1
|
Zheng XQ, Wang DB, Jiang YR, Song CL. Gut microbiota and microbial metabolites for osteoporosis. Gut Microbes 2025; 17:2437247. [PMID: 39690861 DOI: 10.1080/19490976.2024.2437247] [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: 07/11/2024] [Revised: 11/13/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Osteoporosis is an age-related bone metabolic disease. As an essential endocrine organ, the skeletal system is intricately connected with extraosseous organs. The crosstalk between bones and other organs supports this view. In recent years, the link between the gut microecology and bone metabolism has become an important research topic, both in preclinical studies and in clinical trials. Many studies have shown that skeletal changes are accompanied by changes in the composition and structure of the gut microbiota (GM). At the same time, natural or artificial interventions targeting the GM can subsequently affect bone metabolism. Moreover, microbiome-related metabolites may have important effects on bone metabolism. We aim to review the relationships among the GM, microbial metabolites, and bone metabolism and to summarize the potential mechanisms involved and the theory of the gut‒bone axis. We also describe existing bottlenecks in laboratory studies, as well as existing challenges in clinical settings, and propose possible future research directions.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Ding-Ben Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Yi-Rong Jiang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| |
Collapse
|
2
|
Mukhopadhya I, Martin JC, Shaw S, Gutierrez-Torrejon M, Boteva N, McKinley AJ, Gratz SW, Scott KP. Novel insights into carbohydrate utilisation, antimicrobial resistance, and sporulation potential in Roseburia intestinalis isolates across diverse geographical locations. Gut Microbes 2025; 17:2473516. [PMID: 40089923 PMCID: PMC11913394 DOI: 10.1080/19490976.2025.2473516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 02/11/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
Roseburia intestinalis is one of the most abundant and important butyrate-producing human gut anaerobic bacteria that plays an important role in maintaining health and is a potential next-generation probiotic. We investigated the pangenome of 16 distinct strains, isolated over several decades, identifying local and time-specific adaptations. More than 50% of the genes in each individual strain were assigned to the core genome, and 77% of the cloud genes were unique to individual strains, revealing the high level of genome conservation. Co-carriage of the same enzymes involved in carbohydrate binding and degradation in all strains highlighted major pathways in carbohydrate utilization and reveal the importance of xylan, starch and mannose as key growth substrates. A single strain had adapted to use rhamnose as a sole growth substrate, the first time this has been reported. The ubiquitous presence of motility and sporulation gene clusters demonstrates the importance of these phenotypes for gut survival and acquisition of this bacterium. More than half the strains contained functional, potentially transferable, tetracycline resistance genes. This study advances our understanding of the importance of R. intestinalis within the gut ecosystem by elucidating conserved metabolic characteristics among different strains, isolated from different locations. This information will help to devise dietary strategies to increase the abundance of this species providing health benefits.
Collapse
Affiliation(s)
- Indrani Mukhopadhya
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
- Microbiology and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Jennifer C. Martin
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Sophie Shaw
- Centre for Genome Enabled Biology and Medicine, University of Aberdeen, Aberdeen, UK
- All Wales Medical Genomics Service, Institute of Medical Genetics, University Hospital of Wales, Heath Park, Cardiff, UK
| | | | - Nikoleta Boteva
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Aileen J. McKinley
- Department of Surgery, Aberdeen Royal Infirmary Foresterhill, Aberdeen, UK
| | - Silvia W. Gratz
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Karen P. Scott
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
3
|
Hijová E, Bertková I, Štofilová J. Incorporating Postbiotics into Intervention for Managing Obesity. Int J Mol Sci 2025; 26:5362. [PMID: 40508171 PMCID: PMC12154039 DOI: 10.3390/ijms26115362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/20/2025] [Accepted: 05/31/2025] [Indexed: 06/16/2025] Open
Abstract
Obesity is reaching global epidemic proportions worldwide, posing a significant burden on individual health and society. Altered gut microbiota is considered a key factor in the pathogenesis of many diseases, producing metabolites that contribute to the health-beneficial properties of postbiotics. Postbiotics, bioactive microbial components derived from probiotics, are emerging as a valuable strategy in modern medicine and a promising alternative for managing obesity without the need for live bacteria. This work provides a comprehensive overview of the potential health benefits of postbiotics, particularly in relation to obesity, which represents an important health challenge. Despite the encouraging insights into the health benefits of postbiotics, we highlight the need for further research to clarify the mechanisms and the specific roles of different postbiotic components. Integrating postbiotics into health interventions has the potential to enhance preventive care and significantly improve health outcomes in at-risk populations.
Collapse
Affiliation(s)
- Emília Hijová
- Center of Clinical and Preclinical Research-MediPark, Faculty of Medicine, Pavol Jozef Šafárik University, SNP 1, 040 11 Košice, Slovakia; (I.B.); (J.Š.)
| | | | | |
Collapse
|
4
|
Zhao M, Zhou L, Wang S. Immune crosstalk between respiratory and intestinal mucosal tissues in respiratory infections. Mucosal Immunol 2025; 18:501-508. [PMID: 39755173 DOI: 10.1016/j.mucimm.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/11/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
Mucosal tissues, including those in the respiratory and gastrointestinal tracts, are critical barrier surfaces for pathogen invasion. Infections at these sites not only trigger local immune response, but also recruit immune cells from other tissues. Emerging evidence in the mouse models and human samples indicates that the immune crosstalk between the lung and gut critically impacts and determines the course of respiratory disease. Here we summarize the current knowledge of the immune crosstalk between the respiratory and gastrointestinal tracts, and discuss how immune cells are recruited and migrate between these tissues during respiratory infections. We also discuss how commensal bacteria contribute to these processes.
Collapse
Affiliation(s)
- Min Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lei Zhou
- Shanghai Immune Therapy Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China.
| |
Collapse
|
5
|
Willis NB, Cannavale CN, Walk AM, Burd NA, Holscher HD, Khan NA. Inhibitory control is related to fecal short-chain fatty acid concentrations in adults with overweight and obesity. Nutr Res 2025; 138:12-21. [PMID: 40280069 DOI: 10.1016/j.nutres.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/29/2025]
Abstract
Obesity is a pro-inflammatory condition with negative effects on executive functioning. Increased inflammation dysregulates gastrointestinal homeostasis and alters microbiota community composition. The gut microbiota produce immunomodulatory short-chain fatty acids (SCFA) that have been related to cognition in obesity, but the neural effects are not explored. Here, we hypothesized that greater fecal SCFA would be positively related to neuroelectric markers of inhibitory control and conflict monitoring in obesity. A cross-sectional cohort of 87 adults (35 ± 6 years, 53 females) with overweight and obesity (BMI = 32 ± 6 kg/m2) provided fresh fecal samples and participated in cognitive testing to assess response inhibition and conflict monitoring with electroencephalographic recording. Linear regressions, controlling for age, sex, BMI, and energy-adjusted dietary fiber intake, revealed positive relationships between NoGo N2 mean amplitude and fecal SCFA concentrations. Linear discriminant analysis effect size (LEfSe) revealed 16 amplicon sequence variants differentially abundant between high and low butyrate groups with Roseburia and Adlercreutzia individually related to NoGo N2 mean amplitude in MaAsLin2 modeling. Thus, greater fecal SCFA concentrations and SCFA producing microbiota (i.e., Roseburia) were related to markers of superior conflict monitoring in the NoGo task when adjusting for key covariates. These data highlight key associations between bacterial derived gut signaling molecules and neural regulation in cognitive domains particularly relevant to weight status that warrant further investigation.
Collapse
Affiliation(s)
- Nathaniel B Willis
- Division of Nutrition Sciences, University of Illinois Urbana-Champaign, Urbana, IL USA
| | - Corinne N Cannavale
- Department of Health and Kinesiology, University of Illinois Urbana-Champaign, Urbana, IL USA
| | - Anne M Walk
- Department of Psychology, Eastern Illinois University, Charleston, IL USA
| | - Nicholas A Burd
- Department of Health and Kinesiology, University of Illinois Urbana-Champaign, Urbana, IL USA
| | - Hannah D Holscher
- Division of Nutrition Sciences, University of Illinois Urbana-Champaign, Urbana, IL USA; Department of Health and Kinesiology, University of Illinois Urbana-Champaign, Urbana, IL USA; Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL USA
| | - Naiman A Khan
- Division of Nutrition Sciences, University of Illinois Urbana-Champaign, Urbana, IL USA; Department of Health and Kinesiology, University of Illinois Urbana-Champaign, Urbana, IL USA; Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL USA.
| |
Collapse
|
6
|
González A, Fullaondo A, Odriozola A. In Search of Healthy Ageing: A Microbiome-Based Precision Nutrition Approach for Type 2 Diabetes Prevention. Nutrients 2025; 17:1877. [PMID: 40507144 PMCID: PMC12158179 DOI: 10.3390/nu17111877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Revised: 05/23/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025] Open
Abstract
Background/Objectives: Type 2 diabetes (T2D) is a leading cause of morbidity and mortality worldwide and in Spain, particularly in the elderly population, affecting healthy ageing. Nutritional strategies are key to its prevention. The gut microbiota is also implicated in T2D and can be modulated by nutrition. We hypothesize that precision nutrition through microbiota modulation may help prevent T2D. This article aims to (1) describe a gut microbiota bacterial profile associated with T2D prevention, (2) provide precision nutrition tools to optimize this profile, (3) analyze how overweight influences the microbiota composition and precision nutrition response, and (4) address the technical challenges of microbiome-based precision nutrition clinical implementation to prevent T2D. Methods: A review of gut microbiota associated with T2D prevention was conducted. 13 healthy Spanish participants over 62 with optimal blood glucose levels (7 normal weight and 6 overweight) underwent a 3-month precision nutrition intervention to optimize T2D-preventive gut microbiota using a bioinformatics food recommendation system, Phymofood (EP22382095). Fecal microbiota was analyzed pre- and post-intervention using full-length 16S rRNA gene amplification, MinION sequencing, and NCBI taxonomic classification. Results: 31 potentially preventive bacteria against T2D were selected. The intervention increased the relative abundance of beneficial genera (Butyrivibrio and Faecalibacterium) and species (Eshraghiella crossota, and Faecalibacterium prausnitzii). The overweight influenced microbiota composition and intervention response. Conclusions: A gut microbiota profile associated with T2D prevention was identified, and precision nutrition could increase the relative abundance of beneficial bacteria. Confounding factors such as overweight should be considered when designing microbiome-based precision nutrition interventions. These results contribute to a better understanding of the microbiota associated with T2D prevention and address technical challenges for clinical implementation in future healthy ageing strategies.
Collapse
Affiliation(s)
| | | | - Adrian Odriozola
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), 48940 Bilbao, Spain; (A.G.); (A.F.)
| |
Collapse
|
7
|
Zhang J, Zheng W, Zhou Z, An Y, Zheng H, Zhang Y, Wei Y, Zhang Q, Zheng J, Wang F. Aqueous Polygalae Radix extract (PRE) prolongs the lifespan of C. elegans and alleviates D-galactose-induced oxidative stress in the mouse liver and brain by modulating PPARγ/MAPK. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119878. [PMID: 40287113 DOI: 10.1016/j.jep.2025.119878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Polygalae Radix has high pharmacological activity and has been widely used as a sedative and tranquilizer to increase cognitive function, prevent epilepsy, and treat respiratory diseases such as bronchitis. However, its role in delaying aging and decreasing oxidative stress and its main functional factors have not been thoroughly studied. AIM OF THE STUDY The purpose was to investigate the antiaging and antioxidant effects of aqueous Polygalae Radix extract (PRE) and its mechanism of action. MATERIALS AND METHODS The effects of different concentrations (1, 5, and 10 mg/mL) of PRE on the lifespan, body length, reproductive ability, motility, lipofuscin, and reactive oxygen species (ROS) on a model of natural senescence of Caenorhabditis elegans were investigated. The effects of PRE treatment on the expression of body weight, malondialdehyde (MDA), glutathione peroxidase (GSH-PX), and mTERT content were evaluated in a D-galactose (Dgal)-induced mouse model of aging. Histopathological changes in the liver and brain of mice were analyzed by hematoxylin-eosin (HE). The enriched pathways associated with differentially expressed genes in the liver tissues of C. elegans and mice were analyzed via RNA-seq, and the results were verified via RT-qPCR, cell transfection and Western blotting. Abundance of and changes in the mouse intestinal flora were analyzed by 16S rDNA sequencing. RESULTS PRE significantly prolonged the average lifespan of C. elegans and improved the physiological indices related to senescence. In addition, PRE slowed the decrease in weight of senescent model mice; protected serum, liver and brain tissues from oxidative stress damage; increased GSH-PX expression; and reduced MDA expression. The role of PRE in the low- and middle-dose groups was similar to that of vitamin C (VC) in inhibiting oxidative stress, but the effect of PRE in the high-dose group was greater than that of VC. The RNA-seq results suggested that PRE might be related to PPARγ/MAPK, and the subsequent RT-qPCR and cell transfection results indicated that PRE decreased oxidative stress by downregulating the mRNA expression of the Fabp1, Acaa1b, Hmgcs1, Map3K5, and Rac2 genes. The Western blot results revealed that PRE decreased oxidative stress by increasing PPARγ expression and inhibiting p38 protein phosphorylation. 16S rDNA sequencing showed that PRE treatment increased the abundance of the intestinal flora in mice and inhibited the growth of pathogenic bacteria such as Helicobacter pylori and Desulfurization vibrio while promoting the growth of beneficial bacteria such as Bifidobacteria. CONCLUSIONS PRE delays aging and resists oxidative stress in organisms; it may act by regulating the PPARγ/MAPK signaling pathway and the intestinal flora. The efficacy and mechanism of PRE against oxidative stress were elucidated using RNA-seq and 16S rDNA sequencing, providing a reference for antiaging and aging-related diseases.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Wenxue Zheng
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Zhengjie Zhou
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - YiMing An
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Haoyu Zheng
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Yanan Zhang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Yuchen Wei
- The First Bethune Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Qianhua Zhang
- The First Bethune Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Jingtong Zheng
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
| | - Fang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
| |
Collapse
|
8
|
Cao C, Li S, Wang W, Shi L, Ma R, Zhang B, Tian J. Intestinal microbiota improves inflammation and cognitive function in the brain of a7nAChR deficient rat through the gut brain axis. Sci Rep 2025; 15:18381. [PMID: 40419554 DOI: 10.1038/s41598-025-02627-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
To investigate the role of intestinal flora and cholinergic anti-inflammatory pathways in the gut-brain axis, using oral gavage and intraperitoneal injection of methyllycaconitine (MLA). MLA was administered at a dose of 4 mg/kg for 30 days, either orally or via intraperitoneal injection. Rats were then assessed for behavioral changes, inflammatory markers, neurotransmitters, neuroreceptors, and intestinal mucosal barrier integrity. Rats receiving MLA via intraperitoneal injection exhibited significant behavioral abnormalities compared to the control and orally administered MLA groups. The levels of IL-1β were elevated in both intestinal and hippocampal tissues, while IL-10 levels were decreased. Brain-derived neurotrophic factor (BDNF) was significantly lower in hippocampal tissues. Furthermore, α7nAChR expression was reduced in hippocampal tissues, accompanied by an increase in 5-HT3A receptors. The intestinal mucosal barrier was compromised, as evidenced by reduced expression of ZO-1 and Occludin, along with increased IL-1β and decreased IL-10 levels in the gut. Our findings suggest that oral gavage of MLA does not induce cognitive impairment in rats compared to intraperitoneal injection, possibly due to the involvement of intestinal flora in the protective effects of CAP.
Collapse
Affiliation(s)
- Chi Cao
- the General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Shulin Li
- The 942 Hospital of the Joint Logistics Support Force of the Chinese People's Liberation Army, Lanzhou, China
| | - Wencheng Wang
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, China
| | - Lei Shi
- the General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Rui Ma
- The First People's Hospital of Yinchuan, Yinchuan, China
| | - Bin Zhang
- the General Hospital of Ningxia Medical University, Yinchuan, 750004, China.
| | - Jianying Tian
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
9
|
Krauze W, Busz N, Pikuła W, Maternowska M, Prowans P, Maciejewska-Markiewicz D. Effect of Sodium Butyrate Supplementation on Type 2 Diabetes-Literature Review. Nutrients 2025; 17:1753. [PMID: 40507022 PMCID: PMC12156991 DOI: 10.3390/nu17111753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2025] [Revised: 05/16/2025] [Accepted: 05/20/2025] [Indexed: 06/16/2025] Open
Abstract
Background: Type 2 diabetes mellitus (T2DM) represents a major global health burden, with prevalence rates escalating due to rapid urbanization, economic growth, and the obesity epidemic. Despite intensive research, the underlying molecular mechanisms remain incompletely understood, with emerging evidence suggesting multifactorial origins involving genetic, epigenetic, lifestyle, and environmental factors. Methods: This review synthesizes current epidemiological data on T2DM prevalence, risk factors, and demographic patterns from 1990 to 2017, and discusses projected trends through 2030. We examine the role of intestinal barrier dysfunction and gut microbiota dysbiosis in T2DM pathogenesis, highlighting key mechanistic insights. Furthermore, we analyze recent findings on the role of butyrate, a major short-chain fatty acid, in preserving gut integrity and its potential therapeutic effects on metabolic health. Results: Global T2DM prevalence has risen markedly across all age groups, with particularly high rates in Western Europe and Pacific Island nations. Disruption of the intestinal barrier ("leaky gut") and gut microbiota alterations contribute significantly to systemic inflammation and insulin resistance, which are pivotal features in T2DM development. Butyrate plays a central role in maintaining epithelial barrier function, modulating immune responses, and regulating glucose metabolism. Preclinical studies have demonstrated that sodium butyrate supplementation improves gut integrity, reduces systemic endotoxemia, and ameliorates metabolic parameters. Emerging clinical evidence suggests benefits of sodium butyrate, particularly when combined with prebiotic fibers, in improving glycemic control and reducing inflammatory markers in T2DM patients. Conclusions: Gut barrier integrity and microbiota composition are critical factors in T2DM pathogenesis. Sodium butyrate shows promise as a complementary therapeutic agent in T2DM management, although further large-scale, long-term clinical trials are required to confirm its efficacy and safety. Targeting gut health may represent a novel strategy for the prevention and treatment of T2DM.
Collapse
Affiliation(s)
- Wiktoria Krauze
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (W.K.); (N.B.); (W.P.); (M.M.)
| | - Nikola Busz
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (W.K.); (N.B.); (W.P.); (M.M.)
| | - Weronika Pikuła
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (W.K.); (N.B.); (W.P.); (M.M.)
| | - Martyna Maternowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (W.K.); (N.B.); (W.P.); (M.M.)
| | - Piotr Prowans
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland;
| | - Dominika Maciejewska-Markiewicz
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (W.K.); (N.B.); (W.P.); (M.M.)
| |
Collapse
|
10
|
Liu Y, Cheng YY, Thompson J, Zhou Z, Vivas EI, Warren MF, DuClos JM, Anantharaman K, Rey FE, Venturelli OS. Decoding the role of the arginine dihydrolase pathway in shaping human gut community assembly and health-relevant metabolites. Cell Syst 2025; 16:101292. [PMID: 40339579 DOI: 10.1016/j.cels.2025.101292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 12/10/2024] [Accepted: 04/11/2025] [Indexed: 05/10/2025]
Abstract
The arginine dihydrolase pathway (arc operon) provides a metabolic niche by transforming arginine into metabolic byproducts. We investigate the role of the arc operon in probiotic Escherichia coli Nissle 1917 on human gut community assembly and health-relevant metabolite profiles. By stabilizing environmental pH, the arc operon reduces variability in community composition in response to pH perturbations and frequently enhances butyrate production in synthetic communities. We use a tailored machine learning model for microbiomes to predict community assembly in response to variation in initial media pH and arc operon activity. This model uncovers the pH- and arc operon-dependent interactions shaping community assembly. Human gut species display altered colonization dynamics in response to the arc operon in the murine gut. In sum, our framework to quantify the contribution of a specific pathway to microbial community assembly and metabolite production can reveal new engineering strategies. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Yiyi Liu
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yu-Yu Cheng
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jaron Thompson
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Zhichao Zhou
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Eugenio I Vivas
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA; Gnotobiotic Animal Core Facility, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Matthew F Warren
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Julie M DuClos
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Karthik Anantharaman
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ophelia S Venturelli
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
11
|
Throat S, Bhattacharya S. The Role of RS Type 2 (High-Amylose Maize Starch) in the Inhibition of Colon Cancer: A Comprehensive Review of Short-Chain Fatty Acid (SCFA) Production and Anticancer Mechanisms. Mol Nutr Food Res 2025:e70107. [PMID: 40392033 DOI: 10.1002/mnfr.70107] [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: 02/02/2025] [Revised: 03/30/2025] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
Dietary fiber, especially resistant starch (RS) Type 2 (RS2) found in high-amylose maize starch (HAMS), is vital for gut health and helps prevent colon cancer. In contrast to most nutrients, dietary fiber is not degraded by the intestinal enzymes; it reaches the distal parts of the gut, where it is fermented by the gut microbiota into short-chain fatty acids (SCFAs), such as acetate, propionate, and butyrate. SCFAs energize colonocytes, reduce inflammation, and enhance gut immunity. HAMS is absorbed in the colon, where it ferments to create SCFAs that feed good gut flora and have antiinflammatory and antiproliferative effects. RS2 in HAMS modulates gene signaling, activates tumor-suppressor genes like tumor suppressor protein (p53), exhibits antidiabetic, cholesterol-lowering, and antiinflammatory effects. Incorporation of RS2-rich sources enhances gut barriers, decreases colorectal cancer biomarkers, and counteracts the negative impacts of low-fiber Western diets, making HAMS a promising functional food for chronic disease prevention and health promotion.
Collapse
Affiliation(s)
- Siddhi Throat
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, India
| | - Sankha Bhattacharya
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, India
| |
Collapse
|
12
|
Limketkai BN, Godoy-Brewer G, Shah ND, Maas L, White J, Parian AM, Mullin GE. Prebiotics for Induction and Maintenance of Remission in Inflammatory Bowel Disease: Systematic Review and Meta-Analysis. Inflamm Bowel Dis 2025; 31:1220-1230. [PMID: 38781004 DOI: 10.1093/ibd/izae115] [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: 02/15/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Prebiotics are nondigestible carbohydrates fermented by gut bacteria into metabolites that confer health benefits. However, evidence on their role for inflammatory bowel disease (IBD) is unclear. This study systematically evaluated the research on prebiotics for treatment of IBD. METHODS A search was performed in PubMed, Embase, Cochrane, and Web of Science. Eligible articles included randomized controlled trials or prospective observational studies that compared a prebiotic with a placebo or lower-dose control in patients with IBD. Meta-analyses were performed using random-effects models for the outcomes of clinical remission, clinical relapse, and adverse events. RESULTS Seventeen studies were included. For induction of clinical remission in ulcerative colitis (UC), the fructooligosaccharide (FOS) kestose was effective (relative risk, 2.75, 95% confidence interval, 1.05-7.20; n = 40), but oligofructose-enriched inulin (OF-IN) and lactulose were not. For maintenance of remission in UC, germinated barley foodstuff trended toward preventing clinical relapse (relative risk, 0.40; 95% confidence interval, 0.15-1.03; n = 59), but OF-IN, oat bran, and Plantago ovata did not. For Crohn's disease, OF-IN and lactulose were no different than controls for induction of remission, and FOS was no different than controls for maintenance of remission. Flatulence and bloating were more common with OF-IN; reported adverse events were otherwise similar to controls for other prebiotics. CONCLUSION Prebiotics, particularly FOS and germinated barley foodstuff, show potential as effective and safe dietary supplements for induction and maintenance of remission in UC, respectively. The overall certainty of evidence was very low. There would be benefit in further investigation on the role of prebiotics as treatment adjuncts for IBD.
Collapse
Affiliation(s)
- Berkeley N Limketkai
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Division of Clinical Nutrition, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Gala Godoy-Brewer
- Department of Medicine, University of Miami School of Medicine, Miami, FL, USA
| | - Neha D Shah
- Colitis and Crohn's Disease Center, Division of Gastroenterology, UCSF School of Medicine, San Francisco, CA, USA
| | - Laura Maas
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacob White
- Welch Library, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alyssa M Parian
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gerard E Mullin
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
13
|
Inokuma K, Sasaki D, Shintani T, Inoue J, Oyama K, Noda Y, Maeda T, Yamada R, Matsuki Y, Kodama Y, Kondo A. Combination of probiotics enhancing butyrogenesis in colonic microbiota model of patients with ulcerative colitis. Appl Microbiol Biotechnol 2025; 109:117. [PMID: 40347262 PMCID: PMC12065738 DOI: 10.1007/s00253-025-13424-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 05/12/2025]
Abstract
Administering beneficial bacteria as probiotics to restore the intestinal microbiota and its metabolic functions, such as butyrogenesis, is a promising treatment strategy in ulcerative colitis (UC). This study aimed to investigate the effect of a combination of probiotics, consisting of the lactic acid bacterium Weizmannia coagulans SANK70258 and the lactate-utilizing butyrate-producing bacteria Anaerostipes caccae or Clostridium butyricum, on the colonic environment using an in vitro colonic microbiota culture model with fecal inoculums from seven patients with UC. Co-inoculated W. coagulans and A. caccae neither inhibited each other's growth nor significantly affected the relative abundance of other bacterial species; however, the growth of W. coagulans was significantly inhibited when co-inoculated with C. butyricum. The relative abundance of pro-inflammatory bacteria (Escherichia sp. and unclassified Enterobacteriaceae) and Bifidobacterium spp. significantly decreased in W. coagulans-C. butyricum co-inoculated cultures. Inoculation with any of the probiotics alone did not increase butyrate production, whereas co-inoculation of W. coagulans with A. caccae or C. butyricum significantly increased the butyrate levels. Overall, the results suggested that W. coagulans and lactate-utilizing butyrate-producing bacteria in combination have synergistic effects through cross-feeding and can effectively restore butyrogenesis in the colonic environment of patients with UC. KEY POINTS: • Effects of probiotics were evaluated using in vitro microbiota model of UC colon. • W. coagulans and lactate-utilizing butyrate producers have synergistic effects. • Co-inoculation of W. coagulans with A. caccae or C. butyricum enhanced butyrogenesis.
Collapse
Affiliation(s)
- Kentaro Inokuma
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-Cho, Nada-Ku, Kobe, 657-8501, Japan
| | - Daisuke Sasaki
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-Cho, Nada-Ku, Kobe, 657-8501, Japan
| | - Tomoya Shintani
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-Cho, Nada-Ku, Kobe, 657-8501, Japan
| | - Jun Inoue
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Katsuaki Oyama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Yuta Noda
- Science & Innovation Center, Mitsubishi Chemical Corporation, Yokohama, Kanagawa, 227-8502, Japan
| | - Takayuki Maeda
- Science & Innovation Center, Mitsubishi Chemical Corporation, Yokohama, Kanagawa, 227-8502, Japan
| | - Ryouichi Yamada
- Science & Innovation Center, Mitsubishi Chemical Corporation, Yokohama, Kanagawa, 227-8502, Japan
| | - Yasushi Matsuki
- Strategic Planning Office, Kobe University, 1-1 Rokkodai-Cho, Nada-Ku, Kobe, 657-8501, Japan
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Akihiko Kondo
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-Cho, Nada-Ku, Kobe, 657-8501, Japan.
- Biomass Engineering Program, RIKEN, 1-7-22 Suehiro-Cho, Tsurumi-Ku, Yokohama, Kanagawa, 230-0045, Japan.
| |
Collapse
|
14
|
Gao F, Wang F, Wang D, Du G, Gao F. Bibliometric analysis of the S24-7 family and its association with health. Front Microbiol 2025; 16:1571883. [PMID: 40406341 PMCID: PMC12095373 DOI: 10.3389/fmicb.2025.1571883] [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: 02/06/2025] [Accepted: 04/22/2025] [Indexed: 05/26/2025] Open
Abstract
A burgeoning corpus of evidence indicates that S24-7 is integral to human health, with links to obesity, inflammation, metabolism, and dietary interactions. In the present study, we conducted a comprehensive review of the S24-7 literature from the past 10 years, augmented by an evaluation of research trends using both quantitative and qualitative approaches. From the Web of Science (WoS) database, we retrieved 903 research articles and four review articles pertaining to S24-7, also known as Muribaculaceae, that were published between January 1, 2014, and January 1, 2024. Employing software tools such as R, Biblioshiny, VOSviewer, the Bibliometric Analysis Platform, and Pajek, we performed visual mapping and correlation analyses on the collected documents. Our analysis revealed China and the United States as the leading publishers in the field of S24-7 research. The top three academic journals for S24-7 family research are Food and Function, Frontiers in Microbiology, and Nutrition. Among individual contributors, Zhang Y stands out with 31 publications and an h-index of 13, representing 3.42% of the 907 articles analyzed. Jiangnan University leads in institutional output with 46 publications. Keyword analysis underscores that S24-7 research is concentrated on examining the family's associations with obesity, inflammation, metabolism, and diet. This study highlights notable contributions from various countries, institutions, journals, and researchers, shedding light on the influence of the S24-7 family on human health. It serves to inform future research directions and clinical applications concerning the S24-7 flora.
Collapse
Affiliation(s)
- Fangmei Gao
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
- The Second People’s Hospital of Dongfang City, Dongfang, China
| | - Fenfen Wang
- Qunying Health Center of Lingshui Li Autonomous County, Lingshui, China
| | - Dandan Wang
- Yongming Health Center of Ledong Li Autonomous County, Ledong, China
| | - Guankui Du
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
- Biotechnology and Biochemistry Laboratory, Hainan Medical University, Haikou, China
- Department of Biochemistry and Molecular Biology, Hainan Medical University, Haikou, China
| | - Fangfang Gao
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| |
Collapse
|
15
|
He H, Liu F, Yang L, Fan H, Wan H. Structure changes of starch complexed with green tea catechin or lemon peel caffeic acid under thermomechanical-assist low moisture and their prebiotics during in vitro digestion and fermentation. Int J Biol Macromol 2025; 308:142676. [PMID: 40164251 DOI: 10.1016/j.ijbiomac.2025.142676] [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: 11/19/2024] [Revised: 03/02/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
This paper highlighted the structural changes of rice starch complexed with green tea catechin and lemon peel caffeic acid under thermomechanical-assist low moisture, and their prebiotics during in vitro digestibility and fermentation were further explained. Extruded starch with caffeic acid, characterized by enhanced short-range order, a distinct fractal structure, and a V-amylose helical arrangement, exhibited slower glucose release due to V-type inclusion crystalline structures and strong hydrogen bondings. Additionally, extruded starch with catechin or caffeic acid changed the composition of gut microbiota by increasing the proliferation of good bacteria and reducing pathogenic microorganisms, which led to a greater synthesis of short-chain fatty acids. According to a PICRUSt2 analysis, the extruded starch with caffeic acid may trigger metabolic alterations via altering the gut microbiota and increasing bile acid metabolism. Thus, extruded starch with caffeic acid demonstrates significant potential as a prebiotic for developing nutritionally tailored starch-based foods.
Collapse
Affiliation(s)
- Hai He
- Heinz Mehlhorn Academician Workstation, Key Laboratory of Tropical Translational Medicine of Ministry of Education, International Collaborative Research Center for the Development and Utilization of Tropical Food for Special Medical Purpose, School of Public Health, Hainan Medical University, Haikou 571199, Hainan Province, China; Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528300, Guangdong Province, China.
| | - Fanrui Liu
- Heinz Mehlhorn Academician Workstation, Key Laboratory of Tropical Translational Medicine of Ministry of Education, International Collaborative Research Center for the Development and Utilization of Tropical Food for Special Medical Purpose, School of Public Health, Hainan Medical University, Haikou 571199, Hainan Province, China
| | - Lei Yang
- Department of Dermatology, Characteristic Medical Centre of PLA Rocket Force, Beijing 100001, China.
| | - Honghao Fan
- NJUST-YX Artificial Intelligence Biomedical Technology Innovation Center, Nanjing University of Science and Technology, Nanjing 210094, Jiangsu Province, China
| | - Hao Wan
- Department of Medical Laboratory, Qianjiang Central Hospital, Qianjiang 433100, Hubei Province, China
| |
Collapse
|
16
|
Shi Y, Jiang M, Zhu W, Chang K, Cheng X, Bao H, Peng Z, Hu Y, Li C, Fang F, Song J, Jian C, Chen J, Shu X. Cyclosporine combined with dexamethasone regulates hepatic Abca1 and PPARα expression and lipid metabolism via butyrate derived from the gut microbiota. Biomed Pharmacother 2025; 186:118017. [PMID: 40168721 DOI: 10.1016/j.biopha.2025.118017] [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: 01/18/2025] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/03/2025] Open
Abstract
Immunosuppression often leads to drastic metabolic, hormonal, and physiological disorders. Changes in the gut microbiota are believed to be one of the factors contributing to these disorders, but the association remains uncertain. Clinical studies can be complicated by confounding variables, such as diet and other drivers of heterogeneity in human microbiomes. In this study, we identified pronounced gut microbiome signatures in rhesus macaques (RMs) with immunosuppression-induced lipid metabolism disorders following cyclosporine combined with dexamethasone. Furthermore, we observed similar changes in the gut microbiota of mice with immunosuppression-induced lipid metabolism disorders, which were associated with short-chain fatty acid metabolism. ELISA showed that immunosuppression significantly reduced the levels of butyric acid in both feces and serum of mice. Spearman correlation analysis identified a significant correlation between serum butyric acid levels and gut microbial dysbiosis induced by immunosuppression, particularly in relation to f_Lachnospiraceae, g_unidentified_Ruminococcaceae, and s_Clostridium leptum. Additionally, mice transplanted with gut microbiota from immunosuppressed mice exhibited hepatic lipid metabolism disorders, and RNA sequencing revealed significant downregulation of ABC transporters and PPARα in the liver, which was closely associated with lipid transport and metabolism, particularly Abca1. Moreover, butyric acid supplementation alleviated hepatic lipid metabolism disorders and upregulated the expression of Abca1 and PPARα in mice transplanted with immunosuppression-induced gut microbiota. Thus, we propose that the combination of cyclosporine and dexamethasone regulates the expression of hepatic Abca1 and PPARα by modulating the gut microbiota and its derived butyrate, particularly Lachnospiraceae and Clostridium leptum, further regulating hepatic lipid metabolism.
Collapse
Affiliation(s)
- Yongping Shi
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Mi Jiang
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Wenzhong Zhu
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Ke Chang
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xukai Cheng
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Haijun Bao
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Zuojie Peng
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Yuan Hu
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Chao Li
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Feifei Fang
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jia Song
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Chenxing Jian
- Department of anorectal surgery, Affiliated hospital of Putian University, China
| | - Jinhuang Chen
- Department of emergency surgery, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xiaogang Shu
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
17
|
Sessa L, Malavolta E, Sodero G, Cipolla C, Rigante D. The conspiring role of gut microbiota as primer of autoimmune thyroid diseases: A scoping focus. Autoimmun Rev 2025; 24:103780. [PMID: 39971108 DOI: 10.1016/j.autrev.2025.103780] [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: 12/19/2024] [Revised: 01/31/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
The thyroid gland is the body's largest single organ specialized for endocrine hormone production, and still unraveled mechanisms regulate its interaction between the hypothalamic-pituitary-thyroid axis and composition of the gut microbiota: in particular, a disrupted integrity of the intestinal barrier, causing dysbiosis and increasing detrimental substances or reducing beneficial metabolites, such as short-chain fatty acids (SCFAs) with proinflammatory effects, may be crucial for the induction of an autoimmune thyroid disease. More specifically, Lactobacilli and Bifidobacteria have a role in this partnership through a "molecular mimicry" mechanism, as their protein sequences share structural similarity with thyroid peroxidase and thyroglobulin. Lactobacilli can also increase T helper 17 cells, modifying the number of colonic regulatory T cells, largely implicated in the maintenance of immunological tolerance at the gut barrier. Additionally, Blautia and Anaerostipes work beneficially with butyric acid, one of the SCFAs, promoting antimicrobial peptide synthesis from the intestinal cells and bolstering the innate immune system's ability to struggle against pathogens, which can also influence thyroid hormone levels by regulating iodine uptake and metabolism. This review aims to summarize the current knowledge about the contribution of gut microbiota changes in triggering immune abnormalities leading to autoimmune thyroid diseases.
Collapse
Affiliation(s)
- Linda Sessa
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elena Malavolta
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giorgio Sodero
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Clelia Cipolla
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Donato Rigante
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Università Cattolica Sacro Cuore, Rome, Italy.
| |
Collapse
|
18
|
Yi K, Huang Y, Jiang Y, Zhou L. Causal relationship between gut microbiota and laryngeal cancer: a mendelian randomization analysis. Braz J Otorhinolaryngol 2025; 91:101634. [PMID: 40305979 PMCID: PMC12118545 DOI: 10.1016/j.bjorl.2025.101634] [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: 10/23/2024] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
OBJECTIVE Laryngeal cancer incidence is rising globally; the role of gut microbiota remains underexplored. This study aimed to establish a causal link between gut microbiota and laryngeal cancer to inform preventive and therapeutic strategies. METHODS Gut microbiota data from GWAS conducted by the MiBioGen consortium served as the exposure variable, with laryngeal cancer as the outcome variable. the exposure variable and the outcome variable were analyzed using Mendelian Randomization. The primary method was Inverse Variance Weighted analysis, with heterogeneity and pleiotropy assessed through Cochran's Q test, MR-Egger regression, and MR-PRESSO. RESULTS In the study, we identified five bacterial taxa with potential causal relationships with laryngeal cancer risk: Higher levels of Clostridiaceae1 (OR = 0.9993, 95% CI 0.9986-0.9999, p = 0.0463) and Turicibacter (OR = 0.9995, 95% CI 0.9989-0.9999, p = 0.0384) were linked to reduced cancer risk, while Mollicutes RF9 (OR = 1.0010, 95% CI 1.0003-1.0016, p = 0.0027), Euryarchaeota (OR = 1.0004, 95% CI 1.0001-1.0007, p = 0.0234), and Cyanobacteria (OR = 1.0005, 95% CI 1.0000-1.0009, p = 0.0464) were associated with increased risk. CONCLUSION Our findings suggest a causal relationship between gut microbiota composition and laryngeal cancer risk. Clostridiaceae1 and Turicibacter may play a protective role, while Mollicutes RF9, Euryarchaeota, and Cyanobacteria could contribute to increased cancer susceptibility. These insights highlight potential microbiome-based strategies for early detection, prevention, and therapeutic intervention in laryngeal cancer. LEVEL OF EVIDENCE Level 5.
Collapse
Affiliation(s)
- Kaiyan Yi
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Yu Huang
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Yun Jiang
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Lingling Zhou
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
19
|
Rampelotto PH, Taufer CR, da Silva J. The Role of Beneficial Microbiota in COVID-19: Insights from Key Bacterial Genera. Microorganisms 2025; 13:1029. [PMID: 40431202 PMCID: PMC12113938 DOI: 10.3390/microorganisms13051029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 04/17/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
The COVID-19 pandemic has highlighted the need for a comprehensive understanding of the factors influencing disease severity and progression. Emerging research indicates that the human microbiota, particularly beneficial bacteria, significantly impacts immune responses and health outcomes in COVID-19 patients. While existing studies provide general insights into the relationship between the microbiota and probiotics with COVID-19, they often lack a detailed exploration of how specific bacterial taxa might be used as adjunctive treatments. This review aims to address this gap by focusing on ten key genera of beneficial bacteria, discussing their roles in COVID-19 and evaluating their potential as probiotics for prevention and treatment. The review covers the impact of these microbes on human health, their population alterations in COVID-19 patients, and their interactions with other viral infections. Among these microbes, several exhibit distinct patterns of abundance in COVID-19 patients, influencing disease outcomes and highlighting their potential roles in infection dynamics. In COVID-19 patients, populations of Akkermansia, Ruminococcus, and Roseburia are consistently reduced, while those of Faecalibacterium show a significant decline in more severe cases. Bacteroides presents varying effects depending on the species involved. Alterations in the abundance of Blautia and Lachnospiraceae are associated with increased inflammation and disease severity. Likewise, the depletion of Lachnospira and Coprococcus populations, both linked to anti-inflammatory effects, may exacerbate symptom severity. Oscillospira, though less studied, is connected to overall health and could have implications for viral infections. This review synthesizes the current understanding of these beneficial microbes to highlight the importance of maintaining a healthy microbiota to alleviate the impact of COVID-19 and contribute to the development of novel therapeutic strategies involving microbiota modulation.
Collapse
Affiliation(s)
- Pabulo Henrique Rampelotto
- Bioinformatics and Biostatistics Core Facility, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| | - Clarissa Reginato Taufer
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| | - Juliana da Silva
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
- Graduate Program in Health and Human Development, Universidade La Salle, Canoas 92010-000, Brazil
| |
Collapse
|
20
|
Ren Y, He X, Wang L, Chen N. Comparison of the gut microbiota in older people with and without sarcopenia: a systematic review and meta-analysis. Front Cell Infect Microbiol 2025; 15:1480293. [PMID: 40357398 PMCID: PMC12066693 DOI: 10.3389/fcimb.2025.1480293] [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: 08/13/2024] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Sarcopenia, an age-related disorder marked by decreased skeletal muscle mass, strength, and function, is associated with negative health impacts in individuals and financial burdens on families and society. Studies have suggested that age-related alterations in gut microbiota may contribute to the development of sarcopenia in older people through the gut-muscle axis, thus modulation of gut microbiota may be a promising approach for sarcopenia treatment. However, the characteristic gut microbiota for sarcopenia has not been consistent across studies. Therefore, the aim of this study was to compare the diversity and compositional differences in the gut microbiota of older people with and without sarcopenia, and to identify gut microbiota biomarkers with therapeutic potential for sarcopenia. Methods The PubMed, Embase, Web of Science, Cochrane Library, China National Knowledge Infrastructure, and Wanfang Database were searched studies about the gut microbiota characteristics in older people with sarcopenia. The quality of included articles was assessed by the Newcastle-Ottawa Scale (NOS). Weighted standardized mean differences (SMDs) and 95% confidence intervals (CIs) for α-diversity index were estimated using a random effects model. Qualitative synthesis was conducted for β-diversity and the correlation between gut microbiota and muscle parameters. The relative abundance of the gut microbiota was analyzed quantitatively and qualitatively, respectively. Results Pooled estimates showed that α-diversity was significantly lower in older people with sarcopenia (SMD: -0.41, 95% CI: -0.57 to -0.26, I²: 71%, P < 0.00001). The findings of β-diversity varied across included studies. In addition, our study identified gut microbiota showing a potential and negative correlation with sarcopenia, such as Prevotella, Slackia, Agathobacter, Alloprevotella, Prevotella copri, Prevotellaceae sp., Bacteroides coprophilus, Mitsuokella multacida, Bacteroides massiliensis, Bacteroides coprocola Conversely, a potential and positive correlation was observed with opportunistic pathogens like Escherichia-Shigella, Eggerthella, Eggerthella lenta and Collinsella aerofaciens. Discussion This study showed that α-diversity is decreased in sarcopenia, probably predominantly due to diminished richness rather than evenness. In addition, although findings of β-diversity varied across included studies, the overall trend toward a decrease in SCFAs-producing bacteria and an increase in conditionally pathogenic bacteria. This study provides new ideas for targeting the gut microbiota for the prevention and treatment of sarcopenia. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/view/CRD42024573090, identifier CRD42024573090.
Collapse
Affiliation(s)
- Yanqing Ren
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Xiangfeng He
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Ling Wang
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Nan Chen
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
- Department of Rehabilitation, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
He AQ, Xiao WY, Zheng T, Li KY, Li BS, Wang S, Yu QX, Liu G. Efficacy of curcumin supplementation for the treatment and prevention of pouchitis after ileal pouch-anal anastomosis: a randomized controlled trial. Eur J Nutr 2025; 64:167. [PMID: 40295333 DOI: 10.1007/s00394-025-03676-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/02/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Pouchitis is the most common complication after ileal pouch-anal anastomosis (IPAA) for ulcerative colitis. Induction and maintenance of remission is a crucial therapeutic goal. We investigated curcumin's efficacy in treatment of pouchitis. METHODS The double-blind trial included an induction cohort of refractory pouchitis patients and a maintenance cohort of patients without pouchitis after IPAA. Patients received either placebo or curcumin for 8 weeks. The pouchitis activity were assessed before and after and was compared between cohorts or groups. Laboratory inflammation indicators, nutritional status and quality of life were also appraised. RESULTS 52 patients were included, with 39 and 13 patients entering the maintenance cohort and induction cohort, respectively. In maintenance cohort, the proportion of clinical remission elevated from 11 to 89% in curcumin group (p = 0.005), whereas there was no significant difference in placebo group (10% vs 5%, p = 1).In induction cohort, 67% (4/6) patients achieved clinical response after 8 weeks' intervention of curcumin, whereas none treated with placebo (p = 0.021). Patients treated with curcumin appeared less inflammation and there was no significant difference in indicators changes between two cohorts. CONCLUSIONS Curcumin has preventive and therapeutic effects on pouchitis. Curcumin supplementation can reduce the disease activity and improve the nutritional status of patients with after IPAA. TRIAL REGISTRATION ChiCTR, ChiCTR1900022243. Registered 31 March 2019, https://www.chictr.org.cn/historyversionpub.aspx?regno=ChiCTR1900022243.
Collapse
Affiliation(s)
- An-Qi He
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China
| | - Wan-Yi Xiao
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China
| | - Ting Zheng
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China
| | - Kai-Yu Li
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China
| | - Bao-Song Li
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China
| | - Song Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China
| | - Qing-Xiang Yu
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.
| | - Gang Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China.
| |
Collapse
|
22
|
Maaskant A, Lee D, Ngo H, Montijn RC, Bakker J, Langermans JAM, Levin E. AI for rapid identification of major butyrate-producing bacteria in rhesus macaques (Macaca mulatta). Anim Microbiome 2025; 7:39. [PMID: 40275402 PMCID: PMC12020216 DOI: 10.1186/s42523-025-00410-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND The gut microbiome plays a crucial role in health and disease, influencing digestion, metabolism, and immune function. Traditional microbiome analysis methods are often expensive, time-consuming, and require specialized expertise, limiting their practical application in clinical settings. Evolving artificial intelligence (AI) technologies present opportunities for developing alternative methods. However, the lack of transparency in these technologies limits the ability of clinicians to incorporate AI-driven diagnostic tools into their healthcare systems. The aim of this study was to investigate an AI approach that rapidly predicts different bacterial genera and bacterial groups, specifically butyrate producers, from digital images of fecal smears of rhesus macaques (Macaca mulatta). In addition, to improve transparency, we employed explainability analysis to uncover the image features influencing the model's predictions. RESULTS By integrating fecal image data with corresponding metagenomic sequencing information, the deep learning (DL) and machine learning (ML) algorithms successfully predicted 16 individual bacterial genera (area under the curve (AUC) > 0.7) among the 50 most abundant genera in rhesus macaques (Macaca mulatta). The model was successful in predicting functional groups, major butyrate producers (AUC 0.75) and a mixed group including fermenters and short-chain fatty acid (SCFA) producers (AUC 0.81). For both models of butyrate producers and mixed fermenters, the explainability experiments revealed no decline in the AUC when random noise was added to the images. Increased blurring led to a gradual decline in the AUC. The model's performance was robust against the impact of fecal shape from smearing, with a stable AUC maintained until patch 4 for all groups, as assessed through scrambling. No significant correlation was detected between the prediction probabilities and the total fecal weight used in the smear; r = 0.30 ± 0.3 (p > 0.1) and r = 0.04 ± 0.36 (p > 0.8) for the butyrate producers and mixed fermenters, respectively. CONCLUSION Our approach demonstrated the ability to predict a wide range of clinically relevant microbial genera and microbial groups in the gut microbiome based on digital images from a fecal smear. The models proved to be robust to the smearing method, random noise and the amount of fecal matter. This study introduces a rapid, non-invasive, and cost-effective method for microbiome profiling, with potential applications in veterinary diagnostics.
Collapse
Affiliation(s)
- Annemiek Maaskant
- Biomedical Primate Research Centre, Lange Kleiweg 161, Rijswijk, 2288 GJ, Netherlands.
- Department Population Health Sciences, Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, Utrecht, 3584 CM, Netherlands.
| | - Donghyeok Lee
- HORAIZON Technology BV, Marshallaan 2, Delft, 2625 GZ, Netherlands
| | - Huy Ngo
- HORAIZON Technology BV, Marshallaan 2, Delft, 2625 GZ, Netherlands
| | - Roy C Montijn
- HORAIZON Technology BV, Marshallaan 2, Delft, 2625 GZ, Netherlands
| | - Jaco Bakker
- Biomedical Primate Research Centre, Lange Kleiweg 161, Rijswijk, 2288 GJ, Netherlands
| | - Jan A M Langermans
- Biomedical Primate Research Centre, Lange Kleiweg 161, Rijswijk, 2288 GJ, Netherlands
- Department Population Health Sciences, Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, Utrecht, 3584 CM, Netherlands
| | - Evgeni Levin
- HORAIZON Technology BV, Marshallaan 2, Delft, 2625 GZ, Netherlands.
| |
Collapse
|
23
|
Ren L, Cao Q, Ye H, Dong Z, Zhang C, Yan F, Zhou Y, Zhou H, Zuo J, Wang W. The single degree of polymerization influences the efficacy of xylooligosaccharides in shaping microbial and metabolite profiles in chicken gut to combat avian pathogenic Escherichia coli. BMC Microbiol 2025; 25:227. [PMID: 40264018 PMCID: PMC12013008 DOI: 10.1186/s12866-025-03948-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Avian pathogenic Escherichia coli (APEC) threatens both poultry production and human health. Xylooligosaccharides (XOS) may suppress pathogenic bacteria through prebiotic actions. However, the influences of single degree of polymerization (DP) on the inhibition of APEC by XOS remain unknown. This study aimed to probe if XOS and their major monomers (xylobiose, xylotriose and xylotetraose) could differentially combat APEC via prebiotic actions using an in vitro fermentation model with chicken cecal microbiota. METHODS Microbiota were randomly divided into 7 groups (5 replicate tubes/group). Control group (CON) received no treatment; XOS group received commercial XOS mixtures; APEC group received APEC; XA, X2, X3 and X4 groups received APEC combined with commercial XOS mixtures, xylobiose, xylotriose and xylotetraose, respectively. RESULTS XOS and their major monomers mitigated APEC-induced decline (p < 0.05) in gut microbial α-diversity, with xylotetrose showing the least effect. Gut microbiota in XA, X2, X3 and X4 groups clustered together, with a relative separation observed in X4 group. XOS and their monomers elevated (p < 0.05) the abundances of Firmicutes, Bacteroidota and several probiotics (Lactobacillus, Bacteroides and Megamonas), but reduced (p < 0.05) the abundances of Proteobacteria and Escherichia-Shigella, with xylotetraose exhibiting the least efficacy. Besides, xylotriose and xylotetrose had an advantage over xylotetraose in promoting microbial production of short-chain fatty acids. Metabolomics analysis revealed that APEC challenge mainly downregulated (p < 0.05) several amino acids metabolism pathways of gut microbiota, while xylotriose had an inferiority to XOS in upregulating (p < 0.05) histidine metabolism pathway. Furthermore, microbial fermentation metabolites of all XOS monomers lowered (p < 0.05) certain virulence genes expression in APEC, with xylotriose being the most advantageous. CONCLUSIONS XOS and their major monomers differentially improved gut microbiota and metabolite profiles in chicken gut against APEC challenge. Overall, xylotriose exhibited the greatest inhibition against APEC abundance and virulence. Our findings underscore the role of single DP in influencing the prebiotic actions of XOS against APEC, providing a basis for the reasonable application of XOS in diets to combat bacterial challenge.
Collapse
Affiliation(s)
- Lulu Ren
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qingyun Cao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Hui Ye
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zemin Dong
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Changming Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Fei Yan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yuping Zhou
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Huiyun Zhou
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jianjun Zuo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Weiwei Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
24
|
Deng Y, Zhang Y, Xiao J, Cao Y, Ho CT, Lu M. Allicin Improves Diet-Induced Nonalcoholic Steatohepatitis and Gut Microbiota Dysbiosis in Mice via the Involvement of the Circadian Clock Gene Rev-erbα. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9019-9032. [PMID: 40168418 DOI: 10.1021/acs.jafc.4c12566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Nonalcoholic Steatohepatitis (NASH) is a progressive liver disease characterized by inflammation and liver damage. Allicin, a bioactive compound derived from garlic, has demonstrated anti-inflammatory and antioxidant properties. This study explores the effects of allicin on NASH and gut microbiota dysbiosis induced by a high-fat, high-fructose diet (HFFD) in mice. Allicin supplementation significantly alleviated hepatic inflammation, improved glucose metabolism, and modulated the circadian rhythm gene Rev-erbα, which plays a critical role in regulating inflammation. The anti-inflammatory effects of allicin were diminished in Si-Rev-erbα-treated HepG2 cells, highlighting the importance of circadian regulation in mediating these effects. Allicin's anti-inflammatory effects were associated with increased levels of short-chain fatty acids (SCFAs) and the restoration of diurnal oscillations in proinflammatory cytokines and gut microbiota composition, particularly in genera, such as Akkermansia, Bacteroidetes, and Lactobacillus. These findings suggest that allicin could be a promising therapeutic approach for managing NASH, liver dysfunction, and related metabolic disorders through the modulation of circadian rhythms and the gut microbiome.
Collapse
Affiliation(s)
- Yupei Deng
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yiyi Zhang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Muwen Lu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
25
|
Guilmineau C, Tremblay-Franco M, Vialaneix N, Servien R. Phoenics: a novel statistical approach for longitudinal metabolomic pathway analysis. BMC Bioinformatics 2025; 26:105. [PMID: 40240918 PMCID: PMC12001596 DOI: 10.1186/s12859-025-06118-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] [Received: 12/19/2024] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Metabolomics describes the metabolic profile of an organism at a given time by the concentrations of its constituent metabolites. When studied over time, metabolite concentrations can help understand the dynamical evolution of a biological process. However, metabolites are involved into sequences of chemical reactions, called metabolic pathways, related to a given biological function. Accounting for these pathways into statistical methods for metabolomic data is thus a relevant way to directly express results in terms of biological functions and to increase their interpretability. METHODS We propose a new method, phoenics, to perform differential analysis for longitudinal metabolomic data at the pathway level. In short, phoenics proceeds in two steps: First, the matrix of metabolite quantifications is transformed by a dimension reduction approach accounting for pathway information. Then, a mixed linear model is fitted on the transformed data. RESULTS This method was applied to semi-synthetic NMR data and two real NMR datasets assessing the effects of antibiotics and irritable bowel syndrome on feces. Results showed that phoenics properly controls the Type I error rate and has a better ability to detect differential metabolic pathways and to extract new impacted biological functions than alternative methods. The method is implemented in the R package phoenics available on CRAN.
Collapse
Affiliation(s)
- Camille Guilmineau
- INRAE, University of Montpellier, LBE, 102 Avenue des Etangs, 11100, Narbonne, France.
| | - Marie Tremblay-Franco
- INRAE, Université de Toulouse, ENVT, Toxalim, 31027, Toulouse, France
- Axiom Platform, MetaToul-MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, 31027, Toulouse, France
| | | | - Rémi Servien
- INRAE, University of Montpellier, LBE, 102 Avenue des Etangs, 11100, Narbonne, France
| |
Collapse
|
26
|
Xiao M, Zhou N, Tian Z, Sun C. Endogenous Metabolites in Metabolic Diseases: Pathophysiologic Roles and Therapeutic Implications. J Nutr 2025:S0022-3166(25)00227-5. [PMID: 40250565 DOI: 10.1016/j.tjnut.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
Breakthroughs in metabolomics technology have revealed the direct regulatory role of metabolites in physiology and disease. Recent data have highlighted the bioactive metabolites involved in the etiology and prevention and treatment of metabolic diseases such as obesity, nonalcoholic fatty liver disease, type 2 diabetes mellitus, and atherosclerosis. Numerous studies reveal that endogenous metabolites biosynthesized by host organisms or gut microflora regulate metabolic responses and disorders. Lipids, amino acids, and bile acids, as endogenous metabolic modulators, regulate energy metabolism, insulin sensitivity, and immune response through multiple pathways, such as insulin signaling cascade, chemical modifications, and metabolite-macromolecule interactions. Furthermore, the gut microbial metabolites short-chain fatty acids, as signaling regulators have a variety of beneficial impacts in regulating energy metabolic homeostasis. In this review, we will summarize information about the roles of bioactive metabolites in the pathogenesis of many metabolic diseases. Furthermore, we discuss the potential value of metabolites in the promising preventive and therapeutic perspectives of human metabolic diseases.
Collapse
Affiliation(s)
- Mengjie Xiao
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China
| | - Ning Zhou
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China
| | - Zhen Tian
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China.
| | - Changhao Sun
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China.
| |
Collapse
|
27
|
El Boukhari R, Matin M, Bouissane L, Ławiński M, Lushchak O, Singla RK, Mickael M, Mayneris‐Perxachs J, Grafakou ME, Xu S, Liu B, Guan J, Półtorak A, Szpicer A, Wierzbicka A, Tzvetkov NT, Banach M, Horbańczuk JO, Jóźwik A, Cascella M, Shen B, Pirgozliev VR, Wang D, Litvinova O, Adamska O, Kamińska A, Łapiński M, Stolarczyk A, Berindan‐Neagoe I, Milella L, Yeung AWK, Suravajhala P, Bishayee A, Lordan R, Iantovics LB, Lagoa R, Michalczuk M, Stoyanov J, Kinghorn AD, Jalil B, Weckwerth W, Goh BH, Li M, Chaubey G, Russo GL, Frazzini S, Rossi L, Battino M, Jia W, Su Q, Ma X, Rollinger JM, Rittmann SKR, Sheridan H, Walsh JJ, Lizard G, Karpiński TM, Silva AS, Piwowarski J, Xie L, Fan T, Giampieri F, El Midaoui A, Wong K, Gan R, Fatimi A, Atanasov AG. Enhancing human gut health: Global innovations in dysbiosis management. IMETA 2025. [DOI: 10.1002/imt2.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/26/2025] [Indexed: 05/03/2025]
Affiliation(s)
- Reda El Boukhari
- Chemical Science and Engineering Research Team (ERSIC), Department of Chemistry, Polydisciplinary Faculty of Beni Mellal (FPBM) Sultan Moulay Slimane University (USMS) Beni Mellal Morocco
| | - Maima Matin
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Latifa Bouissane
- Molecular Chemistry, Materials and Catalysis Laboratory, Faculty of Sciences and Technologies Sultan Moulay Slimane University Beni Mellal Morocco
| | - Michał Ławiński
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
- Department of General, Gastroenterologic and Oncologic Surgery Medical University of Warsaw Warsaw Poland
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology Vasyl Stefanyk Precarpathian National University Ivano‐Frankivsk Ukraine
- Research and Development University Ivano‐Frankivsk Ukraine
| | - Rajeev K. Singla
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu Sichuan China
- School of Pharmaceutical Sciences Lovely Professional University Phagwara Punjab India
| | - Michel‐Edwar Mickael
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Jordi Mayneris‐Perxachs
- Department of Diabetes, Endocrinology and Nutrition Dr. Josep Trueta University Hospital Girona Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) Madrid Spain
- Integrative Systems Medicine and Biology Group, Girona Biomedical Research Institute (IDIBGI‐CERCA) Parc Hospitalari Martí i Julià Salt Spain
| | - Maria Eleni Grafakou
- Chair of Pharmaceutical Biology, Faculty of Pharmacy and Chemistry University of Regensburg Germany
| | - Shuhua Xu
- Center for Evolutionary Biology, School of Life Sciences Fudan University Shanghai China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center Fudan University Shanghai China
| | - Bowen Liu
- School of Agriculture Yunnan University Kunming China
| | - Jiayi Guan
- Henan Institute of Medical and Pharmaceutical Sciences Zhengzhou University Zhengzhou China
| | - Andrzej Półtorak
- Department of Technique and Food Development, Institute of Human Nutrition Sciences Warsaw University of Life Sciences Warsaw Poland
| | - Arkadiusz Szpicer
- Department of Technique and Food Development, Institute of Human Nutrition Sciences Warsaw University of Life Sciences Warsaw Poland
| | - Agnieszka Wierzbicka
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
- Department of Technique and Food Development, Institute of Human Nutrition Sciences Warsaw University of Life Sciences Warsaw Poland
| | - Nikolay T. Tzvetkov
- Department of Biochemical Pharmacology and Drug Design, Institute of Molecular Biology “Roumen Tsanev” Bulgarian Academy of Sciences Sofia Bulgaria
| | - Maciej Banach
- Faculty of Medicine The John Paul II Catholic University of Lublin (KUL) Lublin Poland
- Department of Cardiology and Adult Congenital Heart Diseases Polish Mother's Memorial Hospital Research Institute (PMMHRI) Lodz Poland
- Department of Preventive Cardiology and Lipidology Medical University of Lodz (MUL) Lodz Poland
- Ciccarone Center for the Prevention of Cardiovascular Disease Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Jarosław Olav Horbańczuk
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Artur Jóźwik
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Marco Cascella
- Anesthesia and Pain Medicine, Department of Medicine, Surgery and Dentistry “Scuola MedicaSalernitana” University of Salerno Baronissi Italy
| | - Bairong Shen
- Department of Critical Care Medicine and Institutes for Systems Genetics Frontiers Science Center for Disease‐Related Molecular Network, West China Hospital,Sichuan University Chengdu Sichuan China
- Center for High Altitude Medicine, West China Hospital Sichuan University Chengdu Sichuan China
| | | | - Dongdong Wang
- Centre for Metabolism, Obesity and Diabetes Research McMaster University Hamilton Ontario Canada
- Division of Endocrinology and Metabolism, Department of Medicine McMaster University Hamilton Ontario Canada
| | - Olena Litvinova
- National University of Pharmacy of the Ministry of Health of Ukraine Kharkiv Ukraine
- Ludwig Boltzmann Institute Digital Health and Patient Safety Medical University of Vienna Vienna Austria
| | - Olga Adamska
- Faculty of Medicine Collegium Medicum Cardinal Stefan Wyszyński University in Warsaw Warsaw Poland
| | - Agnieszka Kamińska
- Faculty of Medicine Collegium Medicum Cardinal Stefan Wyszyński University in Warsaw Warsaw Poland
| | - Marcin Łapiński
- Orthopaedic and Rehabilitation Department Medical University of Warsaw Warsaw Poland
| | - Artur Stolarczyk
- Orthopaedic and Rehabilitation Department Medical University of Warsaw Warsaw Poland
| | - Ioana Berindan‐Neagoe
- Department of Genomics MEDFUTURE ‐ Institute for Biomedical Research“Iuliu Hațieganu” University of Medicine and Pharmacy No. 23 Cluj‐Napoca Romania
| | - Luigi Milella
- Department of Health Sciences University of Basilicata Potenza Italy
| | - Andy Wai Kan Yeung
- Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry The University of Hong Kong Pokfulam Hong Kong SAR
| | - Prashanth Suravajhala
- Amrita School of Biotechnology Amrita Viswa Vidyapeetham Clappana Kerala India
- Department of Biosciences Manipal University Jaipur, Dehmi Kala Jaipur Rajasthan India
| | - Anupam Bishayee
- Department of Pharmacology College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine Bradenton Florida USA
| | - Ronan Lordan
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| | - Laszlo Barna Iantovics
- Department of Electrical Engineering and Information Technology George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures Targu Mures Romania
| | - Ricardo Lagoa
- ESTG‐Polytechnic Institute of Leiria Morro do Lena‐Alto do Vieiro Leiria Portugal
- LSRE‐LCM‐Associate Laboratory in Chemical Engineering University of Porto Porto Portugal
| | - Monika Michalczuk
- Department of Animal Breeding, Institute of Animal Sciences Warsaw University of Life Sciences ‐ SGGW Warsaw Poland
| | - Jivko Stoyanov
- Swiss Paraplegic Research Nottwil Switzerland
- Institute of Social and Preventive Medicine (ISPM) University of Bern Bern Switzerland
| | | | - Banaz Jalil
- Pharmacognosy and Phytotherapy UCL School of Pharmacy London UK
| | - Wolfram Weckwerth
- Molecular Systems Biology Lab (MOSYS), Department of Functional and Evolutionary Ecology University of Vienna Vienna Austria
- Vienna Metabolomics Center (VIME) University of Vienna Vienna Austria
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre (SBMDC) School of Medical and Life Sciences Subang Jaya Malaysia
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy Monash University Malaysia Subang Jaya Malaysia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine University of Technology Sydney Ultimo New South Wales Australia
| | - Meng‐Yao Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
- Department of Biliary‐Pancreatic Surgery, Renji Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Gyaneshwer Chaubey
- Cytogenetics Laboratory, Department of Zoology Banaras Hindu University Varanasi Uttar Pradesh India
| | - Gian Luigi Russo
- National Research Council Institute of Food Sciences Avellino Italy
| | - Sara Frazzini
- Department of Veterinary Medicine and Animal Science (DIVAS) University of Milan Lodi Italy
| | - Luciana Rossi
- Department of Veterinary Medicine and Animal Science (DIVAS) University of Milan Lodi Italy
| | - Maurizio Battino
- Department of Clinical Sciences Polytechnic University of Marche Ancona Italy
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods Polytechnic University of Marche (Italy), Universidad Europea del Atlántico (Spain), and Jiangsu University (China) Ancona Italy
- International Joint Research, Laboratory of Intelligent Agriculture and Agri‐Products Processing Jiangsu University Zhenjiang China
| | - Wei Jia
- Department of Pharmacology and Pharmacy The University of Hong Kong Pokfulam Hong Kong SAR
| | - Qi Su
- Microbiota I‐Center Shatin Hong Kong SAR
- Department of Medicine and Therapeutics The Chinese University of Hong Kong Shatin Hong Kong SAR
| | - Xiaoqiang Ma
- Department of Food Science and Technology, School of Agriculture and Biology Shanghai Jiao Tong University Shanghai China
| | - Judith M. Rollinger
- Division of Pharmacognosy, Department of Pharmaceutical Sciences, Faculty of Life Sciences University of Vienna Vienna Austria
| | - Simon K.‐M. R. Rittmann
- Archaea Physiology & Biotechnology Group, Department of Functional and Evolutionary Ecology University of Vienna Vienna Austria
| | - Helen Sheridan
- The NatPro Centre & School of Pharmacy and Pharmaceutical Sciences Trinity College Dublin Dublin Ireland
- Université Bourgogne Europe/INSERM, 21000 Dijon and PHYNOHA Consulting Fontaine‐lès‐Dijon France
| | - John J. Walsh
- The NatPro Centre & School of Pharmacy and Pharmaceutical Sciences Trinity College Dublin Dublin Ireland
| | - Gérard Lizard
- Université Bourgogne Europe/INSERM, 21000 Dijon and PHYNOHA Consulting Fontaine‐lès‐Dijon France
| | - Tomasz M. Karpiński
- Department of Medical Microbiology Poznań University of Medical Sciences Poznań Poland
| | - Ana Sanches Silva
- University of Coimbra, Faculty of Pharmacy, Polo III, Azinhaga de Santa Comba Coimbra Portugal
- Centre for Animal Science Studies (CECA), ICETA University of Porto Porto Portugal
| | - Jakub Piwowarski
- Microbiota Lab, Department of Pharmaceutical Microbiology and Bioanalysis Medical University of Warsaw Warsaw Poland
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology Institute of Microbiology, Guangdong Academy of Sciences Guangzhou China
- School of Life & Health Sciences Fuyao University of Science & Technology Fuzhou Fujian China
| | - Tai‐Ping Fan
- School of Life & Health Sciences Fuyao University of Science & Technology Fuzhou Fujian China
| | - Francesca Giampieri
- Department of Clinical Sciences Polytechnic University of Marche Ancona Italy
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods Polytechnic University of Marche (Italy), Universidad Europea del Atlántico (Spain), and Jiangsu University (China) Ancona Italy
- Research Group on Food, Nutritional Biochemistry and Health Universidad Europea del Atlántico Santander Spain
- International Research Center for Food Nutrition and Safety Jiangsu University Zhenjiang China
| | - Adil El Midaoui
- Faculty of Sciences and Techniques Errachidia, Moulay Ismail University of Meknes Meknes Morocco
- Department of Pharmacology and Physiology, Faculty of Medicine University of Montreal Montreal Quebec Canada
| | - Ka‐Hing Wong
- Research Institute for Future Food The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
- Department of Food Science and Nutrition The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
| | - Ren‐You Gan
- Research Institute for Future Food The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
- Department of Food Science and Nutrition The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
| | - Ahmed Fatimi
- Chemical Science and Engineering Research Team (ERSIC), Department of Chemistry, Polydisciplinary Faculty of Beni Mellal (FPBM) Sultan Moulay Slimane University (USMS) Beni Mellal Morocco
| | - Atanas G. Atanasov
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
- Ludwig Boltzmann Institute Digital Health and Patient Safety Medical University of Vienna Vienna Austria
- Laboratory of Natural Products and Medicinal Chemistry (LNPMC), Center for Global Health Research, Saveetha Medical College and Hospital Saveetha Institute of Medical and Technical Sciences (SIMATS) Thandalam Chennai India
| |
Collapse
|
28
|
Jena PK, Wakita D, Gomez AC, Carvalho TT, Atici AE, Aubuchon E, Narayanan M, Lee Y, Fishbein MC, Takasato Y, Kurashima Y, Kiyono H, Cani PD, de Vos WM, Underhill DM, Devkota S, Chen S, Shimada K, Crother TR, Arditi M, Rivas MN. Intestinal Microbiota Contributes to the Development of Cardiovascular Inflammation and Vasculitis in Mice. Circ Res 2025; 136:e53-e72. [PMID: 40026151 PMCID: PMC11985309 DOI: 10.1161/circresaha.124.325079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Alterations in the intestinal microbiota contribute to the pathogenesis of various cardiovascular disorders, but how they affect the development of Kawasaki disease (KD) an acute pediatric vasculitis, remains unclear. METHODS We used the Lactobacillus casei cell wall extract (LCWE) murine model of KD vasculitis to assess the contribution of the intestinal microbiota to the development of vascular inflammation. We evaluated the severity of vasculitis in microbiota-depleted mice. 16S rRNA gene sequencing was used to characterize the fecal microbiome composition of LCWE-injected mice. Some groups of mice were orally treated with selected live or pasteurized bacteria, short-chain fatty acids, or Amuc_1100, the Toll-like receptor 2 signaling outer membrane protein from Akkermansia muciniphila, and their impact on vasculitis development was assessed. RESULTS We report that depleting the gut microbiota reduces the development of cardiovascular inflammation in a murine model mimicking KD vasculitis. The development of cardiovascular lesions was associated with alterations in the intestinal microbiota composition and, notably, a decreased abundance of Akkermansia muciniphila and Faecalibacterium prausnitzii. Oral supplementation with either of these live or pasteurized individual bacteria or with short-chain fatty acids produced by them attenuated cardiovascular inflammation, as reflected by decreased local immune cell infiltrations. Treatment with Amuc_1100 also reduced the severity of vascular inflammation. CONCLUSIONS This study reveals an underappreciated gut microbiota-cardiovascular inflammation axis in KD vasculitis pathogenesis and identifies specific intestinal commensals that regulate vasculitis in mice by producing metabolites or via extracellular proteins capable of enhancing and supporting gut barrier function.
Collapse
Affiliation(s)
- Prasant K. Jena
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daiko Wakita
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Angela C. Gomez
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Thacyana T. Carvalho
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Asli E. Atici
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Emily Aubuchon
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Meena Narayanan
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Youngho Lee
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael C. Fishbein
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, CA, USA
| | - Yoshihiro Takasato
- Department of Allergy, Allergy and Immunology Center, Aichi Children’s Health and Medical Center, Obu, Japan
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yosuke Kurashima
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Japan
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Japan
| | - Patrice D. Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, WELBIO department, WEL Research Institute, Wavre, Belgium
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - David M. Underhill
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- F. Widjaja Inflammatory Bowel Diseases Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Suzanne Devkota
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- F. Widjaja Inflammatory Bowel Diseases Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Human Microbiome Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shuang Chen
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kenichi Shimada
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Timothy R. Crother
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
29
|
Kalkan AE, BinMowyna MN, Raposo A, Ahmad MF, Ahmed F, Otayf AY, Carrascosa C, Saraiva A, Karav S. Beyond the Gut: Unveiling Butyrate's Global Health Impact Through Gut Health and Dysbiosis-Related Conditions: A Narrative Review. Nutrients 2025; 17:1305. [PMID: 40284169 PMCID: PMC12029953 DOI: 10.3390/nu17081305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Short-chain fatty acids (SCFAs), mainly produced by gut microbiota through the fermentation process of dietary fibers and proteins, are crucial to human health, with butyrate, a famous four-carbon SCFA, standing out for its inevitably regulatory impact on both gut and immune functions. Within this narrative review, the vital physiological functions of SCFAs were examined, with emphasis on butyrate's role as an energy source for colonocytes and its ability to enhance the gut barrier while exhibiting anti-inflammatory effects. Knowledge of butyrate synthesis, primarily generated by Firmicutes bacteria, can be influenced by diets with specifically high contents of resistant starches and fiber. Butyrate can inhibit histone deacetylase, modulate gene expression, influence immune functionality, and regulate tight junction integrity, supporting the idea of its role in gut barrier preservation. Butyrate possesses systemic anti-inflammatory properties, particularly, its capacity to reduce pro-inflammatory cytokines and maintain immune homeostasis, highlighting its therapeutic potential in managing dysbiosis and inflammatory diseases. Although butyrate absorption into circulation is typically minimal, its broader health implications are substantial, especially regarding obesity and type 2 diabetes through its influence on metabolic regulation and inflammation. Furthermore, this narrative review thoroughly examines butyrate's growing recognition as a modulator of neurological health via its interaction with the gut-brain axis. Additionally, butyrate's neuroprotective effects are mediated through activation of specific G-protein-coupled receptors, such as FFAR3 and GPR109a, and inhibition of histone deacetylases (HDACs). Research indicates that butyrate can alleviate neurological disorders, including Alzheimer's, Parkinson's, autism spectrum disorder, and Huntington's disease, by reducing neuroinflammation, enhancing neurotransmitter modulation, and improving histone acetylation. This focus will help unlock its full therapeutic potential for metabolic and neurological health, rather than exclusively on its well-known benefits for gut health, as these are often interconnected.
Collapse
Affiliation(s)
- Arda Erkan Kalkan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| | - Mona N. BinMowyna
- College of Education, Shaqra University, Shaqra 11911, Saudi Arabia;
| | - António Raposo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Md Faruque Ahmad
- Department of Clinical Nutrition, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; (M.F.A.); (A.Y.O.)
| | - Faiyaz Ahmed
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, P.O. Box 6666, Buraydah 51452, Saudi Arabia;
| | - Abdullah Y. Otayf
- Department of Clinical Nutrition, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; (M.F.A.); (A.Y.O.)
| | - Conrado Carrascosa
- Department of Animal Pathology and Production, Bromatology and Food Technology, Faculty of Veterinary, Universidad de Las Palmas de Gran Canaria, Trasmontaña s/n, 35413 Arucas, Spain;
| | - Ariana Saraiva
- Research in Veterinary Medicine (I-MVET), Faculty of Veterinary Medicine, Lisbon University Centre, Lusófona University, Campo Grande 376, 1749-024 Lisboa, Portugal;
- Veterinary and Animal Research Centre (CECAV), Faculty of Veterinary Medicine, Lisbon University Centre, Lusófona University, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| |
Collapse
|
30
|
Sørum ME, Boulund U, De Pietri S, Weischendorff S, Enevold C, Rathe M, Als-Nielsen B, Hasle H, Pamp S, Stokholm J, Müller K. Changes in gut microbiota predict neutropenia after induction treatment in childhood acute lymphoblastic leukemia. Blood Adv 2025; 9:1508-1521. [PMID: 39561377 PMCID: PMC11985026 DOI: 10.1182/bloodadvances.2024013986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/19/2024] [Accepted: 10/11/2024] [Indexed: 11/21/2024] Open
Abstract
ABSTRACT Delayed neutrophil recovery during acute lymphoblastic leukemia (ALL) treatment increases the risk of infection and causes delay in chemotherapy. Emerging evidence implicates gut microbiota in neutrophil reconstitution after chemotherapy. We explored the interplay between the gut microbiota and neutrophil dynamics, including neutrophil chemoattractants, in 51 children with newly diagnosed ALL. Daily absolute neutrophil count (ANC), weekly plasma chemokines (CXCL1 and CXCL8), granulocyte colony-stimulating factor (G-CSF), and fecal samples were monitored until day 29 during ALL induction treatment. Fecal sequencing using 16S ribosomal RNA revealed an overall significant reduction in bacterial diversity and Enterococcus overgrowth throughout the induction treatment. Prolonged neutropenia (ANC <0.5 × 109 cells per L at day 36) and elevated chemokine levels were associated with a decreased abundance of genera from the Ruminococcaceae and Lachnospiraceae families, decreased Veillonella genus, and Enterococcus overgrowth from diagnosis and throughout induction treatment. G-CSF was upregulated in response to neutropenia but was unrelated to microbiota changes. Overall, this study revealed that a diminished abundance of specific intestinal commensals and Enterococcus overgrowth is associated with delayed neutrophil reconstitution and increased chemokine signaling, indicating that disruption of the microbiota may contribute to prolonged neutropenia. These findings lay the groundwork for future investigations into the mechanisms underlying these associations and their clinical implications for developing gut-sparring strategies to minimize the impact of gut dysbiosis on immune recovery.
Collapse
Affiliation(s)
- Maria Ebbesen Sørum
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ulrika Boulund
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Silvia De Pietri
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sarah Weischendorff
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Christian Enevold
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Mathias Rathe
- Department of Pediatrics and Adolescent Medicine, The Hans Christian Andersen Children’s Hospital, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Bodil Als-Nielsen
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Hasle
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Sünje Pamp
- Research Group for Genomic Epidemiology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Klaus Müller
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
31
|
Cao J, Wang S, Ding R, Liu Y, Yuan B. Comparative analyses of the gut microbiome of two sympatric rodent species, Myodes rufocanus and Apodemus peninsulae, in northeast China based on metagenome sequencing. PeerJ 2025; 13:e19260. [PMID: 40226542 PMCID: PMC11988107 DOI: 10.7717/peerj.19260] [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: 06/04/2024] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
The gut microbiota is integral to an animal's physiology, influencing nutritional metabolism, immune function, and environmental adaptation. Despite the significance of gut microbiota in wild rodents, the Korean field mouse (Apodemus peninsulae) and the gray red-backed vole (Myodes rufocanus) remain understudied. To address this, a metagenomic sequencing analysis of the gut microbiome of these sympatric rodents in northeast China's temperate forests was conducted. Intestinal contents were collected from A. peninsulae and M. rufocanus within the Mudanfeng National Nature Reserve. High-throughput sequencing elucidated the gut microbiome's composition, diversity, and functional pathways. Firmicutes, Bacteroidetes, and Proteobacteria were identified as the dominant phyla, with M. rufocanus showing greater microbiome diversity. Key findings indicated distinct gut bacterial communities between the species, with M. rufocanus having a higher abundance of Proteobacteria. The gut microbiota of A. peninsulae and M. rufocanus differed marginally in functional profiles, specifically in the breakdown of complex carbohydrates, which might reflect their distinct food preferences albeit both being herbivores with a substantial dietary overlap. The investigation further elucidated gut microbiota's contributions to energy metabolism and environmental adaptation mechanisms. This study aligns with information on rodent gut microbiota in literature and highlights the two understudied rodent species, providing comparative data for future studies investigating the role of gut microbiota in wildlife health and ecosystem functioning.
Collapse
Affiliation(s)
- Jing Cao
- College of Biology and Food, Shangqiu Normal University, Shangqiu, Henan, China
| | - Shengze Wang
- School of Life Science, Liaocheng University, Liaocheng, Shandong, China
| | - Ruobing Ding
- College of Biology and Food, Shangqiu Normal University, Shangqiu, Henan, China
| | - Yijia Liu
- College of Biology and Food, Shangqiu Normal University, Shangqiu, Henan, China
| | - Baodong Yuan
- School of Life Science, Liaocheng University, Liaocheng, Shandong, China
| |
Collapse
|
32
|
Mainardi F, Garcia-Garcera M, Nash AK. A bi-variate framework to model microbiome resilience in healthy dogs. Front Vet Sci 2025; 12:1486679. [PMID: 40241810 PMCID: PMC12001528 DOI: 10.3389/fvets.2025.1486679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 02/26/2025] [Indexed: 04/18/2025] Open
Abstract
Introduction Ecological resilience is the capacity of an ecosystem to maintain its state and recover from disturbances. This concept can be applied to the gut microbiome as a marker of health. Methods Several metrics have been proposed to quantify microbiome resilience, based on the prior choice of some salient feature of the trajectories of microbiome change. We propose a data-driven approach based on compositional and functional data analysis to quantify microbiome resilience. We demonstrate the validity of our approach through applications to sled dogs undergoing three types of exercise: running on an exercise wheel, pulling an all-terrain vehicle, and pulling a sled. Results Microbiota composition was clearly impacted by each exercise type. Log-ratio analysis was utilized for dimensionality reduction and identified 33 variables (taxa) explaining 90% of the variance. Functional principal component analysis identified two scores (FPCA 1 and FPCA2) which explained 76% and 19% of the variability of the trajectories, respectively. More resilient trajectories corresponded to low values of FPCA1 and FPCA2 values close to zero. Levels of chemokines MCP-1 and KC-like, which increased significantly after exercise and returned to pre-exercise levels within 24 h, were significantly associated with FPCA scores as well. Discussion To our knowledge, this is the first study proposing a principled approach to quantify microbiome resilience in healthy dogs and associate it with immune response to exercise-related stress.
Collapse
Affiliation(s)
- Fabio Mainardi
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Marc Garcia-Garcera
- Department of Gastrointestinal Health, Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | | |
Collapse
|
33
|
Kulkarni R, Kumari S, Dhapola R, Sharma P, Singh SK, Medhi B, HariKrishnaReddy D. Association Between the Gut Microbiota and Alzheimer's Disease: An Update on Signaling Pathways and Translational Therapeutics. Mol Neurobiol 2025; 62:4499-4519. [PMID: 39460901 DOI: 10.1007/s12035-024-04545-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
Alzheimer's disease (AD) is a cognitive disease with high morbidity and mortality. In AD patients, the diversity of the gut microbiota is altered, which influences pathology through the gut-brain axis. Probiotic therapy alleviates pathological and psychological consequences by restoring the diversity of the gut microbial flora. This study addresses the role of altered gut microbiota in the progression of neuroinflammation, which is a major hallmark of AD. This process begins with the activation of glial cells, leading to the release of proinflammatory cytokines and the modulation of cholinergic anti-inflammatory pathways. Short-chain fatty acids, which are bacterial metabolites, provide neuroprotective effects and maintain blood‒brain barrier integrity. Furthermore, the gut microbiota stimulates oxidative stress and mitochondrial dysfunction, which promote AD progression. The signaling pathways involved in gut dysbiosis-mediated neuroinflammation-mediated promotion of AD include cGAS-STING, C/EBPβ/AEP, RAGE, TLR4 Myd88, and the NLRP3 inflammasome. Preclinical studies have shown that natural extracts such as Ganmaidazao extract, isoorentin, camelia oil, Sparassis crispa-1, and xanthocerasides improve gut health and can delay the worsening of AD. Clinical studies using probiotics such as Bifidobacterium spp., yeast beta-glucan, and drugs such as sodium oligomannate and rifaximine have shown improvements in gut health, resulting in the amelioration of AD symptoms. This study incorporates the most current research on the pathophysiology of AD involving the gut microbiota and highlights the knowledge gaps that need to be filled to develop potent therapeutics against AD.
Collapse
Affiliation(s)
- Rutweek Kulkarni
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Sunil K Singh
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India.
| |
Collapse
|
34
|
Qiu J, Zhang Y, Chen K, Xu J, Chen Y, Li M, Zhu C, Wang S, Zhang L. Integrating serum pharmacochemistry, network pharmacology, metabolomics and 16S rRNA sequencing to explore the mechanism of total flavonoids from Flemingia philippinensis in treating collagen induced arthritis rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156531. [PMID: 39987603 DOI: 10.1016/j.phymed.2025.156531] [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: 09/22/2024] [Revised: 02/15/2025] [Accepted: 02/15/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUD Rheumatoid arthritis (RA) is a prevalent chronic autoimmune disease characterized by symmetric polyarthritis, resulting in pain and swelling in the synovial joints. Flemingia philippinensis, a traditional Chinese medicine, has been shown to be an effective treatment approach for anti-rheumatoid arthritis (RA), which still needs further research in its active ingredient and regulatory mechanisms. PURPOSE This study aimed at investigate the pharmacodynamic basis and intricate mechanism of action of Flemingia philippinensis (FPTF) in the treatment of RA based on integrated omics technologies. METHODS UPLC-Q-Orbitrap HRMS was first established to identify the active components of FPTF in blood and network pharmacology was then used to predict the key therapeutic targets and corresponding pathways of FPTF in treatment of RA. To substantiate the pharmacodynamic effects, a collagen-induced arthritis (CIA) animal model was employed to observe the anti-RA effects of FPTF through a series of indicators, including rat body weight, arthritis scoring, paw swelling, histopathological analysis of synovial tissue, and serum inflammatory factors. Subsequently, the potential mechanisms underlying the anti-RA efficacy of FPTF was elucidated by integrating metabolomics analysis with 16S rRNA gene sequencing. Specifically, the RT-qPCR experiment was further conducted to validate the pathways predicted by serum pharmacochemistry, network pharmacology, metabolomics and 16S rRNA gene sequencing. RESULTS A total of 10 compounds derived from FPTF were identified by serum sample analysis. Utilizing network pharmacology, we identified 117 common targets for FPTF in the treatment of RA. Notably, KEGG analysis highlighted the PI3K/AKT signaling pathway and the IL17 signaling pathway as key pathways associated with the anti-RA effects of FPTF. Pharmacodynamic studies showed that FPTF can significantly alleviate CIA-induced arthritis. Compared with the CIA model group, FPTF treatment significantly improved the expression of mRNA in the PI3K/AKT and IL-17 signaling pathways. Further investigation unveiled a total of 28 differential metabolites in serum samples, among which 21 metabolites were observed to be reversed following FPTF administration. Metabolomic profiling revealed pronounced perturbations in amino acid metabolism, fatty acid metabolism, and glycerophospholipid metabolism pathways in CIA rats, which were partially rectified by FPTF treatment. Additionally, 16S rRNA gene sequencing analysis indicated that FPTF could restore the gut microbiota balance disrupted by RA. RT-qPCR further confirmed that FPTF can modulate key enzymes in metabolic pathway analysis and gut microbiota metabolic pathways. CONCLUSION This study pioneeringly elucidates the potential pharmacodynamic material basis of FPTF for treatment of RA, detailing the regulated metabolic pathways and key gut microbiota genera involved. The findings provide a comprehensive understanding of mechanisms underlying the effects of FPTF in RA treatment.
Collapse
Affiliation(s)
- Jingwen Qiu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China; Guangdong China Resources Shunfeng Pharmaceutical Co., Ltd, Foshan, 528300, PR China
| | - Yingyin Zhang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Kairan Chen
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Junya Xu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Yingnan Chen
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Minjing Li
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Chenchen Zhu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Song Wang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Lei Zhang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| |
Collapse
|
35
|
Horill S, Zhou XK, Jin W. Probiotics as a possible novel therapeutic option to mitigate perioperative neurocognitive disorders: A review exploring the latest research findings. J Clin Anesth 2025; 103:111801. [PMID: 40043583 DOI: 10.1016/j.jclinane.2025.111801] [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: 10/19/2024] [Revised: 01/27/2025] [Accepted: 02/26/2025] [Indexed: 05/16/2025]
Abstract
Perioperative neurocognitive disorders (PND) refer to a constellation of symptoms that primarily affect the elderly and typically manifest as common complications after exposure to surgery and anesthesia. PND is associated with high morbidity, mortality, and progression to neurodegenerative diseases, thus exerting significant financial strains on families as well as the healthcare system. Given that an ageing global population is an inevitable trend and, with the latest advances in the healthcare system, an ever-growing number of elderly people present for surgery and anesthesia, PND is of prominent concern. The two-way communication between the intestinal flora and the brain, also known as the microbiota-gut-brain axis, plays an important role in central nervous system development, and multiple studies have highlighted the influence exerted by gut microbiome in both health and disease. Pertinent studies have corroborated the fact that anesthesia and surgery disrupt the harmony of the gut ecology, which sets off a cascade of events that initiate neuroinflammation, eventually leading to PND. Probiotics, which are live microorganisms that promote the host's health, have been shown as a viable option to restore or minimise the disruption of gut flora. Evidence exists that probiotics exhibit immunomodulatory and anti-inflammatory benefits. Given the effectiveness of probiotics in reducing neuroinflammation, research has also focused on their impact on the development of PND. This review aims to compile the data from relevant clinical trials focusing on the influence of probiotics on PND to determine whether the derived findings might be applied for the prevention and treatment of PND.
Collapse
Affiliation(s)
- Smita Horill
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Xiao-Kai Zhou
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Wenjie Jin
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
36
|
Kovynev A, Charchuta MM, Begtašević A, Ducarmon QR, Rensen PCN, Schönke M. Combination of dietary fiber and exercise training improves fat loss in mice but does not ameliorate MASLD more than exercise alone. Am J Physiol Gastrointest Liver Physiol 2025; 328:G399-G410. [PMID: 40033967 DOI: 10.1152/ajpgi.00317.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/02/2025] [Accepted: 02/20/2025] [Indexed: 03/05/2025]
Abstract
Lifestyle interventions, such as diet and exercise, are currently the main therapies against metabolic dysfunction-associated steatotic liver disease (MASLD). However, not much is known about the combined impact of fiber and exercise on the modulation of gut-liver axis and MASLD amelioration. Here, we studied the impact of the combination of exercise training and a fiber-rich diet on the amelioration of MASLD. Male APOE*3-Leiden.CETP mice were fed a high-fat high-cholesterol diet with or without the addition of fiber (10% inulin) and exercise trained on a treadmill, or remained sedentary. Exercise training and fiber supplementation reduced fat mass gain and lowered plasma glucose levels. Only the combination treatment, however, induced fat loss and decreased plasma triglyceride and cholesterol levels compared with sedentary control mice. Exercise training with and without the addition of fiber had a similar ameliorating effect on the MASLD score. Only exercise without fiber decreased the hepatic expression of inflammatory markers. Fiber diet was mainly responsible for remodeling the gut microbial composition, with an increase in the relative abundance of the short-chain fatty acid (SCFA)-producing genera Anaerostipes and Muribaculaceae, whereas, surprisingly, exercise training alone and with fiber resulted in the highest increase of SCFA production. Overall, the combination of exercise training and dietary fiber decreases fat mass and improves glucose and lipid homeostasis but does not have an additional synergistic positive effect on liver health compared with exercise training alone.NEW & NOTEWORTHY The combination of dietary fiber intake and exercise training has a synergetic beneficial effect on the metabolic health, resulting in fat loss, lowered blood glucose, and lowered plasma lipid levels in mice with steatotic liver disease. However, fiber supplementation, despite a positive remodulation of the gut-liver axis, does not have an additional positive effect on liver health compared with exercise training alone.
Collapse
Affiliation(s)
- Artemiy Kovynev
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Mikołaj M Charchuta
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Amina Begtašević
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Quinten R Ducarmon
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Milena Schönke
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
37
|
He P, Lyu Y, Shen X, Liu W, Zhang Y, Li Y, Huang Y, Xu L, Zhang L, Guo S. Gut microbiome and short-chain fatty acids associated with the efficacy of growth hormone treatment in children with short stature. Front Pediatr 2025; 13:1557878. [PMID: 40230807 PMCID: PMC11994682 DOI: 10.3389/fped.2025.1557878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/14/2025] [Indexed: 04/16/2025] Open
Abstract
Objective To investigate associations between fecal microbiota, short-chain fatty acids (SCFAs), and the efficacy of recombinant human growth hormone (rhGH) treatment in children with growth hormone deficiency (GHD) or idiopathic short stature (ISS). Methods A 2-phase cohort study was conducted. Phase I included 102 participants (GHD: n = 33, ISS: n = 28, controls: n = 41) for cross-sectional analysis using 16S rRNA sequencing and targeted metabolomics to compare microbial diversity, predicted metabolic pathways, and SCFA levels. Phase II longitudinally monitored 61 rhGH-treated children (GHD = 33, ISS = 28) over 2 years, assessing growth velocity, IGF-1 levels, and fecal microbiota/SCFA dynamics. Statistical analyses included alpha/beta diversity metrics, LEfSe, PERMANOVA, and redundancy analysis (RDA) to link microbial/SCFA profiles with clinical outcomes. Results (1). Microbiota Dysbiosis: Untreated GHD/ISS children exhibited reduced beneficial taxa (e.g., Faecalibacterium, Akkermansia) and increased pathobionts (e.g., Streptococcus, Collinsella) compared to controls (PERMANOVA: R 2 = 0.114, P = 0.001). (2). Metabolic Pathways: GHD/ISS groups showed enrichment in xenobiotic degradation (e.g., atrazine) and deficits in nutrient-associated pathways (e.g., carotenoid biosynthesis). (3). rhGH Effects: Treatment increased beneficial taxa (e.g., Bifidobacterium, Faecalibacterium) and modulated amino acid/lipid metabolism pathways (e.g., glycine-serine-threonine metabolism, P = 0.035). (4). SCFAs and Growth Velocity: Higher growth velocity percentiles correlated with elevated acetic acid (GHD-treated: 1952 ± 962.4 vs. untreated: 1290 ± 886.0 μg/g, P = 0.037) and butyric acid levels. Conclusion GHD, ISS, and healthy children have different fecal microbiota compositions and SCFA metabolisms. rhGH therapy partially restores microbial balance and alters metabolic pathways, with SCFA levels associated with treatment efficacy. These findings highlight the gut microbiome as a potential modulator of rhGH response and provide insight into microbiota-targeted therapies to improve growth outcomes (e.g., "probiotic interventions").
Collapse
Affiliation(s)
- Pingsihua He
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongfen Lyu
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyuan Shen
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenxin Liu
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Zhang
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Li
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuying Huang
- Department of Traditional Chinese Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liya Xu
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liang Zhang
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sheng Guo
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
38
|
Zhao T, Wang C, Liu Y, Li B, Shao M, Zhao W, Zhou C. The role of polysaccharides in immune regulation through gut microbiota: mechanisms and implications. Front Immunol 2025; 16:1555414. [PMID: 40230839 PMCID: PMC11994737 DOI: 10.3389/fimmu.2025.1555414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 03/05/2025] [Indexed: 04/16/2025] Open
Abstract
Polysaccharides, as complex carbohydrates, play a pivotal role in immune modulation and interactions with the gut microbiota. The diverse array of dietary polysaccharides influences gut microbial ecology, impacting immune responses, metabolism, and overall well-being. Despite their recognized benefits, there is limited understanding of the precise mechanisms by which polysaccharides modulate the immune system through the gut microbiota. A comprehensive search of Web of Science, PubMed, Google Scholar, and Embase up to May 2024 was conducted to identify relevant studies. This study employs a systematic approach to explore the interplay between polysaccharides and the gut microbiota, focusing on cytokine-mediated and short-chain fatty acid (SCFA)-mediated pathways. The findings underscore the significant role of polysaccharides in shaping the composition and function of the gut microbiota, thereby influencing immune regulation and metabolic processes. However, further research is necessary to elucidate the detailed molecular mechanisms and translate these findings into clinical applications.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Congyue Wang
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Yuhan Liu
- Department of Medical Oncology, Anshan Cancer Hospital, Anshan, China
| | - Bo Li
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Mingjia Shao
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Wuyang Zhao
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Chuang Zhou
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| |
Collapse
|
39
|
Zhang X, Liu X, Xie K, Pan Y, Liu F, Hou F. Effects of different fiber levels of energy feeds on rumen fermentation and the microbial community structure of grazing sheep. BMC Microbiol 2025; 25:180. [PMID: 40165064 PMCID: PMC11956436 DOI: 10.1186/s12866-024-03644-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/08/2024] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Rumen microbial community structure and stability are very important for ruminant health, growth and development, and livestock product yield. Dietary composition and nutritional structure affect microbial diversity and richness. The purpose of this study was to evaluate the effects of different fiber levels of energy feed on the rumen microflora and fermentation function of grazing sheep in salinized sown pasture, to reveal the response of the main microflora of sheep rumen at the phylum and genus levels to different fiber levels of energy feed and to analyze the internal mechanism to provide a reference for the selection of energy feed and the improvement of the production performance of grazing livestock. RESULTS The fiber level of energy feed affects the rumen fermentation and rumen microbial community structure of grazing sheep. Low-fiber-energy feeds significantly increased the relative abundance of Actinobacteria, while the relative abundances of Cyanobacteria, Ruminococcaceae_UCG_010, Ruminococcaceae_NK4A214_group, and Elusimicrobium significantly decreased, adjusting the relationship between the flora toward cooperation. High-fiber-energy feeds significantly increased the concentration of VFAs, significantly decreased the relative abundances of Proteobacteria, Ruminococcaceae_NK4A214_group and Rikenellaceae_RC9_gut_group, adjusted the relationship between the flora to compete, and promoted the enrichment of metabolic pathways such as "Protein Digestion and Absorption," "Nitrogen Metabolism," "Starch and Sucrose Metabolism," and "Degradation of Other Sugars." CONCLUSIONS Supplementary feeding of high and low fiber energy feeds reduced the pH value of rumen fluid and the richness and diversity of microorganisms in grazing sheep, reduced the relative abundance of some harmful microorganisms, affected the metabolic activities of some fiber-digesting bacteria, regulated the interaction and competition between bacteria, increased the content of volatile fatty acids (VFAs) and the relative abundance of metabolic-related microorganisms in the supplementary feeding group, and enriched the metabolic-related pathways. However, further understand the mechanism of the effect of fiber level on the rumen of sheep, it is necessary to conduct in-depth analysis using research methods such as transcriptomics, proteomics and metabolomics.
Collapse
Affiliation(s)
- Xiaoyun Zhang
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Ministry of Agriculture and Rural Affairs, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, 730020, China
| | - Xulei Liu
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Ministry of Agriculture and Rural Affairs, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, 730020, China
| | - Kaili Xie
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Ministry of Agriculture and Rural Affairs, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, 730020, China
| | - Yueting Pan
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Ministry of Agriculture and Rural Affairs, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, 730020, China
| | - Fuyao Liu
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Ministry of Agriculture and Rural Affairs, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, 730020, China
| | - Fujiang Hou
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Ministry of Agriculture and Rural Affairs, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, 730020, China.
- , Lanzhou, P. R. China.
| |
Collapse
|
40
|
Gao G, Cai L, Fan Y, Aroche Ginarte R, Li Y, Sun W, Jiang X, Li X, Pi Y. Effects of Different Hemicellulose Components on Fermentation Kinetics and Microbial Composition in Fecal Inoculum from Suckling Piglets In Vit ro. ACS OMEGA 2025; 10:9120-9131. [PMID: 40092836 PMCID: PMC11904680 DOI: 10.1021/acsomega.4c08848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/08/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
This study investigated the fermentation characteristics of different hemicellulose components using a fecal inoculum derived from suckling piglets. The results showed that after 60 h of fermentation, the arabinogalactan (Ara-gal), glucomannan (Glu-man), galactomannan (Gal-man), and mannan (Man) groups exhibited similar levels of gas production, which were higher than those of the β-glucan (β-Glu) group. The β-Glu group had the lowest pH value. After 48 h of fermentation, the Ara-gal group had the highest microbial crude protein content and the lowest ammonia nitrogen content. The Glu-man, Gal-man, and Man groups produced similar amounts of acetate, propionate, and total short-chain fatty acids (SCFAs), which were higher than those in the Ara-gal and β-Glu groups. Furthermore, the Man and Ara-gal groups showed the highest butyrate production. Significant differences in the microbial community composition were observed among the groups. Correlation analyses further revealed that the abundance of specific bacteria, such as Prevotella_9 and Parabacteroides, was closely related to the production of acetate, propionate, and butyrate. These results suggest that Glu-man, Gal-man, and Man undergo rapid fermentation, with Ara-gal following, while β-Glu ferments the slowest. The distinct fiber compositions and fermentation properties of different hemicellulose components significantly influence the microbial composition and SCFA production. Our findings offer valuable theoretical insights for selecting fiber components in the diets of suckling piglets and potentially in infants.
Collapse
Affiliation(s)
- Ge Gao
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Long Cai
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Yuyang Fan
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Roisbel Aroche Ginarte
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
- Department
of Animal Husbandry, Faculty of Agricultural Science, University of Granma, Bayamo 85100, Cuba
| | - Yanpin Li
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Wenjuan Sun
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Xianren Jiang
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Xilong Li
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Yu Pi
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
41
|
Butowski CF, Dixit Y, Reis MM, Mu C. Metatranscriptomics for Understanding the Microbiome in Food and Nutrition Science. Metabolites 2025; 15:185. [PMID: 40137150 PMCID: PMC11943699 DOI: 10.3390/metabo15030185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
Microbiome science has greatly expanded our understanding of the diverse composition and function of gut microorganisms over the past decades. With its rich microbial composition, the microbiome hosts numerous functionalities essential for metabolizing food ingredients and nutrients, resulting in the production of active metabolites that affect food fermentation or gut health. Most of these processes are mediated by microbial enzymes such as carbohydrate-active enzymes and amino acid metabolism enzymes. Metatranscriptomics enables the capture of active transcripts within the microbiome, providing invaluable functional insights into metabolic activities. Given the inter-kingdom complexity of the microbiome, metatranscriptomics could further elucidate the activities of fungi, archaea, and bacteriophages in the microbial ecosystem. Despite its potential, the application of metatranscriptomics in food and nutrition sciences remains limited but is growing. This review highlights the latest advances in food science (e.g., flavour formation and food enzymology) and nutrition science (e.g., dietary fibres, proteins, minerals, and probiotics), emphasizing the integration of metatranscriptomics with other technologies to address key research questions. Ultimately, metatranscriptomics represents a powerful tool for uncovering the microbiome activity, particularly in relation to active metabolic processes.
Collapse
|
42
|
Wang X, Wang Y, Feng M, Li J, Liu Z, Fu L, Zhang N, Zhang H, Qin J. Herbal formula alleviates heat stress by improving physiological and biochemical attributes and modulating the rumen microbiome in dairy cows. Front Vet Sci 2025; 12:1558856. [PMID: 40125321 PMCID: PMC11925914 DOI: 10.3389/fvets.2025.1558856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Heat stress significantly impacts dairy cow productivity, health, and welfare. This study evaluated a self-developed herbal formula as a dietary intervention to mitigate heat stress. A total of 198 lactating cows were divided into two groups: a Control group receiving standard total mixed rations and a Herbs group supplemented with herbal formula for 60 days. Various parameters were assessed, including milk yield and composition, antioxidant capacity, immune responses, stress-related gene expression, and rumen microbial composition. Compared to the Control group, cows in the Herbs group showed improved feed intake, milk yield and quality, rumination frequency, and enhanced antioxidant activity and immune response. Rumen microbiome analysis revealed a reduced relative abundance of Proteobacteria and Ochrobactrum in the Herbs group, along with an enrichment of beneficial genera such as Lachnospira. Functional predictions indicated that the Herbs group exhibited enhanced glycolysis/gluconeogenesis, pyruvate metabolism, and starch and sucrose metabolism, reflecting improved fermentation efficiency and energy utilization. In conclusion, the herbal formula improved physiological and biochemical attributes, boosted antioxidant and immune responses, and modulated the rumen microbiome, contributing to the alleviation of heat stress in dairy cows. These findings highlight its potential as a natural dietary strategy to support dairy cow health and productivity under heat stress conditions.
Collapse
Affiliation(s)
- Xiaofang Wang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
- Institute of Animal Husbandry and Veterinary Medicine of Hebei, Baoding, China
| | - Yawen Wang
- Institute of Animal Husbandry and Veterinary Medicine of Hebei, Baoding, China
| | - Man Feng
- Chengde Academy of Agriculture and Forestry Sciences, Chengde, China
| | - Jiefeng Li
- Institute of Animal Husbandry and Veterinary Medicine of Hebei, Baoding, China
| | - Ze Liu
- Beijing-Tianjin-Hebei Modern Agriculture Collaborative Innovation and Development Service Center, Baoding, China
| | - Le Fu
- Institute of Animal Husbandry and Veterinary Medicine of Hebei, Baoding, China
| | - Ning Zhang
- Institute of Animal Husbandry and Veterinary Medicine of Hebei, Baoding, China
| | - Huaying Zhang
- Institute of Animal Husbandry and Veterinary Medicine of Hebei, Baoding, China
| | - Jianhua Qin
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| |
Collapse
|
43
|
Liu Y, Li Z, Lee SC, Chen S, Li F. Akkermansia muciniphila: promises and pitfallsfor next-generation beneficial microorganisms. Arch Microbiol 2025; 207:76. [PMID: 40032707 DOI: 10.1007/s00203-025-04263-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 03/05/2025]
Abstract
Akkermansia muciniphila, a microorganism ubiquitously colonizing the mucosal layer of the human gut, has garnered significant scientific interest as a promising candidate for probiotic therapeutics. Its persistent identification in both laboratory and living organism studies underscores its potential physiological benefits, positioning it as a bacterium of paramount importance in promoting host health. This review examines the diversity and abundance of gut microbiota members, emphasizing the identification of microbial species engaged in cross-feeding networks with A. muciniphila. Insightful exploration into the mechanisms of cross-feeding, including mucin-derived nutrient exchange and metabolite production, unveils the intricate dynamics shaping microbial community stability. Such interactions contribute not only to the availability of essential nutrients within the gut environment but also to the production of metabolites influencing microbial community dynamics and host health. In conclusion, the cumulative evidence from in vitro and in vivo perspectives substantiates the notion that A. muciniphila holds tremendous promise as a next-generation probiotic. By leveraging its unique physiological benefits, particularly in mucosal health and metabolic regulation, A. muciniphila stands poised to revolutionize the landscape of probiotic interventions for enhanced host well-being.
Collapse
Affiliation(s)
- Yantong Liu
- Department of Computer and Information Engineering, Kunsan National University, Gunsan, 54150, Republic of Korea
| | - Zonglun Li
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, 453007, China
| | - Sze Ching Lee
- Department of Neurology & Neurosurgery, Mayo clinic, Rochester, MN, 55902, USA
| | - Shurui Chen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Feifei Li
- Department of Biochemistry and molecular biology, Mayo clinic, 200 First St. SW, Rochester, MN, 55902, USA.
| |
Collapse
|
44
|
Zheng T, Meng C, Lv Z, Wu C, Zhou X, Mao W. The Critical Role of Faecalibacterium prausnitzii in Cardiovascular Diseases. Rev Cardiovasc Med 2025; 26:26740. [PMID: 40160596 PMCID: PMC11951488 DOI: 10.31083/rcm26740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 04/02/2025] Open
Abstract
Due to the continued aging of the global population, cardiovascular diseases (CVDs) remain the main cause of death worldwide, with millions of fatalities from diseases, including stroke and coronary artery disease, reported annually. Thus, novel therapeutic approaches and targets are urgently required for diagnosing and treating CVDs. Recent studies emphasize the vital part of gut microbiota in both CVD prevention and management. Among these, Faecalibacterium prausnitzii (F. prausnitzii) has emerged as a promising probiotic capable of improving intestinal health. Although preliminary investigations demonstrate that F. prausnitzii positively enhances cardiovascular health, research specifically connecting this strain to CVD outcomes remains limited. Based on current research and assessment of possible clinical applications, this paper aimed to investigate the positive effects on cardiovascular health using F. prausnitzii and its metabolites. Targeting gut flora is expected to become a mainstay in CVD treatment as research develops.
Collapse
Affiliation(s)
- Tiantian Zheng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Chenchen Meng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Zhengtian Lv
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Chenxia Wu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
- Department of Cardiology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, 310030 Hangzhou, Zhejiang, China
| | - Xinbin Zhou
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 310006 Hangzhou, Zhejiang, China
| | - Wei Mao
- Department of Cardiology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, 310030 Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Integrative Chinese and Western Medicine for Diagnosis and Treatment of Circulatory Diseases, 310030 Hangzhou, Zhejiang, China
| |
Collapse
|
45
|
Tao W, Zhang Y, Wang B, Nie S, Fang L, Xiao J, Wu Y. Advances in molecular mechanisms and therapeutic strategies for central nervous system diseases based on gut microbiota imbalance. J Adv Res 2025; 69:261-278. [PMID: 38579985 PMCID: PMC11954836 DOI: 10.1016/j.jare.2024.03.023] [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: 01/14/2024] [Revised: 03/12/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUD Central nervous system (CNS) diseases pose a serious threat to human health, but the regulatory mechanisms and therapeutic strategies of CNS diseases need to be further explored. It has been demonstrated that the gut microbiota (GM) is closely related to CNS disease. GM structure disorders, abnormal microbial metabolites, intestinal barrier destruction and elevated inflammation exist in patients with CNS diseases and promote the development of CNS diseases. More importantly, GM remodeling alleviates CNS pathology to some extent. AIM OF REVIEW Here, we have summarized the regulatory mechanism of the GM in CNS diseases and the potential treatment strategies for CNS repair based on GM regulation, aiming to provide safer and more effective strategies for CNS repair from the perspective of GM regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW The abundance and composition of GM is closely associated with the CNS diseases. On the basis of in-depth analysis of GM changes in mice with CNS disease, as well as the changes in its metabolites, therapeutic strategies, such as probiotics, prebiotics, and FMT, may be used to regulate GM balance and affect its microbial metabolites, thereby promoting the recovery of CNS diseases.
Collapse
Affiliation(s)
- Wei Tao
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Yanren Zhang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Bingbin Wang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Saiqun Nie
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Li Fang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Jian Xiao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yanqing Wu
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
46
|
Hernández-Rocha C, Turpin W, Borowski K, Stempak JM, Sabic K, Gettler K, Tastad C, Chasteau C, Korie U, Hanna M, Khan A, Mengesha E, Bitton A, Schwartz MB, Barrie A, Datta LW, Lazarev M, Brant SR, Rioux JD, McGovern DPB, Duerr RH, Schumm LP, Cho JH, Silverberg MS. After Surgically Induced Remission, Ileal and Colonic Mucosa-Associated Microbiota Predicts Crohn's Disease Recurrence. Clin Gastroenterol Hepatol 2025; 23:612-620.e10. [PMID: 38969076 PMCID: PMC11979954 DOI: 10.1016/j.cgh.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND & AIMS Investigating the tissue-associated microbiota after surgically induced remission may help to understand the mechanisms initiating intestinal inflammation in Crohn's disease. METHODS Patients with Crohn's disease undergoing ileocolic resection were prospectively recruited in 6 academic centers. Biopsy samples from the neoterminal ileum, colon, and rectosigmoid were obtained from colonoscopies performed after surgery. Microbial DNA was extracted for 16S rRNA gene sequencing. Microbial diversity and taxonomic differential relative abundance were analyzed. A random forest model was applied to analyze the performance of clinical and microbial features to predict recurrence. A Rutgeerts score ≥i2 was deemed as endoscopic recurrence. RESULTS A total of 349 postoperative colonoscopies and 944 biopsy samples from 262 patients with Crohn's disease were analyzed. Ileal inflammation accounted for most of the explained variance of the ileal and colonic mucosa-associated microbiota. Samples obtained from 97 patients who were in surgically induced remission at first postoperative colonoscopy who went on to develop endoscopic recurrence at second colonoscopy showed lower diversity and microbial deviations when compared with patients who remained in endoscopic remission. Depletion of genus Anaerostipes and increase of several genera from class Gammaproteobacteria at the 3 biopsy sites increase the risk of further recurrence. Gut microbiome was able to predict future recurrence better than clinical features. CONCLUSIONS Ileal and colonic mucosa-associated microbiome deviations precede development of new-onset ileal inflammation after surgically induced remission and show good predictive performance for future recurrence. These findings suggest that targeted microbial modulation is a plausible modality to prevent postoperative Crohn's disease recurrence.
Collapse
Affiliation(s)
- Cristian Hernández-Rocha
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica of Chile, Santiago, Chile
| | - Williams Turpin
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Krzysztof Borowski
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Joanne M Stempak
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ksenija Sabic
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kyle Gettler
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Christopher Tastad
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Colleen Chasteau
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ujunwa Korie
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mary Hanna
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, California
| | - Abdul Khan
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, California
| | - Emebet Mengesha
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, California
| | - Alain Bitton
- Division of Gastroenterology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Marc B Schwartz
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Arthur Barrie
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lisa W Datta
- Harvey M. and Lyn P. Meyerhoff Inflammatory Bowel Disease Center, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mark Lazarev
- Harvey M. and Lyn P. Meyerhoff Inflammatory Bowel Disease Center, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven R Brant
- Harvey M. and Lyn P. Meyerhoff Inflammatory Bowel Disease Center, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Division of Gastroenterology and Hepatology, Department of Medicine, Rutgers Robert Wood Johnson Medical School and the Crohn's and Colitis Center of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; Department of Genetics and The Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - John D Rioux
- Research Centre, Montreal Heart Institute, Montréal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Dermot P B McGovern
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, California
| | - Richard H Duerr
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania
| | - L Phil Schumm
- Biostatistics Laboratory & Research Computing Group, Department of Public Health Sciences, University of Chicago, Chicago, Illinois
| | - Judy H Cho
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mark S Silverberg
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
47
|
Caesar R. The impact of novel probiotics isolated from the human gut on the gut microbiota and health. Diabetes Obes Metab 2025; 27 Suppl 1:3-14. [PMID: 39726216 PMCID: PMC11894790 DOI: 10.1111/dom.16129] [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: 10/25/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
The gut microbiota plays a pivotal role in influencing the metabolism and immune responses of the body. A balanced microbial composition promotes metabolic health through various mechanisms, including the production of beneficial metabolites, which help regulate inflammation and support immune functions. In contrast, imbalance in the gut microbiota, known as dysbiosis, can disrupt metabolic processes and increase the risk of developing diseases, such as obesity, type 2 diabetes, and inflammatory disorders. The composition of the gut microbiota is dynamic and can be influenced by environmental factors such as diet, medication, and the consumption of live bacteria. Since the early 1900s, bacteria isolated from food and have been used as probiotics. However, the human gut also offers an enormous reservoir of bacterial strains, and recent advances in microbiota research have led to the discovery of strains with probiotic potentials. These strains, derived from a broad spectrum of microbial taxa, differ in their ecological properties and how they interact with their hosts. For most probiotics bacterial structural components and metabolites, such as short-chain fatty acids, contribute to the maintenance of metabolic and immunological homeostasis by regulating inflammation and reinforcing gut barrier integrity. Metabolites produced by probiotic strains can also be used for bacterial cross-feeding to promote a balanced microbiota. Despite the challenges related to safety, stability, and strain-specific properties, several newly identified strains offer great potential for personalized probiotic interventions, allowing for targeted health strategies.
Collapse
Affiliation(s)
- Robert Caesar
- The Wallenberg Laboratory, Department of Molecular and Clinical MedicineUniversity of GothenburgGothenburgSweden
| |
Collapse
|
48
|
Dwibedi C, Axelsson AS, Abrahamsson B, Fahey JW, Asplund O, Hansson O, Ahlqvist E, Tremaroli V, Bäckhed F, Rosengren AH. Effect of broccoli sprout extract and baseline gut microbiota on fasting blood glucose in prediabetes: a randomized, placebo-controlled trial. Nat Microbiol 2025; 10:681-693. [PMID: 39929977 PMCID: PMC11879859 DOI: 10.1038/s41564-025-01932-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/13/2025] [Indexed: 03/06/2025]
Abstract
More effective treatments are needed for impaired fasting glucose or glucose intolerance, known as prediabetes. Sulforaphane is an isothiocyanate that reduces hepatic gluconeogenesis in individuals with type 2 diabetes and is well tolerated when provided as a broccoli sprout extract (BSE). Here we report a randomized, double-blind, placebo-controlled trial in which drug-naive individuals with prediabetes were treated with BSE (n = 35) or placebo (n = 39) once daily for 12 weeks. The primary outcome was a 0.3 mmol l-1 reduction in fasting blood glucose compared with placebo from baseline to week 12. Gastro-intestinal side effects but no severe adverse events were observed in response to treatment. BSE did not meet the prespecified primary outcome, and the overall effect in individuals with prediabetes was a 0.2 mmol l-1 reduction in fasting blood glucose (95% confidence interval -0.44 to -0.01; P = 0.04). Exploratory analyses to identify subgroups revealed that individuals with mild obesity, low insulin resistance and reduced insulin secretion had a pronounced response (0.4 mmol l-1 reduction) and were consequently referred to as responders. Gut microbiota analysis further revealed an association between baseline gut microbiota and pathophysiology and that responders had a different gut microbiota composition. Genomic analyses confirmed that responders had a higher abundance of a Bacteroides-encoded transcriptional regulator required for the conversion of the inactive precursor to bioactive sulforaphane. The abundance of this gene operon correlated with sulforaphane serum concentration. These findings suggest a combined influence of host pathophysiology and gut microbiota on metabolic treatment response, and exploratory analyses need to be confirmed in future trials. ClinicalTrials.gov registration: NCT03763240 .
Collapse
Affiliation(s)
- Chinmay Dwibedi
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology and Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Annika S Axelsson
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Birgitta Abrahamsson
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jed W Fahey
- Departments of Medicine, Pharmacology and Molecular Sciences, and Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Olof Asplund
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - Ola Hansson
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
- Institute for Molecular Medicine Finland, Helsinki University, Helsinki, Finland
| | - Emma Ahlqvist
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anders H Rosengren
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
49
|
Raehtz KD, Pandrea I, Apetrei C. It's all in the gut: the central role of the gut and microbiome in preventing disease progression in simian immunodeficiency viruses infected African nonhuman primates. Curr Opin HIV AIDS 2025; 20:124-132. [PMID: 39774258 PMCID: PMC11802300 DOI: 10.1097/coh.0000000000000911] [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: 01/11/2025]
Abstract
PURPOSE OF REVIEW Typically, both HIV-infected humans and simian immunodeficiency virus (SIV)-infected Asian nonhuman primates (NHPs) eventually progress to AIDS, while African NHPs that are natural hosts of SIV do not, in spite of life-long, high levels of viral replication. Lack of disease progression in African NHPs is not due to some adaptation by the virus, but rather to host adaptations to the virus. Central to these adaptations is maintenance of the gut integrity during acute viral replication and inflammation, which allows natural hosts to avoid the chronic inflammation characteristic to pathogenic HIV/SIV infection. RECENT FINDINGS It has been recently shown that natural hosts of SIVs, such as the African green monkey (AGM), avoid damage to the mucosal epithelium through wound healing mechanisms, possibly with the contribution of a unique anti-inflammatory microbiome. Furthermore, these mechanisms are independent of viral replication, and CD4 + T-cell activation or depletion. SUMMARY Future SIV research on natural hosts should focus on further elucidating the anti-inflammatory state of their gut, and the role of microbiome/dysbiosis in the pathogenesis of SIV infection, with the goal of development new regiments or treatments to reduce or even halt the vicious cycle of gut damage and inflammation triggered by pathogenic HIV/SIV infection.
Collapse
Affiliation(s)
| | - Ivona Pandrea
- Department of Pathology, School of Medicine
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health
| | - Cristian Apetrei
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
50
|
Xie C, Qi C, Zhang J, Wang W, Meng X, Aikepaer A, Lin Y, Su C, Liu Y, Feng X, Gao H. When short-chain fatty acids meet type 2 diabetes mellitus: Revealing mechanisms, envisioning therapies. Biochem Pharmacol 2025; 233:116791. [PMID: 39894305 DOI: 10.1016/j.bcp.2025.116791] [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: 10/25/2024] [Revised: 01/19/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Evidence is accumulating that short-chain fatty acids (SCFAs) produced by the gut microbiota play pivotal roles in host metabolism. They contribute to the metabolic regulation and energy homeostasis of the host not only by preserving intestinal health and serving as energy substrates but also by entering the systemic circulation as signaling molecules, affecting the gut-brain axis and neuroendocrine-immune network. This review critically summarizes the current knowledge regarding the effects of SCFAs in the fine-tuning of the pathogenesis of type 2 diabetes mellitus (T2DM) and insulin resistance, with an emphasis on the complex relationships among diet, microbiota-derived metabolites, T2DM inflammation, glucose metabolism, and the underlying mechanisms involved. We hold an optimistic view that elucidating how diet can influence gut bacterial composition and activity, SCFA production, and metabolic functions in the host will advance our understanding of the mutual interactions of the intestinal microbiota with other metabolically active organs, and may pave the way for harnessing these pathways to develop novel personalized therapeutics for glucometabolic disorders.
Collapse
Affiliation(s)
- Cong Xie
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Cong Qi
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Jianwen Zhang
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617 China
| | - Wei Wang
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Xing Meng
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617 China
| | - Aifeila Aikepaer
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; Dongzhimen Hospital, the First Clinical Medical School of Beijing University of Chinese Medicine, Beijing 100700 China
| | - Yuhan Lin
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; Dongzhimen Hospital, the First Clinical Medical School of Beijing University of Chinese Medicine, Beijing 100700 China
| | - Chang Su
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730124 China
| | - Yunlu Liu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700 China
| | - Xingzhong Feng
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China.
| | - Huijuan Gao
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China.
| |
Collapse
|