acidophilus KLDS1.0901 ameliorates non-alcoholic fatty liver disease by modulating the tryptophan metabolite indole-3-aldehyde and acting on its receptor AhR. Food Funct 2025. [PMID: 40423690 DOI: 10.1039/d4fo05280c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Our previous study demonstrated that Lactobacillus acidophilus KLDS1.0901 significantly alleviated symptoms of high-fat diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD) and showed a strong association with the gut microbiota; however, the underlying mechanisms remained unclear. In this study, we focused on changes in intestinal metabolic pathways in mice following intervention with Lactobacillus acidophilus KLDS1.0901, using non-targeted metabolomics. Tryptophan metabolism was found to be closely associated with NAFLD alleviation, and indole-3-aldehyde (IAld) was identified as a key target. Animal experiments showed no significant differences in the levels of liver triglycerides, fasting blood glucose, alanine aminotransferase, aspartate aminotransferase, IL-6, IL-1β, TNF-α, and IL-10 between the direct-feeding IAld group and the Lactobacillus acidophilus KLDS1.0901 group. This suggests that the IAId, produced by Lactobacillus acidophilus KLDS1.0901, is the key intermediate mediator responsible for its improvement of NAFLD. The alleviating effect of Lactobacillus acidophilus KLDS1.0901 on NAFLD symptoms was suppressed after the inhibition of the IAld receptor aromatic hydrocarbon receptor (AhR), suggesting that the bacterium relies on the AhR signaling pathway to mediate its effect on NAFLD. Cellular experiments showed that IAld significantly reduced triglyceride content, decreased lipid accumulation, and increased glycogen levels in oleic acid-induced cells. The effects of IAld on gene transcription levels in oleic acid-induced HepG2 cells were further analyzed using high-throughput sequencing. Transcriptomic analysis revealed that IAld regulates key pathways, including the NF-κB, chemokine and AGE-RAGE signaling pathways. Our study identified the ameliorative effects of tryptophan metabolites, particularly IAld, on NAFLD, along with the underlying mechanisms, offering new insights into potential treatment strategies for NAFLD.
Collapse
Affiliation(s)
- Zhengtao Guo
- Food College, Northeast Agricultural University, Harbin 150030, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Shengjun Yang
- Food College, Northeast Agricultural University, Harbin 150030, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Liwen Qi
- Food College, Northeast Agricultural University, Harbin 150030, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Xinming Ma
- Food College, Northeast Agricultural University, Harbin 150030, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Yanbo Wang
- Food College, Northeast Agricultural University, Harbin 150030, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Bailiang Li
- Food College, Northeast Agricultural University, Harbin 150030, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Jian He
- National Center of Technology Innovation for Dairy, Hohhot 010110, China.
| |
Collapse
|
7
|
Hovinen T, Kettunen E, Erkkola M, Suomalainen A, Freese R, Korkalo L. Nutrient intakes and metabolomic profiles associated with animal source energy percentage in children's diets. Sci Rep 2025; 15:18568. [PMID: 40425646 DOI: 10.1038/s41598-025-02114-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
The purpose of the study was to calculate the relative intake of animal source energy (animal source energy percentage, ASEP) and examine its utility in nutrition and metabolic research with a special attention on children. In addition, the capacity of ASEP to identify potential metabolic biomarkers of plant-forward diets was tested. Four-day food records and 7-day dietary screeners of 51 Finnish children aged 1-7 years, consuming a vegan (n = 7), pesco-/lacto-/ovo-vegetarian (n = 11) or omnivorous diet (n = 33), were used for assessing general performance of ASEP as diet measure. Blood samples of 40 children (six vegans, eight vegetarians, 26 omnivores) were analyzed for plasma LDL cholesterol, erythrocyte folate concentration and serum untargeted MS-metabolomics. Estimated ASEPs varied between 0% and 52%. Vegetarians and omnivores showed overlapping ASEPs. ASEP had a strong positive correlation with saturated fatty acid (percent of energy, E%; r = 0.785) and cholesterol intake (r = 0.715) and a strong negative correlation with fibre (r = - 0.811) and polyunsaturated fatty acid (E%; r = - 0.819) intake. ASEP also correlated with plasma LDL cholesterol (r = 0.699) concentration. Serum indoleacrylic acid (IAA) is suggested to be further investigated as a possible biomarker of plant-forward diet. ASEP provides a useful tool for objective and quantitative assessment of diet characteristics in research.
Collapse
Affiliation(s)
- Topi Hovinen
- Research Programs Unit, Stem Cells and Metabolism, University of Helsinki, 00290, Helsinki, Finland
| | - Elina Kettunen
- Department of Food and Nutrition, University of Helsinki, 00014, Helsinki, Finland
| | - Maijaliisa Erkkola
- Department of Food and Nutrition, University of Helsinki, 00014, Helsinki, Finland
| | - Anu Suomalainen
- Research Programs Unit, Stem Cells and Metabolism, University of Helsinki, 00290, Helsinki, Finland
- HUS Diagnostic Center, Helsinki University Hospital, 00290, Helsinki, Finland
- HiLife, University of Helsinki, 00014, Helsinki, Finland
| | - Riitta Freese
- Department of Food and Nutrition, University of Helsinki, 00014, Helsinki, Finland
| | - Liisa Korkalo
- Department of Food and Nutrition, University of Helsinki, 00014, Helsinki, Finland.
| |
Collapse
|
8
|
Ma T, Li Y, Yang N, Wang H, Shi X, Liu Y, Jin H, Kwok LY, Sun Z, Zhang H. Efficacy of a postbiotic and its components in promoting colonic transit and alleviating chronic constipation in humans and mice. Cell Rep Med 2025; 6:102093. [PMID: 40286792 DOI: 10.1016/j.xcrm.2025.102093] [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: 06/26/2024] [Revised: 12/06/2024] [Accepted: 04/01/2025] [Indexed: 04/29/2025]
Abstract
This study evaluates the efficacy of the postbiotic Probio-Eco in alleviating constipation in humans and mice. A randomized, double-blind, placebo-controlled crossover trial involving 110 adults with chronic constipation (Rome IV criteria) demonstrates that a 3-week Probio-Eco intervention significantly improves constipation symptoms, stool straining, and worry scores. Gut microbiota and metabolomic analyses reveal modulations in specific gut microbiota, succinate, tryptophan derivatives, deoxycholate, propionate, butyrate, and cortisol, correlating with symptom relief. A loperamide-induced mouse model confirms that Probio-Eco and its bioactive components (succinate, 3-indoleacrylic acid, and 5-hydroxytryptophan) alleviate constipation by stimulating mucin-2 secretion, regulating intestinal transport hormones, and promoting anti-inflammatory responses. Multi-omics integration identifies key pathways, including succinate-short-chain fatty acid, tryptophan-5-hydroxytryptophan-serotonin, and tryptophan-3-indoleacrylic acid, driving intestinal homeostasis and motility. These findings highlight the comprehensive efficacy of Probio-Eco and provide robust evidence for its clinical application in constipation management. This study was registered at Chinese Clinical Trial Registry (ChiCTR2100054376).
Collapse
Affiliation(s)
- Teng Ma
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Yalin Li
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Ni Yang
- State Key Laboratory of Research and Development of Classical Prescription and Modern Chinese Medicine, 1899 Meiling Road, Nanchang 330103, China
| | - Huan Wang
- Inner Mongolia People's Hospital, Hohhot, China
| | - Xuan Shi
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Yanfang Liu
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Hao Jin
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Lai-Yu Kwok
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Zhihong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Heping Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China.
| |
Collapse
|
9
|
von Ameln Lovison O, Zempulski Volpato FC, Weber LG, Barth AL, Simon Coitinho A, Martins AF. Unveiling the role of the upper respiratory tract microbiome in susceptibility and severity to COVID-19. Front Cell Infect Microbiol 2025; 15:1531084. [PMID: 40433668 PMCID: PMC12106449 DOI: 10.3389/fcimb.2025.1531084] [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: 11/19/2024] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
It is argued that commensal bacteria in the upper respiratory tract (URT) protect against pathogen colonization and infection, including respiratory viruses. Given that the microbiome can mediate immune modulation, a link between the URT microbiome (URTM) and COVID-19 susceptibility and severity is expected. This 16S metagenomics cross-sectional study assessed URTM composition, metabolic prediction, and association with laboratory biomarkers in non-COVID-19 pneumonia (NO-CoV), moderate (M-CoV), severe (S-CoV) COVID-19 patients, as well as COVID-19-negative, asymptomatic (NC) patients. The S-CoV group exhibited reduced URTM diversity, primarily due to a decreased abundance of eubiotic taxa. Some of these taxa (e.g., Haemophilus sp., Neisseria sp.) were also associated with inflammatory biomarkers. Multiple metabolic pathways (e.g., short-chain fatty acids, vitamin B12) linked to immune response, antiviral activity, and host susceptibility showed decreased abundance in S-CoV. These pathways could suggest potential alternatives for the therapeutic arsenal against COVID-19, providing reassurance about the progress in understanding and treating this disease.
Collapse
Affiliation(s)
- Otávio von Ameln Lovison
- Laboratório de Pesquisa em Resistência Bacteriana (LABRESIS), Hospital de Clínicas de Porto Alegre (HCPA), Centro de Pesquisa Experimental, Porto Alegre, Brazil
- Laboratório de Microbiologia e Saúde Única do Instituto de Ciências Básicas da Saúde (ICBS) da Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Núcleo de Bioinformática (Bioinformatics Core), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Fabiana Caroline Zempulski Volpato
- Laboratório de Pesquisa em Resistência Bacteriana (LABRESIS), Hospital de Clínicas de Porto Alegre (HCPA), Centro de Pesquisa Experimental, Porto Alegre, Brazil
| | - Lorenzo Gómez Weber
- Laboratório de Microbiologia e Saúde Única do Instituto de Ciências Básicas da Saúde (ICBS) da Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Núcleo de Bioinformática (Bioinformatics Core), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Afonso Luis Barth
- Laboratório de Pesquisa em Resistência Bacteriana (LABRESIS), Hospital de Clínicas de Porto Alegre (HCPA), Centro de Pesquisa Experimental, Porto Alegre, Brazil
| | - Adriana Simon Coitinho
- Laboratório de Neuroimunologia do Instituto de Ciências Básicas da Saúde (ICBS) da Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Andreza Francisco Martins
- Laboratório de Pesquisa em Resistência Bacteriana (LABRESIS), Hospital de Clínicas de Porto Alegre (HCPA), Centro de Pesquisa Experimental, Porto Alegre, Brazil
- Laboratório de Microbiologia e Saúde Única do Instituto de Ciências Básicas da Saúde (ICBS) da Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Núcleo de Bioinformática (Bioinformatics Core), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| |
Collapse
|
10
|
Fang Y, Min S, Wu Y, Xu F, Chen H, Li Y, Lu Y, Hu J, Zhu L, Shen H. Integration of Multi-Omics and Network Pharmacology Analysis Reveals the Mechanism of Qingchang Huashi Jianpi Bushen Formula in Repairing the Epithelial Barrier of Ulcerative Colitis. J Inflamm Res 2025; 18:6167-6189. [PMID: 40386180 PMCID: PMC12083493 DOI: 10.2147/jir.s510966] [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: 12/20/2024] [Accepted: 04/30/2025] [Indexed: 05/20/2025] Open
Abstract
Purpose Derivation of Qingchang Huashi formula, named Qingchang Huashi Jianpi Bushen (QCHS_JPBS) formula, has shown significant therapeutic effect on patients with ulcerative colitis (UC). In this study, the potential mechanism of QCHS_JPBS formula in repairing mucosal damage was explored from the perspective of intestinal stem cell (ISCs) differentiation, and potential targets of the QCHS_JPBS formula to improve UC were predicted using network pharmacology analysis. Methods The therapeutic efficacy of QCHS_JPBS formula was evaluated in a mouse model of 2.5% dextran sulfate sodium (DSS) induced colitis. The effect of this formula on the ISC differentiation was evaluated using tissue transmission electron microscopy, immunofluorescence, and RT-qPCR. The cecal contents were subjected to 16s RNA sequencing analysis and non-target metabolomics analysis using LC-MS/MS. The fecal microbiota transplantation method verified the essential role of gut microbiota in promoting ISC differentiation and repairing mucosal damage. Results The results indicated that QCHS_JPBS formula suppressed the inflammatory response and repaired the damaged intestinal epithelial barrier in DSS-induced colitis mice. QCHS_JPBS formula promoted ISC differentiation, particularly in the direction of goblet cells. QCHS_JPBS formula restored gut dysbiosis and regulated metabolic disorders in DSS-induced colitis mice. And then, the results of fecal microbiota transplantation indicated that QCHS_JPBS formula promoted differentiation of intestinal stem cells to repair mucosal damage through gut microbiota. Finally, a total of 79 active ingredients of QCHS_JPBS formula were identified based on LC-MS analysis and EGFR, STAT3, SRC, AKT1, and HSP90AA1 were considered as potential therapeutic UC targets of QCHS_JPBS formula based on network pharmacology analysis. Conclusion The present study demonstrated that QCHS_JPBS formula promoted the differentiation of ISCs through gut microbiota to repair the damaged intestinal epithelial barrier in UC mice.
Collapse
Affiliation(s)
- Yulai Fang
- Digestive Disease Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Shichen Min
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yuguang Wu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Feng Xu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Hongxin Chen
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yanan Li
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yizhou Lu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Jingyi Hu
- Digestive Disease Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Lei Zhu
- Digestive Disease Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Hong Shen
- Digestive Disease Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| |
Collapse
|
11
|
Fang L, Peng H, Tan Z, Deng N, Peng X. The Role of Gut Microbiota on Intestinal Fibrosis in Inflammatory Bowel Disease and Traditional Chinese Medicine Intervention. J Inflamm Res 2025; 18:5951-5967. [PMID: 40357383 PMCID: PMC12067688 DOI: 10.2147/jir.s504827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, relapsing inflammatory disorder of the intestine, frequently complicated by intestinal fibrosis. As fibrosis progresses, it can result in luminal stricture and compromised intestinal function, significantly diminishing patients' quality of life. Emerging evidence suggests that gut microbiota and their metabolites contribute to the pathogenesis of IBD-associated intestinal fibrosis by influencing inflammation and modulating immune responses. This review systematically explores the mechanistic link between gut microbiota and intestinal fibrosis in IBD and evaluates the therapeutic potential of traditional Chinese medicine (TCM) interventions. Relevant studies were retrieved from PubMed, Web of Science, Embase, Scopus, CNKI, Wanfang, and VIP databases. Findings indicate that TCM, including Chinese herbal prescriptions and bioactive constituents, can modulate gut microbiota composition and microbial metabolites, ultimately alleviating intestinal fibrosis through anti-inflammatory, immunemodulatory, and anti-fibrotic mechanisms. These insights highlight the potential of TCM as a promising strategy for targeting gut microbiota in the management of IBD-associated fibrosis.
Collapse
Affiliation(s)
- Leyao Fang
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Huiyi Peng
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Zhoujin Tan
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Na Deng
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Xinxin Peng
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| |
Collapse
|
12
|
Zhang S, Zhao R, Wang R, Lu Y, Xu M, Lin X, Lan R, Zhang S, Tang H, Fan Q, Yang J, Liu L, Xu J. Weissella viridescens Attenuates Hepatic Injury, Oxidative Stress, and Inflammation in a Rat Model of High-Fat Diet-Induced MASLD. Nutrients 2025; 17:1585. [PMID: 40362894 PMCID: PMC12073722 DOI: 10.3390/nu17091585] [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: 04/17/2025] [Revised: 05/02/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Metabolic-dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disorder globally. Probiotic supplementation has shown promise in its prevention and treatment. Although Weissella viridescens, a lactic acid bacterium with immunomodulatory effects, has antibacterial and anti-inflammatory activities, there is a lack of direct evidence for its role in alleviating MASLD. This study aimed to investigate the protective effects of W. viridescens strain Wv2365, isolated from healthy human feces, in a high-fat diet (HFD)-induced rat model of MASLD. Methods: Rats were randomly assigned to a normal chow diet (NC), high-fat diet (HFD), and HFD supplemented with W. viridescens Wv2365 (Wv2365) groups. All groups were fed their respective diets for 8 weeks. During this period, the NC and HFD groups received a daily oral gavage of PBS, while the Wv2365 group received a daily oral gavage of Wv2365. Results: Wv2365 supplementation significantly reduced HFD-induced body weight gain, improved NAFLD activity scores, alleviated hepatic injury, and restored lipid metabolism. A liver transcriptomic analysis revealed the downregulation of inflammation-related pathways, along with decreased serum levels of TNF-α, IL-1β, IL-6, MCP-1, and LPS. Wv2365 also activated the Nrf2/HO-1 antioxidant pathway, enhanced hepatic antioxidant enzyme activities and reduced malondialdehyde levels. A gut microbiota analysis showed the enrichment of beneficial genera, including Butyricicoccus, Akkermansia, and Blautia. Serum metabolomic profiling revealed increased levels of metabolites including indole-3-propionic acid, indoleacrylic acid, and glycolithocholic acid. Conclusions: Wv2365 attenuates hepatic injury, oxidative stress, and inflammation in a rat model of high-fat-diet-induced MASLD, supporting its potential as a probiotic candidate for the modulation of MASLD.
Collapse
Affiliation(s)
- Shuwei Zhang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ruiqing Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ruoshi Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yao Lu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Mingchao Xu
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shijiazhuang 050010, China
| | - Xiaoying Lin
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ruiting Lan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Suping Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Huijing Tang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Qianhua Fan
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Jing Yang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing 102206, China
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| | - Liyun Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing 102206, China
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| | - Jianguo Xu
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing 102206, China
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| |
Collapse
|
13
|
Chaves LS, Oliveira ACP, Oliveira AP, Lopes ALF, Araujo AKS, Pacheco G, Silva KC, Martins FEC, Gomes IAB, Ramos SVS, Viana HTMC, Batista AVF, Oliveira BC, Nicolau LAD, Ribeiro FOS, Castro AV, de Araujo-Nobre AR, Silva DA, Cordeiro LMC, Góis MB, Medeiros JVR. Cashew gum fractions protect intestinal mucosa against shiga toxin-producing Escherichia coli infection: Characterization and insights into microbiota modulation. Int J Biol Macromol 2025; 311:143916. [PMID: 40324507 DOI: 10.1016/j.ijbiomac.2025.143916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Diarrheal diseases remain a major public health concern, particularly in regions with poor sanitation. Polysaccharides extracted from natural gums have been investigated as functional agents for intestinal health, and their fractionation enables the production of oligosaccharides with potential prebiotic activity. This study aimed to produce cashew gum (CG) fractions through Smith degradation (CGD48) and partial hydrolysis (CGD24) and to evaluate their ability to modulate and protect the intestinal microbiota. Balb/c mice were administered CG (1200 mg/kg), CGD24 (800 mg/kg), or CGD48 (800 mg/kg) for 10 and 26 days, followed by infection with Shiga toxin-producing Escherichia coli (STEC) (5 × 1010 CFU/mL) for three days. Characterization assays confirmed the fragmentation of CG. Both CGD24 and CGD48 promoted the growth of beneficial bacteria with and without infection and reduced STEC colonization. Furthermore, they preserved mucin levels in the cecum and large intestine and maintained baseline levels of superoxide dismutase (SOD), suggesting protection of the intestinal mucosa. These findings indicate that CG fractions exhibit microbiota-modulating and protective effects against STEC, highlighting their therapeutic potential and the need for further studies to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Letícia S Chaves
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Antonio C P Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Ana P Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - André L F Lopes
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Andreza K S Araujo
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Gabriella Pacheco
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Katriane C Silva
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Francisco E C Martins
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Isaac A B Gomes
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Sabrine V S Ramos
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Hémilly T M C Viana
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Ana V F Batista
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Beatriz C Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Lucas A D Nicolau
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Fábio O S Ribeiro
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Auricélia V Castro
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Alyne Rodrigues de Araujo-Nobre
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Durcilene A Silva
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Lucimara M C Cordeiro
- Department of Biochemistry and Molecular Biology, Federal University of Parana, Curitiba, PR, Brazil
| | - Marcelo B Góis
- Post-Graduation Program in Biosciences and Health, Federal University of Rondonópolis, Rondonópolis, Brazil
| | - Jand V R Medeiros
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil; Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil.
| |
Collapse
|
14
|
Wang J, Cheng W, Yang R. Nervous system-gut microbiota-immune system axis: future directions for preventing tumor. Front Immunol 2025; 16:1535955. [PMID: 40376000 PMCID: PMC12078214 DOI: 10.3389/fimmu.2025.1535955] [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] [Received: 11/28/2024] [Accepted: 04/01/2025] [Indexed: 05/18/2025] Open
Abstract
Tumor is one of the leading causes of death worldwide. The occurrence and development of tumors are related to multiple systems and factors such as the immune system, gut microbiota, and nervous system. The immune system plays a critical role in tumor development. Studies have also found that the gut microbiota can directly or indirectly affect tumorigenesis and tumor development. With increasing attention on the tumor microenvironment in recent years, the nervous system has emerged as a novel regulator of tumor development. Some tumor therapies based on the nervous system have also been tested in clinical trials. However, the nervous system can not only directly interact with tumor cells but also indirectly affect tumor development through the gut microbiota. The nervous system-mediated gut microbiota can regulate tumorigenesis, growth, invasion, and metastasis through the immune system. Here, we mainly explore the potential effects of the nervous system-gut microbiota-immune system axis on tumorigenesis and tumor development. The effects of the nervous system-gut microbiota-immune system axis on tumors involve the nervous system regulating immune cells through the gut microbiota, which can prevent tumor development. Meanwhile, the direct effects of the gut microbiota on tumors and the regulation of the immune system by the nervous system, which can affect tumor development, are also reviewed.
Collapse
Affiliation(s)
- Juanjuan Wang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Wenyue Cheng
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
| |
Collapse
|
15
|
Hu W, Garrison C, Prasad R, Boulton M, Grant M. Indole metabolism and its role in diabetic macrovascular and microvascular complications. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2025; 53:100532. [PMID: 40230659 PMCID: PMC11995707 DOI: 10.1016/j.ahjo.2025.100532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/03/2025] [Accepted: 03/21/2025] [Indexed: 04/16/2025]
Abstract
Tryptophan (Trp), an essential amino acid obtained through dietary sources, plays a crucial role in various physiological processes. The metabolism of Trp branches into three principal pathways: the serotonin pathway, the kynurenine pathway, and the indole pathway. The kynurenine and serotonin pathways are host pathways while the indole pathway is solely the result of bacterial metabolism. Trp metabolites extend their influence beyond protein biosynthesis to affect a spectrum of pathophysiological mechanisms including, but not limited to, neuronal function, immune modulation, inflammatory responses, oxidative stress regulation, and maintenance of intestinal health. This review focuses on indole derivatives and their impact on vascular health. Trp-containing dipeptides are highlighted as a targeted nutraceutical approach to modulate Trp metabolism, enhance beneficial metabolite production, and mitigate risk factors for vascular diseases. The importance of optimizing Trp intake and dietary strategies to harness the benefits of Trp-derived metabolites for vascular health is underscored, bringing to light the need for further research to refine these therapeutic approaches.
Collapse
Affiliation(s)
- W. Hu
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Food Science and Technology, National University of Singapore, Singapore
| | - C. Garrison
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - R. Prasad
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - M.E. Boulton
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - M.B. Grant
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
16
|
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
|
17
|
Chakraborty N, Holmes-Hampton G, Rusling M, Kumar VP, Hoke A, Lawrence AB, Gautam A, Ghosh SP, Hammamieh R. Delayed Impact of Ionizing Radiation Depends on Sex: Integrative Metagenomics and Metabolomics Analysis of Rodent Colon Content. Int J Mol Sci 2025; 26:4227. [PMID: 40362462 PMCID: PMC12071923 DOI: 10.3390/ijms26094227] [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: 02/26/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
There is an escalating need to comprehend the long-term impacts of nuclear radiation exposure since the permeation of ionizing radiation has been frequent in our current societal framework. A system evaluation of the microbes that reside inside a host's colon could meet this knowledge gap since the microbes play major roles in a host's response to stress. Indeed, our past study suggested that these microbes might break their symbiotic association with moribund hosts to form a pro-survival condition exclusive to themselves. In this study, we undertook metagenomics and metabolomics assays regarding the descending colon content (DCC) of adult mice. DCCs were collected 1 month and 6 months after 7 Gy or 7.5 Gy total body irradiation (TBI). The assessment of the metagenomic diversity profile in DCC found a significant sex bias caused by TBI. Six months after 7.5 Gy TBI, decreased Bacteroidetes were replaced by increased Firmicutes in males, and these alterations were reflected in the functional analysis. For instance, a larger number of networks linked to small chain fatty acid (SCFA) synthesis and metabolism were inhibited in males than in females. Additionally, bioenergy networks showed regression dynamics in females at 6 months post-TBI. Increased accumulation of glucose and pyruvate, which are typical precursors of beneficial SCFAs coupled with the activated networks linked to the production of reactive oxygen species, suggest a cross-sex energy-deprived state. Overall, there was a major chronic adverse implication in male mice that supported the previous literature in suggesting females are more radioresistant than males. The sex-biased chronic effects of TBI should be taken into consideration in designing the pertinent therapeutics.
Collapse
Affiliation(s)
- Nabarun Chakraborty
- Medical Readiness Systems Biology, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (N.C.); (M.R.); (A.H.); (A.B.L.); (A.G.)
| | - Gregory Holmes-Hampton
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD 20889-5603, USA; (G.H.-H.); (V.P.K.)
| | - Matthew Rusling
- Medical Readiness Systems Biology, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (N.C.); (M.R.); (A.H.); (A.B.L.); (A.G.)
| | - Vidya P. Kumar
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD 20889-5603, USA; (G.H.-H.); (V.P.K.)
| | - Allison Hoke
- Medical Readiness Systems Biology, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (N.C.); (M.R.); (A.H.); (A.B.L.); (A.G.)
| | - Alexander B. Lawrence
- Medical Readiness Systems Biology, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (N.C.); (M.R.); (A.H.); (A.B.L.); (A.G.)
- Vysnova, Inc., Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Aarti Gautam
- Medical Readiness Systems Biology, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (N.C.); (M.R.); (A.H.); (A.B.L.); (A.G.)
| | - Sanchita P. Ghosh
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD 20889-5603, USA; (G.H.-H.); (V.P.K.)
| | - Rasha Hammamieh
- Medical Readiness Systems Biology, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (N.C.); (M.R.); (A.H.); (A.B.L.); (A.G.)
| |
Collapse
|
18
|
Yuan Y, Liu Y, Wang S, Zhang J, Gao X, Li Y, Yu Z, Zhou Y. Tryptophan-Derived Metabolites and Glutamate Dynamics in Fatal Insulin Poisoning: Mendelian Randomization of Human Cohorts and Experimental Validation in Rat Models. Int J Mol Sci 2025; 26:4152. [PMID: 40362391 PMCID: PMC12072148 DOI: 10.3390/ijms26094152] [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] [Received: 03/02/2025] [Revised: 04/04/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Insulin overdose may cause hypoglycemic encephalopathy. In this study, Mendelian randomization was employed to analyze changes in the serum metabolites of patients with hypoglycemic encephalopathy, and metabolomics analysis was conducted to detect differential metabolites in the serum of a rat model of hypoglycemic encephalopathy induced by insulin overdose. The results indicated an overall upward trend in the tryptophan metabolism pathway in patients with hypoglycemic encephalopathy and rats with hypoglycemic encephalopathy caused by insulin overdose, while serum glutamate levels declined. The metabolic changes in the tryptophan pathway provide new insights into the impact of hypoglycemia on brain function. The related products of the tryptophan metabolism pathway have a certain diagnostic value for hypoglycemic encephalopathy and forensic identification of insulin overdose-induced hypoglycemic encephalopathy death.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zhonghao Yu
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (Y.Y.); (Y.L.); (S.W.); (J.Z.); (X.G.); (Y.L.)
| | - Yiwu Zhou
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (Y.Y.); (Y.L.); (S.W.); (J.Z.); (X.G.); (Y.L.)
| |
Collapse
|
19
|
Taher RF, Abd El ghany EM, El-Gendy ZA, Elghonemy MM, Hassan HA, Abdel Jaleel GA, Hassan A, Sarker TC, Abd-ElGawad AM, Farag MA, Elshamy AI. In vivo anti-ulceration effect of Pancratium maritimum extract against ethanol-induced rats via NLRP3 inflammasome and HMGB1/TLR4/MYD88/NF-κβ signaling pathways and its extract metabolite profile. PLoS One 2025; 20:e0321018. [PMID: 40238859 PMCID: PMC12002509 DOI: 10.1371/journal.pone.0321018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 02/27/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Gastric ulcer is a multifaceted ailment of multiple causes and is chronic warranting the discovery of remedies to alleviate its symptoms and severity. Pancratium maritimum L. is recognized for its several health benefits, although its potential against gastric ulcers has yet to be reported. METHODS AND FINDINGS This study reports on the effects of P. maritimum L. whole plant (PM-EtOH) ethanol extract at a dose of 25, 50, and 100 mg/kg body weight orally for managing ethanol-induced peptic ulcer in rats. The anti-ulceration capacity of PM-EtOH was determined against ethanol (EtOH)-induced rats via biochemical, histological, immunohistochemical, and western blotting assays. The profiling of the bioactive metabolites in P. maritimum extract was based on Ultra-high-performance liquid chromatography coupled with electrospray ionization quadrupole time-of-flight mass spectrometry (UHPLC-ESI-qTOF-MS/MS) analysis. Following PM-EtOH treated group, the gastric glutathione (GSH) level dropped in the ulcer group receiving ethanol was restored to normal levels. Additionally, following PM-EtOH, elevated malondialdehyde (MDA) content in the stomach tissues diminished. PM-EtOH treated group displayed recovery and comparable morphology compared with normal group, concurrent with lower levels of Tumor Necrosis Factor α (TNF-α), MyD88, and NLRP3, along with low expression of Nuclear Factor kappa β (NF-кβ) and high-mobility group box protein 1 (HMGB1) proteins. Immune-histochemicals of caspase-3 and toll-like receptors-4 (TLR-4) showed their normalization. These findings imply that PM-EtOH exerts a protective effect on rat stomach damage that has yet to be further tested in clinical trials for treatment of stomach ulcers. Phytochemical profiling of PM-EtOH via UHPLC-ESI-qTOF-MS/MS led to the identification of 84 metabolites belonging to amino acids, organic acids, phenolic acids, alkaloids, flavonoids, and fatty acids to likely mediate for the observed effects. CONCLUSIONS These outcomes provided evidence for the potential of PM-EtOH in gastric ulcers management.
Collapse
Affiliation(s)
- Rehab F. Taher
- Department of Natural Compounds Chemistry, National Research Centre, Dokki, Giza, Egypt
| | | | - Zeinab A. El-Gendy
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Mai M. Elghonemy
- Department of Natural Compounds Chemistry, National Research Centre, Dokki, Giza, Egypt
| | - Heba A. Hassan
- Therapeutic Chemistry Department, National Research Centre, Dokki, Giza, Egypt
| | - Gehad A. Abdel Jaleel
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Azza Hassan
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Tushar C. Sarker
- Texs A&M AgriLife Research Center, Overton, Texas, United States of America
| | - Ahmed M. Abd-ElGawad
- Plant Production Department, College of Food & Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed A. Farag
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Healthcare Department, Saxony Egypt University (SEU), Badr City, Egypt
| | - Abdelsamed I. Elshamy
- Department of Natural Compounds Chemistry, National Research Centre, Dokki, Giza, Egypt
| |
Collapse
|
20
|
Joja M, Grant ET, Desai MS. Living on the edge: Mucus-associated microbes in the colon. Mucosal Immunol 2025:S1933-0219(25)00041-8. [PMID: 40233878 DOI: 10.1016/j.mucimm.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/28/2025] [Accepted: 04/09/2025] [Indexed: 04/17/2025]
Abstract
The colonic mucus layer acts as a physicochemical barrier to pathogen invasion and as a habitat for mucus-associated microbes. This mucosal microbiome plays a crucial role in moderating mucus production, maintaining barrier integrity, and shaping the host immune response. However, unchecked mucin foraging may render the host vulnerable to disease. To better understand these dynamics in the mucus layer, it is essential to advance fundamental knowledge on how commensals bind to and utilize mucin as well as their interactions with both the host and their microbial neighbors. We present an overview of approaches for surveying mucus-associated bacteria and assessing their mucin-utilizing capacity, alongside a discussion of the limitations of existing methods. Additionally, we highlight how diet and host secretory immunoglobulin A interact with the mucosal bacterial community in the colon. Insights into this subset of the microbial community can guide therapeutic strategies to optimally support and modulate mucosal barrier integrity.
Collapse
Affiliation(s)
- Mihovil Joja
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| | - Erica T Grant
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
21
|
Sharma S, Tiwari N, Tanwar SS. The current findings on the gut-liver axis and the molecular basis of NAFLD/NASH associated with gut microbiome dysbiosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04069-z. [PMID: 40202676 DOI: 10.1007/s00210-025-04069-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025]
Abstract
Recent research has highlighted the complex relationship between gut microbiota, metabolic pathways, and nonalcoholic fatty liver disease (NAFLD) progression. Gut dysbiosis, commonly observed in NAFLD patients, impairs intestinal permeability, leading to the translocation of bacterial products like lipopolysaccharides, short-chain fatty acids, and ethanol to the liver. These microbiome-associated mechanisms contribute to intestinal and hepatic inflammation, potentially advancing NAFLD to NASH. Dietary habits, particularly those rich in saturated fats and fructose, can modify the microbiome composition, leading to dysbiosis and fatty liver development. Metabolomic approaches have identified unique profiles in NASH patients, with specific metabolites like ethanol linked to disease progression. While bariatric surgery has shown promise in preventing NAFLD progression, the role of gut microbiome and metabolites in this improvement remains to be proven. Understanding these microbiome-related pathways may provide new diagnostic and therapeutic targets for NAFLD and NASH. A comprehensive review of current literature was conducted using multiple medical research databases, including PubMed, Scopus, Web of Science, Embase, Cochrane Library, ClinicalTrials.gov, ScienceDirect, Medline, ProQuest, and Google Scholar. The review focused on studies that examine the relationship between gut microbiota composition, metabolic pathways, and NAFLD progression. Key areas of interest included microbial dysbiosis, endotoxin production, and the influence of diet on gut microbiota. The analysis revealed that gut dysbiosis contributes to NAFLD through several mechanisms, diet significantly influences gut microbiota composition, which in turn affects liver function through the gut-liver axis. High-fat diets can lead to dysbiosis, altering microbial metabolic activities and promoting liver inflammation. Specifically, gut microbiota-mediated generation of saturated fatty acids, such as palmitic acid, can activate liver macrophages and increase TNF-α expression, contributing to NASH development. Different dietary components, including cholesterol, fiber, fat, and carbohydrates, can modulate the gut microbiome and influence NAFLD progression. This gut-liver axis plays a crucial role in maintaining immune homeostasis, with the liver responding to gut-derived bacteria by activating innate and adaptive immune responses. Microbial metabolites, such as bile acids, tryptophan catabolites, and branched-chain amino acids, regulate adipose tissue and intestinal homeostasis, contributing to NASH pathogenesis. Additionally, the microbiome of NASH patients shows an elevated capacity for alcohol production, suggesting similarities between alcoholic steatohepatitis and NASH. These findings indicate that targeting the gut microbiota may be a promising approach for NASH treatment and prevention. Recent research highlights the potential of targeting gut microbiota for managing nonalcoholic fatty liver disease (NAFLD). The gut-liver axis plays a crucial role in NAFLD pathophysiology, with dysbiosis contributing to disease progression. Various therapeutic approaches aimed at modulating gut microbiota have shown promise, including probiotics, prebiotics, synbiotics, fecal microbiota transplantation, and dietary interventions. Probiotics have demonstrated efficacy in human randomized controlled trials, while other interventions require further investigation in clinical settings. These microbiota-targeted therapies may improve NAFLD outcomes through multiple mechanisms, such as reducing inflammation and enhancing metabolic function. Although lifestyle modifications remain the primary recommendation for NAFLD management, microbiota-focused interventions offer a promising alternative for patients struggling to achieve weight loss targets.
Collapse
Affiliation(s)
- Seema Sharma
- Department of Pharmacy, Shri Vaishnav Vidyapeeth Vishwavidyalaya, Indore, M.P, India
| | - Nishant Tiwari
- Acropolis Institute of Pharmaceutical Education and Research, Indore, M.P, India
| | - Sampat Singh Tanwar
- Department of Pharmacy, Shri Vaishnav Vidyapeeth Vishwavidyalaya, Indore, M.P, India.
| |
Collapse
|
22
|
Luo D, Luo G, Xu H, Li K, Li Z, Zhang C. Inorganic dietary nanoparticles in intestinal barrier function of inflammatory bowel disease: allies or adversaries? Front Immunol 2025; 16:1563504. [PMID: 40270957 PMCID: PMC12014688 DOI: 10.3389/fimmu.2025.1563504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/24/2025] [Indexed: 04/25/2025] Open
Abstract
Inorganic dietary nanoparticles (IDNPs) are frequently utilized as food additives and in packaging, resulting in their exposure becoming a substantial yet often overlooked concern for patients with inflammatory bowel disease (IBD). Considering that impaired intestinal barrier function plays a central role in the pathogenesis of IBD, this review concentrates on the roles and mechanisms of IDNPs in the intestinal barrier (physical, chemical, biological, and immune barriers) of IBD patients. Previous studies have shown that different types of nanoparticles have varying effects on animals in diverse states. In this context, factors such as the source, size, shape, dosage, and duration of action of the nanoparticles, as well as the species, gender, dietary habits, and age of the animals, significantly influence research outcomes. Future studies should undertake more comprehensive explorations into the effects and mechanisms of IDNPs with diverse sources and properties in IBD patients.
Collapse
Affiliation(s)
- Duo Luo
- Department of Geriatrics, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Guifang Luo
- Department of Geriatrics, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Haoming Xu
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Kangbao Li
- Department of Geriatrics, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhaotao Li
- Department of Gastroenterology, The First People’s Hospital of Foshan, Foshan, China
| | - Cong Zhang
- Department of Gastroenterology, The First People’s Hospital of Foshan, Foshan, China
| |
Collapse
|
23
|
Xu Y, Li LN, He XJ, Wang S, Li X, Feng H, Zhang HF, Song L, Shi HS, Tian XY. Exogenous GABA Alleviates Tourette Syndrome-Like Behavior in Sprague-Dawley Rats by Altering Gut Microbiota and Striatum Metabolism. Neuropsychiatr Dis Treat 2025; 21:711-727. [PMID: 40200938 PMCID: PMC11977633 DOI: 10.2147/ndt.s512191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Context Tourette syndrome (TS) is a common chronic neuropsychiatric disorder with a prevalence of approximately 1% in children and adolescents. TS is characterized by sudden involuntary motor tics along with vocal tics. A pathological study on postmortem patients has reported a 50-60% reduction in striatal gamma-aminobutyric acidergic (GABAergic) interneurons, suggesting a role for GABAergic system imbalances in tic disorder development. However, the effect of exogenous GABA administration on tic alleviation remains unreported. Objective In this study, we aim to investigate the therapeutic effects of exogenous GABA on TS-like behaviors in Sprague-Dawley rats and explore its potential mechanisms, including gut microbiota regulation, oxidative stress mitigation, and restoration of GABA-glutamate balance, to provide insights into TS pathogenesis and alternative treatment strategies. Materials and Methods A TS model rat was established through intraperitoneal administration of 3,3-Iminodipropionitrile (150 mg/kg/day), followed by GABA (20 mg/kg/day) administration by gavage. 15 minutes of behavioral testing (stereotypical behavior and head twitching behavior) was then conducted. 16S rRNA sequencing identified microbiome changes, and LC-MS assessed striatal metabolite changes. Results The results showed that a 4-week GABA treatment alleviated TS-like behavior in rats. GABA treatment led to an increase in Acinetobacter and other beneficial bacteria. GABA also significantly upregulated 15 striatal metabolites compared with TS group. By correlation analysis of striatal metabolites and intestinal bacteria, statistical analysis showed that Clostridium_sensu_stricto_1 was negatively correlated with metabolites on the top 20 differential gut microbiota and metabolites. Moreover, changes in gut microbiota correlated with alterations in striatal metabolites, suggesting a gut-brain axis involvement. Conclusion Exogenous GABA alleviated TS-like behavior in rats by reducing harmful gut flora and modulating striatal GABA-glutamate metabolism. Despite challenges like low blood-brain barrier permeability and dose safety in humans, GABA's therapeutic potential may be realized through prodrug development and optimized dosing. These findings are preliminary and require further clinical validation.
Collapse
Affiliation(s)
- Ying Xu
- The Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Li-Na Li
- The Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Xiang-Jun He
- The Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Shuang Wang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Xincheng Li
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Hao Feng
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Hui-Feng Zhang
- The Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
| | - Li Song
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Hai-Shui Shi
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
- Nursing School, Hebei Medical University, Shijiazhuang, 050031, People’s Republic of China
- Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, People’s Republic of China
| | - Xiao-Yu Tian
- The Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
| |
Collapse
|
24
|
Roessler J, Zimmermann F, Heidecker B, Landmesser U, Haghikia A. Gut microbiota-related modulation of immune mechanisms in post-infarction remodelling and heart failure. ESC Heart Fail 2025; 12:942-954. [PMID: 39385474 PMCID: PMC11911630 DOI: 10.1002/ehf2.14991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 10/12/2024] Open
Abstract
The immune system has long been recognized as a key driver in the progression of heart failure (HF). However, clinical trials targeting immune effectors have consistently failed to improve patient outcome across different HF aetiologies. The activation of the immune system in HF is complex, involving a broad network of pro-inflammatory and immune-modulating components, which complicates the identification of specific immune pathways suitable for therapeutic targeting. Increasing attention has been devoted to identifying gut microbial pathways that affect cardiac remodelling and metabolism and, thereby impacting the development of HF. In particular, gut microbiota-derived metabolites, absorbed by the host and transported to the peripheral circulation, can act as signalling molecules, influencing metabolism and immune homeostasis. Recent reports suggest that the gut microbiota plays a crucial role in modulating immune processes involved in HF. Here, we summarize recent advances in understanding the contributory role of gut microbiota in (auto-)immune pathways that critically determine the progression or alleviation of HF. We also thoroughly discuss potential gut microbiota-based intervention strategies to treat or decelerate HF progression.
Collapse
Affiliation(s)
- Johann Roessler
- University Hospital St Josef‐Hospital Bochum, Cardiology and RhythmologyRuhr University BochumBochumGermany
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
| | - Friederike Zimmermann
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
| | - Bettina Heidecker
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
- Berlin Institute of Health at Charité – Universitätsmedizin BerlinBerlinGermany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
- Berlin Institute of Health at Charité – Universitätsmedizin BerlinBerlinGermany
- Friede Springe‐Cardiovascular Prevention Center at Charité, Charité‐Universitätsmedizin, Berlin Institute of Health (BIH)BerlinGermany
| | - Arash Haghikia
- University Hospital St Josef‐Hospital Bochum, Cardiology and RhythmologyRuhr University BochumBochumGermany
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
- Berlin Institute of Health at Charité – Universitätsmedizin BerlinBerlinGermany
- Friede Springe‐Cardiovascular Prevention Center at Charité, Charité‐Universitätsmedizin, Berlin Institute of Health (BIH)BerlinGermany
| |
Collapse
|
25
|
Hassan A, Azid A, Hamid FS, Pariatamby A, Ossai IC, Aboudi-Mana SC. Field application of arbuscular mycorrhizal fungi and Alocasia calidora (Schott) G.Don for effective remediation of heavy metal/metalloid-polluted landfill soil. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2025; 32:11443-11465. [PMID: 40220159 DOI: 10.1007/s11356-025-36374-8] [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: 10/03/2024] [Accepted: 04/02/2025] [Indexed: 04/14/2025]
Abstract
The research aimed to assess the influence of arbuscular mycorrhizal fungi (AMF) inoculation on the growth, tolerance, and phytoremediation capacity of Alocasia calidora (Schott) G.Don used in heavy metal/metalloid-polluted soil under field conditions. Five new, young A. calidora plants were planted in each treatment and control plot (1 m × 1 m). AMF inoculum was supplemented on days 0, 30, 60, and 90 in the treatment plot, and the experiment was conducted for 120 days. Substantial growth was observed in the roots and shoots of AMF-inoculated A. calidora. In the shoots of treated A. calidora, a high accumulation of Cu, As, Ni, Mn, and Pb was observed. The AMF-treated plant exhibited a higher accumulation of Cu, Fe, and Ni in the roots (P < 0.05). The removal efficiencies in the AMF-treated soil were 84.67, 74.82, 81.61, 80.77, 88.21, 92.26, 92.35, and 67.32% for As, Cr, Cu, Fe, Mn, Ni, Pb, and Zn, correspondingly, while, for control, the corresponding values were 61.24, 52.42, 62.95, 33.46, 57.89, 61.45, 71.19, and 54.98%. AMF-treated A. calidora demonstrated an increased tolerance and metal/metalloid accumulation in response to a variety of compounds, including tryptophan, S-(4-nitrobenzyl) glutathione, 5,7,2',3'-tetrahydroxy flavone, 5,2-dihydroxy flavone, indole-acrylic acid, and various tripeptides. A. calidora proliferation, tolerance, and metal/metalloid accumulation were enhanced by the inoculated AMF. Therefore, treating soil contaminated with heavy metals/metalloids can be accomplished by combining AMF with plants.
Collapse
Affiliation(s)
- Auwalu Hassan
- Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, 22000, Besut, Terengganu, Malaysia
- Department of Biological Sciences, Faculty of Science, Federal University of Kashere, Kashere, Gombe State, Nigeria
| | - Azman Azid
- Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, 22000, Besut, Terengganu, Malaysia.
| | - Fauziah Shahul Hamid
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
- Center for Research in Waste Management, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Agamuthu Pariatamby
- Jeffrey Sachs Center On Sustainable Development, Sunway University, Sunway, Malaysia
| | - Innocent Chukwunonso Ossai
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
- Center for Research in Waste Management, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Suzanne Christine Aboudi-Mana
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
- Center for Research in Waste Management, University of Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
26
|
Mu X, Feng L, Wang Q, Li H, Zhou H, Yi W, Sun Y. Decreased gut microbiome-derived indole-3-propionic acid mediates the exacerbation of myocardial ischemia/reperfusion injury following depression via the brain-gut-heart axis. Redox Biol 2025; 81:103580. [PMID: 40058066 PMCID: PMC11930714 DOI: 10.1016/j.redox.2025.103580] [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] [Received: 02/06/2025] [Revised: 02/11/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Despite the increasing recognition of the interplay between depression and cardiovascular disease (CVD), the precise mechanisms by which depression contributes to the pathogenesis of cardiovascular disease remain inadequately understood. The involvement of gut microbiota and their metabolites to health and disease susceptibility has been gaining increasing attention. In this study, it was found that depression exacerbated cardiac injury, impaired cardiac function (EF%: P < 0.01; FS%: P < 0.05), hindered long-term survival (P < 0.01), and intensified adverse cardiac remodeling (WGA: P < 0.01; MASSON: P < 0.0001) after myocardial ischemia/reperfusion (MI/R) in mice. Then we found that mice receiving microbiota transplants from chronic social defeat stress (CSDS) mice exhibited worse cardiac function (EF%: P < 0.01; FS%: P < 0.01) than those receiving microbiota transplants from non-CSDS mice after MI/R injury. Moreover, impaired tryptophan metabolism due to alterations in gut microbiota composition and structure was observed in the CSDS mice. Mechanistically, we analyzed the metabolomics of fecal and serum samples from CSDS mice and identified indole-3-propionic acid (IPA) as a protective agent for cardiomyocytes against ferroptosis after MI/R via NRF2/System xc-/GPX4 axis, played a role in mediating the detrimental influence of depression on MI/R. Our findings provide new insights into the role of the gut microbiota and IPA in depression and CVD, forming the basis of intervention strategies aimed at mitigating the deterioration of cardiac function following MI/R in patients experiencing depression.
Collapse
Affiliation(s)
- Xingdou Mu
- Department of Geriatric, XiJing Hospital, Xi'an, Shaanxi, 710000, China
| | - Lele Feng
- Department of Cardiovascular Surgery, XiJing Hospital, Xi'an, Shaanxi, 710000, China
| | - Qiang Wang
- Department of Geriatric, XiJing Hospital, Xi'an, Shaanxi, 710000, China
| | - Hong Li
- Department of Geriatric, XiJing Hospital, Xi'an, Shaanxi, 710000, China
| | - Haitao Zhou
- Department of Geriatric, XiJing Hospital, Xi'an, Shaanxi, 710000, China
| | - Wei Yi
- Department of Cardiovascular Surgery, XiJing Hospital, Xi'an, Shaanxi, 710000, China.
| | - Yang Sun
- Department of Geriatric, XiJing Hospital, Xi'an, Shaanxi, 710000, China.
| |
Collapse
|
27
|
Li G, Dong S, Liu C, Yang J, Rensen PCN, Wang Y. Serotonin signaling to regulate energy metabolism: a gut microbiota perspective. LIFE METABOLISM 2025; 4:loae039. [PMID: 39926388 PMCID: PMC11803461 DOI: 10.1093/lifemeta/loae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/11/2024] [Accepted: 11/21/2024] [Indexed: 02/11/2025]
Abstract
Serotonin is one of the most potent gastrointestinal, peripheral, and neuronal signaling molecules and plays a key role in regulating energy metabolism. Accumulating evidence has shown the complex interplay between gut microbiota and host energy metabolism. In this review, we summarize recent findings on the role of gut microbiota in serotonin metabolism and discuss the complicated mechanisms by which serotonin, working in conjunction with the gut microbiota, affects total body energy metabolism in the host. Gut microbiota affects serotonin synthesis, storage, release, transport, and catabolism. In addition, serotonin plays an indispensable role in regulating host energy homeostasis through organ crosstalk and microbe-host communication, particularly with a wide array of serotonergic effects mediated by diverse serotonin receptors with unique tissue specificity. This fresh perspective will help broaden the understanding of serotonergic signaling in modulating energy metabolism, thus shedding light on the design of innovative serotonin-targeting strategies to treat metabolic diseases.
Collapse
Affiliation(s)
- Guoli Li
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Sijing Dong
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Chunhao Liu
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jing Yang
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Patrick C N Rensen
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Yanan Wang
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
28
|
Wang L, Zhang Y, Ran Y, Li L, Mei L, Ye F, Sun Y, Wang T, Quan X, Shi H, Dai F. Association between AHR in EGCs and IBS-D patients: the indole pathway of tryptophan metabolism. Front Nutr 2025; 12:1566595. [PMID: 40225339 PMCID: PMC11985470 DOI: 10.3389/fnut.2025.1566595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Background The pathophysiological mechanisms of irritable bowel syndrome (IBS) are intricate, and associated with tryptophan metabolites. This study was designed to investigate the relationship between indole metabolites in the feces and intestinal function in patients with IBS. Methods In this study, 42 patients with diarrhea-predominant IBS (IBS-D) and 36 healthy controls were recruited. The symptom severity was evaluated using IBS-quality of life (IBS-QOL) and IBS symptom severity system (IBS-SSS). The levels of indole metabolite in fecal samples were determined by means of mass spectrometry. Colon mucosal tissues were collected during colonoscopy procedures. Immunohistochemistry or immunofluorescence techniques were employed to analyze the expressions of the aryl hydrocarbon receptor (AHR), cytochrome P450 1A1 (CYP1A1), glial fibrillary acidic protein (GFAP), S100 calcium-binding protein B (S100B), zonula occludens-1 (Zo-1), occludin, substance P (SP), nerve growth factor (NGF), NOD-like receptor family pyrin domain containing 3 (NLRP3), and nuclear factor kappa B (NF-κB) in the mucosal tissues. Results Compared with healthy controls, the concentrations of the main indole metabolites (p = 0.020), and the expressions of CYP1A1 (p < 0.001), and Zo-1 (p = 0.017) were decreased in patients with IBS-D, but the expressions of S100B (p < 0.001), NF-κB (p = 0.006), and NRLP3 (p = 0.041) were increased. Immunofluorescence analysis demonstrated the co-expression of AHR with GFAP or S100B. Moreover, the ratio of S100B/AHR (p = 0.011) was higher in IBS-D patients than in health controls. This ratio was positively correlated with IBS-SSS score (r = 0.47, p = 0.006), as well as with the expression levels of NRLP3 (r = 0.505, p = 0.019), NF-κB (r = 0.548, p = 0.01), and SP (r = 0.832, p < 0.01). Conclusion Patients with IBS-D exhibited low-grade inflammation in the colon mucosal tissues, compromised intestinal barrier function, and abnormal visceral sensation. This may be attributed to the decreased levels of tryptophan indole metabolites, the heightened activity of enteric glial cells (EGCs), and the inhibition of AHR/CPY1A1 signaling pathway.
Collapse
Affiliation(s)
- Lianli Wang
- Division of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yue Zhang
- Division of Gastroenterology, Honghui Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, China
| | - Yan Ran
- Division of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Laifu Li
- Division of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lin Mei
- Division of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Fangchen Ye
- Division of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yating Sun
- Division of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ting Wang
- Division of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaojing Quan
- Division of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Haitao Shi
- Division of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Fei Dai
- Division of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
29
|
Feng A, Zhao H, Qiu C, Luo D, Wu H, Meng X, Li L, Zou H. Gut microbiota metabolites impact immunologic responses to antiretroviral therapy in HIV-infected men who have sex with men. Infect Dis Poverty 2025; 14:21. [PMID: 40098016 PMCID: PMC11917012 DOI: 10.1186/s40249-025-01291-y] [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: 12/08/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND The association between gut microbial metabolites and immunologic non-response among people living with HIV (PLHIV) receiving antiretroviral therapy (ART) has not been well established. We aimed to characterize gut microbial metabolites among HIV-infected men who have sex with men (MSM) with different immunologic responses. METHODS We recruited HIV-infected MSM from Guangzhou Eighth People's Hospital and HIV-uninfected MSM (healthy controls, HC) from a local MSM community-based organization in Guangzhou between June and October 2021. HIV-infected MSM were grouped into good immunological responders (GIR) (CD4 + T cell count ≥ 350 cells/μl) and poor immunological responders (PIR) (CD4 + T cell count < 350 cells/μl) after 24 months of ART treatment. Online questionnaires and stool samples were collected. Microbial metabolites in stool were obtained through ultra-performance liquid chromatography coupled to a tandem mass spectrometry (UPLC-MS/MS) system. Differential metabolites were identified and analyzed using the Kruskal-Wallis test, followed by pairwise comparisons with the Wilcoxon rank-sum test. The least absolute selection and shrinkage operator was used to select potential metabolites biomarkers. RESULTS A total of 51 HC, 56 GIR, and 42 PIR were included. No statistically significant differences were observed in the median time since HIV diagnosis and ART duration between GIR and PIR. Among the 174 quantified metabolites, 81 significantly differed among HC, GIR, and PIR (P < 0.05). Among differential metabolites, indole-3-propionic acid significantly decreased from HC (11.39 nmol/g) and GIR (8.16 nmol/g) to PIR (6.50 nmol/g). The pathway analysis showed that tryptophan metabolism differed significantly between GIR and PIR (P < 0.05). Four potential metabolites biomarkers (dimethylglycine, cinnamic acid, 3-hydroxyisovaleric acid, and propionic acid) that distinguish GIR and PIR were identified, and the corresponding area under the curve based on potential biomarkers was 0.773 (95% CI: 0.675-0.871). CONCLUSIONS This study identified significant differences in gut microbial metabolites among HIV-infected MSM with different immunologic responses. These results indicate the potential of gut microbial metabolites as novel disease progression markers and therapeutic targets.
Collapse
Affiliation(s)
- Anping Feng
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Heping Zhao
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
- Infectious Disease Center, Guangzhou Eighth People's Hospital, Guangzhou Medical University, No 8 Huaying Road, Guangzhou, 510060, Guangdong, China
| | - Chunting Qiu
- Department of Infectious Diseases, Tianjin Second People's Hospital, Tianjin, 300192, China
| | - Dan Luo
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Hao Wu
- Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Xiaojun Meng
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi, 214023, Jiangsu, China.
| | - Linghua Li
- Infectious Disease Center, Guangzhou Eighth People's Hospital, Guangzhou Medical University, No 8 Huaying Road, Guangzhou, 510060, Guangdong, China.
| | - Huachun Zou
- School of Public Health, Fudan University, Room 435, Bld #8, 130 Dongan Road, Xuhui District, Shanghai, 200032, China.
- School of Public Health, Southwest Medical University, Luzhou, China.
- Kirby Institute, University of New South Wales, Sydney, Australia.
| |
Collapse
|
30
|
Flanagan K, Gassner K, Lang M, Ozelyte J, Hausmann B, Crepaz D, Pjevac P, Gasche C, Berry D, Vesely C, Pereira FC. Human-derived microRNA 21 regulates indole and L-tryptophan biosynthesis transcripts in the gut commensal Bacteroides thetaiotaomicron. mBio 2025; 16:e0392824. [PMID: 39878512 PMCID: PMC11898669 DOI: 10.1128/mbio.03928-24] [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: 12/17/2024] [Accepted: 01/08/2025] [Indexed: 01/31/2025] Open
Abstract
In the gut, microRNAs (miRNAs) produced by intestinal epithelial cells are secreted into the lumen and can shape the composition and function of the gut microbiome. Crosstalk between gut microbes and the host plays a key role in irritable bowel syndrome (IBS) and inflammatory bowel diseases, yet little is known about how the miRNA-gut microbiome axis contributes to the pathogenesis of these conditions. Here, we investigate the ability of miR-21, a miRNA that we found decreased in fecal samples from IBS patients, to associate with and regulate gut microbiome function. When incubated with the human fecal microbiota, miR-21 revealed a rapid internalization or binding to microbial cells, which varied in extent across different donor samples. Fluorescence-activated cell sorting and sequencing of microbial cells incubated with fluorescently labeled miR-21 identified organisms belonging to the genera Bacteroides, Limosilactobacillus, Ruminococcus, or Coprococcus, which predominantly interacted with miR-21. Surprisingly, these and other genera also interacted with a miRNA scramble control, suggesting that physical interaction and/or uptake of these miRNAs by gut microbiota is not sequence-dependent. Nevertheless, transcriptomic analysis of the gut commensal Bacteroides thetaiotaomicron revealed a miRNA sequence-specific effect on bacterial transcript levels. Supplementation of miR-21, but not of small RNA controls, resulted in significantly altered levels of many cellular transcripts and increased transcription of a biosynthetic operon for indole and L-tryptophan, metabolites known to regulate host inflammation and colonic motility. Our study identifies a novel putative miR-21-dependent pathway of regulation of intestinal function through the gut microbiome with implications for gastrointestinal conditions. IMPORTANCE The mammalian gut represents one of the largest and most dynamic host-microbe interfaces. Host-derived microRNAs (miRNAs), released from the gut epithelium into the lumen, have emerged as important contributors to host-microbe crosstalk. Levels of several miRNAs are altered in the stool of patients with irritable bowel syndrome or inflammatory bowel disease. Understanding how miRNAs interact with and shape gut microbiota function is crucial as it may enable the development of new targeted treatments for intestinal diseases. This study provides evidence that the miRNA miR-21 can rapidly associate with diverse microbial cells form the gut and increase levels of transcripts involved in tryptophan synthesis in a ubiquitous gut microbe. Tryptophan catabolites regulate key functions, such as gut immune response or permeability. Therefore, this mechanism represents an unexpected host-microbe interaction and suggests that host-derived miR-21 may help regulate gut function via the gut microbiota.
Collapse
Affiliation(s)
- Kayla Flanagan
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Kirsten Gassner
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Michaela Lang
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Jurgita Ozelyte
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Bela Hausmann
- Joint Microbiome Facility, Medical University of Vienna and University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Division of Clinical Microbiology, Medical University of Vienna, Vienna, Austria
| | - Daniel Crepaz
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Petra Pjevac
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Joint Microbiome Facility, Medical University of Vienna and University of Vienna, Vienna, Austria
| | - Christoph Gasche
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - David Berry
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Joint Microbiome Facility, Medical University of Vienna and University of Vienna, Vienna, Austria
| | - Cornelia Vesely
- Center of Anatomy and Cell Biology, Division of Cell and Developmental Biology, Medical University of Vienna, Vienna, Austria
| | - Fatima C. Pereira
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
31
|
Sun Y, Huang S, Li M, Yang Y, Ma J, Xie R, Wang J, Zhao Q, Qin S, He L, Jiang J, Zhao Q, Jin G, Liu X, Huang H, Yang Y, Wei J, Liu W, Wang B, Yang R, Su X, Cao H. Maternal high-fat diet disrupts intestinal mucus barrier of offspring by regulating gut immune receptor LRRC19. Commun Biol 2025; 8:420. [PMID: 40075219 PMCID: PMC11903762 DOI: 10.1038/s42003-025-07836-z] [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: 03/17/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Maternal high fat diet (MHFD) increased colitis susceptibility in adulthood. However, the mechanism remains unclear. We sought to explore whether novel gut immune receptor leucine-rich repeat C19 (LRRC19) contributed to the impaired mucus barrier of offspring exposed to MHFD via gut immune response and microbiota. The results showed that MHFD significantly impaired the intestinal mucus barrier of offspring, and up-regulated the expression of LRRC19. Lrrc19 deletion alleviated the mucus barrier disruption. Mechanistically, metagenome sequencing revealed that the MHFD-induced gut microbiota alteration was partly restored in Lrrc19-/- offspring. Muc2-associated bacteria were decreased in the MHFD group, such as Akkermansia_muciniphila_CAG_154, which increased in the Lrrc19-deficient offspring. Moreover, Lrrc19-/- offspring had a higher rate of indole-3-acetic acid (IAA)-producing bacterium, such as Lactobacillus reuteri. A targeted metabolomics analysis revealed that IAA emerged as the top candidate that might mediate the protective effects. IAA was found to improve the mucus barrier function by increasing the ratio of interleukin-22 (IL-22)+ ILC3 cells in an aryl hydrocarbon receptor (AhR)-dependent manner. These results suggest that MHFD disrupts the intestinal mucus barrier of offspring through regulating gut immune receptor LRRC19 and inducing an imbalance of gut microbiota and microbiota-derived metabolites.
Collapse
Affiliation(s)
- Yue Sun
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
- Department of Endoscopy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Shumin Huang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Mengfan Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Yunwei Yang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Jiahui Ma
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Runxiang Xie
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Jingyi Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Qianjing Zhao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Siqi Qin
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Linlin He
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Jiaying Jiang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Qing Zhao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Ge Jin
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Xiang Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Huan Huang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Yazheng Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jianmei Wei
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China
| | - Wentian Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiaomin Su
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China.
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Key Medical Discipline (Specialty), Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China.
| |
Collapse
|
32
|
He Y, Qin X, Liao C, Lima RLS, Hou Q, Lei J, Lai Y, Jiang Q, Wang B, Zhang B. Genistein alleviates colitis by suppressing inflammation and modulating colonic Marvinbryantia formatexigens abundance and metabolites. Curr Res Food Sci 2025; 10:101016. [PMID: 40207203 PMCID: PMC11979476 DOI: 10.1016/j.crfs.2025.101016] [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: 12/31/2024] [Revised: 02/28/2025] [Accepted: 03/01/2025] [Indexed: 04/11/2025] Open
Abstract
As an active ingredient of leguminous plants, genistein is extremely important for alleviating various human diseases. However, the regulatory effect of genistein on intestinal microbiota in alleviating enteritis is still unclear. In this study, the effect of genistein in alleviating dextran sodium sulfate (DSS)-induced colitis and the potential microbial metabolic regulation mechanism were explored. First, the effect of genistein on DSS-induced colitis was studied in mice. Then antibiotics were used to inhibit intestinal bacteria to verify that intestinal microorganisms play an important role in alleviating colitis of genistein. Finally, mice were administrated with live differential bacterium to confirm that genistein can regulate intestinal microorganisms to treat colitis. The results indicated that genistein alleviated DSS-induced colonic inflammation by inhibiting the Nuclear factor kappa-B and Cyclooxygenase-2/Prostaglandin E2 pathway. Genistein alleviated DSS-induced intestinal injury and decreased Mucin 2 secretion. Supplementation with genistein attenuated the DSS-induced decrease in the alpha diversity of gut bacteria. Genistein increased the abundance of Lachnospiraceae and Marvinbryantia formatexigens, increased the concentration of short chain fatty acids in colitis. After antibiotics depleted the intestinal bacteria, genistein lost the effect of relieving colitis, indicating that genistein must relieve colitis through the intestinal bacteria. Mice fed with living Marvinbryantia formatexigens increased short-chain fatty acids and relieved colitis. The present study demonstrates that genistein alleviated colonic inflammation by regulating intestinal bacterium of Marvinbryantia formatexigens and increasing short-chain fatty acid production.
Collapse
Affiliation(s)
- Yang He
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing 100193, China
| | - Xiaoli Qin
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing 100193, China
| | - Chaoyong Liao
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing 100193, China
| | - Rafaela Lameira Souza Lima
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing 100193, China
| | - Qihang Hou
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing 100193, China
| | - Jiaqi Lei
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing 100193, China
| | - Yujiao Lai
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing 100193, China
| | - Qiuyu Jiang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing 100193, China
| | - Bo Wang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing 100193, China
| | - Bingkun Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing 100193, China
| |
Collapse
|
33
|
de Luca Silva B, Cendoroglo MS, Colleoni GWB. Gut Microbiota and Metabolic Biomarkers Associated With Longevity. Nutr Rev 2025:nuaf027. [PMID: 40036950 DOI: 10.1093/nutrit/nuaf027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025] Open
Abstract
The dynamic balance between pro- and anti-inflammatory networks decreases as individuals age, and intestinal dysbiosis can initiate and maintain low-grade systemic inflammation. Interactions between the microbiota and humans occur from the beginning of life and, in general, the diversity of microbiota decreases with aging. The microbiome produces different metabolites with systemic effects, including immune system regulation. This understanding will be useful in controlling inflammation and preventing metabolic changes. Therefore, this review aims to identify the main metabolites synthesized by the intestinal microbiota to be used as biomarkers associated with longevity. This is a narrative review using scientific articles published in the last 10 years in the following databases: PubMed, Scielo, and Lilacs, using the Boolean operators "and" or "or." For this review, we identified 5 articles. The main metabolites described in the literature to date are organic acids, bile acids (BAs), short-chain fatty acids, branched-chain amino acids, trimethylamine N-oxide (TMAO), and derivatives of tryptophan and indole. Among these, the only ones not yet well characterized in studies on longevity were BAs and TMAO. Glutamate and p-cresol were also highlighted in the literature, with a negative association with longevity. The others showed an association, mostly positive, and can be used as potential biomarkers correlated with healthy aging and, if better studied, as targets for intervention to promote health and well-being.
Collapse
Affiliation(s)
- Beatriz de Luca Silva
- Geriatrics and Gerontology Discipline, Paulista School of Medicine, Federal University of São Paulo, São Paulo, SP 04025-002, Brazil
| | - Maysa Seabra Cendoroglo
- Geriatrics and Gerontology Discipline, Paulista School of Medicine, Federal University of São Paulo, São Paulo, SP 04025-002, Brazil
| | - Gisele W B Colleoni
- Geriatrics and Gerontology Discipline, Paulista School of Medicine, Federal University of São Paulo, São Paulo, SP 04025-002, Brazil
| |
Collapse
|
34
|
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
|
35
|
Jin S, Wu J, Wang C, He Y, Tang Y, Huang L, Zhou H, Liu D, Wu Z, Feng Y, Chen H, He X, Yang G, Peng C, Qiu J, Li T, Yin Y, He L. Aspartate Metabolism-Driven Gut Microbiota Dynamics and RIP-Dependent Mitochondrial Function Counteract Oxidative Stress. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404697. [PMID: 39874197 PMCID: PMC11923965 DOI: 10.1002/advs.202404697] [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: 05/01/2024] [Revised: 12/18/2024] [Indexed: 01/30/2025]
Abstract
Aspartate (Asp) metabolism-mediated antioxidant functions have important implications for neonatal growth and intestinal health; however, the antioxidant mechanisms through which Asp regulates the gut microbiota and influences RIP activation remain elusive. This study reports that chronic oxidative stress disrupts gut microbiota and metabolite balance and that such imbalance is intricately tied to the perturbation of Asp metabolism. Under normal conditions, in vivo and in vitro studies reveal that exogenous Asp improves intestinal health by regulating epithelial cell proliferation, nutrient uptake, and apoptosis. During oxidative stress, Asp reduces Megasphaera abundance while increasing Ruminococcaceae. This reversal effect depends on the enhanced production of the antioxidant eicosapentaenoic acid mediated through Asp metabolism and microbiota. Mechanistically, the application of exogenous Asp orchestrates the antioxidant responses in enterocytes via the modulation of the RIP3-MLKL and RIP1-Nrf2-NF-κB pathways to eliminate excessive reactive oxygen species and maintain mitochondrial functionality and cellular survival. These results demonstrate that Asp signaling alleviates oxidative stress by dynamically modulating the gut microbiota and RIP-dependent mitochondrial function, providing a potential therapeutic strategy for oxidative stress disease treatment.
Collapse
Affiliation(s)
- Shunshun Jin
- Hunan Provincial Key Laboratory of Animal Intestinal Function and RegulationHunan international joint laboratory of Animal Intestinal Ecology and HealthLaboratory of Animal Nutrition and Human HealthCollege of Life SciencesHunan Normal UniversityChangsha410081China
- Department of Animal ScienceUniversity of ManitobaWinnipegManitobaR3T2N2Canada
| | - Jian Wu
- Key Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessChangsha410125China
| | - Chenyu Wang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and RegulationHunan international joint laboratory of Animal Intestinal Ecology and HealthLaboratory of Animal Nutrition and Human HealthCollege of Life SciencesHunan Normal UniversityChangsha410081China
- Key Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessChangsha410125China
| | - Yiwen He
- Hunan Provincial Key Laboratory of Animal Intestinal Function and RegulationHunan international joint laboratory of Animal Intestinal Ecology and HealthLaboratory of Animal Nutrition and Human HealthCollege of Life SciencesHunan Normal UniversityChangsha410081China
- Key Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessChangsha410125China
| | - Yulong Tang
- Key Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessChangsha410125China
| | - Le Huang
- Key Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessChangsha410125China
| | - Hui Zhou
- Hunan Provincial Key Laboratory of Animal Intestinal Function and RegulationHunan international joint laboratory of Animal Intestinal Ecology and HealthLaboratory of Animal Nutrition and Human HealthCollege of Life SciencesHunan Normal UniversityChangsha410081China
- Key Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessChangsha410125China
| | - Di Liu
- Heilongjiang Academy of Agricultural SciencesHarbin150086China
| | - Ziping Wu
- Agricultural and Food EconomicsQueen's University BelfastNorthern IrelandBT95PXUK
| | - Yanzhong Feng
- Heilongjiang Academy of Agricultural SciencesHarbin150086China
| | - Heshu Chen
- Heilongjiang Academy of Agricultural SciencesHarbin150086China
| | - Xinmiao He
- Hunan Provincial Key Laboratory of Animal Intestinal Function and RegulationHunan international joint laboratory of Animal Intestinal Ecology and HealthLaboratory of Animal Nutrition and Human HealthCollege of Life SciencesHunan Normal UniversityChangsha410081China
- Heilongjiang Academy of Agricultural SciencesHarbin150086China
| | - Guan Yang
- Department of Infectious Diseases and Public HealthCity University of Hong KongKowloonHong Kong SAR999077China
| | - Can Peng
- Key Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessChangsha410125China
| | - Jiazhang Qiu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infections DiseaseKey Laboratory for Zoonosis Research of the Ministry of EducationCollege of Veterinary MedicineJilin UniversityChangchun130025China
| | - Tiejun Li
- Key Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessChangsha410125China
| | - Yulong Yin
- Key Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessChangsha410125China
- Yuelushan LaboratoryNo. 246 Hongqi Road, Furong DistrictChangsha410128China
| | - Liuqin He
- Hunan Provincial Key Laboratory of Animal Intestinal Function and RegulationHunan international joint laboratory of Animal Intestinal Ecology and HealthLaboratory of Animal Nutrition and Human HealthCollege of Life SciencesHunan Normal UniversityChangsha410081China
- Key Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessChangsha410125China
| |
Collapse
|
36
|
Baral T, Johnson AS, Unnikrishnan MK, Manu MK, Saravu K, Udyavara Kudru C, Abdulsalim S, Singh J, Mukhopadhyay C, Rao M, Miraj SS. Potential role of indole-3-propionic acid in tuberculosis: current perspectives and future prospects. Expert Opin Ther Targets 2025; 29:171-178. [PMID: 40160109 DOI: 10.1080/14728222.2025.2482548] [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: 04/15/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Indole-3-propionic acid (IPA), a tryptophan catabolite derived from gut bacterial metabolism, has been identified as a functional link between the gut microbiome and tuberculosis. AREA COVERED IPA has gained ample attention over the past two decades on account of its multiple physiological roles, besides being both detectable and quantifiable. IPA is well studied across different health conditions, including cardiovascular and neurological conditions. IPA blocks tryptophan synthesis in Mycobacterium by binding to the allosteric tryptophan-binding site of TrpE, thereby threatening Mycobacterium survival due to tryptophan deficit. EXPERT OPINION Characterizing IPA would enable its use as a tool to investigate the pathophysiology of tuberculosis. Integrating 'OMICS' techniques (through next-generation sequencing) along with targeted microbial metabolomics may help explore the possible association of serum IPA levels with TB in patients. This will aid in identifying IPA-producing gut microbes and selecting probiotic strains as a microbiome-targeting adjunct therapy, eventually enhancing our understanding of the molecular dynamics of the pathophysiology of tuberculosis in the context of the microbiome.
Collapse
Affiliation(s)
- Tejaswini Baral
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Aieshel Serafin Johnson
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | | | - Mohan K Manu
- Department of Respiratory Medicine, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Kavitha Saravu
- Department of Infectious Diseases, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | | | - Suhaj Abdulsalim
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Buraidah, Saudi Arabia
| | - Jitendra Singh
- Department of Translational Medicine, All India Institute of Medical Sciences, Bhopal, India
| | - Chiranjay Mukhopadhyay
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Sonal Sekhar Miraj
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
37
|
Qin L, Fan B, Zhou Y, Zheng J, Diao R, Wang F, Liu J. Targeted gut microbiome therapy: Applications and prospects of probiotics, fecal microbiota transplantation and natural products in the management of type 2 diabetes. Pharmacol Res 2025; 213:107625. [PMID: 39875017 DOI: 10.1016/j.phrs.2025.107625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/21/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is considered as one of the most pressing public health challenges worldwide. Studies have shown significant differences in the gut microbiota between healthy individuals and T2DM patients, suggesting that gut microorganisms may play a key role in the onset and progression of T2DM. This review systematically summarizes the relationship between gut microbiota and T2DM, and explores the mechanisms through which gut microorganisms may alleviate T2DM. Additionally, it evaluates the potential of probiotics, fecal microbiota transplantation (FMT)/virome transplantation (FVT), and natural products in modulating gut microbiota to treat T2DM. Although existing studies have suggested that these interventions may delay or even halt the progression of T2DM, most research remained limited to animal models and observational clinical studies, with a lack of high-quality clinical data. This has led to an imbalance between theoretical research and clinical application. Although some studies have explored the regulatory role of the gut virome on the gut microbiota, research in this area remains in its early stages. Based on these current studies, future research should be focused on large-scale, long-term clinical studies and further investigation on the potential role of the gut virome in T2DM. In conclusion, this review aims to summarize the current evidence and explore the applications of gut microbiota in T2DM treatment, as well as providing recommendations for further investigation in this field.
Collapse
Affiliation(s)
- Luqi Qin
- Key Laboratory of Agro-Products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No.2, Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China
| | - Bei Fan
- Key Laboratory of Agro-Products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No.2, Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China
| | - Yixia Zhou
- Key Laboratory of Agro-Products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No.2, Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China
| | - Jiahuan Zheng
- Key Laboratory of Agro-Products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No.2, Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China
| | - Rao Diao
- Key Laboratory of Agro-Products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No.2, Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China
| | - Fengzhong Wang
- Key Laboratory of Agro-Products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No.2, Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China.
| | - Jiameng Liu
- Key Laboratory of Agro-Products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No.2, Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China.
| |
Collapse
|
38
|
Lu Z, Zhang C, Zhang J, Su W, Wang G, Wang Z. The Kynurenine Pathway and Indole Pathway in Tryptophan Metabolism Influence Tumor Progression. Cancer Med 2025; 14:e70703. [PMID: 40103267 PMCID: PMC11919716 DOI: 10.1002/cam4.70703] [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: 09/19/2024] [Revised: 01/22/2025] [Accepted: 02/04/2025] [Indexed: 03/20/2025] Open
Abstract
Tryptophan (Trp), an essential amino acid, is solely acquired through dietary intake. It is vital for protein biosynthesis and acts as a precursor for numerous key bioactive compounds. The Kynurenine Pathway and the Indole Pathway are the main metabolic routes and are extensively involved in the occurrence and progression of diseases in the digestive, nervous, and urinary systems. In the Kynurenine Pathway, enzymes crucial to tryptophan metabolism, indoleamine-2,3-dioxygenase 1 (IDO1), IDO2, and Trp-2,3-dioxygenase (TDO), trigger tumor immune resistance within the tumor microenvironment and nearby lymph nodes by depleting Trp or by activating the Aromatic Hydrocarbon Receptor (AhR) through its metabolites. Furthermore, IDO1 can influence immune responses via non-enzymatic pathways. The Kynurenine Pathway exerts its effects on tumor growth through various mechanisms, including NAD+ regulation, angiogenesis promotion, tumor metastasis enhancement, and the inhibition of tumor ferroptosis. In the Indole Pathway, indole and its related metabolites are involved in gastrointestinal homeostasis, tumor immunity, and drug resistance. The gut microbiota related to indole metabolism plays a critical role in determining the effectiveness of tumor treatment strategies and can influence the efficacy of immunochemotherapy. It is worth noting that there are conflicting effects of the Kynurenine Pathway and the Indole Pathway on the same tumor phenotype. For example, different tryptophan metabolites affect the cell cycle differently, and indole metabolism has inconsistent protective effects on tumors in different regions. These differences may hold potential for enhancing therapeutic efficacy.
Collapse
Affiliation(s)
- Zhanhui Lu
- Department of Medical Oncology, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
- Shanghai University of Traditional Chinese MedicineShanghaiChina
- Cancer Institute, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Chengcheng Zhang
- Department of Medical Oncology, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
- Shanghai University of Traditional Chinese MedicineShanghaiChina
- Cancer Institute, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Jia Zhang
- Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Wan Su
- Department of Medical Oncology, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Guoying Wang
- Department of Critical Care MedicineThe Second People's Hospital of DongyingDongyingShandongChina
| | - Zhongqi Wang
- Department of Medical Oncology, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
39
|
Tangon N, Kumfu S, Chattipakorn N, Chattipakorn SC. Links between oropharyngeal microbiota and IgA nephropathy: A paradigm shift from isolated microbe to microbiome. Microbiol Res 2025; 292:128005. [PMID: 39675141 DOI: 10.1016/j.micres.2024.128005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
Immunoglobulin A nephropathy (IgAN) is the most prevalent form of primary glomerulonephritis globally, yet its pathogenesis remains incompletely understood. While much research has focused on the gut microbiome in the development of the disease, emerging evidence suggests that the oropharyngeal microbiota may also be a potential contributor. Studies have revealed significant alterations in oropharyngeal microbial diversity and specific bacterial taxa in IgAN patients, correlating with disease severity and progression. This review aims to comprehensively summarize and discuss the key findings from in vitro, in vivo, and clinical studies into the oropharyngeal bacteria and microbiome alterations in IgAN. Clinical studies have identified associations between certain oropharyngeal bacteria, particularly Cnm+Streptococcus mutans, Campylobacter rectus, and Porphyromonas gingivalis with IgAN patients and severe clinical outcomes with. In vitro and in vivo studies further establish a causal relationship between IgAN and oropharyngeal bacteria such as Streptococcus and Haemophilus. Microbiome analyses demonstrate dysbiotic patterns in IgAN patients and identify new potential bacterial genera that have yet to be explored experimentally but may potentially contribute to the disease's pathogenesis. Additionally, the use of these bacterial genera as diagnostic and prognostic biomarkers of IgAN has achieved promising performance. Overall, the evidence highlights the strong connection between oropharyngeal bacteria and IgAN through both causal and non-causal associations. Further investigation into these newly identified bacterial genera and integration of multi-omics data are necessary to uncover mechanisms, validate their role in IgAN, and potentially develop novel diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Narongsak Tangon
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sirinart Kumfu
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand; The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
40
|
Wang N, Pei Z, Wang H, Zhao J, Lu W. Bifidobacterium longum Ameliorates Intestinal Inflammation and Metabolic Biomarkers in Mice Fed a High-Fat Diet with Gliadin by Indoleacrylic Acid. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10486-6. [PMID: 39982644 DOI: 10.1007/s12602-025-10486-6] [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] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
Gliadin, abundant in flour-based foods and processed foods, has been widely researched for allergies. However, the impact of gliadin on the intestinal barrier of healthy individuals and the intervention effect of Bifidobacterium longum (B. longum) are rarely explored. Three strains (JCM1217, CCFM1216, CCFM1218) of B. longum with strong gliadin hydrolysis were screened from 18 strains. This study explored the effects of B. longum on mice with a 10-week high-fat diet and 6% gliadin (HFD + 6%G), assessing duodenal health, lipid metabolism, metabolomics, and gut microbiota in the duodenum and colon changes. Three B. longum strains were screened for gliadin hydrolysis to produce minimal R5 immunopeptide production. All three B. longum strains improved duodenal morphology, reduced intestinal permeability, reduced inflammation (IL-15), and activated tryptophan metabolism. Additionally, alterations in the microbiota of the duodenum and colon were also observed. Linear discriminant analysis (LDA) showed that the HFD + 6% G group significantly increased the abundance of Ileibacterium, Alistipes, Bacteroides, Candidatus, Saccharimonas, Streptococcus, Sediminibacterium, and Odoribacterium in the duodenum. The abundance of Blautia, Butyricimonas, Ruminococcaceae UCG-010, Parabacterioids, and Eubacterium nodatum in the colon was also increased. The B. longum CCFM1216 and B. longum CCFM1218 reversed the abundance of these strains. Specifically, B. longum CCFM1216 enhanced the duodenal barrier with indoleacrylic acid, beneficial for blood lipids and glucose. These strains may be used as probiotics for gliadin-related diseases.
Collapse
Affiliation(s)
- Ning Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Zhangming Pei
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hongchao Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, China.
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China.
| |
Collapse
|
41
|
Qi Z, Cao J, Liu J, Chen J, Chen S, Zhang L, Xu J, Wu D, Wu Y, Li G. Toxicological mechanisms of carbon polymers in accelerating cognitive decline in Alzheimer's disease. J Adv Res 2025:S2090-1232(25)00115-8. [PMID: 39983830 DOI: 10.1016/j.jare.2025.02.017] [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: 12/20/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/23/2025] Open
Abstract
INTRODUCTION Alzheimer's disease (AD) is the primary cause of dementia and is emerging as a global threat to human health. Increased availability of processed food is identified as a crucial environmental risk factor underlying the prevalence of Alzheimer's disease. Carbon polymers (CPs), as neo-formed substances and ubiquitous in thermally processed foods, the relationship between them and AD onset is remains unclear. OBJECTIVES The effect of CPs on AD onset was examined and the toxicological mechanisms of prolonged exposure to CPs derived from thermal processed foods on AD progression were comprehensively investigated using a scopolamine-induced neuroinflammatory cell models and the transgenic APPswe/PSEN1dE9 (APP/PS1) AD mouse. METHODS The CPs were extracted from thermally processed foods and the effects of CPs exposure on oxidative stress in neuroinflammatory cells were evaluated using scopolamine-induced PC12 cells as a neuroinflammation model. Furthermore, APP/PS1 AD mice were used to validate the potential adverse impacts of prolonged exposure to CPs on AD progression through the Morris water maze and open field test. In addition, histopathological examination, including immunofluorescence, immunohistochemistry, Nissl staining, and H&E, of the brain tissue in AD mice after chronic CPs treatment was performed to elucidate the underlying risk of dietary exposure to CPs on AD progression. RESULTS Exposure to CPs enhanced oxidative damage in neuroinflammatory cells, as demonstrated by impaired mitochondrial function and activated NF-κB/MAPK signaling pathways. Further results from electron spin resonance substantiated the catalytic properties of CPs, which accelerated oxidative damage through promoting free radical generation. Using transgenic AD mice model, our findings also demonstrated that prolonged CPs exposure aggravated AD-associated pathology, as evidenced by increased amyloid-beta deposition and glial cell activation, ultimately accelerating cognitive decline. CONCLUSION These findings provide compelling evidence of the potential health risks associated with long-term dietary exposure to CPs and provide insight into the relationship between foodborne risk factors and neurodegenerative diseases.
Collapse
Affiliation(s)
- Zihe Qi
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, People's Republic of China
| | - Juanjuan Cao
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, People's Republic of China
| | - Jianghua Liu
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, People's Republic of China
| | - Jian Chen
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, People's Republic of China
| | - Shasha Chen
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, People's Republic of China
| | - Luyao Zhang
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, People's Republic of China
| | - Jingwen Xu
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, People's Republic of China
| | - Di Wu
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast BT9 5DL, United Kingdom
| | - Yongning Wu
- NHC Key Laboratory of Food Safety Risk Assessment, Food Safety Research Unit (2019RU014) of Chinese Academy of Medical Science, China National Center for Food Safety Risk Assessment, Beijing 100021, China
| | - Guoliang Li
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, People's Republic of China.
| |
Collapse
|
42
|
Blok L, Hanssen N, Nieuwdorp M, Rampanelli E. From Microbes to Metabolites: Advances in Gut Microbiome Research in Type 1 Diabetes. Metabolites 2025; 15:138. [PMID: 39997763 PMCID: PMC11857261 DOI: 10.3390/metabo15020138] [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: 02/01/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Type 1 diabetes (T1D) is a severe chronic T-cell mediated autoimmune disease that attacks the insulin-producing beta cells of the pancreas. The multifactorial nature of T1D involves both genetic and environmental components, with recent research focusing on the gut microbiome as a crucial environmental factor in T1D pathogenesis. The gut microbiome and its metabolites play an important role in modulating immunity and autoimmunity. In recent years, studies have revealed significant alterations in the taxonomic and functional composition of the gut microbiome associated with the development of islet autoimmunity and T1D. These changes include reduced production of short-chain fatty acids, altered bile acid and tryptophan metabolism, and increased intestinal permeability with consequent perturbations of host (auto)immune responses. Methods/Results: In this review, we summarize and discuss recent observational, mechanistic and etiological studies investigating the gut microbiome in T1D and elucidating the intricate role of gut microbes in T1D pathogenesis. Moreover, we highlight the recent advances in intervention studies targeting the microbiota for the prevention or treatment of human T1D. Conclusions: A deeper understanding of the evolution of the gut microbiome before and after T1D onset and of the microbial signals conditioning host immunity may provide us with essential insights for exploiting the microbiome as a prognostic and therapeutic tool.
Collapse
Affiliation(s)
- Lente Blok
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Nordin Hanssen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Elena Rampanelli
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
- Amsterdam Institute for Infection and Immunity (AII), Amsterdam, The Netherlands
| |
Collapse
|
43
|
Cheng C, Xu F, Pan XF, Wang C, Fan J, Yang Y, Liu Y, Sun L, Liu X, Xu Y, Zhou Y, Xiao C, Gou W, Miao Z, Yuan J, Shen L, Fu Y, Sun X, Zhu Y, Chen Y, Pan A, Zhou D, Zheng JS. Genetic mapping of serum metabolome to chronic diseases among Han Chinese. CELL GENOMICS 2025; 5:100743. [PMID: 39837327 PMCID: PMC11872534 DOI: 10.1016/j.xgen.2024.100743] [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: 05/01/2024] [Revised: 10/31/2024] [Accepted: 12/24/2024] [Indexed: 01/23/2025]
Abstract
Serum metabolites are potential regulators for chronic diseases. To explore the genetic regulation of metabolites and their roles in chronic diseases, we quantified 2,759 serum metabolites and performed genome-wide association studies (GWASs) among Han Chinese individuals. We identified 184 study-wide significant (p < 1.81 × 10-11) metabolite quantitative trait loci (metaboQTLs), 88.59% (163) of which were novel. Notably, we identified Asian-ancestry-specific metaboQTLs, including the SNP rs2296651 for taurocholic acid and taurochenodesoxycholic acid. Leveraging the GWAS for 37 clinical traits from East Asians, Mendelian randomization analyses identified 906 potential causal relationships between metabolites and clinical traits, including 27 for type 2 diabetes and 38 for coronary artery disease. Integrating genetic regulation of the transcriptome and proteome revealed putative regulators of several metabolites. In summary, we depict a landscape of the genetic architecture of the serum metabolome among Han Chinese and provide insights into the role of serum metabolites in chronic diseases.
Collapse
Affiliation(s)
- Chunxiao Cheng
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou 310058, Zhejiang, China
| | - Fengzhe Xu
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou 310024, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
| | - Xiong-Fei Pan
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & National Medical Products Administration Key Laboratory for Technical Research on Drug Products In Vitro and In Vivo Correlation, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Shuangliu Institute of Women's and Children's Health, Shuangliu Maternal and Child Health Hospital, Chengdu 610200, China; West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Cheng Wang
- Department of Clinical Nutrition, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510012, China
| | - Jiayao Fan
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou 310058, Zhejiang, China
| | - Yunhaonan Yang
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & National Medical Products Administration Key Laboratory for Technical Research on Drug Products In Vitro and In Vivo Correlation, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yuanjiao Liu
- Department of Epidemiology & Biostatistics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lingyun Sun
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou 310058, Zhejiang, China
| | - Xiaojuan Liu
- Department of Laboratory Medicine, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yue Xu
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou 310058, Zhejiang, China
| | - Yuan Zhou
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou 310058, Zhejiang, China
| | - Congmei Xiao
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou 310024, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
| | - Wanglong Gou
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou 310024, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
| | - Zelei Miao
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou 310024, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
| | - Jiaying Yuan
- Department of Science and Education & Shuangliu Institute of Women's and Children's Health, Shuangliu Maternal and Child Health Hospital, Chengdu, Sichuan 610200, China
| | - Luqi Shen
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou 310024, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
| | - Yuanqing Fu
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou 310024, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
| | - Xiaohui Sun
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yimin Zhu
- Department of Epidemiology & Biostatistics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuming Chen
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - An Pan
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Dan Zhou
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou 310058, Zhejiang, China.
| | - Ju-Sheng Zheng
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou 310024, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China; Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou 310024, China; Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| |
Collapse
|
44
|
Elmassry MM, Sugihara K, Chankhamjon P, Kim Y, Camacho FR, Wang S, Sugimoto Y, Chatterjee S, Chen LA, Kamada N, Donia MS. A meta-analysis of the gut microbiome in inflammatory bowel disease patients identifies disease-associated small molecules. Cell Host Microbe 2025; 33:218-234.e12. [PMID: 39947133 DOI: 10.1016/j.chom.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 08/14/2024] [Accepted: 01/06/2025] [Indexed: 02/19/2025]
Abstract
Gut microbiome changes have been associated with several human diseases, but the molecular and functional details underlying these associations remain largely unknown. Here, we performed a meta-analysis of small molecule biosynthetic gene clusters (BGCs) in metagenomic samples of the gut microbiome from inflammatory bowel disease (IBD) patients and matched healthy subjects and identified two Clostridia-derived BGCs that are significantly associated with Crohn's disease (CD), a main IBD type. Using synthetic biology, we discovered and solved the structures of six fatty acid amides as the products of the CD-enriched BGCs, which we subsequently detected in fecal samples from IBD patients. Finally, we show that the discovered molecules disrupt gut permeability and exacerbate disease in chemically or genetically susceptible mouse models of colitis. These findings suggest that microbiome-derived small molecules may play a role in the etiology of IBD and represent a generalizable approach for discovering molecular mediators of disease-relevant microbiome-host interactions.
Collapse
Affiliation(s)
- Moamen M Elmassry
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Kohei Sugihara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Yeji Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Francine R Camacho
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Shuo Wang
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Yuki Sugimoto
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Seema Chatterjee
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Lea Ann Chen
- Department of Medicine, Division of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mohamed S Donia
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
45
|
Chi J, Patterson JS, Jin Y, Kim KJ, Lalime N, Hawley D, Lewis F, Li L, Wang X, Campen MJ, Cui JY, Gu H. Metabolic Reprogramming in Gut Microbiota Exposed to Polystyrene Microplastics. Biomedicines 2025; 13:446. [PMID: 40002859 PMCID: PMC11853289 DOI: 10.3390/biomedicines13020446] [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: 12/23/2024] [Revised: 01/26/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Microplastics (MPs) are small plastic fragments with diameters less than 5 mm in size and are prevalent in everyday essentials and consumables. Large global plastic production has now led to a flooding of MPs in our natural environment. Due to their detrimental impacts on the planet's ecosystems and potentially our health, MPs have emerged as a significant public health concern. In this pilot study, we hypothesize that MPs exposure will negatively affect gut microbiota composition and function, in which metabolic reprogramming plays an important role. Methods: Using in vitro experiments, three bacterial strains (Escherichia coli MG1655, Nissle 1917, and Lactobacillus rhamnosus) were selected to investigate the impacts of MPs exposure. The bacterial strains were individually cultured in an anaerobic chamber and exposed to 1 µm polystyrene MPs at various concentrations (0, 10, 20, 50, 100, and 500 µg/mL) in the culture medium. Results: MPs exposure reduced the growth of all three bacterial strains in a dose-dependent manner. Liquid chromatography mass spectrometry (LC-MS)-based untargeted metabolomics revealed significant differences in multiple metabolic pathways, such as sulfur metabolism and amino sugar and nucleotide sugar metabolism. In addition, we extracted gut microbiota from C57BL/6 mice, and 16S rRNA sequencing results showed a significant upregulation of Lactobacillales and a significant reduction in Erysipelotrichales due to MPs exposure. Furthermore, targeted and untargeted metabolomics corroborated the in vitro results and revealed alterations in microbial tryptophan metabolism and energy producing pathways, such as glycolysis/gluconeogenesis and the pentose phosphate pathway. Conclusions: These findings provide evidence that MPs exposure causes comprehensive changes to healthy gut microbiota, which may also provide insights into the mechanistic effects of MPs exposure in humans.
Collapse
Affiliation(s)
- Jinhua Chi
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (J.C.); (J.S.P.); (L.L.)
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA;
| | - Jeffrey S. Patterson
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (J.C.); (J.S.P.); (L.L.)
| | - Yan Jin
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA;
| | - Kyle Joohyung Kim
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA; (K.J.K.); (J.Y.C.)
| | - Nicole Lalime
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA;
| | - Daniella Hawley
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA; (D.H.); (X.W.)
| | - Freeman Lewis
- Environmental Health Sciences, Florida International University, Miami, FL 33199, USA;
| | - Lingjun Li
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (J.C.); (J.S.P.); (L.L.)
| | - Xuan Wang
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA; (D.H.); (X.W.)
| | - Matthew J. Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences, Albuquerque, NM 87106, USA;
| | - Julia Yue Cui
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA; (K.J.K.); (J.Y.C.)
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (J.C.); (J.S.P.); (L.L.)
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA;
| |
Collapse
|
46
|
Bian Z, Li Z, Chang H, Luo J, Jian S, Zhang J, Lin P, Deng B, Deng J, Zhang L. Resveratrol Ameliorates Chronic Stress in Kennel Dogs and Mice by Regulating Gut Microbiome and Metabolome Related to Tryptophan Metabolism. Antioxidants (Basel) 2025; 14:195. [PMID: 40002382 PMCID: PMC11851397 DOI: 10.3390/antiox14020195] [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: 01/06/2025] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Chronic stress poses threats to the physical and psychological well-being of dogs. Resveratrol (Res) is a polyphenol with antidepressant properties and has rarely been studied in dogs. This study aimed to investigate the stress-relieving effects and underlying mechanism of Res in dogs. Dogs were fed a basal diet supplemented with Res for 35 days. The fecal microbiota of the dogs was cultured with Res in vitro. The results show that Res improved the stress-related behaviors and increased the serum levels of 5-hydroxytryptamine (5-HT), brain-derived neurotrophic factor (BDNF), immunoglobulin A, and antioxidant capacity in dogs. Res downregulated the hormones of the hypothalamic-pituitary-adrenal axis. The abundance of butyric-producing bacteria, like Blautia, increased, while the growth of Fusobacterium related to gut inflammation was inhibited in the Res group. A higher content of fecal butyric acid was observed in the Res group. The metabolome indicated that Res increased the fecal and serum levels of tryptophan (Trp) and decreased the consumption of Trp by microorganisms. A chronic unpredictable mild stress mouse model was established, and Res was administered for 35 days. The results show that Res ameliorated the stress-related behavior and increased the levels of Trp and 5-HT in the whole brains of mice. The relative mRNA expression of genes associated with the tight junction protein, aryl hydrocarbon receptor, and Trp transporters in the colon were upregulated. In conclusion, Res could ameliorate canine stress by increasing 5-HT, BDNF, and the antioxidant capacity and improving the immune function and stress response, which was attributed to the role of Res in the restructuring of gut microbiota and the modulation of tryptophan metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jinping Deng
- Laboratory of Companion Animal Science, Department of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Z.B.); (Z.L.); (H.C.); (J.L.); (S.J.); (J.Z.); (P.L.); (B.D.)
| | - Lingna Zhang
- Laboratory of Companion Animal Science, Department of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Z.B.); (Z.L.); (H.C.); (J.L.); (S.J.); (J.Z.); (P.L.); (B.D.)
| |
Collapse
|
47
|
Zarei P, Sedeh PA, Vaez A, Keshteli AH. Using metabolomics to investigate the relationship between the metabolomic profile of the intestinal microbiota derivatives and mental disorders in inflammatory bowel diseases: a narrative review. Res Pharm Sci 2025; 20:1-24. [PMID: 40190827 PMCID: PMC11972020 DOI: 10.4103/rps.rps_273_23] [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: 12/20/2023] [Revised: 04/30/2024] [Accepted: 05/28/2024] [Indexed: 04/09/2025] Open
Abstract
Individuals with inflammatory bowel disease (IBD) are at a higher risk of developing mental disorders, such as anxiety and depression. The imbalance between the intestinal microbiota and its host, known as dysbiosis, is one of the factors, disrupting the balance of metabolite production and their signaling pathways, leading to disease progression. A metabolomics approach can help identify the role of gut microbiota in mental disorders associated with IBD by evaluating metabolites and their signaling comprehensively. This narrative review focuses on metabolomics studies that have comprehensively elucidated the altered gut microbial metabolites and their signaling pathways underlying mental disorders in IBD patients. The information was compiled by searching PubMed, Web of Science, Scopus, and Google Scholar from 2005 to 2023. The findings indicated that intestinal microbial dysbiosis in IBD patients leads to mental disorders such as anxiety and depression through disturbances in the metabolism of carbohydrates, sphingolipids, bile acids, neurotransmitters, neuroprotective, inflammatory factors, and amino acids. Furthermore, the reduction in the production of neuroprotective factors and the increase in inflammation observed in these patients can also contribute to the worsening of psychological symptoms. Analyzing the metabolite profile of the patients and comparing it with that of healthy individuals using advanced technologies like metabolomics, aids in the early diagnosis and prevention of mental disorders. This approach allows for the more precise identification of the microbes responsible for metabolite production, enabling the development of tailored dietary and pharmaceutical interventions or targeted manipulation of microbiota.
Collapse
Affiliation(s)
- Parvin Zarei
- Department of Bioinformatics, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Peyman Adibi Sedeh
- Isfahan Gastroenterology and Hepatology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ahmad Vaez
- Department of Bioinformatics, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Epidemiology, University of Groningen, University Medical Centre Groningen, 9713 GZ Groningen, The Netherlands
| | | |
Collapse
|
48
|
Shin HK, Bang YJ. Aromatic Amino Acid Metabolites: Molecular Messengers Bridging Immune-Microbiota Communication. Immune Netw 2025; 25:e10. [PMID: 40078785 PMCID: PMC11896664 DOI: 10.4110/in.2025.25.e10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Aromatic amino acid (AAA) metabolites, derived from tryptophan, phenylalanine, and tyrosine through coordinated host and microbial metabolism, have emerged as critical modulators of immune function. We examine the complex journey of AAAs from dietary intake through intestinal absorption and metabolic transformation, highlighting the crucial role of host-microbe metabolic networks in generating diverse immunomodulatory compounds. This review provides a unique integrative perspective by mapping the molecular mechanisms through which these metabolites orchestrate immune responses. Through detailed analysis of metabolite-receptor and metabolite-transporter interactions, we reveal how specific molecular recognition drives cell type-specific immune responses. Our comprehensive examination of signaling networks-from membrane receptor engagement to nuclear receptor activation to post-translational modifications- demonstrates how the same metabolite can elicit distinct functional outcomes in different immune cell populations. The context-dependent nature of these molecular interactions presents both challenges and opportunities for therapeutic development, particularly in inflammatory conditions where metabolite signaling pathways are dysregulated. Understanding the complexity of these regulatory networks and remaining knowledge gaps is fundamental for advancing metabolite-based therapeutic strategies in immune-mediated disorders.
Collapse
Affiliation(s)
- Hyun-Ki Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ye-Ji Bang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul 03080, Korea
| |
Collapse
|
49
|
Rocha CS, Alexander KL, Herrera C, Weber MG, Grishina I, Hirao LA, Kramer DJ, Arredondo J, Mende A, Crakes KR, Fenton AN, Marco ML, Mills DA, Kappes JC, Smythies LE, Ziprin P, Sankaran-Walters S, Smith PD, Dandekar S. Microbial remodeling of gut tryptophan metabolism and indole-3-lactate production regulate epithelial barrier repair and viral suppression in human and simian immunodeficiency virus infections. Mucosal Immunol 2025:S1933-0219(25)00011-X. [PMID: 39894082 DOI: 10.1016/j.mucimm.2025.01.011] [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: 07/30/2024] [Revised: 01/02/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
Gut inflammatory diseases cause microbial dysbiosis. Human immunodeficiency virus-1 (HIV) infection disrupts intestinal integrity, subverts repair/renewal pathways, impairs mucosal immunity and propels microbial dysbiosis. However, microbial metabolic mechanisms driving repair mechanisms in virally inflamed gut are not well understood. We investigated the capability and mechanisms of gut microbes to restore epithelial barriers and mucosal immunity in virally inflamed gut by using a multipronged approach: an in vivo simian immunodeficiency virus (SIV)-infected nonhuman primate model of HIV/AIDS, ex vivo HIV-exposed human colorectal explants and primary human intestinal epithelial cells. SIV infection reprogrammed tryptophan (TRP) metabolism, increasing kynurenine catabolite levels that are associated with mucosal barrier disruption and immune suppression. Administration of Lactiplantibacillus plantarum or Bifidobacterium longum subsp. infantis into the SIV-inflamed gut lumen in vivo resulted in rapid reprogramming of microbial TRP metabolism towards indole-3-lactic acid (ILA) production. This shift accelerated epithelial repair and enhanced anti-viral defenses through induction of IL-22 signaling in mucosal T cells and aryl hydrocarbon receptor activation. Additionally, ILA treatment of human colorectal tissue explants ex vivo inhibited HIV replication by reducing mucosal inflammatory cytokine production and cell activation. Our findings underscore the therapeutic potential of microbial metabolic reprogramming of TRP-to-ILA and mechanisms in mitigating viral pathogenic effects and bolstering mucosal defenses for HIV eradication.
Collapse
Affiliation(s)
- Clarissa Santos Rocha
- Department of Medical Microbiology & Immunology, University of California Davis, Davis, CA, 95616, United States
| | - Katie L Alexander
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, UK
| | - Carolina Herrera
- Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, United States
| | - Mariana G Weber
- Department of Medical Microbiology & Immunology, University of California Davis, Davis, CA, 95616, United States
| | - Irina Grishina
- Department of Medical Microbiology & Immunology, University of California Davis, Davis, CA, 95616, United States
| | - Lauren A Hirao
- Department of Medical Microbiology & Immunology, University of California Davis, Davis, CA, 95616, United States
| | - Dylan J Kramer
- Department of Medical Microbiology & Immunology, University of California Davis, Davis, CA, 95616, United States
| | - Juan Arredondo
- Department of Medical Microbiology & Immunology, University of California Davis, Davis, CA, 95616, United States
| | - Abigail Mende
- Department of Medical Microbiology & Immunology, University of California Davis, Davis, CA, 95616, United States
| | - Katti R Crakes
- Department of Medical Microbiology & Immunology, University of California Davis, Davis, CA, 95616, United States
| | - Anne N Fenton
- Department of Medical Microbiology & Immunology, University of California Davis, Davis, CA, 95616, United States
| | - Maria L Marco
- Department of Food Science and Technology, University of California Davis, Davis, CA, 95616, United States
| | - David A Mills
- Department of Food Science and Technology, University of California Davis, Davis, CA, 95616, United States
| | - John C Kappes
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, UK
| | - Lesley E Smythies
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, UK; Birmingham Veterans Affairs Medical Center, Research Service, Birmingham, AL 35233, UK
| | - Paul Ziprin
- Department of Surgery and Cancer, St. Mary's Hospital, Imperial College London, London, UK
| | - Sumathi Sankaran-Walters
- Department of Medical Microbiology & Immunology, University of California Davis, Davis, CA, 95616, United States
| | - Phillip D Smith
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, UK
| | - Satya Dandekar
- Department of Medical Microbiology & Immunology, University of California Davis, Davis, CA, 95616, United States.
| |
Collapse
|
50
|
Sampson TR, Tansey MG, West AB, Liddle RA. Lewy body diseases and the gut. Mol Neurodegener 2025; 20:14. [PMID: 39885558 PMCID: PMC11783828 DOI: 10.1186/s13024-025-00804-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/21/2025] [Indexed: 02/01/2025] Open
Abstract
Gastrointestinal (GI) involvement in Lewy body diseases (LBDs) has been observed since the initial descriptions of patients by James Parkinson. Recent experimental and human observational studies raise the possibility that pathogenic alpha-synuclein (⍺-syn) might develop in the GI tract and subsequently spread to susceptible brain regions. The cellular and mechanistic origins of ⍺-syn propagation in disease are under intense investigation. Experimental LBD models have implicated important contributions from the intrinsic gut microbiome, the intestinal immune system, and environmental toxicants, acting as triggers and modifiers to GI pathologies. Here, we review the primary clinical observations that link GI dysfunctions to LBDs. We first provide an overview of GI anatomy and the cellular repertoire relevant for disease, with a focus on luminal-sensing cells of the intestinal epithelium including enteroendocrine cells that express ⍺-syn and make direct contact with nerves. We describe interactions within the GI tract with resident microbes and exogenous toxicants, and how these may directly contribute to ⍺-syn pathology along with related metabolic and immunological responses. Finally, critical knowledge gaps in the field are highlighted, focusing on pivotal questions that remain some 200 years after the first descriptions of GI tract dysfunction in LBDs. We predict that a better understanding of how pathophysiologies in the gut influence disease risk and progression will accelerate discoveries that will lead to a deeper overall mechanistic understanding of disease and potential therapeutic strategies targeting the gut-brain axis to delay, arrest, or prevent disease progression.
Collapse
Affiliation(s)
- Timothy R Sampson
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30329, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Malú Gámez Tansey
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
- Normal Fixel Institute of Neurological Diseases, Gainesville, FL, 32608, USA
| | - Andrew B West
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
- Duke Center for Neurodegeneration and Neurotherapeutic Research, Department of Pharmacology and Cancer Biology, Durham, NC, 27710, USA.
| | - Rodger A Liddle
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
- Duke Institute for Brain Sciences, Duke University, Durham, NC, 27710, USA.
- Department of Medicine, Duke University and Department of Veterans Affairs Health Care System, Durham, NC, 27710, USA.
| |
Collapse
|