1
|
Xu Y, Gao Z, Liu J, Yang Q, Xu S. Role of gut microbiome in suppression of cancers. Gut Microbes 2025; 17:2495183. [PMID: 40254597 PMCID: PMC12013426 DOI: 10.1080/19490976.2025.2495183] [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: 12/14/2024] [Revised: 03/23/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025] Open
Abstract
The pathogenesis of cancer is closely related to the disruption of homeostasis in the human body. The gut microbiome plays crucial roles in maintaining the homeostasis of its host throughout lifespan. In recent years, a large number of studies have shown that dysbiosis of the gut microbiome is involved in the entire process of cancer initiation, development, and prognosis by influencing the host immune system and metabolism. Some specific intestinal bacteria promote the occurrence and development of cancers under certain conditions. Conversely, some other specific intestinal bacteria suppress the oncogenesis and progression of cancers, including inhibiting the occurrence of cancers, delaying the progression of cancers and boosting the therapeutic effect on cancers. The promoting effects of the gut microbiome on cancers have been comprehensively discussed in the previous review. This article will review the latest advances in the roles and mechanisms of gut microbiome in cancer suppression, providing a new perspective for developing strategies of cancer prevention and treatment.
Collapse
Affiliation(s)
- Yao Xu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Zhaoyu Gao
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, P. R. China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| | - Jiaying Liu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Qianqian Yang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Shunjiang Xu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, P. R. China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| |
Collapse
|
2
|
Zöggeler T, Kavallar AM, Pollio AR, Aldrian D, Decristoforo C, Scholl-Bürgi S, Müller T, Vogel GF. Meta-analysis of shotgun sequencing of gut microbiota in obese children with MASLD or MASH. Gut Microbes 2025; 17:2508951. [PMID: 40396204 PMCID: PMC12101585 DOI: 10.1080/19490976.2025.2508951] [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: 07/31/2024] [Revised: 03/25/2025] [Accepted: 05/14/2025] [Indexed: 05/22/2025] Open
Abstract
Alterations in the gut microbiome affect the development and severity of metabolic dysfunction-associated steatotic liver disease (MASLD) or metabolic dysfunction-associated steatohepatitis (MASH). We analyzed microbiomes of obese children with and without MASLD, MASH, and healthy controls. Electronic databases were searched for studies on the gut microbiome in children with obesity with/without MASLD or MASH, providing shotgun-metagenomic-sequencing data. Nine studies and an additionally recruited cohort were included. Fecal microbiomes of children with MASLD (n = 153) and MASH (n = 70) were significantly different in alpha- and beta-diversity (p < 0.001) compared to obese (n = 58) and healthy (n = 132). Species Faecalibacterium_prausnitzii and Prevotella_copri are differentially abundant between obese, MASLD and MASH groups. XGBoost and random forest-models accurately predict MASLD over obesity with an AUROC of 87% and MASH over MASLD with 89%. Pathway-abundance-based models accurately predict MASLD over obesity with an AUROC of 81% and MASH over MASLD with 88%. The composition of the gut microbiome is altered with increasing hepatic fibrosis and concomitant species-abundance increase of Prevotella_copri (p = 0.0082). Machine-learning models discriminate pediatric from adult MASH with an AUROC of 97%. The gut microbial composition is increasingly altered in children with the progression of MASLD toward MASH. This can be utilized as a fecal biomarker and highlights the impact of diet on the gut microbiome for disease intervention.
Collapse
Affiliation(s)
- Thomas Zöggeler
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Maria Kavallar
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Adam Robert Pollio
- Institute of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Denise Aldrian
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Sabine Scholl-Bürgi
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Müller
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Friedrich Vogel
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
- Institute of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
3
|
Zheng W, Lin X, Chen H, Yang Z, Zhao H, Li S, Song T, Sun Y. Gut microbiota and endometrial cancer: research progress on the pathogenesis and application. Ann Med 2025; 57:2451766. [PMID: 39810645 PMCID: PMC11737052 DOI: 10.1080/07853890.2025.2451766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/02/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
As one of the three major malignant tumors in women, the morbidity of endometrial cancer is second only to that of cervical cancer and is increasing yearly. Its etiological mechanism is not clear, and the risk factors are numerous and common and are closely related to obesity, hypertension, diabetes, etc. The gut microbiota has many strains, which play a considerable part in normal digestion and absorption in the human body and the regulation of the immune response. In the last few years, research on the gut microbiota has been unprecedentedly popular, and it has been confirmed that the gut microbiota closely correlates with the occurrence and development of all kinds of benign and malignant diseases. In this article, the effects of the gut microbiota and its metabolites on the occurrence and development of endometrial cancer is reviewed, and its application in the prevention, diagnosis and treatment of endometrial cancer is explored.
Collapse
Affiliation(s)
- Weiqin Zheng
- Department of Gynecology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaowen Lin
- Department of Gynecology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huixin Chen
- Department of Gynecology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ziling Yang
- Department of Gynecology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Han Zhao
- Department of Gynecology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shibo Li
- Department of Gynecology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tao Song
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuhui Sun
- Department of Gynecology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
4
|
Driuchina A, Isola V, Hulmi JJ, Salmi VM, Hintikka J, Ahtiainen JP, Pekkala S. Unveiling the impact of competition weight loss on gut microbiota: alterations in diversity, composition, and predicted metabolic functions. J Int Soc Sports Nutr 2025; 22:2474561. [PMID: 40033182 PMCID: PMC11881659 DOI: 10.1080/15502783.2025.2474561] [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: 03/26/2024] [Accepted: 02/26/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Competitive sports and sports nutrition, popular among amateur athletes aiming for a lean physique, have limited research on gut microbiota. METHODS We conducted a 46-week study to analyze the consequences of fat loss and diet restrictions in 23 fitness athletes who prepared for a physique competition. Body composition, dietary intakes, serum cytokines and chemokines, and fecal samples were analyzed. RESULTS Fat loss through caloric restriction and aerobic exercise led to an increased phylogenetic diversity of gut microbiota and changes in the composition of gut microbiota, with Faecalibacterium, Lachnospiraceae, Bacteroides, and Intestinimonas showing altered abundances. Fat loss also changed the predicted microbial functions responsible for the metabolism of carbohydrates and amino acids. Consumption of energy, carbohydrates, fiber, vitamins and minerals, and various fatty acids decreased during the preparation for the competition, which was partly associated with changes in gut microbiota. Several cytokine levels decreased (IL1a, IL1b, IL10, and TFNα), and certain chemokine levels increased (GROa and RANTES). During the 23-week regain period after the competition, gut microbiota showed signs of recovery, with increased diversity compared to pre- and post-competition measurements. Most taxonomic changes returned to their baseline levels after the regain period. CONCLUSIONS The study highlights the dynamic nature of gut microbiota and its response to fat loss and regain in non-obese fitness/physique competitors and provides novel insights into how competitive sports and sports nutrition can influence the gut ecosystem.
Collapse
Affiliation(s)
- Anastasiia Driuchina
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Ville Isola
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Juha J Hulmi
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Vera M Salmi
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Jukka Hintikka
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Juha P Ahtiainen
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Satu Pekkala
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
- Turku University Hospital, Department of Clinical Microbiology, Turku, Finland
| |
Collapse
|
5
|
Loison L, Huré M, Lefranc B, Leprince J, Bôle-Feysot C, Coëffier M, Ribet D. Staphylococcus warneri dampens SUMOylation and promotes intestinal inflammation. Gut Microbes 2025; 17:2446392. [PMID: 39819277 DOI: 10.1080/19490976.2024.2446392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 11/28/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025] Open
Abstract
Gut bacteria play key roles in intestinal physiology, via the secretion of diversified bacterial effectors. Many of these effectors remodel the host proteome, either by altering transcription or by regulating protein post-translational modifications. SUMOylation, a ubiquitin-like post-translational modification playing key roles in intestinal physiology, is a target of gut bacteria. Mutualistic gut bacteria can promote SUMOylation, via the production of short- or branched-chain fatty acids (SCFA/BCFA). In contrast, several pathogenic bacteria were shown to dampen SUMOylation in order to promote infection. Here, we demonstrate that Staphylococcus warneri, a natural member of the human gut microbiota, decreases SUMOylation in intestinal cells. We identify that Warnericin RK, a hemolytic toxin secreted by S. warneri, targets key components of the host SUMOylation machinery, leading to the loss of SUMO-conjugated proteins. We further demonstrate that Warnericin RK promotes inflammation in intestinal and immune cells using both SUMO-dependent and SUMO-independent mechanisms. We finally show that Warnericin RK regulates the expression of genes involved in intestinal tight junctions. Together, these results highlight the diversity of mechanisms used by bacteria from the gut microbiota to manipulate host SUMOylation. They further highlight that changes in gut microbiota composition may impact intestinal inflammation, by altering the equilibrium between bacterial effectors promoting or dampening SUMOylation.
Collapse
Affiliation(s)
- Léa Loison
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, Rouen, France
| | - Marion Huré
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, Rouen, France
| | - Benjamin Lefranc
- Univ Rouen Normandie, INSERM, Normandie Univ, NorDiC, UMR 1239, PRIMACEN, Rouen, France
| | - Jérôme Leprince
- Univ Rouen Normandie, INSERM, Normandie Univ, NorDiC, UMR 1239, PRIMACEN, Rouen, France
| | - Christine Bôle-Feysot
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, Rouen, France
| | - Moïse Coëffier
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, CHU Rouen, Department of Nutrition, CIC-CRB1404, Rouen, France
| | - David Ribet
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, Rouen, France
| |
Collapse
|
6
|
Qiao S, Wang T, Sun J, Han J, Dai H, Du M, Yang L, Guo CJ, Liu C, Liu SJ, Liu H. Cross-feeding-based rational design of a probiotic combination of Bacterides xylanisolvens and Clostridium butyricum therapy for metabolic diseases. Gut Microbes 2025; 17:2489765. [PMID: 40190016 PMCID: PMC11980479 DOI: 10.1080/19490976.2025.2489765] [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: 04/29/2024] [Revised: 12/13/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
The human gut microbiota has gained interest as an environmental factor that contributes to health or disease. The development of next-generation live biotherapeutic products (LBPs) is a promising strategy to modulate the gut microbiota and improve human health. In this study, we identified a novel cross-feeding interaction between Bacteroides xylanisolvens and Clostridium butyricum and developed their combination into a novel LBP for treating metabolic syndrome. Using in-silico analysis and in vitro experiments, we demonstrated that B. xylanisolvens supported the growth and butyrate production of C. butyricum by supplying folate, while C. butyricum reciprocated by providing pABA for folate biosynthesis. Animal gavage experiments showed that the two-strain combination LBP exhibited superior therapeutic efficacy against metabolic disorders in high-fat diet-induced obese (DIO) mice compared to either single-strain treatment. Further omics-based analyses revealed that the single-strain treatments exhibited distinct taxonomic preferences in modulating the gut microbiota, whereas the combination LBP achieved more balanced modulation to preserve taxonomic diversity to a greater extent, thereby enhancing the stability and resilience of the gut microbiome. Moreover, the two-strain combinations more effectively restored gut microbial functions by reducing disease-associated pathways and opportunistic pathogen abundance. This work demonstrates the development of new LBP therapy for metabolic diseases from cross-feeding microbial pairs which exerted better self-stability and robust efficacy in complex intestinal environments compared to conventional single-strain LBPs.
Collapse
Affiliation(s)
- Shanshan Qiao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Tao Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Jingzu Sun
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Junjie Han
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Huanqin Dai
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Mengxuan Du
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Lan Yang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Chun-Jun Guo
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Chang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
| | - Hongwei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| |
Collapse
|
7
|
Martinez-Medina JN, Ghazisaeedi F, Kramer C, Ziegler JF, McParland V, Mönch PW, Siegmund B, Jarquín-Díaz VH, Fulde M, Forslund-Startceva SK. Mucosal washes are useful for sampling intestinal mucus-associated microbiota despite low biomass. Gut Microbes 2025; 17:2464296. [PMID: 39980334 PMCID: PMC11849919 DOI: 10.1080/19490976.2025.2464296] [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: 12/26/2023] [Revised: 10/24/2024] [Accepted: 02/04/2025] [Indexed: 02/22/2025] Open
Abstract
Understanding the dynamic relationship between mucus-associated microbiota and host health is critical. However, studies predominantly using stool samples may not accurately represent these bacterial communities. Here, we investigated the mucus-associated microbiota in the gastrointestinal tract of mice and the terminal ileum of humans using different sample types: mucosal washes, brushes, scrapings, and intestinal contents in mice and biopsies, brushes and mucosal washes in humans. We used DNA quantification and 16S rRNA amplicon sequencing to evaluate the comparability of the information yielded from the different sample types under a controlled benchmark. In mice, mucosal washes and brushes had comparative bacterial DNA and host DNA contamination than scraping samples. Similarly, in humans, washes outperformed biopsies in bacterial DNA content. Read counts and microbiota alpha diversity remained remarkably similar in mice and between brushes and washes in humans. The composition of the microbiota varied based on the subsegment and sample type in mice and sample type in humans. We conclude that washes and brushes reduce host contamination without inducing substantial compositional bias when sampling mucosal microbiota. Our findings suggest that mucosal washes and brushes are a viable alternative to biopsies in humans and scrapings in mice, thereby improving the transferability of results across hosts. Our study highlights the importance of focusing on mucus-associated microbiota to better capture host-microbiome interactions at their closer interface.
Collapse
Affiliation(s)
- Jennifer N. Martinez-Medina
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
| | - Fereshteh Ghazisaeedi
- Institute of Microbiology and Epizootics, School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
| | - Catharina Kramer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Jörn F Ziegler
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
| | - Victoria McParland
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
| | - Paul W. Mönch
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Britta Siegmund
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Víctor Hugo Jarquín-Díaz
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
| | - Marcus Fulde
- Institute of Microbiology and Epizootics, School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
| | - Sofia K. Forslund-Startceva
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Structural and Computational Biology, European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
8
|
Geng P, Zhao N, Zhou Y, Harris RS, Ge Y. Faecalibacterium prausnitzii regulates carbohydrate metabolic functions of the gut microbiome in C57BL/6 mice. Gut Microbes 2025; 17:2455503. [PMID: 39841201 DOI: 10.1080/19490976.2025.2455503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/08/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
The probiotic impact of microbes on host metabolism and health depends on both host genetics and bacterial genomic variation. Faecalibacterium prausnitzii is the predominant human gut commensal emerging as a next-generation probiotic. Although this bacterium exhibits substantial intraspecies diversity, it is unclear whether genetically distinct F. prausnitzii strains might lead to functional differences in the gut microbiome. Here, we isolated and characterized a novel F. prausnitzii strain (UT1) that belongs to the most prevalent but underappreciated phylogenetic clade in the global human population. Genome analysis showed that this butyrate-producing isolate carries multiple putative mobile genetic elements, a clade-specific defense system, and a range of carbohydrate catabolic enzymes. Multiomic approaches were used to profile the impact of UT1 on the gut microbiome and associated metabolic activity of C57BL/6 mice at homeostasis. Both 16S rRNA and metagenomic sequencing demonstrated that oral administration of UT1 resulted in profound microbial compositional changes including a significant enrichment of Lactobacillus, Bifidobacterium, and Turicibacter. Functional profiling of the fecal metagenomes revealed a markedly higher abundance of carbohydrate-active enzymes (CAZymes) in UT1-gavaged mice. Accordingly, UT1-conditioned microbiota possessed the elevated capability of utilizing starch in vitro and exhibited a lower availability of microbiota-accessible carbohydrates in the gut. Further analysis uncovered a functional network wherein UT1 reduced the abundance of mucin-degrading CAZymes and microbes, which correlated with a concomitant reduction of fecal mucin glycans. Collectively, our results reveal a crucial role of UT1 in facilitating the carbohydrate metabolism of the gut microbiome and expand our understanding of the genetic and phenotypic diversity of F. prausnitzii.
Collapse
Affiliation(s)
- Peiling Geng
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Ni Zhao
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yufan Zhou
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Reuben S Harris
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yong Ge
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
9
|
Cortés-Martín A, Buttimer C, Maier JL, Tobin CA, Draper LA, Ross RP, Kleiner M, Hill C, Shkoporov AN. Adaptations in gut Bacteroidales facilitate stable co-existence with their lytic bacteriophages. Gut Microbes 2025; 17:2507775. [PMID: 40407098 PMCID: PMC12118408 DOI: 10.1080/19490976.2025.2507775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/07/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025] Open
Abstract
Bacteriophages (phages) and bacteria within the gut microbiome persist in long-term stable coexistence. These interactions are driven by eco-evolutionary dynamics, where bacteria employ a variety of mechanisms to evade phage infection, while phages rely on counterstrategies to overcome these defenses. Among the most abundant phages in the gut are the crAss-like phages that infect members of the order Bacteroidales, in particular, genus Bacteroides. In this study, we explored some of the mechanisms enabling the co-existence of four phage-Bacteroidales host pairs in vitro using a multi-omics approach (transcriptomics, proteomics and metabolomics). These included three Bacteroides species paired with three crAss-like phages (Bacteroides intestinalis and фcrAss001, Bacteroides xylanisolvens and фcrAss002, and an acapsular mutant of Bacteroides thetaiotaomicron with DAC15), and Parabacteroides distasonis paired with the siphovirus фPDS1. We show that phase variation of individual capsular polysaccharides (CPSs) is the primary mechanism promoting phage co-existence in Bacteroidales, but this is not the only strategy. Alternative resistance mechanisms, while potentially less efficient than CPS phase variation, can be activated to support bacterial survival by regulating gene expression and resulting in metabolic adaptations, particularly in amino acid degradation pathways. These mechanisms, also likely regulated by phase variation, enable bacterial populations to persist in the presence of phages, and vice versa. An acapsular variant of B. thetaiotaomicron demonstrated broader transcriptomic, proteomic, and metabolomic changes, supporting the involvement of additional resistance mechanisms beyond CPS variation. This study advances our understanding of long-term phage-host interaction, offering insights into the long-term persistence of crAss-like phages and extending these observations to other phages, such as фPDS1. Knowledge of the complexities of phage-bacteria interactions is essential for designing effective phage therapies and improving human health through targeted microbiome interventions.
Collapse
Affiliation(s)
- Adrián Cortés-Martín
- APC Microbiome Ireland & School of Microbiology, University College Cork, Cork, Ireland
| | - Colin Buttimer
- APC Microbiome Ireland & School of Microbiology, University College Cork, Cork, Ireland
| | - Jessie L. Maier
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, North Carolina, USA
| | - Ciara A. Tobin
- APC Microbiome Ireland & School of Microbiology, University College Cork, Cork, Ireland
| | - Lorraine A. Draper
- APC Microbiome Ireland & School of Microbiology, University College Cork, Cork, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland & School of Microbiology, University College Cork, Cork, Ireland
| | - Manuel Kleiner
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, North Carolina, USA
| | - Colin Hill
- APC Microbiome Ireland & School of Microbiology, University College Cork, Cork, Ireland
| | - Andrey N. Shkoporov
- APC Microbiome Ireland & School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
10
|
Steinert RE, Rehman A, Sadabad MS, Milanese A, Wittwer-Schegg J, Burton JP, Spooren A. Microbial micronutrient sharing, gut redox balance and keystone taxa as a basis for a new perspective to solutions targeting health from the gut. Gut Microbes 2025; 17:2477816. [PMID: 40090884 PMCID: PMC11913388 DOI: 10.1080/19490976.2025.2477816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 02/05/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025] Open
Abstract
In health, the gut microbiome functions as a stable ecosystem maintaining overall balance and ensuring its own survival against environmental stressors through complex microbial interaction. This balance and protection from stressors is maintained through interactions both within the bacterial ecosystem as well as with its host. As a consequence, the gut microbiome plays a critical role in various physiological processes including maintaining the structure and function of the gut barrier, educating the gut immune system, and modulating the gut motor, digestive/absorptive, as well as neuroendocrine system all of which are crucial for human health and disease pathogenesis. Pre- and probiotics, widely available and clinically established, offer various health benefits primarily by beneficially modulating the gut microbiome. However, their clinical outcomes can vary significantly due to differences in host physiology, diets, individual microbiome compositions, and other environmental factors. This perspective paper highlights emerging scientific insights into the importance of microbial micronutrient sharing, gut redox balance, keystone species, and the gut barrier in maintaining a diverse and functional microbial ecosystem, and their relevance to human health. We propose a novel approach that targets microbial ecosystems and keystone taxa performance by supplying microbial micronutrients in the form of colon-delivered vitamins, and precision prebiotics [e.g. human milk oligosaccharides (HMOs) or synthetic glycans] as components of precisely tailored ingredient combinations to optimize human health. Such a strategy may effectively support and stabilize microbial ecosystems, providing a more robust and consistent approach across various individuals and environmental conditions, thus, overcoming the limitations of current single biotic solutions.
Collapse
Affiliation(s)
- Robert E. Steinert
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
- Department of Surgery and Transplantation, University Hospital Zurich (USZ) and University of Zurich (UZH), Zürich, Switzerland
| | - Ateequr Rehman
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
| | | | - Alessio Milanese
- Data Science, Science & Research, Dsm-Firmenich, Delft, Netherlands
| | | | - Jeremy P. Burton
- Department of Microbiology and Immunology, The University of Western Ontario, London, Canada
| | - Anneleen Spooren
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
| |
Collapse
|
11
|
Li H, Huang J, Zhao D, Zhu L, Zhang Z, Yi M, Peng W. Longitudinal assessment of peripheral organ metabolism and the gut microbiota in an APP/PS1 transgenic mouse model of Alzheimer's disease. Neural Regen Res 2025; 20:2982-2997. [PMID: 39610107 PMCID: PMC11826447 DOI: 10.4103/nrr.nrr-d-23-01979] [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: 12/03/2023] [Revised: 03/25/2024] [Accepted: 05/06/2024] [Indexed: 11/30/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202510000-00028/figure1/v/2024-11-26T163120Z/r/image-tiff Alzheimer's disease not only affects the brain, but also induces metabolic dysfunction in peripheral organs and alters the gut microbiota. The aim of this study was to investigate systemic changes that occur in Alzheimer's disease, in particular the association between changes in peripheral organ metabolism, changes in gut microbial composition, and Alzheimer's disease development. To do this, we analyzed peripheral organ metabolism and the gut microbiota in amyloid precursor protein-presenilin 1 (APP/PS1) transgenic and control mice at 3, 6, 9, and 12 months of age. Twelve-month-old APP/PS1 mice exhibited cognitive impairment, Alzheimer's disease-related brain changes, distinctive metabolic disturbances in peripheral organs and fecal samples (as detected by untargeted metabolomics sequencing), and substantial changes in gut microbial composition compared with younger APP/PS1 mice. Notably, a strong correlation emerged between the gut microbiota and kidney metabolism in APP/PS1 mice. These findings suggest that alterations in peripheral organ metabolism and the gut microbiota are closely related to Alzheimer's disease development, indicating potential new directions for therapeutic strategies.
Collapse
Affiliation(s)
- Hongli Li
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Metabolic Diseases, Changsha, Hunan Province, China
| | - Jianhua Huang
- Hunan Academy of Chinese Medicine, Changsha, Hunan Province, China
| | - Di Zhao
- Hunan Academy of Chinese Medicine, Changsha, Hunan Province, China
| | - Lemei Zhu
- Academician Workstation, Changsha Medical University, Changsha, Hunan Province, China
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Min Yi
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Metabolic Diseases, Changsha, Hunan Province, China
| |
Collapse
|
12
|
Castagnoli R, Pala F, Subramanian P, Oguz C, Schwarz B, Lim AI, Burns AS, Fontana E, Bosticardo M, Corsino C, Angelova A, Delmonte OM, Kenney H, Riley D, Smith G, Ott de Bruin L, Oikonomou V, Dos Santos Dias L, Fink D, Bohrnsen E, Kimzey CD, Marseglia GL, Alva-Lozada G, Bergerson JR, Brett A, Brigatti KW, Dimitrova D, Dutmer CM, Freeman AF, Ale H, Holland SM, Licciardi F, Pasic S, Poskitt LE, Potts DE, Dasso JF, Sharapova SO, Strauss KA, Ward BR, Yilmaz M, Kuhns DB, Lionakis MS, Daley SR, Kong HH, Segre JA, Villa A, Pittaluga S, Walter JE, Vujkovic-Cvijin I, Belkaid Y, Notarangelo LD. Immunopathological and microbial signatures of inflammatory bowel disease in partial RAG deficiency. J Exp Med 2025; 222:e20241993. [PMID: 40314722 PMCID: PMC12047384 DOI: 10.1084/jem.20241993] [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: 10/26/2024] [Revised: 03/16/2025] [Accepted: 04/10/2025] [Indexed: 05/03/2025] Open
Abstract
Partial RAG deficiency (pRD) can manifest with systemic and tissue-specific immune dysregulation, with inflammatory bowel disease (IBD) in 15% of the patients. We aimed at identifying the immunopathological and microbial signatures associated with IBD in patients with pRD and in a mouse model of pRD (Rag1w/w) with spontaneous development of colitis. pRD patients with IBD and Rag1w/w mice showed a systemic and colonic Th1/Th17 inflammatory signature. Restriction of fecal microbial diversity, abundance of pathogenic bacteria, and depletion of microbial species producing short-chain fatty acid were observed, which were associated with impaired induction of lamina propria peripheral Treg cells in Rag1w/w mice. The use of vedolizumab in Rag1w/w mice and of ustekinumab in a pRD patient were ineffective. Antibiotics ameliorated gut inflammation in Rag1w/w mice, but only bone marrow transplantation (BMT) rescued the immunopathological and microbial signatures. Our findings shed new light in the pathophysiology of gut inflammation in pRD and establish a curative role for BMT to resolve the disease phenotype.
Collapse
Affiliation(s)
- Riccardo Castagnoli
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Francesca Pala
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Poorani Subramanian
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cihan Oguz
- Integrated Data Sciences Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Schwarz
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Ai Ing Lim
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrew S. Burns
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cristina Corsino
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Angelina Angelova
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ottavia M. Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Heather Kenney
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Deanna Riley
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Grace Smith
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lisa Ott de Bruin
- Willem-Alexander Children’s Hospital, Department of Pediatrics, Pediatric Stem Cell Transplantation Program, Leiden University Medical Center, Leiden, Netherlands
| | - Vasileios Oikonomou
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lucas Dos Santos Dias
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Danielle Fink
- Neutrophil Monitoring Lab, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Eric Bohrnsen
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Cole D. Kimzey
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Gian Luigi Marseglia
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Guisela Alva-Lozada
- Allergy and Immunology Division Hospital Nacional Edgardo Rebagliati Martins, Lima, Peru
| | - Jenna R.E. Bergerson
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ana Brett
- Hospital Pediátrico, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
- Clínica Universitária de Pediatria, Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
| | | | - Dimana Dimitrova
- Experimental Transplantation and Immunotherapy Branch, National Cancer Institute of the National Institutes of Health, Bethesda, MD, USA
| | - Cullen M. Dutmer
- Allergy and Immunology, Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hanadys Ale
- Division of Immunology, Allergy and Rheumatology, Joe DiMaggio Children’s Hospital, Memorial Healthcare System, Hollywood, FL, USA
| | - Steven M. Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Francesco Licciardi
- Immuno-reumatologia, Pediatria Specialistica Universitaria, Ospedale Infantile Regina Margherita, Torino, Italy
| | - Srdjan Pasic
- Department of Pediatric Immunology, Mother and Child Health Institute, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | | | - David E. Potts
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Joseph F. Dasso
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Svetlana O. Sharapova
- Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | | | - Brant R. Ward
- Division of Allergy and Immunology, Children’s National Hospital, Washington, DC, USA
| | - Melis Yilmaz
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Douglas B. Kuhns
- Neutrophil Monitoring Lab, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Michail S. Lionakis
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stephen R. Daley
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Heidi H. Kong
- Cutaneous Microbiome and Inflammation Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julia A. Segre
- Microbial Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anna Villa
- San Raffaele-Telethon Institute for Gene Therapy (SR-Tiget), IRCSS San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Stefania Pittaluga
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jolan E. Walter
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Ivan Vujkovic-Cvijin
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Immunology, Institut Pasteur, Paris, France
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
13
|
Gu J, Shen Y, Guo L, Chen Z, Zhou D, Ji G, Gu A. Investigation of the mechanisms of liver injury induced by emamectin benzoate exposure at environmental concentrations in zebrafish: A multi-omics approach to explore the role of the gut-liver axis. JOURNAL OF HAZARDOUS MATERIALS 2025; 491:138008. [PMID: 40132265 DOI: 10.1016/j.jhazmat.2025.138008] [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: 12/01/2024] [Revised: 02/27/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Emamectin benzoate (EMB) is a lipophilic pesticide that enters aquatic systems and adversely affects non-target organisms. This study investigated the long-term effects of EMB on zebrafish, exposing them to concentrations of 0, 0.1, 1, and 10 μg/L from the 4-hour post-fertilization (hpf) embryo stage to the 120-day post-fertilisation (dpf) adult stage. We found that exposure to 1 μg/L EMB induced liver damage, manifested as impaired liver function (elevated aspartate aminotransferase (AST) and alanine aminotransferase (ALT)), histopathological damage (lipid accumulation), as well as inflammatory and oxidative damage, with a dose - dependent effect. Non-targeted metabolomic analysis revealed an increase in lipid molecules in the liver, affecting the pathways related to glycerophospholipid metabolism. In addition, EMB exposure resulted in damage to the intestinal barrier and inflammatory responses in zebrafish. 16S rRNA sequencing demonstrated that EMB exposure resulted in notable alterations in the gut microbiota composition. Notably, the abundance of Plesiomonas and Cetobacterium increased in the EMB exposure group and exhibited a positive correlation with the majority of liver lipid metabolites. In contrast, reductions in Muribaculaceae and Alloprevotella were negatively correlated. The results of this study indicate that long-term exposure to EMB disrupts the gut microbiota, leading to the dysregulation of hepatic phospholipid metabolism. These findings provide new insights into the health risks associated with EMB and highlight its potential threats to higher organisms, including mammals.
Collapse
Affiliation(s)
- Jie Gu
- Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment of the People's Republic of China, Nanjing Institute of Environmental science, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Yuehong Shen
- Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment of the People's Republic of China, Nanjing Institute of Environmental science, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Liguo Guo
- Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment of the People's Republic of China, Nanjing Institute of Environmental science, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Zhicheng Chen
- Department of Toxicology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology,Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, China
| | - Dingyu Zhou
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, China
| | - Guixiang Ji
- Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment of the People's Republic of China, Nanjing Institute of Environmental science, Ministry of Ecology and Environment, Nanjing 210042, China.
| | - Aihua Gu
- Jiangsu Environmental Health Risk Assessment Engineering Research Center, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
14
|
Xue M, Deng Q, Deng L, Xun T, Huang T, Zhao J, Wei S, Zhao C, Chen X, Zhou Y, Liang Y, Yang X. Alterations of gut microbiota for the onset and treatment of psoriasis: A systematic review. Eur J Pharmacol 2025; 998:177521. [PMID: 40107339 DOI: 10.1016/j.ejphar.2025.177521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Psoriasis is a chronic, recurrent and systemic inflammatory skin disease which is mediated by immunoreaction. Its pathogenesis is multifactorial, and the exact driving factor remains unclear. Recent studies showed that gut microbiota, which maintain immune homeostasis of our bodies, is closely related with occurrence, development and prognosis of psoriasis. The intestinal microbial abundance and diversity in patients with psoriasis have changed significantly, including intestinal microbiota disorders and reduced production of short chain fatty acids (SCFAs), abnormalities in Firmicutes/Bacteroidetes (F/B), etc. Besides, the intestinal microbiota of psoriasis patients has also changed after treatment of systemic drugs, biologics and small molecule chemical drugs, suggesting that the intestinal microbiota may be a potential response-to-treatment biomarker for evaluating treatment effectiveness. Oral probiotics and prebiotics administration as well as fecal microbial transplantation were also reported to benefit well in psoriasis patients. Additionally, we also discussed the microbial changes from the skin and other organs, which regulated both the onset and treatment of psoriasis together with gut microbiota. Herein, we reviewed recent studies on the psoriasis-related microbiota in an attempt to confidently identify the "core" microbiota of psoriatic patients, understand how microbiota influence psoriasis through the gut-skin axis, and explore potential therapeutic strategies for psoriasis.
Collapse
Affiliation(s)
- Man Xue
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - QuanWen Deng
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Li Deng
- Department of Dermatology, Cosmetology and Venereology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - TianRong Xun
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - TingTing Huang
- Department of Clinical Laboratory Medicine, Shenzhen Hospital, Southern Medical University, China
| | - JingQian Zhao
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Sui Wei
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - ChenYu Zhao
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Xi Chen
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - YiWen Zhou
- Department of Clinical Laboratory Medicine, Shenzhen Hospital, Southern Medical University, China
| | - YanHua Liang
- Department of Dermatology, Cosmetology and Venereology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - XiXiao Yang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China.
| |
Collapse
|
15
|
Zhao Q, Liu Y, Li M, Zhao L, Wang T, Xiao Y, Wei S, Wu K, Yang J, Wang Y, Zhao Y, Du F, Chen Y, Deng S, Shen J, Xiao Z, Li Z, Wang S, Wu X. Hawthorn pectin mitigates high-fat diet induced hyperlipidemia in mice through promoting Dubosiella newyorkensis. Carbohydr Polym 2025; 359:123570. [PMID: 40306777 DOI: 10.1016/j.carbpol.2025.123570] [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: 07/02/2024] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 05/02/2025]
Abstract
Hawthorn fruit is rich in pectins; however, the gut microbiota-modulating effects of hawthorn pectin remain unclear. In this study, enzyme-extracted hawthorn pectin (Mw: 126.24 kDa; HG/RG-I ratio: 55.64 %/37.93 %; branching degree: 2.44) was fractionated into two distinct components: D1 (spherical, 345.76 kDa, HG/RG-I 57.48 %/37.25 %, branching degree 2.16) and D2 (flexible, 17.20 kDa, HG/RG-I 78.24 %/21.76 %, branching degree 1.33). Hawthorn pectin directly stimulated Dubosiella newyorkensis (D. newyorkensis) and Bifidobacterium bifidum (B. bifidum) growth in vitro. In hyperlipidemic mice, it alleviated dyslipidemia and enriched the abundance of Lactobacillus and Dubosiella. Parallel experiments confirmed that D. newyorkensis supplementation similarly alleviated dyslipidemia and increased Lactobacillus abundance, which was mediated by bacterial acetate production. Notably, only D1 emulated the pectin's prebiotic activity, promoting D. newyorkensis and B. bifidum growth. The high-RG-I D1, with elevated branching and larger Mw, was identified as the key driver of hawthorn pectin's prebiotic effect. These findings highlight that hawthorn pectin, particularly D1, specifically targets probiotic D. newyorkensis to synergize with Lactobacillus against hyperlipidemia.
Collapse
Affiliation(s)
- Qianyun Zhao
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Department of Pharmacy, Maternal and Child Health Care And Family Planning Service Center, 620000 Meishan, Sichuan, China
| | - Yubin Liu
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Mingxing Li
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; South Sichuan Institute of Translation Medicine, 646000 Luzhou, Sichuan, China
| | - Long Zhao
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Tiangang Wang
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yaqin Xiao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, 999078 Macao, China
| | - Shulin Wei
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Ke Wu
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Jiayue Yang
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yaqiu Wang
- Aulin College, Northeast Forestry University, 150040 Ha'erbin, China
| | - Yueshui Zhao
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; South Sichuan Institute of Translation Medicine, 646000 Luzhou, Sichuan, China
| | - Fukuan Du
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; South Sichuan Institute of Translation Medicine, 646000 Luzhou, Sichuan, China
| | - Yu Chen
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; South Sichuan Institute of Translation Medicine, 646000 Luzhou, Sichuan, China
| | - Shuai Deng
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; South Sichuan Institute of Translation Medicine, 646000 Luzhou, Sichuan, China
| | - Jing Shen
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; South Sichuan Institute of Translation Medicine, 646000 Luzhou, Sichuan, China
| | - Zhangang Xiao
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; South Sichuan Institute of Translation Medicine, 646000 Luzhou, Sichuan, China; School of Pharmacy, Sichuan College of Traditional Chinese Medicine, 621000 Mianyang, Sichuan, China
| | - Zhi Li
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, 646000 Luzhou, Sichuan, China; The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou city, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, 646000 Luzhou, Sichuan, China.
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, 999078 Macao, China.
| | - Xu Wu
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Department of Paediatrics, Luzhou People's Hospital, 646000 Luzhou, Sichuan, China; Department of Paediatric Care, Luzhou People's Hospital, 646000 Luzhou, Sichuan, China.
| |
Collapse
|
16
|
Duan M, Che L, Wu X, Quek SY, Zhang B, Lin H, He N. Incorporation of probiotics with pressure-sensitive pectin-fructooligosaccharide hydrogel for potential intestinal delivery. Carbohydr Polym 2025; 359:123566. [PMID: 40306774 DOI: 10.1016/j.carbpol.2025.123566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/09/2025] [Accepted: 03/29/2025] [Indexed: 05/02/2025]
Abstract
Probiotics and prebiotics serve as vital tools in managing gut microecology and enhancing immune responses. However, the effectiveness of non-encapsulated probiotics often diminishes during processing, storage, and transport to the gastrointestinal tract, especially at elevated temperatures. To address this challenge, a novel loading strategy for Lactobacillus reuteri DPC16 (L. reuteri) is proposed in this work, using pressure-sensitive high-methoxy pectin (HMP)/fructooligosaccharides (FOS) hydrogel. The HMP/FOS hydrogel melted at 600 MPa to form a sol. The resulting sol was mixed with L. reuteri immediately at ambient conditions, which underwent a sol-to-gel transition subsequently to form a composite hydrogel with a continuous porous structure. The resulting HMP/FOS@L. reuteri hydrogel achieved a loading concentration of viable bacteria at 109 CFU/mL. In vitro assessments reveal that the hydrogel demonstrates good biocompatibility and targeted release of probiotics within the intestine. Furthermore, the hydrogel substantially boosted the short-chain fatty acids levels and increased the amounts of acetic and isovaleric acids, respectively. This work underscores the unique advantages of employing a pressure-sensitive HMP/FOS hydrogel for loading and targeted delivery of probiotics and prebiotics to improve intestinal health.
Collapse
Affiliation(s)
- Mengwen Duan
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China
| | - Liming Che
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China.
| | - Xuee Wu
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China
| | - Siew Young Quek
- Food Science, School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Bangzhou Zhang
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350000, PR China
| | - Hao Lin
- Xiamen Treatgut Biotechnology Co. Ltd., Xiamen 361005, PR China
| | - Ning He
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China.
| |
Collapse
|
17
|
Mahmoud AA, Wang X, Liao X, Zhang S, Ding T, Ahn J. Impact of prophages on gut microbiota and disease associations. Microb Pathog 2025; 204:107642. [PMID: 40300731 DOI: 10.1016/j.micpath.2025.107642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 04/01/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
The gut microbiota plays an important role in maintaining host health by affecting various physiological functions. Among the diverse microbial communities in the gut, prophages are integral components of bacterial genomes, contributing significantly to bacterial evolution, ecology and pathogenicity. Prophages are capable of switching to lytic cycles in response to various internal and external factors. Factors that induce prophage induction include DNA damage, oxidative stress, nutrient availability, host immune response, quorum sensing, diet, secondary metabolites, antibiotics, and lifestyle changes. Prophage induction could contribute to both gut homeostasis and dysbiosis. Importantly, the connections between prophage induction and disorders such as inflammatory bowel disease, ulcerative colitis, and bacterial vaginosis highlight the dual roles of prophages in both health and disease. Although therapeutic approaches such as phage therapy (PT), fecal microbiota transplants (FMT), and fecal virome transplants (FVT) have gained attention, the concept of dietary prophage induction therapy offers a novel, targeted method to modulate gut microbiota. In spite of recent advances in understanding the role of prophages in gut health, the exact mechanisms by which they influence gut health remain only partially understood. Therefore, further research is needed to elucidate additional molecular mechanisms of prophage induction pathways and to explore their implications for gut microbiota dynamics and disease associations. This review discusses the molecular mechanisms and key factors that trigger prophage induction in the gut. Insights into these processes could lead to innovative therapeutic strategies that utilize prophages to support gut health.
Collapse
Affiliation(s)
- Aminu Abdullahi Mahmoud
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China; Department of Food Science and Nutrition, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiaoyu Wang
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China; Department of Food Science and Nutrition, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xinyu Liao
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China
| | - Song Zhang
- Department of Biomedical Science, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Tian Ding
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China; Department of Food Science and Nutrition, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Juhee Ahn
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China; Department of Food Science and Nutrition, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Biomedical Science, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea.
| |
Collapse
|
18
|
Lotankar M, Houttu N, Mokkala K, Laitinen K. Diet-Gut Microbiota Relations: Critical Appraisal of Evidence From Studies Using Metagenomics. Nutr Rev 2025; 83:e1917-e1938. [PMID: 39718602 PMCID: PMC12166168 DOI: 10.1093/nutrit/nuae192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
Diet may influence the gut microbiota and subsequently affect the host's health. Recent developments in methods analyzing the composition and function of the gut microbiota allow a deeper understanding of diet-gut microbiota relationships. A state-of-the-art methodology, shotgun metagenomics sequencing, offers a higher taxonomic resolution of the gut microbiota at the bacterial species and strain levels, and more accurate information regarding the functional potential of gut microbiota. Here, the available evidence on the relationship between diet and gut microbiota was critically reviewed, focusing on results emerging from recent metagenomics sequencing studies applied in randomized controlled trials and observational studies. The PubMed and Embase databases were used to search publications between January 2011 and September 2023. Thus far, the number of studies is limited, and the study designs and methods utilized have been variable. Nevertheless, the cumulative evidence from interventions relates to dietary fiber as a modifier of bacterial species, such as Anaerostipes hadrus and Faecalibacterium prausnitzii. Furthermore, observational studies have detected associations between different dietary patterns and food groups with certain microbial species. Utilization of metagenomics sequencing is becoming more common and will undoubtedly provide further insights into diet-gut microbiota relationships at the species level as well as their functional pathways in the near future. For reproducible results and to draw reliable conclusions across various studies on diet-gut microbiota relationships, there is a need for harmonization of the study designs and standardized ways of reporting.
Collapse
Affiliation(s)
- Mrunalini Lotankar
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, Faculty of Medicine, University of Turku, 20520 Turku, Finland
| | - Noora Houttu
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, Faculty of Medicine, University of Turku, 20520 Turku, Finland
| | - Kati Mokkala
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, Faculty of Medicine, University of Turku, 20520 Turku, Finland
- Nutrition and Food Research Center, Faculty of Medicine, University of Turku, 20520 Turku, Finland
| | - Kirsi Laitinen
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, Faculty of Medicine, University of Turku, 20520 Turku, Finland
- Nutrition and Food Research Center, Faculty of Medicine, University of Turku, 20520 Turku, Finland
- Department of Obstetrics and Gynecology, Turku University Hospital, Wellbeing Services County of Southwest Finland, 20520 Turku, Finland
| |
Collapse
|
19
|
Yin X, Shi Y, Sheng T, Ji C. Early-Life Gut Microbiota: A Possible Link Between Maternal Exposure to Non-Nutritive Sweeteners and Metabolic Syndrome in Offspring. Nutr Rev 2025; 83:e1954-e1969. [PMID: 39348276 DOI: 10.1093/nutrit/nuae140] [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] [Indexed: 10/02/2024] Open
Abstract
Metabolic syndrome (MetS) is recognized as a group of metabolic abnormalities, characterized by clustered interconnected traits that elevate the risks of obesity, cardiovascular and atherosclerotic diseases, hyperlipidemia, and type 2 diabetes mellitus. Non-nutritive sweeteners (NNS) are commonly consumed by those with imbalanced calorie intake, especially in the perinatal period. In the past, accumulating evidence showed the transgenerational and mediated roles of human microbiota in the development of early-life MetS. Maternal exposure to NNS has been recognized as a risk factor for filial metabolic disturbance through various mechanisms, among which gut microbiota and derived metabolites function as nodes linking NNS and MetS in early life. Despite the widespread consumption of NNS, there remain growing concerns about their transgenerational impact on metabolic health. There is growing evidence of NNS being implicated in the development of metabolic abnormalities. Intricate complexities exist and a comprehensive understanding of how the gut microbiota interacts with mechanisms related to maternal NNS intake and disrupts metabolic homeostasis of offspring is critical to realize its full potential in preventing early-life MetS. This review aims to elucidate the effects of early-life gut microbiota and links to maternal NNS exposure and imbalanced offspring metabolic homeostasis and discusses potential perspectives and challenges, which may provide enlightenment and understanding into optimal perinatal nutritional management.
Collapse
Affiliation(s)
- Xiaoxiao Yin
- Nanjing Medical Research Center for Women and Children, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu 210004, China
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yujie Shi
- Nanjing Medical Research Center for Women and Children, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu 210004, China
| | - Tongtong Sheng
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Chenbo Ji
- Nanjing Medical Research Center for Women and Children, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu 210004, China
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
20
|
Kumar R, Sykes DJ, Band VI, Schaller ML, Patel R, Vitvitsky V, Sajjakulnukit P, Singhal R, Wong HKA, Hourigan SK, Ichinose F, Lyssiotis CA, Shah YM, Banerjee R. Gut sulfide metabolism modulates behavior and brain bioenergetics. Proc Natl Acad Sci U S A 2025; 122:e2503677122. [PMID: 40526718 DOI: 10.1073/pnas.2503677122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 05/16/2025] [Indexed: 06/19/2025] Open
Abstract
The host-microbiome interface is rich in metabolite exchanges and exquisitely sensitive to diet. Hydrogen sulfide (H2S) is present at high concentrations at this interface and is a product of both microbial and host metabolism. The mitochondrial enzyme, sulfide quinone oxidoreductase (SQOR), couples H2S detoxification to oxidative phosphorylation; its inherited deficiency presents as Leigh disease. Since an estimated two-thirds of systemic H2S metabolism originates in the gut, it raises questions as to whether impaired sulfide clearance in this compartment contributes to disease and whether it can be modulated by dietary sulfur content. In this study, we report that SQOR deficiency confined to murine intestinal epithelial cells perturbs colon bioenergetics that is reversed by antibiotics, revealing a significant local contribution of microbial H2S to host physiology. We also find that a 2.5-fold higher methionine intake, mimicking the difference between animal and plant proteins, synergizes with intestinal SQOR deficiency to adversely impact colon architecture and alter microbiome composition. In serum, increased thiosulfate, a biomarker of H2S oxidation, reveals that intestinal SQOR deficiency combined with higher dietary methionine affects sulfide metabolism globally and perturbs energy metabolism as indicated by higher ketone bodies. The mice exhibit lower exploratory locomotor activity while brain MRI reveals an atypical reduction in ventricular volume, which is associated with lower aquaporin 1 that is important for cerebrospinal fluid secretion. Our study reveals the dynamic interaction between dietary sulfur intake and sulfide metabolism at the host-microbe interface, impacting gut health, and the potential for lower dietary methionine intake to modulate pathology.
Collapse
Affiliation(s)
- Roshan Kumar
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109
| | | | - Victor I Band
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Megan L Schaller
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109
| | - Romel Patel
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109
| | - Victor Vitvitsky
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109
| | - Peter Sajjakulnukit
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109
| | - Rashi Singhal
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109
| | - Harrison K A Wong
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109
| | - Suchitra K Hourigan
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Fumito Ichinose
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Ruma Banerjee
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
21
|
Chen HY, Wu PS, Li ZY, Liu YC, Yeh SR, Duan BC, Cheng KW, Hsu CC, Chiu YL, Lee WT, Fan SZ, Wang PY. Gut microbiome and host TOR pathway interact to regulate predator-induced aversive memory in Drosophila melanogaster. Proc Natl Acad Sci U S A 2025; 122:e2422928122. [PMID: 40540603 DOI: 10.1073/pnas.2422928122] [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: 11/07/2024] [Accepted: 05/28/2025] [Indexed: 06/22/2025] Open
Abstract
The gut microbiome has emerged as a key factor influencing a wide range of host physiological processes and behaviors, though the mechanisms behind these effects remain only partially understood. In this study, we explored the role of the gut microbiome in memory regulation using a parasitoid wasp-induced oviposition depression paradigm in Drosophila melanogaster. Our findings show that flies with depleted gut microbiota, either through axenic culture or antibiotic treatment, exhibited significant memory impairments. However, reintroducing the commensal bacterium Lactobacillus plantarum alone was sufficient to restore memory, while coinoculation with Acetobacter pomorum further enhanced memory performance. Hemolymph metabolomic analyses revealed reduced amino acid levels in antibiotic-treated flies, which were linked to impaired Drosophila target of rapamycin (dTOR) signaling. Additionally, genetic manipulation of dTOR or dietary supplementation with branched-chain amino acids either mimicked or rescued the memory deficits caused by antibiotic treatments. These results suggest that the gut microbiome is essential for regulating memory function by maintaining amino acid homeostasis and proper dTOR signaling, with profound implications for advancing knowledge of cognitive regulation.
Collapse
Affiliation(s)
- Hao-Yu Chen
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Po-Sheng Wu
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Zong-You Li
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - You-Cheng Liu
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Sheng-Rong Yeh
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department of Anesthesiology, National Taiwan University Hospital, National Taiwan University, Taipei 100, Taiwan
- Department of Anesthesiology, En Chu Kong Hospital, Taipei 237, Taiwan
| | - Bi-Chun Duan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department of Pediatrics, Lotung Poh-Ai Hospital, Yilan 265, Taiwan
| | - Kai-Wen Cheng
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
- Leeuwenhoek Laboratories Co. Ltd, Taipei 106, Taiwan
| | - Yen-Ling Chiu
- Division of Nephrology, Department of Medicine, Far Eastern Memorial Hospital, Taipei 220, Taiwan
| | - Wang-Tso Lee
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department of Pediatrics, National Taiwan University Children's Hospital, Taipei 100, Taiwan
| | - Shou-Zen Fan
- Department of Anesthesiology, National Taiwan University Hospital, National Taiwan University, Taipei 100, Taiwan
- Department of Anesthesiology, En Chu Kong Hospital, Taipei 237, Taiwan
| | - Pei-Yu Wang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei 106, Taiwan
- Ph.D. Program in Translational Medicine, National Taiwan University and Academia Sinica, Taipei 100, Taiwan
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Taiwan University and Academia Sinica, Taipei 115, Taiwan
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 235, Taiwan
| |
Collapse
|
22
|
Kadharusman MM, Syahputra RA, Kurniawan R, Hadinata E, Tjandrawinata RR, Taslim NA, Romano R, Santini A, Nurkolis F. Seagrass Enhalus acoroides extract mitigates obesity and diabetes via GLP-1, PPARγ, SREBP-1c modulation and gut microbiome restoration in diabetic zebrafish. Diabetol Metab Syndr 2025; 17:235. [PMID: 40542443 DOI: 10.1186/s13098-025-01823-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2025] [Accepted: 06/14/2025] [Indexed: 06/22/2025] Open
Abstract
BACKGROUND The global rise in obesity and type 2 diabetes highlights the need for safe and effective therapeutic interventions. Enhalus acoroides is a tropical seagrass rich in carotenoids and other bioactives. Its potential for metabolic regulation has been suggested in vitro, but in vivo efficacy and molecular mechanisms remain unexplored. This study aimed to evaluate the anti-obesity and anti-diabetic effects of Enhalus acoroides extract (SEAE) in a zebrafish model of diet- and glucose-induced metabolic dysfunction. METHODS Adult zebrafish were subjected to overfeeding and glucose immersion, after overfeeding and 14 days of glucose immersion to induce diabetes, adult zebrafish were randomized into three groups: untreated diabetic, SEAE-treated (5 mg/L), and metformin-treated (3.3 mg/L) for 20 days. Body weight, fasting blood glucose, lipid profile, gene expression (GLP-1, PPARγ, SREBP-1c), and gut microbiota profiles via 16 S rRNA sequencing were assessed. RESULTS SEAE significantly reduced body weight and blood glucose in diabetic zebrafish (p < 0.05), with efficacy comparable to or exceeding Metformin. It upregulated GLP-1 and downregulated PPARγ and SREBP-1c. SEAE also reduced total cholesterol, triglycerides, and LDL levels, while increasing HDL levels. Moreover, SEAE restored the Firmicutes/Bacteroidetes ratio, increased alpha diversity, and shifted beta diversity toward healthy controls. SEAE-treated fish showed microbial profiles closer to normal than those treated with Metformin. CONCLUSIONS SEAE exhibits strong anti-obesity and anti-hyperglycemic effects by modulating key metabolic pathways and restoring gut microbial homeostasis. These findings highlight SEAE as a promising marine-derived therapeutic candidate for metabolic syndrome and warrant further investigation as a functional food or nutraceutical. CLINICAL TRIAL Not applicable.
Collapse
Affiliation(s)
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, 20155, Indonesia
| | - Rudy Kurniawan
- Graduate School of Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Edwin Hadinata
- Faculty of Medicine, Ciputra University of Surabaya, Surabaya, 60219, Indonesia
| | - Raymond Rubianto Tjandrawinata
- Center for Pharmaceutical and Nutraceutical Research and Policy, Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, Jakarta, 12930, Indonesia
| | - Nurpudji Astuti Taslim
- Division of Clinical Nutrition, Department of Nutrition, Faculty of Medicine, Hasanuddin University, Makassar, 90245, Indonesia
| | - Raffaele Romano
- Department of Agricultural Sciences, University of Naples Federico II, Piazza Carlo di Borbone I, Portici, NA, Italy.
| | - Antonello Santini
- Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano, 49, 80131, Napoli, Italy.
| | - Fahrul Nurkolis
- State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta, 55281, Indonesia.
- Master of Basic Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
- Medical Research Center of Indonesia, Surabaya, East Java, Indonesia.
| |
Collapse
|
23
|
Chen K, Wang H, Yang Y, Tang C, Sun X, Zhou J, Liu S, Li Q, Zhao L, Gao Z. Common mechanisms of Gut microbe-based strategies for the treatment of intestine-related diseases: based on multi-target interactions with the intestinal barrier. Cell Commun Signal 2025; 23:288. [PMID: 40528179 DOI: 10.1186/s12964-025-02299-5] [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: 01/06/2025] [Accepted: 06/07/2025] [Indexed: 06/20/2025] Open
Abstract
The concurrent occurrence and exacerbation of multiple diseases, including geriatric diseases and chronic diseases, impose a heavy burden on human health and medical expenses. Clarifying the common mechanisms of related multifarious diseases and developing preventive and therapeutic strategies with synergistic effects for multiple diseases are of great significance in alleviating the burden on the medical system and reducing patients' burden of drug metabolism. Recent studies have revealed that gut microbiota disorders and intestinal barrier damage, which consequently cause metabolic and immunological disorders, may be a common pathological basis underlying various intestinal-related diseases. In this review, we focus on the intestinal barrier function, summarizing the multi-target interactions and common mechanisms involved in diseases related to the gut such as ulcerative colitis, colorectal cancer, and type 2 diabetes. We identified gut microbe-based strategies, including probiotics, prebiotics, synbiotics, postbiotics, as well as potential targets in faecal microbiota transplant and berberine. The common mechanisms and key targets in the treatment of these diseases mainly include increasing the abundance of beneficial genera Bifidobacterium and Lactobacillus, increasing the levels of Short Chain Fatty Acids, restoring the intestinal mechanical barrier, and suppressing gut inflammation infiltration. We aim to provide a crucial basis and direction for the development of novel drugs with therapeutic effects for multiple diseases, thereby alleviating the patients' burden of medication and enhancing the efficacy of treatment.
Collapse
Affiliation(s)
- Keyu Chen
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
- Department of Endocrinology, Academy of Chinese Medical Sciences, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Han Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yingying Yang
- China-Japan Friendship Hospital, Beijing, 100029, China
| | - Cheng Tang
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaowei Sun
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Jie Zhou
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Shixu Liu
- Department of Traditional Chinese Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266003, China
| | - Qingwei Li
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Zezheng Gao
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
24
|
You X, Yang W, Li X, Li X, Huang Y, Huang C. Dietary modulation of pubertal timing: gut microbiota-derived SCFAs and neurotransmitters orchestrate hypothalamic maturation via the gut-brain axis. J Endocrinol Invest 2025:10.1007/s40618-025-02615-3. [PMID: 40526265 DOI: 10.1007/s40618-025-02615-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Accepted: 05/19/2025] [Indexed: 06/19/2025]
Abstract
BACKGROUND The global rise in early pubertal activation is closely linked to dietary patterns and gut microbiota (GM) dysbiosis. This review synthesizes evidence on how GM-derived metabolites modulate hypothalamic maturation and pubertal timing through the gut-brain axis. METHODS Following PRISMA guidelines, we conducted a systematic review of human and animal studies (PubMed, Medline, CNKI, Wanfang) up to October 2024, focusing on dietary impacts (high-fat/high-sugar) on GM composition and puberty onset. Inclusion criteria prioritized studies linking GM metabolites to HPGA activation. RESULTS High-fat/high-sugar diets reduce GM diversity and short-chain fatty acid (SCFA) production (e.g., butyrate, acetate), impair gut barrier integrity, and promote systemic inflammation. Dysbiosis in SCFA-producing taxa (Roseburia, Faecalibacterium) and neurotransmitter-modulating genera (Bifidobacterium, Lactobacillus) disrupts leptin/insulin signaling and kisspeptin-GnRH interactions, accelerating HPGA activation. Animal studies demonstrate SCFA supplementation delays puberty by reducing hypothalamic inflammation, while human data reveal ethnic and dietary variability in GM profiles. Western diets heighten altered pubertal timing risk via GM-mediated HPGA dysregulation, whereas fiber-rich Mediterranean diets exhibit protective effects. CONCLUSION GM dysbiosis and SCFA depletion are pivotal in diet-driven alterations of pubertal timing. Culturally adapted interventions targeting microbiota-metabolite interactions may mitigate risks of early puberty onset.
Collapse
Affiliation(s)
- Xiaoqing You
- Department of Pediatrics, Fuzhou First General Hospital Affiliated With Fujian Medical University, Fuzhou, Fujian, China
| | - Wei Yang
- Department of Pediatrics, Shenzhen Baoan Clinical Medical College of Guangdong Medical University (The People's Hospital of Baoan Shenzhen), Shenzhen, China
| | - Xiuyun Li
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Longgang Maternity and Child Institute of Shantou University Medical College, Shenzhen, China
| | - Xiaoli Li
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Longgang Maternity and Child Institute of Shantou University Medical College, Shenzhen, China
| | - Ying Huang
- The Second Clinical College of Southern Medical University, Guangzhou, China
| | - Congfu Huang
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Longgang Maternity and Child Institute of Shantou University Medical College, Shenzhen, China.
| |
Collapse
|
25
|
Zheng L, Liu L, An S, Chen X, Hua H, Zhao J. Integrated analysis of metabolomics, network pharmacology, and intestinal microbiota reveals Tibetan herb E'se ameliorate disorders of glycolipid metabolism in db/db mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 145:156979. [PMID: 40541118 DOI: 10.1016/j.phymed.2025.156979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/29/2025] [Accepted: 06/11/2025] [Indexed: 06/22/2025]
Abstract
BACKGROUND E'se (Malus toringoides (Rehd.) Hughes or Malus transitoria (Batal.) Schneid) is widely used as a drug for the treatment of diabetes mellitus in China, but the mechanism by which E 'se regulates disorders of glucose-lipid metabolism has lacked in-depth study. The intestinal microbiota also plays a crucial role in lipid metabolism, but whether E 'se regulates this process by modulating the intestinal microbiota needs to be further investigated. OBJECTIVE Effects of aqueous extract of E'se decoction lyophilized powder (EMT) on metabolic disorders of glucose and lipid, and its mechanism of glucose regulation mediated through intestinal microbiota in db/db mice. METHODS UPLC-Q-TOF-MS was used to analyze the chemical composition of EMT and predict potential therapeutic targets in combination with network pharmacology and molecular docking. The pharmacological effects of EMT were evaluated in six groups of db/db mice (db/db group, rosiglitazone group, low, medium, and high dose EMT of 0.75 g, 1.5 g, 3.0 g/kg/d) and db/m (WT) for 4 weeks' treatment. ELISA was performed to determine serum concentrations of glycated hemoglobin (HbA1C), glycated serum protein (GSP), free fatty acids (FFA), fasting insulin (FINS), lipopolysaccharide (LPS), and glucagon-like peptide-1 (GLP-1), feces was used for microbial 16S rRNA sequencing and short-chain fatty acid (SCFA) quantification, and organs were used for pathologic assessment and subsequent mechanistic studies. RESULTS Based on network pharmacology and molecular docking predictions, the role of EMT in regulating glycolipid metabolism mainly involves pathways such as G protein-coupled receptor(GPR) activity and GLP-1 secretion. Subsequently, it was demonstrated in animal experiments that EMT significantly ameliorated the abnormalities of glycolipid metabolism in db/db mice. Further microbial 16S r RNA sequencing analysis revealed significant changes in the composition of the intestinal microbiota, with increased abundance of Muribaculacea, Alloprevotella, Rikenella, and Parabacteroides, associated with enhanced SCFA secretion. Increased SCFA activated hepatic GPR, promoted GLP-1 secretion, modulated secretion of inflammatory factors and oxidative factors in the intestine, and down-regulated the NF-κB pathway in db/db mice. CONCLUSION Studies have demonstrated that E'se can effectively alleviate abnormalities of glucose-lipid metabolism and intestinal barrier inflammation, making it a novel drug with great therapeutic potential.
Collapse
Affiliation(s)
- Luyao Zheng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Sichuan Institute for Translational Chinese Medicine, Key Lab.: Biological Evaluation of TCM Quality of the State Administration of Traditional Chinese Medicine Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Chengdu 610041, PR China
| | - Li Liu
- Sichuan Institute for Translational Chinese Medicine, Key Lab.: Biological Evaluation of TCM Quality of the State Administration of Traditional Chinese Medicine Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Chengdu 610041, PR China
| | - Shangxiao An
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Sichuan Institute for Translational Chinese Medicine, Key Lab.: Biological Evaluation of TCM Quality of the State Administration of Traditional Chinese Medicine Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Chengdu 610041, PR China
| | - Xue Chen
- Sichuan Institute for Translational Chinese Medicine, Key Lab.: Biological Evaluation of TCM Quality of the State Administration of Traditional Chinese Medicine Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Chengdu 610041, PR China
| | - Hua Hua
- Sichuan Institute for Translational Chinese Medicine, Key Lab.: Biological Evaluation of TCM Quality of the State Administration of Traditional Chinese Medicine Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Chengdu 610041, PR China.
| | - Junning Zhao
- Sichuan Institute for Translational Chinese Medicine, Key Lab.: Biological Evaluation of TCM Quality of the State Administration of Traditional Chinese Medicine Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Chengdu 610041, PR China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, PR China.
| |
Collapse
|
26
|
Zheng XQ, Guan ZY, Zhang YD, Song CL. Different role of the gut microbiota in postmenopausal and senile osteoporosis. Bone Res 2025; 13:63. [PMID: 40514392 PMCID: PMC12166039 DOI: 10.1038/s41413-025-00432-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/13/2025] [Accepted: 03/17/2025] [Indexed: 06/16/2025] Open
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Zhi-Yuan Guan
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Yun-Di Zhang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
27
|
Yao D, Ma C, Ke C, Wang D, Xu K, Liu Y, Qu L. Integrating transcriptomics, metabolomics, and microbiomics to explore the mechanism of action of bran-fried Atractylodes lancea rhizome polysaccharide in ameliorating the enhanced pharmacological effects of dextran sodium sulfate-induced colitis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119805. [PMID: 40324703 DOI: 10.1016/j.jep.2025.119805] [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: 01/03/2025] [Revised: 04/01/2025] [Accepted: 04/12/2025] [Indexed: 05/07/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Atractylodes lancea (Thunb.) DC. is used in China as a folk medicine for gastrointestinal disorder treatment, and its effect on treating gastrointestinal disorders is enhanced when it is fried in bran. Atractylodes lancea rhizome polysaccharide (ALP) are a group of active substances in Atractylodes lancea rhizome,ALP has good anti-inflammatory, oxidative, immunological, and intestinal flora-regulating activities, suggesting that it is a potential drug option for treating ulcerative colitis (UC). However, the effects and mechanisms of raw Atractylodes lancea rhizome polysaccharide (SALP) as well as bran-fried Atractylodes lancea rhizome polysaccharide (FALP) on dextran sodium sulfate (DSS)-induced UC in mice are unclear. AIM OF THE STUDY This study aimed to investigate the comparative therapeutic effects of SALP and FALP in mice with UC and assess their potential mechanisms of action. MATERIALS AND METHODS BALB/c mice were regularly administered 3.5 % DSS to develop and establish an acute UC model, following which SALP-L, SALP-H, FALP-L, FALP-H and sulfasalazine (SASP) were administered for 10 days continuously. The body weight, disease activity index (DAI), organ index, colon length, histopathological damage, proinflammatory cytokine expression level, tight junction protein expression, transcriptome, metabolomics, and 16S rDNA of the mouse model were examined to compare the efficacy and mechanisms of action of SALP and FALP in UC treatment. RESULTS Both SALP and FALP significantly alleviated clinical signs (increased body weight, decreased DAI scores, reduced colonic pathological damage, and others), improved intestinal barrier (promoted Occludin and ZO-1 expression), and reduced intestinal inflammation (inhibited IL-1β and TNF-α [proinflammatory cytokines] expression) in DSS-induced acute UC mice. Metabolomics revealed that both SALP and FALP reversed arachidonic acid, lactic acid, ethanolamine, 9,12-octadecadienoic acid, phosphoric acid, and 1-monopalmitin levels in colonic tissues. In addition, they attenuated intestinal flora disorders in DSS-treated mice by increasing the relative abundance of the beneficial bacteria Lachnospiraceae_NK4A136_group and Alistipes while decreasing that of the harmful bacteria Alloprevotella and Prevotellaceae_UCG_001. Of note, the improvement effect of FALP was better than that of SALP in these results. CONCLUSIONS Both SALP and FALP reduced colitis symptoms by repairing the intestinal barrier, modulating intestinal flora, and improving the metabolism of compounds in colonic tissues. Of note, The therapeutic effects of FALP were all stronger than those of SALP.
Collapse
Affiliation(s)
- Ding Yao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Chaoyang Ma
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Chang Ke
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Dongpeng Wang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Kang Xu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Center for Hubei TCM Processing Technology Engineering, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China.
| | - Yanju Liu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Center for Hubei TCM Processing Technology Engineering, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China.
| | - Linghang Qu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Center for Hubei TCM Processing Technology Engineering, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China.
| |
Collapse
|
28
|
Ren T, Yin Y, Zhou X, Chen L, Shen Y, Feng H, Zhou C, Xu Y, Ling Y. Dahuang Huanglian Decoction alleviates dysbiosis by inhibiting GBP5/NLRP3 signaling pathway-mediated pyroptosis of colonic epithelial cells. JOURNAL OF ETHNOPHARMACOLOGY 2025; 351:120143. [PMID: 40513920 DOI: 10.1016/j.jep.2025.120143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 06/09/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Currently, the prevalence of dysbiosis is increasing, but its treatment options are limited. Dahuang Huanglian Xiexin Decoction (XXD) is a traditional herbal prescription recorded in the Treatise on Typhoid Fever, with a longstanding application in the treatment of digestive system diseases. It consists mainly of three classical chinese medicinal herbs: Dahuang (Rheum palmatum L.), Huangqin (Scutellaria baicalensis Georgi), and Huanglian (Coptis chinensis Franch.). Previous studies demonstrated the efficacy of XXD in treating dysbiosis. However, the exact underlying mechanism requires further investigation. AIM OF THE STUDY The effects of XXD were evaluated in this study to determine its impact on dysbiosis and to reveal the potential mechanisms underlying its alleviation using proteomics and transcriptomics. MATERIALS AND METHODS The components of XXD were identified through UPLC-Q-TOF-MS. Dysbiosis mice were established by mixing antibiotic solutions, and XXD was employed as the therapeutic agent in the intervention. Body weight changes, diarrhea rates, and histopathology were evaluated to determine the therapeutic effects of XXD. Proteomics and transcriptomics were subsequently employed to further elucidate the mechanisms underlying the therapeutic effects of XXD on dysbiosis. Meanwhile, TEM was used to observe tight junctions and pyroptosis in the mouse colon. Furthermore, IF, western blotting, RT-qPCR, and ELISA were employed to investigate the mechanism of XXD. RESULTS This study indicates that XXD promoted the recovery from dysbiosis and repair of the intestinal barrier. Integrative proteomic and transcriptomic analyses identified the NOD-like receptor signaling pathway as a potential key mechanism, with GBP5 as a possible key protein or gene. In the verification of the prediction results, XXD could significantly inhibit the protein expression of GBP5, NLRP3, ASC, Pro-Caspase1/Cleaved-Caspase1, and N-GSDMD/GSDMD; upregulate the protein levels of ZO-1 and occludin; and reduce the mRNA levels of GBP5 and NLRP3. In addition, it reduced the secretion of IL-1β and IL-18. IF confirmed the co-localization of EpCAM-GSDMD, GBP5-NLRP3, and NLRP3-ASC in colon tissues, whereas TEM suggested that XXD alleviated the ultrastructural damage caused by pyroptosis. CONCLUSION This study demonstrated that XXD can repair the intestinal mucosal barrier and regulate dysbiosis and its associated symptoms. Multiomics approaches have been used to predict the potential mechanisms of XXD. Evidence from experiments indicates that the regulation of dysbiosis by XXD may involve alleviating pyroptosis via inhibition of the GBP5/NLRP3 pathway.
Collapse
Affiliation(s)
- Tianyi Ren
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100083, China
| | - Yanyun Yin
- Department of Reproductive Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Xibin Zhou
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Liwei Chen
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yiming Shen
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hui Feng
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chunxiang Zhou
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yong Xu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yun Ling
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
29
|
Lu Z, Yu L, Bai Y, Cui Y, Shi M, Li Z, Li X, Zhong X, Jin Y, Wei C. The potential mechanisms of reciprocal regulation of gut microbiota-liver immune signaling in metabolic dysfunction-associated steatohepatitis revealed in multi-omics analysis. mSystems 2025:e0051825. [PMID: 40492756 DOI: 10.1128/msystems.00518-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Accepted: 05/13/2025] [Indexed: 06/12/2025] Open
Abstract
As a commonly known aggressive liver-related manifestation within the spectrum of metabolic syndrome with a significant risk of progressing to cirrhosis and hepatocellular carcinoma, metabolic dysfunction-associated steatohepatitis (MASH) is closely intertwined with obesity, insulin resistance, and dyslipidemia. Although the gut microbiota is implicated in MASH progression, the underlying mechanisms require further investigation. In this study, we sought to combine the analysis of the liver transcriptome, circulating metabolome, and gut microbiota to investigate the potential molecular mechanisms underlying the reciprocal regulation between gut microbiota and liver immune signaling. We utilized a high-fat and methionine/choline-deficient diet (HFMCD)-induced MASH model in a db/db mouse. Following annotation analysis using KEGG and Metorigin, a comprehensive correlation analysis was conducted among these genes and specific metabolites (such as L-glutamine, isocitric acid, putrescine, pyroglutamic acid, rhamnose) and gut microbiota genera (Enteroccus and Romboutsia). The results revealed intricate interactions among the liver's immune microenvironment, the metabolome, and the gut microbiota. These interactions suggest a potential regulatory mechanism for metabolic disorders and immune responses.IMPORTANCEOur multi-omics analysis showed that the interactions between gut microbiota and liver immune responses mediated by the disorders in lipid, amino acid, and glucose metabolism are associated with activation of the JAK-STAT and NF-κB signaling pathway in MASH. The multi-omics analysis provides valuable insights into the interactions among microbiota, circulating metabolites, and immune signaling. These insights can be harnessed to enhance the management of MASH.
Collapse
Affiliation(s)
- Zhaoyang Lu
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ligen Yu
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yun Bai
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yifeng Cui
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Meixin Shi
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhitao Li
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoxue Li
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xin Zhong
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Ye Jin
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Can Wei
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
30
|
Tan J, Taitz JJ, Ni D, Potier-Villette C, Pinget G, Pulpitel T, Stanley D, Nanan R, Macia L. Gut microbiota-derived extracellular vesicles exhibit diurnal regulation and activate hepatic gluconeogenesis. Mol Metab 2025; 98:102180. [PMID: 40484172 DOI: 10.1016/j.molmet.2025.102180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2025] [Revised: 05/30/2025] [Accepted: 06/02/2025] [Indexed: 06/16/2025] Open
Abstract
The circadian clock regulates tissue-specific homeostasis, and its disruption is associated with metabolic disorders. Both host metabolic processes and the gut microbiota exhibit diurnal regulation, and both contribute to the maintenance of glucose homeostasis (Thaiss et al., 2014; Bishehsari et al., 2020; Frazier et al., 2023) [1-3]. However, how the gut microbiota and the circadian rhythm interplay to control host glucose homeostasis is not fully understood. Here, we identified gut microbiota-derived extracellular vesicles (MEV) as a potential peripheral Zeitgeber (time cue) for the hepatic circadian clock, controlling hepatic gluconeogenesis. Host feeding patterns influence the gut microbiota, driving the diurnal production of MEV. Gut MEV levels coincide with the activity of hepatic gluconeogenesis, with overnight fasting associated with increased production of MEV by gut bacteria. MEV directly activates hepatic gluconeogenesis and chronic increase in MEV exposure impairs glucose homeostasis in vivo. Our finding highlights a mechanism by which the gut microbiota has co-evolved with the host to support its glucose needs during periods of energy demands (such as during fasting or starvation). On the contrary, an abnormal increase in MEV production, leading to dysregulated gluconeogenesis, may underlie various glucose-associated disorders, such as type 2 or gestational diabetes. Together, our data reconcile the gut microbiota and circadian rhythm in the control of host glucose homeostasis and metabolic health.
Collapse
Affiliation(s)
- Jian Tan
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; School of Medical Sciences, Faculty of Medicine and Health, the University of Sydney, Sydney, NSW, Australia; Sydney Medical School Nepean, the University of Sydney, Sydney, NSW, Australia; Nepean Hospital, Nepean Blue Mountains Local Health District, Sydney, New South Wales, Australia
| | - Jemma Justine Taitz
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; School of Medical Sciences, Faculty of Medicine and Health, the University of Sydney, Sydney, NSW, Australia
| | - Duan Ni
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; School of Medical Sciences, Faculty of Medicine and Health, the University of Sydney, Sydney, NSW, Australia; Sydney Medical School Nepean, the University of Sydney, Sydney, NSW, Australia
| | - Camille Potier-Villette
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; School of Medical Sciences, Faculty of Medicine and Health, the University of Sydney, Sydney, NSW, Australia
| | - Gabriela Pinget
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; School of Medical Sciences, Faculty of Medicine and Health, the University of Sydney, Sydney, NSW, Australia
| | - Tamara Pulpitel
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; School of Life and Environmental Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Dragana Stanley
- School of Health, Medical and Applied Science, Central Queensland University, Rockhampton, QLD, Australia
| | - Ralph Nanan
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; Sydney Medical School Nepean, the University of Sydney, Sydney, NSW, Australia; Nepean Hospital, Nepean Blue Mountains Local Health District, Sydney, New South Wales, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; School of Medical Sciences, Faculty of Medicine and Health, the University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
31
|
Pisoni L, Donini L, Gagni P, Pennuto M, Ratti A, Verde F, Ticozzi N, Mandrioli J, Calvo A, Basso M. Barriers in the Nervous System: Challenges and Opportunities for Novel Biomarkers in Amyotrophic Lateral Sclerosis. Cells 2025; 14:848. [PMID: 40498024 PMCID: PMC12155447 DOI: 10.3390/cells14110848] [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: 05/11/2025] [Revised: 06/02/2025] [Accepted: 06/03/2025] [Indexed: 06/19/2025] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a complex neurodegenerative disorder characterized by wide phenotypic heterogeneity. Despite efforts to carefully define and stratify ALS patients according to their clinical and genetic features, prognosis prediction still remains unreliable. Biomarkers that reflect changes in the central nervous system would be useful, but the physical impossibility of direct sampling and analysis of the nervous system makes them challenging to validate. A highly explored option is the identification of neuronal-specific markers that could be analyzed in peripheral biofluids. This review focuses on the description of the physical and biological barriers to the central nervous system and of the composition of biofluids in which ALS disease biomarkers are actively searched. Finally, we comment on already validated biomarkers, such as the neurofilament light chain, and show the potential of extracellular vesicles (EVs) and cell-free DNA as additional biomarkers for disease prediction.
Collapse
Affiliation(s)
- Lorena Pisoni
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy;
| | - Luisa Donini
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy;
| | - Paola Gagni
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimiche “Giulio Natta” (SCITEC-CNR), 20131 Milano, Italy;
| | - Maria Pennuto
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy;
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Antonia Ratti
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20133 Milan, Italy;
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy; (F.V.); (N.T.)
| | - Federico Verde
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy; (F.V.); (N.T.)
- Department of Pathophysiology and Transplantation, “Dino Ferrari” Center, Università degli Studi di Milano, 20122 Milan, Italy
| | - Nicola Ticozzi
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy; (F.V.); (N.T.)
- Department of Pathophysiology and Transplantation, “Dino Ferrari” Center, Università degli Studi di Milano, 20122 Milan, Italy
| | - Jessica Mandrioli
- Department of Neurosciences, Ospedale Civile Baggiovara, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy;
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Andrea Calvo
- ALS Centre, “Rita Levi Montalcini” Department of Neuroscience, University of Torino, 10120 Torino, Italy;
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, SC Neurologia 1U, 10120 Torino, Italy
| | - Manuela Basso
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy;
| |
Collapse
|
32
|
Lee J, McClure S, Weichselbaum RR, Mimee M. Designing live bacterial therapeutics for cancer. Adv Drug Deliv Rev 2025; 221:115579. [PMID: 40228606 PMCID: PMC12067981 DOI: 10.1016/j.addr.2025.115579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Humans are home to a diverse community of bacteria, many of which form symbiotic relationships with their host. Notably, tumors can also harbor their own unique bacterial populations that can influence tumor growth and progression. These bacteria, which selectively colonize hypoxic and acidic tumor microenvironments, present a novel therapeutic strategy to combat cancer. Advancements in synthetic biology enable us to safely and efficiently program therapeutic drug production in bacteria, further enhancing their potential. This review provides a comprehensive guide to utilizing bacteria for cancer treatment. We discuss key considerations for selecting bacterial strains, emphasizing their colonization efficiency, the delicate balance between safety and anti-tumor efficacy, and the availability of tools for genetic engineering. We also delve into strategies for precise spatiotemporal control of drug delivery to minimize adverse effects and maximize therapeutic impact, exploring recent examples of engineered bacteria designed to combat tumors. Finally, we address the underlying challenges and future prospects of bacterial cancer therapy. This review underscores the versatility of bacterial therapies and outlines strategies to fully harness their potential in the fight against cancer.
Collapse
Affiliation(s)
- Jaehyun Lee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Sandra McClure
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago 60637, USA; The Ludwig Center for Metastasis Research, University of Chicago, Chicago 60637, USA
| | - Mark Mimee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
33
|
Huang W, Jiang T, He J, Ruan J, Wu B, Tao R, Xu P, Wang Y, Chen R, Wang H, Yang Q, Zhang K, Jin L, Sun D, You J. Modulation of Intestinal Flora: a Novel Immunotherapeutic Approach for Enhancing Thyroid Cancer Treatment. Probiotics Antimicrob Proteins 2025; 17:1038-1063. [PMID: 39890752 DOI: 10.1007/s12602-025-10471-z] [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] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Over the past 3 years, there has been a growing interest in clinical research regarding the potential involvement of intestinal flora in thyroid cancer (TC). This review delves into the intricate connection between intestinal flora and TC, focusing on the particular intestinal flora that is directly linked to the disease and identifying which may be able to predict potential microbial markers of TC. In order to shed light on the inflammatory pathways connected to the onset of TC, we investigated the impact of intestinal flora on immune modulation and the connection between chronic inflammation when investigating the role of intestinal flora in the pathogenesis of TC. Furthermore, the potential role of intestinal flora metabolites in the regulation of thyroid function was clarified by exploring the effects of short-chain fatty acids and lipopolysaccharide on thyroid hormone synthesis and metabolism. Based on these findings, we further explore the effects of probiotics, prebiotics, postbiotics, vitamins, and trace elements.
Collapse
Affiliation(s)
- Weiqiang Huang
- Department of General Surgery, The First People's Hospital of Jiashan, Jiashan Hospital Afliated of Jiaxing University, Jiaxing, 314100, China
| | - Tao Jiang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jiaxuan He
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jing Ruan
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Baihui Wu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Runchao Tao
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Peiye Xu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Yongpan Wang
- Department of General Surgery, The First People's Hospital of Jiashan, Jiashan Hospital Afliated of Jiaxing University, Jiaxing, 314100, China
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, SAR 999077, China
| | - Hanbing Wang
- The University of Hong Kong School of Biomedical Sciences, Hong Kong, 999077, SAR, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Kun Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Jinfeng You
- Department of Obstetrics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China.
| |
Collapse
|
34
|
Støy S, Schnabl B. Role of Intestinal Microbiome in Potentiating Inflammation and Predicting Outcomes in Alcohol-Associated Cirrhosis. Gastroenterol Clin North Am 2025; 54:453-467. [PMID: 40348498 PMCID: PMC12066832 DOI: 10.1016/j.gtc.2024.12.001] [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] [Indexed: 05/14/2025]
Abstract
In patients with alcohol-associated cirrhosis, the intestinal microbiome composition is disturbed with a loss of beneficial functions and an increase in pathobionts. These changes are associated with disease severity and decompensation, due in part to the exacerbation of liver inflammation by an altered microbiome. Microbes or their antigens may translocate to the liver to potentiate the activation of immune cells and thereby contribute to inflammatory injury. Moreover, microbes may aggravate liver disease through the production of toxins or metabolites, via the effects on bile acids or the intestinal immune system.
Collapse
Affiliation(s)
- Sidsel Støy
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
35
|
Okunlola FO, Okunlola AR, Adetuyi BO, Soliman MES, Alexiou A, Papadakis M, Fawzy MN, El-Saber Batiha G. Beyond the gut: Unraveling the multifaceted influence of microbiome on cardiovascular health. Clin Nutr ESPEN 2025; 67:71-89. [PMID: 40064239 DOI: 10.1016/j.clnesp.2025.03.002] [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: 10/09/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Cardiovascular disease is one of the leading causes of death worldwide. Even while receiving adequate pharmacological treatment for their hypertension, people are nonetheless at greater risk for cardiovascular disease. There is growing evidence that the gut microbiota may have major positive and negative effects on blood pressure and illnesses related with it as more study into this topic is conducted. Trimethylamine n-oxide (TMAO) and short-chain fatty acids (SCFA) are two major by-products of the gut microbiota. TMAO is involved in the formation of other coronary artery diseases, including atherosclerosis and hypertension, while SCFAs play an important role in controlling blood pressure. Numerous investigations have confirmed the established link between dietary salt intake and hypertension. Reducing sodium in the diet is linked to lower rates of cardiovascular disease morbidity and mortality as well as lower rates of blood pressure and hypertension. In both human and animal research, high salt diets increase local and systemic tissue inflammation and compromise gut architecture. Given that the gut microbiota constantly interacts with the immune system and is required for the correct maturation of immune cells, it is scientifically conceivable that it mediates the inflammatory response. This review highlights the therapeutic possibilities for focusing on intestinal microbiomes as well as the potential functions of the gut microbiota and its metabolites in the development of hypertension.
Collapse
Affiliation(s)
- Felix Oladele Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Abimbola Rafiat Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Babatunde Oluwafemi Adetuyi
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research & Development, Funogen, Athens, 11741, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohamed N Fawzy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt.
| |
Collapse
|
36
|
Tang W, Long Z, Xiao Y, Du J, Tang C, Chen J, Hou C. Dietary butyric acid intake, kidney function, and survival: The National Health and Nutrition Examination Surveys, 2005-2018. Clin Nutr ESPEN 2025; 67:453-462. [PMID: 40147762 DOI: 10.1016/j.clnesp.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 02/23/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Although preclinical data support the hypothesis that butyric acid supplementation improves kidney health, the clinical significance of dietary butyric acid intake in patients with chronic kidney disease (CKD) remains unconfirmed in large-sample studies. This study aimed to investigate the association between dietary butyric acid intake and all-cause mortality in the United States population, stratified by kidney function. METHODS We examined the relationship between dietary butyric acid intake, assessed through a 24-h dietary recall, and all-cause mortality among 23,008 consecutive adult participants from the National Health and Nutrition Examination Surveys (NHANES, 2005-2018), categorized by impaired versus normal kidney function (estimated glomerular filtration rate <60 vs ≥ 60 mL/min/1.72 m2), using multivariable Cox models. We also employed a restricted cubic spline based on Cox regression models to elucidate the nonlinear relationship between dietary butyric acid intake and mortality in patients. RESULT In participants with impaired kidney function, high dietary butyric acid intake was associated with lower mortality, while lower intake levels (reference) showed no such association: adjusted HRs (aHRs) were 0.67 (95 % CI: 0.45, 1.00), 0.65 (95 % CI: 0.45, 0.94), and 0.58 (95 % CI: 0.38, 0.89) for intake levels of the square root of butyric acid 0.25-0.45, 0.45-0.75, and >0.75 g/day, respectively. However, in participants with normal kidney function, no association between butyric acid levels and mortality was observed. Additionally, we identified an L-shaped association between the levels of the square root of dietary butyric acid intake and all-cause mortality in the CKD population, reaching a plateau at 0.52 g/day (butyric acid intake of approximately 0.27 g/day). CONCLUSION This study revealed a nonlinear association between high dietary butyric acid intake and reduced all-cause mortality in patients with chronic kidney disease. A plateau occurs after 0.27 g/day, and for individuals with CKD whose butyric acid intake is below approximately 0.27 g/day, increasing a butyrate-rich diet or supplementing with butyric acid preparations may help prevent progression to renal failure and associated adverse outcomes in CKD patients, thereby reducing mortality. Therefore, it can be considered a new therapeutic strategy for the treatment of chronic kidney disease.
Collapse
Affiliation(s)
- Wei Tang
- Hunan Key Laboratory of Kidney Disease and Blood Purification, and Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhengyi Long
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yang Xiao
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jingyun Du
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Chenyuan Tang
- Hunan Key Laboratory of Kidney Disease and Blood Purification, and Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - JunXiang Chen
- Hunan Key Laboratory of Kidney Disease and Blood Purification, and Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Can Hou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
37
|
Procopio AC, Soggiu A, Urbani A, Roncada P. Interactions between microplastics and microbiota in a One Health perspective. One Health 2025; 20:101002. [PMID: 40123919 PMCID: PMC11927730 DOI: 10.1016/j.onehlt.2025.101002] [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: 12/05/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
Microplastics are recognised as ubiquitous pollutants as they are now found in all terrestrial and marine ecosystems. The interactions between microbiota and microplastics are an issue of fundamental importance in studying and maintaining global health. Microplastics alter the structures and functions of microbial communities, resulting in adverse health effects. A comprehensive understanding of these effects through interdisciplinary research is essential to mitigate pollution and protect the health of ecosystems. The review aims to explore these interactions within a One Health framework. Indeed, a deeper understanding of the processes involved in the interaction between microbiota and microplastics could pave the way for new and promising strategies to mitigate the harmful effects of microplastics on ecosystems and human health.
Collapse
Affiliation(s)
- Anna Caterina Procopio
- Department of Health Science, University Magna Graecia of Catanzaro, viale Europa, 88100 Catanzaro, Italy
| | - Alessio Soggiu
- Department of Biomedical, Surgical and Dental Sciences, University of Milano, Via Celoria n.10, 20133 Milano, Italy
| | - Andrea Urbani
- Department of Diagnostic and Laboratory Medicine, Unity of Chemistry, Biochemistry and Clinical Molecular Biology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics Research, Catholic University of the Sacred Heart, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Paola Roncada
- Department of Health Science, University Magna Graecia of Catanzaro, viale Europa, 88100 Catanzaro, Italy
| |
Collapse
|
38
|
Zhu B, Liang L, Huang Y, Wang H, Zhou J, Xiong D, Li S, Li H, Li X, Chen S, Ning Y, Wu F, Wu K. Exploring the relationship between the gut microbiota and cognitive function in schizophrenia patients with distinct weights. Schizophr Res 2025; 280:103-113. [PMID: 40279867 DOI: 10.1016/j.schres.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 04/07/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The gut microbiota is disrupted in schizophrenia (SZ) patients and is associated with cognitive function. This study aimed to investigate the gut microbiota composition in SZ patients with different body mass index (BMI) levels and their associations with cognitive function. METHODS We analyzed 16S rRNA sequencing data from 156 SZ patients, including 88 with overweight/obesity (OW) and 68 with normal weight (NW), and 156 normal control (NC), including 48 with OW and 108 with NW. We analyzed differences in microbial diversity and gut microbiota composition between SZ patients and NC at different BMI levels. Additionally, we explored the correlations between microbial communities, and symptom severity, as well as cognitive function. Furthermore, we examined between-group differences in metabolic pathways. RESULTS The abundance of Turicibacter was higher in the SZ_OW group but lower in the SZ_NW group compared to the NC groups at the same BMI level, respectively. In the SZ_OW group, increased Collinsella was significantly negatively associated with cognitive function, whereas decreased Clostridium and Butyricicoccus were significantly positively associated with cognitive function. Additionally, the functional analysis revealed enrichment of "metabolism of other amino acids" and "neurodegenerative disease" pathways, associated with non-standard amino acid metabolism and oxidative stress in the SZ_OW group compared to the NC_OW group. CONCLUSIONS Our findings revealed significant differences in the gut microbiota between SZ patients and NC with different BMI levels and identified microbial associations with clinical characteristics, providing new insights into the mechanism of how the gut microbiota could impact cognitive deficits in SZ patients with obesity.
Collapse
Affiliation(s)
- Baoyuan Zhu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Liqin Liang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Yuanyuan Huang
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou 510370, China
| | - Haiyuan Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Jing Zhou
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Dongsheng Xiong
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Shaochuan Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Hehua Li
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou 510370, China
| | - Xiaobo Li
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Shuhao Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Yuping Ning
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou 510370, China
| | - Fengchun Wu
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou 510370, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou 510370, China.
| | - Kai Wu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan.
| |
Collapse
|
39
|
Jiang J, Liu Y, Yang H, Ma Z, Liu W, Zhao M, Peng X, Qin X, Xia Y. Dietary fiber intake, genetic predisposition of gut microbiota, and the risk of metabolic dysfunction-associated steatotic liver disease. Food Res Int 2025; 211:116497. [PMID: 40356189 DOI: 10.1016/j.foodres.2025.116497] [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: 06/17/2024] [Revised: 04/16/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025]
Abstract
This study aimed to explore the association between dietary fiber intake and the risk of metabolic dysfunction-associated steatotic liver disease (MASLD), as well as liver fat content, while considering genetic predispositions of MASLD, gut microbial abundance, and butyrate levels. This study analyzed data from 190,276 participants in the UK Biobank. Dietary fiber intake was assessed using 24-h dietary recall. MASLD cases were diagnosed through hospital admission records and death registries, and liver fat content was measured via magnetic resonance imaging. The genetic predispositions of MASLD, gut microbial abundance, and butyrate levels were evaluated using single nucleotide polymorphisms. Cox proportional hazards models were used to calculate hazard ratios (HRs) and 95 % confidence intervals (CIs). Over a median follow-up of 10.49 years, 1423 MASLD cases were recorded. Elevated dietary fiber intake was associated with a reduced risk of MASLD (HR: 0.72; 95 % CI: 0.58, 0.90) and a lower level of liver fat content (β: -0.97; 95 % CI: -1.21, -0.73) (all P for trend <0.05). Restricted cubic spline analyses further confirmed the linear inverse associations between fiber intake and the risk of MASLD. Notably, the negative associations between dietary fiber intake and both MASLD and liver fat content were consistent across different genetic predispositions of gut microbial abundance and butyrate levels. Moreover, the inverse association between dietary fiber intake and liver fat was strengthened by high genetic susceptibility of MASLD and elevated body mass index (both P for interaction <0.05). Overall, increased dietary fiber consumption was associated with a lower MASLD risk and decreased liver fat content regardless of genetic predispositions of gut microbial abundance and butyrate levels.
Collapse
Affiliation(s)
- Jinguo Jiang
- School of Public Health, Shenyang Medical College, Shenyang, China; Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China.
| | - Yang Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, 38# Xueyuan Road, Haidian District, Beijing 100191, China.
| | - Honghao Yang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China; Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Liaoning Province, Shenyang, China.
| | - Zheng Ma
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China; Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Liaoning Province, Shenyang, China.
| | - Wenqi Liu
- School of Public Health, Shenyang Medical College, Shenyang, China; Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China.
| | - Maoxiang Zhao
- Interventional Center of Valvular Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing 100011, China.
| | - Xinyi Peng
- Hypertension Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Center for Cardiovascular Diseases of China, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China.
| | - Xueying Qin
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, 38# Xueyuan Road, Haidian District, Beijing 100191, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, China.
| | - Yang Xia
- School of Public Health, Shenyang Medical College, Shenyang, China; Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
40
|
Zhou S, Jiang H, Wang Y, Yan A, Liu G, Liu S, Chen B. Dietary supplementation with isochlorogenic acid improves growth performance and intestinal health of broilers. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 21:472-486. [PMID: 40519248 PMCID: PMC12166709 DOI: 10.1016/j.aninu.2024.11.012] [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: 01/28/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 06/18/2025]
Abstract
This study evaluated the effects of dietary supplementation with isochlorogenic acid (ICA) on growth performance and intestinal health of broilers. A total of 400 Cobb broilers (44.20 ± 0.16 g, one day old) were randomly assigned to 5 treatments (ICA supplementation at 0, 500, 1000, 2000, and 3000 mg/kg diet) with 8 replicates of 10 birds each. The experimental trial lasted for 42 d. The feed-to-gain ratio (F/G) exhibited a quadratic decrease in response to ICA supplementation during both the 1 to 42 d (P = 0.048) and 22 to 42 d (P = 0.039) periods, with the lowest F/G observed at a dietary ICA concentration of 2000 mg/kg. The apparent digestibility of calcium (P = 0.038) and crude protein (P < 0.001) exhibited a linear upward trend, and both the villus height and the villus height-to-crypt depth ratio showed a quadratic increase (P = 0.027) with ICA supplementation. Meanwhile, the relative mRNA expression of ileum claudin-1 (P = 0.003) and occludin (P = 0.048), along with the gastrointestinal pH (P < 0.05), decreased linearly, whereas the concentration of ileum secretory immunoglobulin A (P = 0.005) increased linearly with ICA supplementation. Additionally, the concentration of total volatile fatty acids (P = 0.038) in the ileum, trypsin activity (P = 0.016), the serum concentration of immunoglobulin G (P = 0.005), and the activities of serum glutathione peroxidase (GSH-Px) (P = 0.005) and superoxide dismutase (SOD) (P = 0.040) increased quadratically with ICA supplementation. Additionally, the relative mRNA expression of SOD-1 (P = 0.040) and GSH-Px (P = 0.040) in the ileum increased linearly with ICA supplementation. The abundance of Streptococcus alactolyticus was significantly higher in broilers supplemented with ICA at 2000 mg/kg compared to the control treatment (P = 0.030), and the concentration of metabolites, such as 15-deoxy-delta-12,14- prostaglandin J2, was increased by ICA supplementation. Dietary ICA enhanced broilers growth performance via increased digestive enzyme activity, leading to improved feed digestibility. Additionally, ICA improved health status by maintaining gut pH and enhancing antioxidant and immune functions. The optimal supplemental level of ICA of improving growth performance and intestinal immune function of broilers was 2000 mg/kg.
Collapse
Affiliation(s)
- Siyuan Zhou
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Haotian Jiang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Yujia Wang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - An Yan
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Guohua Liu
- Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100080, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Shudong Liu
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Baojiang Chen
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071001, China
| |
Collapse
|
41
|
Totter E, von Einsiedel E, Regazzoni L, Schuerle S. Paving the way for bacteria-based drug delivery: biohybrid microrobots emerging from microrobotics and synthetic biology. Adv Drug Deliv Rev 2025; 221:115577. [PMID: 40250568 DOI: 10.1016/j.addr.2025.115577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 04/20/2025]
Abstract
Advances in microrobotics and synthetic biology are paving the way for innovative solutions to long-standing challenges in drug delivery. Both fields have independently worked on engineering bacteria as a therapeutic system, focusing on enhancing propulsion, cargo delivery, detection, and biocompatibility. Bacteria, with their inherent adaptability and functional versatility, serve as an ideal foundation for these efforts, enabling them to navigate complex biological environments such as the human body. This review explores the convergence of microrobotics and synthetic biology, which has catalysed the development of biohybrid bacterial microrobots that integrate the strengths of both disciplines. By incorporating external control modalities - such as light, ultrasound, and magnetic fields - these hybrid systems address the limitations of purely microrobotic or biological approaches, offering new opportunities to enhance precision and efficacy in targeted therapies. However, realising the full potential of biohybrid bacterial microrobots requires overcoming critical challenges, such as ensuring compatibility between biological and synthetic components, scaling manufacturing processes, and defining regulatory pathways tailored to living therapeutics. Addressing these hurdles through joint, interdisciplinary research efforts, can unlock the transformative possibilities of these systems in modern medicine.
Collapse
Affiliation(s)
- Elena Totter
- ETH Zurich, Institute of Translational Medicine, Gloriastrasse 37/39, 8092 Zurich, Switzerland
| | - Emilie von Einsiedel
- ETH Zurich, Institute of Translational Medicine, Gloriastrasse 37/39, 8092 Zurich, Switzerland
| | - Lisa Regazzoni
- ETH Zurich, Institute of Translational Medicine, Gloriastrasse 37/39, 8092 Zurich, Switzerland
| | - Simone Schuerle
- ETH Zurich, Institute of Translational Medicine, Gloriastrasse 37/39, 8092 Zurich, Switzerland.
| |
Collapse
|
42
|
Luo Y, Tang R, Huang Y. Differences in structure, antioxidant capacity and gut microbiota modulation of red raspberry pectic polysaccharides extracted by different methods. Food Res Int 2025; 211:116474. [PMID: 40356136 DOI: 10.1016/j.foodres.2025.116474] [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: 01/08/2025] [Revised: 03/19/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
Red raspberries are associated with various health benefits, with pectic polysaccharides as their primary component and potential key contributor to these effects. This study aimed to evaluate the antioxidant and prebiotic potential of four red raspberry pectic polysaccharides (RP)-EN-RP (enzyme-assisted extraction), AC-RP (acid-assisted extraction), AL-RP (alkali-assisted extraction), and US-RP (ultrasound-assisted extraction)-and to elucidate the relationship between their structure and function. AC-RP and US-RP contained higher proportions of homogalacturonan (HG) at 50.92 % and 53.10 %, respectively, while EN-RP and AL-RP exhibited higher proportions of rhamnogalacturonan-I (RG-I) at 63.89 % and 43.37 %, respectively. All four polysaccharides demonstrated significant antioxidant and prebiotic properties. AL-RP exhibited the strongest DPPH radical scavenging activity, while US-RP showed the highest hydroxyl radical scavenging ability. These pectic polysaccharides were highly fermentable, significantly modulating gut microbiota composition and promoting the production of propionic acid, particularly EN-RP and AL-RP. Compared to the blank group, RP intervention significantly enriched Bacteroides, Phocaeicola, Bifidobacterium, Limosilactobacillus, and Paraprevotella. Carbohydrate-active enzyme genes in metagenomes revealed that glycoside hydrolases played a vital role in the degradation and utilization of red raspberry polysaccharides. Furthermore, correlation analysis indicated that a higher RG-I proportion and an elevated Rha/GalA ratio enhanced the abundance of certain beneficial microbial species and increased propionic acid production. These findings advance the understanding of the structure-function relationship of natural pectic polysaccharides and highlight their potential for tailoring gut microbiota and promoting health through precise dietary interventions.
Collapse
Affiliation(s)
- You Luo
- School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China; Centre for Nutrition and Food Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Indooroopilly, QLD, 4068, Australia.
| | - Ruling Tang
- School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China
| | - Yongguang Huang
- School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China
| |
Collapse
|
43
|
Burananat T, Wilantho A, Kulalert P, Nanthapisal S, Tonglim J, Deetienin W, Wangkumhang P, Tongsima S, Thaweekul P. The role of gut microbiota in obesity severity and metabolic risk in pediatric populations. Nutr Metab Cardiovasc Dis 2025; 35:103970. [PMID: 40180829 DOI: 10.1016/j.numecd.2025.103970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/12/2025] [Accepted: 03/04/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND AND AIMS Childhood obesity is a considerable public health issue. Recent research has shown that alterations in gut microbiota can have an impact on developing obesity and other metabolic health problems in children. This study aimed to investigate whether the characteristics of gut microbiota in obese children and adolescents are associated with the severity of obesity and any metabolic complications. METHODS AND RESULTS During May 2022 to May 2023, a total of 56 children and adolescents with obesity, aged 6-18 years, were recruited at Thammasat Hospital, situated in provincial Pathumthani in central Thailand. Participants were allocated into two groups, characterized by the severity of their obesity. Demographic data, body composition, along with resting energy expenditures were determined. Serum samples were collected for the metabolic profile and inflammatory markers. Fecal samples were obtained for gut microbiota analysis via 16S rRNA Illumina. The obese group exhibited notably greater relative abundance of Actinobacteriota in comparison to the severely obese group, along with a lower abundance of Bacteroidota. There were no statistically significant differences in the relative abundance of Firmicutes and the Firmicutes to Bacteroidota ratio between the two cohorts. Bacteroidota positively correlated with FMI, while Actinobacteriota showed a negative correlation with FMI. CONCLUSION The data gathered from this study illustrated that children and adolescents with obesity and severe obesity in Thailand showed differences in the relative abundance of Actinobacteriota and Bacteroidota. Certain microbiome taxa showed correlations with various body and metabolic parameters.
Collapse
Affiliation(s)
- Thanyamas Burananat
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Alisa Wilantho
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Prapasri Kulalert
- Department of Clinical Epidemiology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Sira Nanthapisal
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | | | | | - Pongsakorn Wangkumhang
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Sissades Tongsima
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Patcharapa Thaweekul
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand.
| |
Collapse
|
44
|
Schoultz I, Claesson MJ, Dominguez‐Bello MG, Fåk Hållenius F, Konturek P, Korpela K, Laursen MF, Penders J, Roager H, Vatanen T, Öhman L, Jenmalm MC. Gut microbiota development across the lifespan: Disease links and health-promoting interventions. J Intern Med 2025; 297:560-583. [PMID: 40270478 PMCID: PMC12087861 DOI: 10.1111/joim.20089] [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] [Indexed: 04/25/2025]
Abstract
The gut microbiota plays a pivotal role in human life and undergoes dynamic changes throughout the human lifespan, from infancy to old age. During our life, the gut microbiota influences health and disease across life stages. This review summarizes the discussions and presentations from the symposium "Gut microbiota development from infancy to old age" held in collaboration with the Journal of Internal Medicine. In early infancy, microbial colonization is shaped by factors such as mode of delivery, antibiotic exposure, and milk-feeding practices, laying the foundation for subsequent increased microbial diversity and maturation. Throughout childhood and adolescence, microbial maturation continues, influencing immune development and metabolic health. In adulthood, the gut microbiota reaches a relatively stable state, influenced by genetics, diet, and lifestyle. Notably, disruptions in gut microbiota composition have been implicated in various inflammatory diseases-including inflammatory bowel disease, Type 1 diabetes, and allergies. Furthermore, emerging evidence suggests a connection between gut dysbiosis and neurodegenerative disorders such as Alzheimer's disease. Understanding the role of the gut microbiota in disease pathogenesis across life stages provides insights into potential therapeutic interventions. Probiotics, prebiotics, and dietary modifications, as well as fecal microbiota transplantation, are being explored as promising strategies to promote a healthy gut microbiota and mitigate disease risks. This review focuses on the gut microbiota's role in infancy, adulthood, and aging, addressing its development, stability, and alterations linked to health and disease across these critical life stages. It outlines future research directions aimed at optimizing the gut microbiota composition to improve health.
Collapse
Affiliation(s)
- Ida Schoultz
- School of Medical SciencesFaculty of Medicine and Health Örebro UniversityOrebroSweden
| | | | - Maria Gloria Dominguez‐Bello
- Department of Biochemistry & Microbiology and of AnthropologyRutgers University–New BrunswickNew BrunswickNew JerseyUSA
| | - Frida Fåk Hållenius
- Department of Food Technology, Engineering and NutritionLund UniversityLundSweden
| | - Peter Konturek
- Department of Medicine, Thuringia Clinic SaalfeldTeaching Hospital of the University JenaJenaGermany
| | - Katri Korpela
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | | | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtthe Netherlands
| | - H. Roager
- Department of Nutrition, Exercise and SportsUniversity of CopenhagenFrederiksbergDenmark
| | - Tommi Vatanen
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFinland
- Department of Microbiology, Faculty of Agriculture and ForestryUniversity of HelsinkiHelsinkiFinland
- Research Program for Clinical and Molecular Metabolism, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
- Liggins InstituteUniversity of AucklandAucklandNew Zealand
| | - Lena Öhman
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Maria C. Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden
| |
Collapse
|
45
|
Liu W, Chen S, Yang J, Chen Y, Yang Q, Lu L, Li J, Yang T, Zhang G, Hu J. Characterization of blood and urine microbiome temporal variability in patients with acute myeloid leukemia. Microb Pathog 2025:107734. [PMID: 40449763 DOI: 10.1016/j.micpath.2025.107734] [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: 02/10/2025] [Revised: 05/09/2025] [Accepted: 05/21/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND Investigating the microbiota of blood and urine from acute myeloid leukemia (AML) patients is essential to unravel the complex role of microbiota in systemic host-microbe interactions and implications. METHODS We conducted a longitudinal observational study to characterize the temporal dynamics of blood and urine microbiota in 27 AML patients, utilizing metagenomic analysis pipeline for microbial identification to identify disease-associated microbial signatures. RESULTS The composition of blood and urine microbiota of AML was dominated by Proteobacteria phylum in blood, Firmicutes phylum in urine. The species and diversity of blood and urine microbiota did not have difference between AML patients and healthy controls. Restitution of alpha and beta diversity of blood microbiota and urine microbiota to resemble that of healthy controls occurred after cessation of treatment. Temporal variation of urine microbiome was higher than blood after treatment which was closely related to pathogenic bacteria and beneficial bacteria measured by coefficient of variation (CV) of alpha diversity. The temporal variability of urine microbiota was significantly correlated with platelet and exposure of levofloxacin. The variation of microbiome of AML patients with infection was found that the relative abundance of Burkholderia significantly enriched in blood and urine which had high accuracy and sensitivity. The correlation between blood microbiota and serum amino acid metabolites was similar to that between gut microbiota and serum metabolites. CONCLUSION This study represents the first comprehensive investigation to quantify the longitudinal variability of blood and urine microbiota in AML patients, revealing distinct patterns compared to gut microbiota and associations with adverse clinical outcomes. Our findings highlight the potential of leveraging stabilizing taxa as a target for microbiome restoration.
Collapse
Affiliation(s)
- Wanying Liu
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China; Fujian Medical University Union Hospital, Fuzhou, China
| | - Shaozhen Chen
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China; Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiajie Yang
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China; Fujian Medical University Union Hospital, Fuzhou, China
| | - Yanxin Chen
- Fujian Medical University Union Hospital, Fuzhou, China
| | - Qinwen Yang
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China; Fujian Medical University Union Hospital, Fuzhou, China
| | - Lihua Lu
- Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiazheng Li
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China; Fujian Medical University Union Hospital, Fuzhou, China
| | - Ting Yang
- Department of Hematology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Hematology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Institute of Precision Medicine, Fujian Medical University, Fuzhou, China
| | - Guanbin Zhang
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Laboratory Medicine, Fujian Medical University, Fuzhou, China; Institute of Precision Medicine, Fujian Medical University, Fuzhou, China; Mianyang People's Hospital, Mianyang, China
| | - Jianda Hu
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China; Fujian Medical University Union Hospital, Fuzhou, China; Institute of Precision Medicine, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
46
|
Zhang L, Wu C, Wang Q. Toxicity of Engineered Nanoparticles in Food: Sources, Mechanisms, Contributing Factors, and Assessment Techniques. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40418745 DOI: 10.1021/acs.jafc.5c01550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
The increasing prevalence of engineered nanoparticles (ENPs) in food systems has raised concerns about their toxicity and potential health risks. To provide a comprehensive evaluation, a structured literature search was conducted using databases such as Web of Science and PubMed, focusing on studies published in the past ten years that examine ENP exposure pathways, toxicity mechanisms, contributing factors, and risk assessment strategies. This review first explores the diverse sources of ENPs, including food additives, nanocarriers, packaging, agricultural practices, and environmental contamination. Upon ingestion, ENPs undergo complex transformations within the human gastrointestinal tract (GIT), causing oxidative stress, cellular dysfunction, inflammation, and gut microbiota dysbiosis, potentially leading to systemic toxicity in vital organs. The toxicity of ENPs is influenced by their physicochemical properties, food matrix effects, GIT conditions, and host-specific factors. This review further discusses current toxicity assessment methodologies, including in silico, in vitro, in vivo, and emerging technologies. Finally, we identify critical research gaps, such as the lack of long-term exposure studies and limited evaluations of organic ENPs. By providing a comprehensive analysis of ingested ENP toxicity, this review aims to guide safer ENP applications and mitigate potential health risks.
Collapse
Affiliation(s)
- Liping Zhang
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, Maryland 20742, United States
| | - Changqing Wu
- Department of Animal and Food Sciences, University of Delaware, Newark, Delaware 19716, United States
| | - Qin Wang
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
47
|
Xie X, Yang H, Zhao X, Teng L, Yang Y, Luo H. Potential role of key rumen microbes in regulating host health and growth performance in Hu sheep. Anim Microbiome 2025; 7:51. [PMID: 40414888 PMCID: PMC12103811 DOI: 10.1186/s42523-025-00412-0] [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: 12/03/2024] [Accepted: 04/16/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Average daily gain (ADG) is an important component affecting the profitability of sheep. However, research on the relationship between rumen microbes and sheep growth phenotype is still very lacking. Therefore, in this study, 16 Hu sheep were selected from a cohort of 318 sheep assigned to the same feeding and management conditions, and divided into high growth rate (HADG, n = 8) group and low growth rate (LADG, n = 8) group according to the extreme ADG value. Then, the differences in rumen microbes, rumen fermentation and animal immune parameters were further compared between groups to explore the potential role of rumen key microbes in regulating the health and growth performance of Hu sheep hosts. RESULTS The results showed that specific pathogenic bacteria associated with ADG, including Anaerotruncus, Sediminibacterium and Glaesserella, exhibited significant correlations with interleukin-6 (IL-6) and immunoglobulin G (IgG). These interactions disrupt immune homeostasis in the host, leading to a metabolic prioritization of energy resources toward immune responses, thereby impairing growth and development. Succinivibrio_dextrinosolvens was enriched in HADG sheep and exhibited a significant positive correlation with propionate levels. This promoted propionate production in the rumen, enhancing the metabolic activity of carbohydrate, amino acid and energy metabolism, ultimately contributing to higher ADG in sheep. Importantly, random forest analysis results showed that Succinivibrio_dextrinosolvens could classify sheep into HADG and LADG with a prediction accuracy of 81.2%. Additionally, we identified 34 bacteria belonged to connectors in the HADG co-occurrence network, including Alloprevotella, Phascolarctobacterium, Anaerovibrio, Butyricicoccus, Ruminococcaceae_noname, and Roseburia, etc., which play an important role in the degradation of carbohydrates and convert them into short-chain fatty acids (SCFAs), maintaining rumen health, and modulating inflammation. CONCLUSIONS In summary, key microbes in the rumen affect the overall healthy homeostasis and rumen fermentation of the host, leading to changes in energy utilization, which in turn affects the average daily gain of Hu sheep. Succinivibrio_dextrinosolvens is a promising biomarker for selecting high growth rate sheep in the future. This study provides a new method to manipulate rumen bacteria to improve growth performance in sheep.
Collapse
Affiliation(s)
- Ximei Xie
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Huan Yang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xingang Zhao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Li Teng
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yuze Yang
- Beijing General Station of Animal Husbandry Service, Beijing, 100101, China.
| | - Hailing Luo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
48
|
Fang J, Dong W, Zheng J, Han B, Zhang Y, Wang J, Zeng X. Antiaging Effect of 2- O-β-D-Glucopyranosyl Ascorbic Acid Derived from Lycium barbarum L. Through Modulating the IIS Pathway and Gut Microbiota in Caenorhabditis elegans. Foods 2025; 14:1875. [PMID: 40509403 PMCID: PMC12154002 DOI: 10.3390/foods14111875] [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: 04/10/2025] [Revised: 05/22/2025] [Accepted: 05/23/2025] [Indexed: 06/16/2025] Open
Abstract
2-O-β-D-Glucopyranosyl ascorbic acid (AA-2βG), a bioactive ascorbic acid derivative isolated from the fruits of Lycium barbarum L., exhibited significant antiaging effects in Caenorhabditis elegans. It significantly extended their lifespan, enhanced stress resistance, reduced lipofuscin accumulation, and improved their healthspan, while strengthening antioxidant defenses. Transcriptomic analysis identified the insulin/insulin-like growth factor (IGF)-1 signaling pathway as a key regulator, with quantitative real-time polymerase chain reaction confirming the upregulation of longevity-associated genes. Functional studies showed that the transcription factors DAF-16, HSF-1, and SIR-2.1 were essential for the lifespan-extending effects of AA-2βG, as mutations in these genes abolished lifespan extension. Moreover, 16S rRNA sequencing revealed that AA-2βG modulated gut microbiota by increasing longevity-associated taxa and reducing pro-aging species, with these alterations linked to metabolic pathways. These findings suggest that AA-2βG exerts antiaging effects through the coordinated regulation of the IIS pathway and gut microbiota composition, highlighting its potential as a natural geroprotective compound.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (J.F.); (W.D.); (J.Z.); (B.H.); (Y.Z.); (J.W.)
| |
Collapse
|
49
|
Zhang JM, Li P, Chen CZ, Liu L, Li ZH. Toxic effects of emerging pollutants on mucosal organs of teleost fish: A review focusing on mucosal microbiota, physical barrier and immune barrier. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 978:179431. [PMID: 40245518 DOI: 10.1016/j.scitotenv.2025.179431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/02/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025]
Abstract
The urgency of emerging pollutants driven by human activities presents an increasing threat to the health of fish. The mucosal system, serving as a primary barrier against environmental pollutants, has emerged as a central focus in toxicological research. Alterations in the mucosal microbiota can impact health at both local and systemic levels. This review explores the toxic effects of emerging pollutants on the mucosal immunity of teleost fish, reflects on the reasons behind the limited focus on adaptive immunity studies, and highlights changes in microbial composition, gene expression, histology, and overall mucosal organ function. Furthermore, we summarize the mechanisms through which these pollutants disrupt the mucosal barriers of teleosts, emphasizing interactions between the mucosal microbiota, physical barriers, and immune defenses. The relevant methodologies and potential solutions to the current challenges have been summarized. While current research predominantly centers on the intestines and gills, further studies are needed to investigate the toxic effects of emerging pollutants on other mucosal organs and to elucidate how microbiota influence host health through neuro-immune communication. This review aims to provide a comprehensive overview of mucosal immunity, serving as a theoretical foundation for the assessment of related ecological risks.
Collapse
Affiliation(s)
- Jia-Ming Zhang
- Marine College, Shandong University, Weihai, Shandong 264209, China
| | - Ping Li
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | | | - Ling Liu
- Marine College, Shandong University, Weihai, Shandong 264209, China
| | - Zhi-Hua Li
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| |
Collapse
|
50
|
Wang W, Lu Y, Wu B, Peng S, Cai W, Xiao Y. Multi-omics analysis reveals Lactobacillus and Indolelactic acid involved in small intestinal adaptation of piglet with short bowel syndrome. Nutr Metab (Lond) 2025; 22:47. [PMID: 40405192 PMCID: PMC12096513 DOI: 10.1186/s12986-025-00938-9] [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: 02/06/2025] [Accepted: 05/05/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Short bowel syndrome (SBS) is a condition characterized by malabsorption that occurs when a patient loses a significant amount of bowel length or function, often necessitating lifelong parenteral nutrition support. This study utilized multi-omics analysis to investigate alterations in gut microbiota, metabolism, and transcriptome during the progression of intestinal adaptation in SBS using a piglet model. METHODS We established a model of SBS in Bama mini piglets by performing a 75% jejunoileal resection. Fifteen piglets were randomized into EN, PN, and PN-SBS groups. Fecal samples were collected for 16 S rRNA gene-based microbiota analysis. Ileal mucosa and serum were collected for untargeted liquid chromatography-mass spectrometry. Transcriptomic analysis on ileal mucosa was performed. RESULTS The PN-SBS model was established in the newborn piglets. A significant decrease in species-level diversity was observed in piglets with SBS, accompanied by alterations in their microbiome compositions. The beneficial anaerobes from Bacillota and Bacteroidota were depleted while microorganisms from Verrucomicrobiota and Fusobacteriota were enriched in feces from SBS piglets. The dysregulation of metabolites and metabolic pathways was observed in the metabolic profiles of ileal mucosa and serum in SBS piglets. Indolelactic acid (ILA) levels were found to be reduced in the ileal mucosa and serum of SBS piglets. Transcriptomic analysis revealed an extensive functional alteration in SBS, primarily manifested as metabolic changes and intestinal proliferation. The multi-omics analysis revealed that the decreased abundance of Lactobacillus may result in a diminished production of their metabolite ILA, thereby influencing intestinal proliferation and anti-inflammatory responses. CONCLUSION Disrupted homeostasis of gut microbiota, metabolism, and transcriptome were reported in the SBS piglets. Multi-omics analysis demonstrated Lactobacillus and its metabolite ILA may be involved in small intestinal adaptation of SBS. These alterations may contribute to the proinflammatory state and the delay of intestinal adaptation in SBS, which in turn provide promising targets for therapies.
Collapse
Affiliation(s)
- Weipeng Wang
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1665, Kong Jiang Road, Shanghai, China
| | - Ying Lu
- Division of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Bo Wu
- Division of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shicheng Peng
- Shanghai Institute of Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Wei Cai
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1665, Kong Jiang Road, Shanghai, China
- Division of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Yongtao Xiao
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1665, Kong Jiang Road, Shanghai, China.
- Division of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Institute of Pediatric Research, Shanghai, China.
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.
| |
Collapse
|