1
|
Loison L, Huré M, Lefranc B, Leprince J, Bôle-Feysot C, Coëffier M, Ribet D. Staphylococcus warneri dampens SUMOylation and promotes intestinal inflammation. Gut Microbes 2025; 17:2446392. [PMID: 39819277 DOI: 10.1080/19490976.2024.2446392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 11/28/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025] Open
Abstract
Gut bacteria play key roles in intestinal physiology, via the secretion of diversified bacterial effectors. Many of these effectors remodel the host proteome, either by altering transcription or by regulating protein post-translational modifications. SUMOylation, a ubiquitin-like post-translational modification playing key roles in intestinal physiology, is a target of gut bacteria. Mutualistic gut bacteria can promote SUMOylation, via the production of short- or branched-chain fatty acids (SCFA/BCFA). In contrast, several pathogenic bacteria were shown to dampen SUMOylation in order to promote infection. Here, we demonstrate that Staphylococcus warneri, a natural member of the human gut microbiota, decreases SUMOylation in intestinal cells. We identify that Warnericin RK, a hemolytic toxin secreted by S. warneri, targets key components of the host SUMOylation machinery, leading to the loss of SUMO-conjugated proteins. We further demonstrate that Warnericin RK promotes inflammation in intestinal and immune cells using both SUMO-dependent and SUMO-independent mechanisms. We finally show that Warnericin RK regulates the expression of genes involved in intestinal tight junctions. Together, these results highlight the diversity of mechanisms used by bacteria from the gut microbiota to manipulate host SUMOylation. They further highlight that changes in gut microbiota composition may impact intestinal inflammation, by altering the equilibrium between bacterial effectors promoting or dampening SUMOylation.
Collapse
Affiliation(s)
- Léa Loison
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, Rouen, France
| | - Marion Huré
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, Rouen, France
| | - Benjamin Lefranc
- Univ Rouen Normandie, INSERM, Normandie Univ, NorDiC, UMR 1239, PRIMACEN, Rouen, France
| | - Jérôme Leprince
- Univ Rouen Normandie, INSERM, Normandie Univ, NorDiC, UMR 1239, PRIMACEN, Rouen, France
| | - Christine Bôle-Feysot
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, Rouen, France
| | - Moïse Coëffier
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, CHU Rouen, Department of Nutrition, CIC-CRB1404, Rouen, France
| | - David Ribet
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN, UMR 1073 Nutrition, Inflammation and Microbiota-Gut-Brain axis, Rouen, France
| |
Collapse
|
2
|
Piao YH, Luo J, Ren T, Wang A, Li YY, Pan LJ, Li XZ, Li FT, Bao YW, Zheng F, Yue H. Integrating Intestinal Biotransformation and Gut Microbiota to Uncover the Influence of Tongfu Xiexia Decoction in Rats With Constipation. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2025; 39:e10065. [PMID: 40329014 DOI: 10.1002/rcm.10065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/24/2025] [Accepted: 04/27/2025] [Indexed: 05/08/2025]
Abstract
RATIONALE Tongfu Xiexia Decoction (TFXXD), a traditional Chinese medicine formula, comprises six herbs: semen raphani, Rehmannia glutinosa, rhubarb, Magnolia officinalis, trifoliate orange, and mirabilite. The aim of this study was to investigate the effects of TFXXD on its biotransformation and microbial abundance in the gut of constipated rats. METHODS Ultrahigh-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF-MS/MS) was used to analyze the biotransformation of TFXXD in the colonic contents, and 16S rRNA sequencing was used to assess the structure and diversity of the gut microbiota across various rat groups. RESULTS We identified and analyzed 25 biotransformations of TFXXD in the colonic contents, which undergo various reactions such as deglycosylation, ring opening, and hydration in intestinal bacteria, with 14 originating from trifoliate orange, six from rhubarb, three from Rehmannia glutinosa, one from Semen raphani, and one from M. officinalis. 16S rRNA analysis revealed that TFXXD significantly enhanced the relative abundance of beneficial bacteria, such as Lactobacillus and Bacteroides while significantly reducing Oscillospira abundance. Moreover, TFXXD considerably affected the carbohydrate and amino acid metabolism. Correlation analyses revealed a significant negative correlation between Bacteroides and ACH (Acetylcholine), NO (nitric oxide), D-Lac (D-Lactate), IL-6 (Interleukin-6), TNF-α (tumor necrosis factor-α), and IL-1β (Interleukin-1β) and a significant positive correlation between Bacteroides, hesperetin, and rhein. CONCLUSIONS In conclusion, our findings indicate that TFXXD can modulate the structure and diversity of the gut microbiota and enhance the metabolic balance in constipated rats.
Collapse
Affiliation(s)
- Yu-Han Piao
- Changchun University of Chinese Medicine, Changchun, China
| | - Jing Luo
- Changchun University of Chinese Medicine, Changchun, China
| | - Tao Ren
- Changchun University of Chinese Medicine, Changchun, China
| | - Ao Wang
- Changchun University of Chinese Medicine, Changchun, China
| | - Yu-Yun Li
- Jilin City Hospital of Chemical Industry, Jilin, China
| | - Li-Jia Pan
- Changchun University of Chinese Medicine, Changchun, China
| | - Xin-Ze Li
- Changchun University of Chinese Medicine, Changchun, China
| | - Fang-Tong Li
- Changchun University of Chinese Medicine, Changchun, China
| | - Yu-Wen Bao
- Changchun University of Chinese Medicine, Changchun, China
| | - Fei Zheng
- Changchun University of Chinese Medicine, Changchun, China
| | - Hao Yue
- Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
3
|
Hetta HF, Ahmed R, Ramadan YN, Fathy H, Khorshid M, Mabrouk MM, Hashem M. Gut virome: New key players in the pathogenesis of inflammatory bowel disease. World J Methodol 2025; 15:92592. [DOI: 10.5662/wjm.v15.i2.92592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/28/2024] [Accepted: 07/23/2024] [Indexed: 11/27/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory illness of the intestine. While the mechanism underlying the pathogenesis of IBD is not fully understood, it is believed that a complex combination of host immunological response, environmental exposure, particularly the gut microbiota, and genetic susceptibility represents the major determinants. The gut virome is a group of viruses found in great frequency in the gastrointestinal tract of humans. The gut virome varies greatly among individuals and is influenced by factors including lifestyle, diet, health and disease conditions, geography, and urbanization. The majority of research has focused on the significance of gut bacteria in the progression of IBD, although viral populations represent an important component of the microbiome. We conducted this review to highlight the viral communities in the gut and their expected roles in the etiopathogenesis of IBD regarding published research to date.
Collapse
Affiliation(s)
- Helal F Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
- Division of Microbiology, Immunology and Biotechnology, Faculty of pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Rehab Ahmed
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Yasmin N Ramadan
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Hayam Fathy
- Department of Internal Medicine, Division Hepatogastroenterology, Assiut University, Assiut 71515, Egypt
| | - Mohammed Khorshid
- Department of Clinical Research, Egyptian Developers of Gastroenterology and Endoscopy Foundation, Cairo 11936, Egypt
| | - Mohamed M Mabrouk
- Department of Internal Medicine, Faculty of Medicine. Tanta University, Tanta 31527, Egypt
| | - Mai Hashem
- Department of Tropical Medicine, Gastroenterology and Hepatology, Assiut University Hospital, Assiut 71515, Egypt
| |
Collapse
|
4
|
Sartor RB. Beyond Random Fecal Microbial Transplants: Next Generation Personalized Approaches to Normalize Dysbiotic Microbiota for Treating IBD. Gastroenterol Clin North Am 2025; 54:333-350. [PMID: 40348491 DOI: 10.1016/j.gtc.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
This review and commentary outline the strong rationale for normalizing the abnormal microbiota of patients with ulcerative colitis, Crohn's disease, and pouchitis and focus on strategies to improve current variable outcomes of fecal microbial transplant (FMT) in ulcerative colitis. Applying lessons from successful FMT therapy of recurrent Clostridioides difficile and insights from basic scientific understanding of host/microbial interactions provide strategies to enhance clinical outcomes in IBD. We outline promising approaches to develop novel-defined consortia of live biotherapeutic products and combination treatments to improve current results and to optimize and personalize treatment approaches in individual patients and disease subsets.
Collapse
Affiliation(s)
- R Balfour Sartor
- Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina - Chapel Hill, Chapel Hill, NC 27517, USA; Department of Microbiology & Immunology, Center for Gastrointestinal Biology and Disease, University of North Carolina - Chapel Hill, Chapel Hill, NC 27517, USA.
| |
Collapse
|
5
|
Wang W, Wu H, Li J, Chen Z, Liu J, Xu E, Hassanin AA, Rahman SU, Chen L, Zheng R. The effect of anthocyanin extract from Lycium ruthenicum Murray on intestinal barrier function in Bamei ternary pigs. PROTOPLASMA 2025:10.1007/s00709-025-02075-9. [PMID: 40410595 DOI: 10.1007/s00709-025-02075-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 05/14/2025] [Indexed: 05/25/2025]
Abstract
The intestinal barrier is a critical defense against external pathogens and plays a central role in immune regulation and nutrient absorption. Oxidative stress and chronic inflammation in high-altitude environments can exacerbate the damage to the intestinal barrier in Baimei ternary pigs. Anthocyanin extract of Lycium ruthenicum Murray (AEL), has garnered widespread attention due to its rich anthocyanin flavonoid content, which exhibits antioxidant and anti-inflammatory properties. These properties help alleviate inflammation and oxidative stress, thereby enhancing gut function in animals. Based on this, the study employed Bamei ternary pigs and supplemented their basic diet with varying concentrations of AEL to investigate its impact on gut barrier function. The results demonstrated that AEL inhibited key factors of the intestinal Toll-like receptor pathway, including Toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), tumor necrosis factor receptor-associated protein 6 (TRAF6), and nuclear factor kappa B (NF-κB), affecting gene transcription and protein expression levels. This led to a reduction in pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), an increase in anti-inflammatory IL-10 production, and improved antioxidant capacity by enhancing total antioxidant capacity (T-AOC), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) activity, while decreasing malondialdehyde (MDA) production. Additionally, AEL improved intestinal morphology and facilitated the transcription and expression of tight junction proteins, including zonula occludens-1 (ZO-1), claudin-1 (CLDN-1), and occludin (OCLN). AEL also elevated the transcription levels of mucin 1 (MUC1) and mucin 2 (MUC2), as well as the secretion levels of polymeric immunoglobulin receptor (pIgR) and secretory immunoglobulin A (SIgA), while increased the number of intestinal goblet cells. Furthermore, dietary supplementation with AEL altered the structure of the intestinal microbiota, enhancing the abundance of beneficial bacterial genera such as Verrucomicrobiaceae, Rikenellaceae, Butyricicoccaceae, UCG-005、Rikenellaceae_RC9_gut_group、norank_f_Ruminococcaceae、Eubacterium_oxidoreducens_group, thereby promoting the production of intestinal short-chain fatty acids (SCFAs). In conclusion, AEL inhibits the Toll-like receptor pathway, reduces the production of inflammatory factors, enhances antioxidant levels, improves intestinal morphology and microbiota structure,, thereby reinforcing intestinal barrier function.
Collapse
Affiliation(s)
- Wensheng Wang
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Hua Wu
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China.
| | - Jinming Li
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Zixin Chen
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Jiayi Liu
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Enron Xu
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Abdallah A Hassanin
- Genetics Department, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Siddiq Ur Rahman
- Department of Computer Science and Bioinformatics, Khushal Khan Khattak University, Karak, Pakistan
| | - Lin Chen
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Runtao Zheng
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| |
Collapse
|
6
|
Aminian-Dehkordi J, Dickson A, Valiei A, Mofrad MRK. MetaBiome: a multiscale model integrating agent-based and metabolic networks to reveal spatial regulation in gut mucosal microbial communities. mSystems 2025; 10:e0165224. [PMID: 40183581 PMCID: PMC12090770 DOI: 10.1128/msystems.01652-24] [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: 12/09/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Mucosal microbial communities (MMCs) are complex ecosystems near the mucosal layers of the gut essential for maintaining health and modulating disease states. Despite advances in high-throughput omics technologies, current methodologies struggle to capture the dynamic metabolic interactions and spatiotemporal variations within MMCs. In this work, we present MetaBiome, a multiscale model integrating agent-based modeling (ABM), finite volume methods, and constraint-based models to explore the metabolic interactions within these communities. Integrating ABM allows for the detailed representation of individual microbial agents each governed by rules that dictate cell growth, division, and interactions with their surroundings. Through a layered approach-encompassing microenvironmental conditions, agent information, and metabolic pathways-we simulated different communities to showcase the potential of the model. Using our in-silico platform, we explored the dynamics and spatiotemporal patterns of MMCs in the proximal small intestine and the cecum, simulating the physiological conditions of the two gut regions. Our findings revealed how specific microbes adapt their metabolic processes based on substrate availability and local environmental conditions, shedding light on spatial metabolite regulation and informing targeted therapies for localized gut diseases. MetaBiome provides a detailed representation of microbial agents and their interactions, surpassing the limitations of traditional grid-based systems. This work marks a significant advancement in microbial ecology, as it offers new insights into predicting and analyzing microbial communities.IMPORTANCEOur study presents a novel multiscale model that combines agent-based modeling, finite volume methods, and genome-scale metabolic models to simulate the complex dynamics of mucosal microbial communities in the gut. This integrated approach allows us to capture spatial and temporal variations in microbial interactions and metabolism that are difficult to study experimentally. Key findings from our model include the following: (i) prediction of metabolic cross-feeding and spatial organization in multi-species communities, (ii) insights into how oxygen gradients and nutrient availability shape community composition in different gut regions, and (iii) identification of spatiallyregulated metabolic pathways and enzymes in E. coli. We believe this work represents a significant advance in computational modeling of microbial communities and provides new insights into the spatial regulation of gut microbiome metabolism. The multiscale modeling approach we have developed could be broadly applicable for studying other complex microbial ecosystems.
Collapse
Affiliation(s)
- Javad Aminian-Dehkordi
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California, USA
| | - Andrew Dickson
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California, USA
| | - Amin Valiei
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California, USA
| | - Mohammad R. K. Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California, USA
- Molecular Biophysics and Integrative Bioimaging Division, Lawrence Berkeley National Lab, Berkeley, California, USA
| |
Collapse
|
7
|
Gu M, Hong Q, Cui J, Li W, Zhang J, Sun Y, Jiang J, Hu Y. Harnessing lactobacillus: a probiotic revolution in combating metabolic dysfunction-associated steatotic liver disease. Crit Rev Food Sci Nutr 2025:1-18. [PMID: 40370039 DOI: 10.1080/10408398.2025.2504162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) involves fat buildup in the liver and is connected to a disruption in gut microbiota and a reduction in beneficial microbiota. Lactobacillus is an important probiotic that can grow in anaerobic or low-oxygen environment and is widely used in food industry and health fields. In recent years, the improvement of MASLD by Lactobacillus has been extensively studied. However, the detailed mechanisms by which different species of Lactobacillus improve MASLD have not been summarized. In this review, we present the potential of Lactobacillus as a non-drug approach to mitigate MASLD. We will discuss the preclinical and clinical research backing this method and the ways these probiotics improve MASLD. This review mainly presents the different species of Lactobacillus that improve MASLD, and various mechanisms by which Lactobacillus strains alleviate MASLD, including improving intestinal permeability and inflammation, reducing oxidative stress, and restoring of gut metabolites. Future research into mechanisms and larger clinical trials will help confirm the effectiveness of using Lactobacillus and related genera in the treatment of MASLD.
Collapse
Affiliation(s)
- Minwen Gu
- College of Biological and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Qing Hong
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd, Shanghai, China
| | - Jie Cui
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, China
| | - Wenhui Li
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, China
| | - Jian Zhang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, China
| | - Ye Sun
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, China
| | - Jinchi Jiang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, China
| | - Yonghong Hu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, China
| |
Collapse
|
8
|
Chen C, Qi M, Xu Z, Wen J, Tang W, Diao H, Li Z, Chu Y, Feng F, Tang Z. Sesamin improved growth and overall health in young animals by enhancing gut-liver axis function. Food Funct 2025. [PMID: 40351157 DOI: 10.1039/d4fo05933f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
The immature gastrointestinal development of infants and young animals made them more vulnerable to stress-related damage, which affected the gut-liver axis and consequently impaired their health and growth. This study used weaned piglets as a model to investigate how dietary sesamin regulated the gut-liver axis and impacted young animal health. We assessed gut-liver tissue morphology, measured key indicators of intestinal barrier damage, mucosal repair, antioxidant and immune pathways in the gut-liver system and serum, and analyzed microbial composition. We further explored the interactions between sesamin and the gut-liver axis through PLS-PM and molecular docking analysis. Results showed that sesamin enhanced intestinal barrier function, reduced liver damage, decreased oxidative stress, promoted anti-inflammatory immune responses, and enriched beneficial microbes, thereby promoting overall growth. Sesamin can enhance the health of young animals by regulating the function of the gut-liver axis.
Collapse
Affiliation(s)
- Chen Chen
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Min Qi
- Yunnan Animal Husbandry Station, Kunming 650225, China
| | - Zhiran Xu
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Jincheng Wen
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Wenjie Tang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtche Group Co., Ltd., Chengdu 610066, China
| | - Hui Diao
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtche Group Co., Ltd., Chengdu 610066, China
| | - Zhangcheng Li
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Yunyun Chu
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Fu Feng
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Zhiru Tang
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
9
|
Ye J, Raman M, Taylor LM, Yousuf M, Panaccione R, Turbide C, Sinha SR, Haskey N. Reduced Sulfur Diet Reshapes the Microbiome and Metabolome in Mild-Moderate Ulcerative Colitis. Int J Mol Sci 2025; 26:4596. [PMID: 40429741 DOI: 10.3390/ijms26104596] [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: 04/07/2025] [Revised: 05/01/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
This pilot study investigated the effects of a reduced sulfur (RS) diet on the gut microbiome composition and fecal metabolome in individuals with remitted or active ulcerative colitis (UC). Thirteen participants maintained their habitual diet (control), while nine followed an RS diet for eight weeks (Wk8). Stool and plasma samples were collected at the baseline and Wk8. The sulfur intake decreased in the RS group (-28 g/1000 kcal) versus the control group (-1.7 g/1000 kcal; p < 0.001). The RS group exhibited a significant decrease in lipopolysaccharide-binding protein (-5280 ng/mL), while these levels increased in the control group (620 ng/mL; p < 0.05). The microbiome analysis showed an increased alpha diversity at Wk8 (p < 0.01), suggesting a microbial shift with a RS intake. The metabolic alterations indicated enhanced nitrogen disposal (increased uric acid, methyluric acid, N-acetyl-L-glutamate) and a higher energy demand (elevated ubiquinol and glucose-pyruvate). The RS diet increased beneficial microbes Collinsella stercoris, Asaccharobacter celatus, and Alistipes finegoldii, while decreasing pathobionts Eggerthella lenta and Romboutsia ilealis. Methyluric acid correlated positively with C. stercoris (β = 0.70) and negatively with E. lenta (β = -0.77) suggesting these microbes utilized this metabolite and influenced the microbiome composition. In conclusion, a RS diet promoted microbial diversity, metabolic adaptations, and reduced inflammation, highlighting its potential as a novel strategy for UC management.
Collapse
Affiliation(s)
- Jiayu Ye
- Division of Gastroenterology and Hepatology, Stanford Medicine, Stanford University, 300 Pasteur Dr., Palo Alto, CA 94305, USA
| | - Maitreyi Raman
- Department of Medicine, Cumming School of Medicine, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Lorian M Taylor
- Department of Medicine, Cumming School of Medicine, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Munazza Yousuf
- Department of Medicine, Cumming School of Medicine, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Remo Panaccione
- Department of Medicine, Cumming School of Medicine, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Christian Turbide
- Department of Medicine, Cumming School of Medicine, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Sidhartha R Sinha
- Division of Gastroenterology and Hepatology, Stanford Medicine, Stanford University, 300 Pasteur Dr., Palo Alto, CA 94305, USA
| | - Natasha Haskey
- Department of Biology, Irving K Barber Faculty of Science, University of British Columbia-Okanagan, 3187 University Way, Kelowna, BC V1V 1V7, Canada
| |
Collapse
|
10
|
Rivera K, González L, Parra L, Oyarzún JE, Concepción-Alvarez A, de Camargo AC, Bridi R, Rigotti A, Andia ME. Red Wine Grape Pomace Restores Gut Barrier Function and Improves Survival in Diet-Induced Ischemic Heart Disease. Antioxidants (Basel) 2025; 14:574. [PMID: 40427456 DOI: 10.3390/antiox14050574] [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: 04/15/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Red wine grape pomace (RWGP), a winemaking by-product rich in phenolics, flavonoids, and dietary fiber, has shown promise in mitigating cardiovascular disease (CVD), however, its mechanisms of action remain incompletely understood. This study comprehensively profiled the phenolic composition of RWGP-including free, esterified, etherified, and insoluble-bound fractions-and evaluated the effects of RWGP dietary supplementation on gut barrier integrity, inflammation, oxidative stress, and survival in SR-B1-/-ApoE-R61h/h mice, a model of diet-induced lethal ischemic heart disease. RWGP supplementation significantly improved survival rates and restored gut barrier function, as evidenced by lower plasma FITC-dextran and LPS levels, increased circulating ZO-1 levels, and reduced histopathological colon damage. In addition, RWGP reduced pro-inflammatory cytokines (IL-1β) and showed a trend toward attenuating systemic oxidative stress (TBARS). Analysis of phenolic compounds indicated a significant presence of insoluble-bound phenolics. Nevertheless, the beneficial effects observed are likely attributable to the synergistic actions of RWGP's complex phytochemical and fiber composition. These results highlight RWGP's potential as a sustainable, gut-targeted functional food ingredient for CVD prevention and management.
Collapse
Affiliation(s)
- Katherine Rivera
- Doctoral Program in Medical Sciences, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
- Radiology Department and Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago 7820436, Chile
| | - Leticia González
- Radiology Department and Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Laura Parra
- Doctoral Program in Medical Sciences, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
- Radiology Department and Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago 7820436, Chile
| | - Juan E Oyarzún
- Radiology Department and Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago 7820436, Chile
| | | | | | - Raquel Bridi
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 8380494, Chile
| | - Attilio Rigotti
- Centro de Nutrición Molecular y Enfermedades Crónicas, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Marcelo E Andia
- Radiology Department and Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago 7820436, Chile
| |
Collapse
|
11
|
Chen L, Yan H, Di S, Guo C, Zhang H, Zhang S, Gold A, Wang Y, Hu M, Wu D, Johnson CH, Wang X, Zhu J. Mapping pesticide-induced metabolic alterations in human gut bacteria. Nat Commun 2025; 16:4355. [PMID: 40348778 PMCID: PMC12065874 DOI: 10.1038/s41467-025-59747-6] [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: 10/24/2024] [Accepted: 04/30/2025] [Indexed: 05/14/2025] Open
Abstract
Pesticides can modulate gut microbiota composition, but their specific effects on it remain largely elusive. In our study, we show that pesticides inhibit or promote the growth of various gut microbial species and can be accumulated to prolong their presence in the host. Pesticide exposure also induces significant alterations in gut bacterial metabolism, as reflected by changes in hundreds of metabolites. We generate a pesticide-gut microbiota-metabolite network that not only reveals pesticide-sensitive gut bacteria species but also reports specific metabolic changes in 306 pesticide-gut microbiota pairs. Using an in vivo mouse model, we further demonstrate the interactions of a representative pesticide-gut microbiota pair and verify the inflammation-inducing effects of pesticide exposure on the host, mediated by microbially dysregulated lipid metabolism. Taken together, our findings generate a comprehensive atlas of pesticide-gut microbiota-metabolite interactions atlas and shed light on the molecular mechanisms by which pesticides affect host health via gut microbiota-modulated metabolism.
Collapse
Affiliation(s)
- Li Chen
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Hong Yan
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
| | - Shanshan Di
- State Key Laboratory of Agricultural Products Safety/ Key Laboratory of Detection for Pesticide Residues and Control of Zhejiang, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Chao Guo
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Huan Zhang
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Shiqi Zhang
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Andrew Gold
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Yu Wang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ming Hu
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Dayong Wu
- Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Caroline H Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Xinquan Wang
- State Key Laboratory of Agricultural Products Safety/ Key Laboratory of Detection for Pesticide Residues and Control of Zhejiang, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Jiangjiang Zhu
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA.
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
12
|
Karukayil Gopalakrishnan N, Pappuswamy M, Meganathan G, Shanmugam S, Pushparaj K, Balasubramanian B, Kim IH. Influence of Probiotic Administration in Canine Feed: A Comprehensive Review. Vet Sci 2025; 12:449. [PMID: 40431542 DOI: 10.3390/vetsci12050449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/30/2025] [Accepted: 05/04/2025] [Indexed: 05/29/2025] Open
Abstract
Dogs are cherished companions, and in today's world, pets are increasingly regarded as family members. Pet owners are placing growing emphasis on their animals' health, particularly for dogs. Probiotics, which are living bacteria that benefit the host when given in sufficient quantities, have drawn a lot of interest in the veterinary nutrition community due to their beneficial effects on companion animals, including dogs. This study emphasizes the advantages of adding probiotics to canine diets in order to enhance the health of the gut flora and the technologies used to incorporate probiotics into canine feed. It looks at the best ways to deal with common dog health problems, highlighting probiotics as a helpful substitute for antibiotics, which can have serious adverse effects, encourage bacterial resistance, and disturb the gut's microbial ecology, which is necessary for digesting. Such disruptions are linked to chronic inflammatory enteropathy and obesity in dogs. This paper also examines biotechnological advancements in probiotic incorporation methods in dog feed, aiming to optimize their health benefits. Probiotic feed supplements may thus represent a promising approach to advancing canine health care, providing a natural adjunct to conventional treatments and preventive measures.
Collapse
Affiliation(s)
| | - Manikantan Pappuswamy
- Department of Life Sciences, School of Sciences, Christ University, Bangalore 560029, Karnataka, India
| | - Gomathy Meganathan
- Department of Life Sciences, School of Sciences, Christ University, Bangalore 560029, Karnataka, India
| | - Sureshkumar Shanmugam
- Department of Animal Resource and Science, Dankook University, Cheonan-si 31116, Chungnam, Republic of Korea
| | - Karthika Pushparaj
- Department of Zoology, School of Biosciences, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, Tamil Nadu, India
| | | | - In Ho Kim
- Department of Animal Resource and Science, Dankook University, Cheonan-si 31116, Chungnam, Republic of Korea
| |
Collapse
|
13
|
Zou T, Tang X, Wang H, Shang X, Liang X, Ma X. Nanocrystalline cellulose-geniposide complex enhances gut-brain axis modulation for depression treatment. Commun Biol 2025; 8:667. [PMID: 40287572 PMCID: PMC12033350 DOI: 10.1038/s42003-025-07934-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: 05/30/2024] [Accepted: 03/13/2025] [Indexed: 04/29/2025] Open
Abstract
Depression, a major global health issue, is closely associated with imbalances in gut microbiota and altered intestinal functions. This study investigates the antidepressant potential of a composite of Geniposide (GP) and Nanocrystalline Cellulose (NCC), focusing on its effects on the gut-brain axis. Utilizing network pharmacology, GP was identified as a key compound targeting the BCL2 gene in depression management. Experimental approaches, including a chronic unpredictable mild stress (CUMS) model in mice, cellular assays, and fecal microbiota transplantation (FMT), were used to evaluate the composite's effectiveness. Results indicate that GP activates the adenosine monophosphate-activated protein kinase (AMPK) pathway by upregulating BCL2, enhancing intestinal barrier integrity, and balancing gut flora. These mechanisms contribute to its positive effects on hippocampal function and depressive-like behaviors in mice, suggesting that the GP-NCC composite could be a promising avenue for developing depression therapies that target gut health.
Collapse
Affiliation(s)
- Tianyu Zou
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China.
| | - Xiang Tang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Haiping Wang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xiaolong Shang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xiaoyu Liang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xuemiao Ma
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| |
Collapse
|
14
|
Zhang X, Pan Q, Yao G, Kong D, Chen H, Zhang Q, Wang Z. Bacteroides fragilis and propionate synergize with low-dose methimazole to treat Graves' disease. Microbiol Spectr 2025:e0318624. [PMID: 40265938 DOI: 10.1128/spectrum.03186-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
Graves' disease (GD) is a common autoimmune thyroid disease with limited treatment efficacy, high relapse rates, and severe adverse effects. Gut microbiota dysbiosis is thought to play a critical role in GD, with the potential for microbiota-based therapies in GD treatment. However, experimental evidence is needed to validate this hypothesis. In this study, we evaluated the therapeutic effects of the commensal bacterium B. fragilis and its metabolite, propionate, in a GD mouse model. We found that oral supplementation with B. fragilis or propionate significantly reduced serum levels of inflammatory cytokines, total thyroxine (TT4), and TSH receptor antibodies (TRAb). It also decreased the proportion of circulating Th17 cells while increasing the proportion of circulating regulatory T cells (Tregs), thus mitigating systemic inflammation, hyperthyroidism, and the autoimmune response against TSHR. In addition, B. fragilis and propionate significantly decreased the levels of inflammatory cytokines and the proportion of M1 macrophages in thyroid tissue, while increasing the proportions of Treg cells and M2 macrophages, thereby reducing thyroid inflammation and size. Notably, the combination of B. fragilis or propionate with methimazole (MMI) significantly ameliorated pathological changes in GD mice and markedly reduced MMI dosage requirements, demonstrating a synergistic therapeutic effect. Our findings suggest that B. fragilis and propionate could serve as effective adjuvant agents in combination with MMI, thereby enhancing therapeutic efficacy while reducing drug dosage and minimizing side effects. This study opens a new avenue for microbiota-based treatments in managing GD.IMPORTANCEThis study explores a new approach to treat Graves' disease (GD), a major type of hyperthyroidism. Traditional treatments for GD often come with significant side effects and high relapse rates. Researchers found that Bacteroides fragilis, a gut commensal bacterium, and its metabolite propionate can improve the condition of GD mice. When combined with methimazole, a conventional medication for GD treatment, these natural agents demonstrated enhanced therapeutic efficacy, enabling dose reduction of methimazole and consequently reducing adverse effects. This research suggests that combining gut microbiota-based treatments with standard therapies may offer a more effective and safer way to manage GD.
Collapse
Affiliation(s)
- Xinjie Zhang
- Department of Geriatrics, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Biology, University College London, London, United Kingdom
| | - Qingxin Pan
- Shandong First Medical University Second Affiliated Hospital, Tai'an, Shandong, China
| | - Guixiang Yao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, Shandong, China
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Jinan, Shandong, China
| | - Danxia Kong
- Department of Occupational Lung Disease, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong, China
| | - Haiyan Chen
- Department of Geriatrics, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qunye Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, Shandong, China
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Jinan, Shandong, China
| | - Zhe Wang
- Department of Geriatrics, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
15
|
Mahgoup EM. "Gut Microbiota as a Therapeutic Target for Hypertension: Challenges and Insights for Future Clinical Applications" "Gut Microbiota and Hypertension Therapy". Curr Hypertens Rep 2025; 27:14. [PMID: 40261509 DOI: 10.1007/s11906-025-01331-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE OF REVIEW Systemic hypertension is a major risk factor for cardiovascular disease and remains challenging to manage despite the widespread use of antihypertensive medications and lifestyle modifications. This review explores the role of gut microbiota in hypertension development and regulation, highlighting key mechanisms such as inflammation, gut-brain axis modulation, and bioactive metabolite production. We also assess the potential of microbiota-targeted therapies for hypertension management. RECENT FINDINGS Emerging evidence indicates that microbial dysbiosis, high-salt diets, and gut-derived metabolites such as short-chain fatty acids (SCFAs) and bile acids significantly influence blood pressure regulation. Preclinical and early clinical studies suggest that interventions targeting gut microbiota, including probiotics, prebiotics, synbiotics, fecal microbiota transplantation (FMT), and dietary modifications, may help modulate hypertension. However, variability in gut microbiota composition among individuals and limited human trial data pose challenges to translating these findings into clinical practice. While microbiota-based therapies show promise for hypertension management, further research is needed to establish their efficacy and long-term effects. Large-scale, standardized clinical trials are crucial for understanding the therapeutic potential and limitations of gut microbiota interventions. A deeper understanding of the gut-hypertension axis could lead to novel, personalized treatment strategies for hypertension.
Collapse
Affiliation(s)
- Elsayed M Mahgoup
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt.
- Department of Internal Medicine, Division of Cardiovascular Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
16
|
Abdu SMN, Abdalla IM, Zhen Y, Zhang C, Xi Z, Ma J, Zhong Y, Lin J, Ali R, Wang M. Gastric Infusion of Short-Chain Fatty Acids Improves Health via Enhance Liver and Intestinal Immune Response and Antioxidant Capacity in Goats. Vet Sci 2025; 12:395. [PMID: 40431488 DOI: 10.3390/vetsci12050395] [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: 02/11/2025] [Revised: 03/17/2025] [Accepted: 04/04/2025] [Indexed: 05/29/2025] Open
Abstract
In the present study, we comprehensively investigated the impacts of the infusion of three short-chain fatty acids (SCFAs), sodium acetate (SA), propionate (SP), and butyrate (SB), to examine their respective roles in the gastrointestinal tract (GIT) health and innate immunity of twenty adult Guanzhong milk goats of 1.5 years of age. Infusion of SCFAs resulted in upregulating the activity of certain antioxidant enzymes in comparison with the control group. The SA group significantly (p < 0.05) increased the activity of the catalase (CAT) in the liver, glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD) in the colon, and maleic dialdehyde (MDA) in the jejunum. SP significantly (p < 0.05) upregulated the activity of the total antioxidant capacity (T-AOC) in the ileum, CAT and MDA in the jejunum, CAT in the colon, and SOD in the liver. SB was significantly (p < 0.05) upregulated the activity of the T-AOC in the ileum, CAT in the jejunum, and T-AOC, CAT, SOD, and GSH-Px in the colon. Infusion of SCFAs resulted in significant (p < 0.05) increases in pro-inflammatory and anti-inflammatory cytokines in the intestine compared to the control group. We found that the SA group significantly (p < 0.05) upregulated the level of interleukin-1 beta (IL-1β) in the ileum and jejunum, as well as the levels of IL-6 and TNF-α in the colon, while the SP group significantly (p < 0.05) increased the level of IL-1β in the jejunum and the level of interleukin-10 (IL-10) in the colon. Furthermore, the SB group significantly (p < 0.05) upregulated levels of IL-1β in the jejunum, interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α) in the colon, and IL-10 in the cecum. Furthermore, some intestinal tight-junction proteins were significantly increased by SCFA infusion. SA significantly (p < 0.05) increased the claudin level in the ileum and occludin in the colon, while the SP group significantly (p < 0.05) upregulated the level of occludin in the jejunum and the claudin level in the ileum. Moreover, SB significantly (p < 0.05) increased the occludin level in the jejunum, claudin level in the ileum, and zonula occludens-1 (ZO-1) level in the colon and cecum. There are many positive associations among antioxidant, inflammatory cytokine, and tight-junction protein indexes in the liver and intestine. In conclusion, our results suggest that the gastric infusion of SA, SP, and SB might improve goat intestinal health through the positive influence on the antioxidant capacity, pro-inflammatory and anti-inflammatory cytokines, and tight-junction proteins.
Collapse
Affiliation(s)
- Shaima Mohmed Nasr Abdu
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | | | - Yongkang Zhen
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Chong Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zanna Xi
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jianjun Ma
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yuhong Zhong
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jiaqi Lin
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Rahmat Ali
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
17
|
Ma X, Duan C, Wang X, Tao Y, Yang L, Teng Y, Pan Y, Zhang M, Xu J, Sheng J, Wang X, Jin P. Human gut microbiota adaptation to high-altitude exposure: longitudinal analysis over acute and prolonged periods. Microbiol Spectr 2025:e0291624. [PMID: 40257273 DOI: 10.1128/spectrum.02916-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025] Open
Abstract
This study investigated the longitudinal effects of acute (7-day) and prolonged (3-month) high-altitude exposure on gut microbiota in healthy adult males, addressing the limited data available in human populations. A cohort of 406 healthy adult males was followed, and fecal samples were collected at three time points: baseline at 800 m (406 samples), 7 days after ascending to 4,500 m (406 samples), and 2 weeks post-return to 800 m following 3 months at high altitude (186 samples). High-throughput 16S ribosomal DNA sequencing was employed to analyze microbiota composition and diversity. Results revealed significant changes in alpha- and beta-diversity, with acute high-altitude exposure inducing more pronounced effects compared to prolonged exposure. Specifically, acute exposure increased opportunistic pathogens (Ruminococcus and Oscillibacter) but decreased beneficial short-chain fatty acid producers (Faecalibacterium and Bifidobacterium). Notably, these changes in microbiota persisted even after returning to low altitude, indicating long-term remodeling. Functional analyses revealed substantial changes in metabolic pathways, suggesting microbiota-driven adaptations to energy utilization under high-altitude hypoxic conditions. In summary, acute high-altitude exposure caused dramatic changes in gut microbiota, while prolonged exposure led to structural and functional reshaping. These findings enhance our understanding of how high-altitude environments reshape gut microbiota. IMPORTANCE This study is the first to investigate the impact of high-altitude exposure on gut microbiota adaptation in a large-scale longitudinal cohort. It seeks to enhance understanding of how high-altitude environments reshape gut microbiota. Acute exposure to high altitude significantly affected both α-diversity and β-diversity of gut microbiota, with acute exposure causing more pronounced changes than prolonged adaptation, indicating temporary disruptions in microbial communities. Notable shifts in microbial abundance were observed, including increased levels of genera linked to hypoxic stress (e.g., Gemmiger, Ruminococcus, and Parabacteroides) and decreased levels of beneficial bacteria (e.g., Faecalibacterium, Roseburia, and Bifidobacterium), suggesting possible adverse health effects. Functional analysis indicated changes in metabolism-related pathways post-exposure, supporting the idea that high-altitude adaptations involve metabolic adjustments for energy management. These findings enhance understanding of high-altitude physiology, illustrating the role of gut microbiota in hypoxic health.
Collapse
Affiliation(s)
- Xianzong Ma
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | | | - Xiaoying Wang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yurong Tao
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lang Yang
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yongsheng Teng
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Yuanming Pan
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Mingjie Zhang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Junfeng Xu
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jianqiu Sheng
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Xin Wang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Peng Jin
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
18
|
Chang L, Thaiss CA, Knight Z, Khalsa S. Gut Feelings: The Critical Role of Interoception in Obesity and Disorders of Gut-Brain Interaction. Gastroenterology 2025:S0016-5085(25)00637-7. [PMID: 40250772 DOI: 10.1053/j.gastro.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/29/2025] [Accepted: 04/10/2025] [Indexed: 04/20/2025]
Affiliation(s)
- Lin Chang
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Christoph A Thaiss
- Department of Pathology, Stanford University, Stanford, CA; Arc Institute, Palo Alto, CA
| | - Zachary Knight
- Department of Physiology, University of California, San Francisco, San Francisco, CA
| | - Sahib Khalsa
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
19
|
Jamali F, Mousavi S, Homayouni-Rad A, Meshkini A, Alikhah H, Houshyar J, Kamalledin Moghadam S, Yaghoubi SM, Motlagh Asghari K, Torbati Ilkhchi M, Naseri Alavi SA. Exploring Innovative Approaches for Managing Spinal Cord Injury: A Comprehensive Review of Promising Probiotics and Postbiotics. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10513-6. [PMID: 40232596 DOI: 10.1007/s12602-025-10513-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2025] [Indexed: 04/16/2025]
Abstract
Spinal cord injury (SCI) affects millions of people worldwide annually, presenting significant challenges in functional recovery despite therapeutic advancements. Current treatment strategies predominantly focus on stabilizing the spinal cord and facilitating neural repair, yet their effectiveness remains uncertain and controversial. Recent scientific investigations have explored the potential of probiotics and postbiotics to modulate inflammation, influence neurotransmitters, and aid in tissue repair, marking a potential paradigm shift in SCI management. This review critically evaluates these innovative approaches, emphasizing their ability to harness the natural properties of microorganisms within the body to potentially enhance outcomes in SCI treatment. By analyzing the latest research findings, this review provides valuable insights into how probiotics and postbiotics can revolutionize inflammation management and neurological recovery following SCI, underscoring their promising role in future therapeutic strategies aimed at improving the quality of life of SCI patients globally.
Collapse
Affiliation(s)
- Fereshteh Jamali
- Neurosurgery Department, Children'S Hospital at Montefiore, New York City, USA
| | - Safa Mousavi
- Department of Public Health, College of Health and Human Services, California State University, Fresno, CA, USA
| | - Aziz Homayouni-Rad
- Department of Food Science and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Meshkini
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Jalil Houshyar
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Kamalledin Moghadam
- Department of Food Science and Technology, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | - Kimia Motlagh Asghari
- Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | |
Collapse
|
20
|
Oami T, Yamamoto A, Ishida S, Kondo K, Hata N, Oshima T. Critical Care Nutrition from a Metabolic Point of View: A Narrative Review. Nutrients 2025; 17:1352. [PMID: 40284216 PMCID: PMC12029973 DOI: 10.3390/nu17081352] [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: 03/05/2025] [Revised: 04/08/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Critical illness induces profound metabolic alterations, characterized by a hypermetabolic state, insulin resistance, protein catabolism, and gut barrier dysfunction, which contribute to increased morbidity and mortality. Emerging evidence highlights the role of the gut microbiome and its metabolites in modulating systemic inflammation and immune responses during critical illness. This narrative review explores the metabolic evolution of critically ill patients, the impact of gut dysbiosis on disease progression, and the potential role of nutrition in modulating metabolism and improving patient outcomes. Methods: A comprehensive literature search was conducted across PubMed and Google Scholar for articles published up to February 2025. Search terms included "critical illness", "metabolism", "gut microbiota", "nutrition", and related keywords. Articles published in English addressing metabolic alterations, microbiome changes, and nutritional strategies in critically ill patients were included. After screening for eligibility, relevant articles were synthesized to outline current knowledge and identify gaps. Results: Metabolic changes in critical illness progress through distinct phases, from catabolism-driven hypermetabolism to gradual recovery. Gut dysbiosis, characterized by a loss of microbial diversity and increased gut permeability, contributes to systemic inflammation and organ dysfunction. Nutritional strategies, including enteral nutrition, probiotics, prebiotics, and metabolomics-driven interventions, may help restore microbial balance, preserve gut barrier integrity, and modulate immune and metabolic responses. Future nutrition therapy should focus on metabolic modulation rather than solely addressing nutrient deficits. Conclusions: Advances in gut microbiome research and metabolomics offer new avenues for personalized nutrition strategies tailored to the metabolic demands of critically ill patients. Integrating these approaches may improve clinical and functional recovery while mitigating the long-term consequences of critical illness.
Collapse
Affiliation(s)
- Takehiko Oami
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan; (T.O.)
| | - Akiyuki Yamamoto
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan; (T.O.)
| | - Shigenobu Ishida
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan; (T.O.)
| | - Kengo Kondo
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan; (T.O.)
| | - Nanami Hata
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan; (T.O.)
| | - Taku Oshima
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan; (T.O.)
- Institute for Advanced Academic Research, Chiba University, Chiba 263-8522, Japan
- Research Institute of Disaster Medicine, Chiba University, Chiba 263-8522, Japan
| |
Collapse
|
21
|
Zhang Z, Jiang C, Xing YQ, Yang T, Zou L, Jia Z, Zhao L, Han X, Qu X, Zhang Z, Zong J, Wang S. Unveiling the interplay among skin microbiota, cytokines, and T2DM: an insightful Mendelian randomization study. Nutr Metab (Lond) 2025; 22:29. [PMID: 40211330 PMCID: PMC11987181 DOI: 10.1186/s12986-025-00922-3] [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: 10/03/2024] [Accepted: 03/26/2025] [Indexed: 04/13/2025] Open
Abstract
BACKGROUND Previous observational studies have indicated a correlation between the skin microbiome and Type 2 diabetes (T2DM). It is hypothesized that this causal relationship may be influenced by inflammatory responses. However, these factors as determinants of T2DM remain largely unexplored. METHOD This study incorporated data from the GWAS database on the skin microbiome, 91 types of inflammatory cytokines, and T2DM. We employed two-sample MR and multivariable MR methods to assess the correlation between the skin microbiome and T2DM, and to investigate whether this correlation is affected by inflammatory cytokines. RESULTS The results of the two-sample MR analysis indicate that within the skin microbiome, genetically predicted genus: Acinetobacter, class: Alphaproteobacteria, genus: Bacteroides, ASV005[Propionibacterium granulosum], and ASV072[Rothia mucilaginosa] are associated with an increased risk of T2DM, while phylum: Proteobacteria, genus: Enhydrobacter, family: Clostridiales, ASV006[Staphylococcus hominis] serve as protective factors against T2DM. Among the inflammatory cytokines, levels of Macrophage colony-stimulating factor 1, Tumor necrosis factor receptor superfamily member 9, Urokinase-type plasminogen activator, and C-C motif chemokine 28 are associated with an increased risk of T2DM. Multivariable MR analysis further revealed that Macrophage colony-stimulating factor 1 levels act as a mediating factor between ASV072[Rothia mucilaginosa] and T2DM. CONCLUSION In this study, we found a connection between the skin microbiome and T2DM, with inflammatory cytokines playing a key role in this relationship. This research helps us better understand this complex link and shows that addressing inflammation is important for preventing and treating diabetes. This could greatly benefit public health by reducing the impact of diabetes and its complications. Our results suggest that future studies should explore the specific biological interactions between the skin microbiome and diabetes to develop more effective risk management and treatment strategies from a microbial perspective.
Collapse
Grants
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
- College of Integrative Medicine, Dalian Medical University, Dalian, China.
| | - Chunyu Jiang
- Department of Trauma Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yi-Qi Xing
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tianke Yang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- College of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Linxuan Zou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhuqiang Jia
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lin Zhao
- Department of Quality Management, Dalian Municipal Central Hospital, Dalian, China
| | - Xin Han
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xueling Qu
- Pelvic Floor Repair Center, Dalian Women and Children Medical Center (Group), Dalian, China
| | - Zhen Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Junwei Zong
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Shouyu Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
22
|
Zhra M, Elahi MA, Tariq A, Abu-Zaid A, Yaqinuddin A. Sirtuins and Gut Microbiota: Dynamics in Health and a Journey from Metabolic Dysfunction to Hepatocellular Carcinoma. Cells 2025; 14:466. [PMID: 40136715 PMCID: PMC11941559 DOI: 10.3390/cells14060466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction leading to non-alcoholic fatty liver disease (NAFLD) exhibits distinct molecular and immune signatures that are influenced by factors like gut microbiota. The gut microbiome interacts with the liver via a bidirectional relationship with the gut-liver axis. Microbial metabolites, sirtuins, and immune responses are pivotal in different metabolic diseases. This extensive review explores the complex and multifaceted interrelationship between sirtuins and gut microbiota, highlighting their importance in health and disease, particularly metabolic dysfunction and hepatocellular carcinoma (HCC). Sirtuins (SIRTs), classified as a group of NAD+-dependent deacetylases, serve as crucial modulators of a wide spectrum of cellular functions, including metabolic pathways, the inflammatory response, and the process of senescence. Their subcellular localization and diverse functions link them to various health conditions, including NAFLD and cancer. Concurrently, the gut microbiota, comprising diverse microorganisms, significantly influences host metabolism and immune responses. Recent findings indicate that sirtuins modulate gut microbiota composition and function, while the microbiota can affect sirtuin activity. This bidirectional relationship is particularly relevant in metabolic disorders, where dysbiosis contributes to disease progression. The review highlights recent findings on the roles of specific sirtuins in maintaining gut health and their implications in metabolic dysfunction and HCC development. Understanding these interactions offers potential therapeutic avenues for managing diseases linked to metabolic dysregulation and liver pathology.
Collapse
Affiliation(s)
- Mahmoud Zhra
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Muhammad Affan Elahi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.A.E.); (A.A.-Z.)
| | - Aamira Tariq
- Department of Biosciences, COMSATS University Islamabad, Islamabad Campus, Islamabad 45550, Pakistan
| | - Ahmed Abu-Zaid
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.A.E.); (A.A.-Z.)
| | - Ahmed Yaqinuddin
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| |
Collapse
|
23
|
Wang D, Sun S, Zhao Q, Zhao B, Ma L, Su T, Xu L, Gui M, Xu D, Chen W, Zeng Y, Shen Y, Liu Y, Jiang C, Ni Q, Cui Y, Lu Y, Lu Q, Dong D, Peng Y, Mao E. Metabolic shifts in tryptophan pathways during acute pancreatitis infections. JCI Insight 2025; 10:e186745. [PMID: 40059826 PMCID: PMC11949050 DOI: 10.1172/jci.insight.186745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/14/2025] [Indexed: 03/29/2025] Open
Abstract
Infectious complications (ICs) in acute pancreatitis (AP) are primarily driven by intestinal bacterial translocation, significantly increasing mortality and hospital stays. Despite this, the role of the gut microenvironment, particularly its metabolic aspects, in AP remains poorly understood. In this study, we investigated a cohort of patients with AP, and conducted supplemental murine studies, to explore the relationship between the gut metabolome and the development of ICs. Metabolomic analysis revealed that disruptions in gut tryptophan metabolism - especially reductions in serotonin and indole pathways - are key features associated with IC occurrence. Additionally, elevated plasma levels of tryptophan metabolites within the kynurenine pathway were identified as valuable predictive biomarkers for ICs. Mechanistic studies in murine models demonstrated that an impaired intestinal Th17 response, modulated by these tryptophan metabolites, plays a critical role in IC development. Serotonin supplementation enhanced Th17 responses, reducing IC incidence, while administration of kynurenic acid, a kynurenine metabolite, exacerbated pancreatic infections, potentially through immunosuppressive effects. These findings highlight the pivotal role of tryptophan metabolites in AP pathogenesis, emphasizing their potential as both predictive markers and therapeutic targets in IC management.
Collapse
Affiliation(s)
- Daosheng Wang
- Department of Emergency
- Department of Laboratory Medicine, and
| | | | | | | | | | | | - Lili Xu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | - Wei Chen
- Department of Laboratory Medicine, and
| | - Yu Zeng
- Department of Laboratory Medicine, and
| | | | - Yiyue Liu
- Department of Laboratory Medicine, and
| | - Cen Jiang
- Department of Laboratory Medicine, and
| | - Qi Ni
- Department of Laboratory Medicine, and
| | | | - Yide Lu
- Department of Laboratory Medicine, and
| | - Qiuya Lu
- Department of Laboratory Medicine, and
| | | | - Yibing Peng
- Department of Laboratory Medicine, and
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | |
Collapse
|
24
|
Amarasiri RPGSK, Hyun J, Lee SW, Kim JI, Lee HG, Ryu B, Jeon YJ. Therapeutic potential of tryptophan metabolite indoleacrylic acid in inflammatory bowel disease: From cellular mechanisms to zebrafish stress-like behavior. Int Immunopharmacol 2025; 149:114207. [PMID: 39904043 DOI: 10.1016/j.intimp.2025.114207] [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: 12/05/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/06/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic condition associated with elevated rates of anxiety and depression and ultimately reduces the quality of life. Thus, preventive care addressing both physical and psychological health is essential. In this study we aimed to explore the protective effects of Indoleacrylic Acid (IA) against lipopolysaccharide (LPS)-induced inflammation using human colorectal adenocarcinoma cells (HT-29) and dextran sulfate sodium (DSS)-induced zebrafish to assess its potential as a novel therapeutic agent for IBD. IA exhibited substantial anti-inflammatory properties in HT-29 cells and zebrafish models. It significantly reduced the production of pro-inflammatory mediators, including PGE2, TNF-α, IL-6, and IL-8, while upregulating MUC2, AhR, and tight junction proteins (ZO-1, occludin, and claudin-1), thereby enhancing mucosal barrier integrity. In zebrafish larvae, IA improved survival rates, boosted mucin production, and reduced macrophage infiltration and heartbeat rate. Behavioral analyses of adult zebrafish revealed that IA alleviated anxiety-like behaviors, as shown by increased locomotion and improved performance in zone preference and light-dark transition tests. By targeting inflammation and anxiety-like symptoms, IA demonstrates a dual benefit by addressing both intestinal inflammation and the psychological burden of IBD. These findings highlight IA's potential as a novel therapeutic agent for managing IBD, offering a comprehensive approach to improving patient outcomes.
Collapse
Affiliation(s)
- R P G S K Amarasiri
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Jimin Hyun
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Sang-Woon Lee
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Jae-Il Kim
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Hyoung-Gon Lee
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Bomi Ryu
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea.
| | - You-Jin Jeon
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
25
|
Wan X, Zhang C, Lei P, Wang H, Chen R, Yang Q, Cheng Y, Wu W, Sun D, Hong X. Precision therapeutics for inflammatory bowel disease: advancing ROS-responsive nanoparticles for targeted and multifunctional drug delivery. J Mater Chem B 2025; 13:3245-3269. [PMID: 39905851 DOI: 10.1039/d4tb02868f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Inflammatory bowel disease (IBD) is a severe chronic intestinal disorder with a rising global incidence. Current therapies, including the delivery of anti-inflammatory drugs and probiotics, face significant challenges in terms of safety, stability, and efficacy. In IBD patients, the activity of antioxidant enzymes (e.g., superoxide dismutase, glutathione peroxidase, and glutathione reductase) is reduced at the site of intestinal inflammation, leading to the accumulation of reactive oxygen species (ROS). This accumulation damages the intestinal mucosa, disrupts tight junctions between cells, and compromises the integrity of the intestinal barrier, exacerbating IBD symptoms. Therefore, nanoparticles responsive to ROS and capable of mimicking antioxidant enzyme activity, such as boronates, polydopamine, sulfides, and metal nanozymes, have emerged as promising tools. These nanoparticles can respond to elevated ROS levels in inflamed intestinal regions and release drugs to effectively neutralize ROS, making them ideal candidates for IBD treatment. This review discusses the application of various ROS-responsive nanomaterial delivery systems in IBD therapy, highlights current challenges, and outlines future research directions. Furthermore, we explore the "layered programmable delivery" strategy, which combines ROS-responsive nanoparticles with pH-responsive and cell membrane-targeted nanoparticles. This strategy has the potential to overcome the limitations of single-mechanism targeted drug delivery, enabling multi-range and multi-functional treatment approaches that significantly enhance delivery efficiency, providing new insights for the future of localized IBD treatment.
Collapse
Affiliation(s)
- Xiuping Wan
- Department of Gastroenterology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Caijie Zhang
- The People's Hospital of Yuhuan (Yuhuan People's Hospital Health Community Group), Taizhou 317600, China
| | - Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| | - Hanbing Wang
- Department of biotechnology, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR 999077, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Yongwei Cheng
- National Engineering Research Center of Cell Growth Factor Drugs and Protein Biologics, Wenzhou Medical University, Wenzhou 325000, China
- MedTech (Wenzhou) Health Innovation Achievement Transformation Institute, Wenzhou Institute of Industry & Science, Wenzhou 325000, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing 400044, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| | - Xiaofei Hong
- Department of Gastroenterology, the Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu Central Hospital, Yiwu 322000, China.
| |
Collapse
|
26
|
Wang Z, Liu T, Liu L, Xie J, Tang F, Pi Y, Zhong Y, He Z, Zhang W, Zheng C. Lactobacillus vaginalis alleviates DSS induced colitis by regulating the gut microbiota and increasing the production of 3-indoleacrylic acid. Pharmacol Res 2025; 213:107663. [PMID: 39961405 DOI: 10.1016/j.phrs.2025.107663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disorder, and its incidence is experiencing an upward trend worldwide. UC can result in gut microbiota dysbiosis, impaired intestinal epithelial barrier, and systemic inflammation, for all of which there is presently no definitive treatment available. Lactobacillus is known to regulate gut microbiota and related metabolites to intervene in the development of UC. The objective of this study was to explore the underlying mechanism through which a novel probiotic, Lactobacillus vaginalis, alleviates DSS-induced colitis. Specifically, L. vaginalis were found to ameliorate the DSS-induced UC phenotype, restore intestinal microbiota balance and intestinal barrier function, and elevate the levels of 3-indoleacrylic acid (IAA) in mouse feces. Furthermore, fecal microbiota transplantation and fecal filtrate transplantation provide additional evidence that L. vaginalis alleviate DSS-induced colitis through metabolic products. Additionally, IAA has been shown to alleviate DSS-induced colitis symptoms, decrease inflammatory responses, and enhance intestinal barrier function. Finally, our findings confirm that L. vaginal and metabolites possess the capability to regulate the immune microenvironment in mice with colitis. And the RNA-seq analysis suggests that L. vaginal may play a pivotal role in alleviating colitis by modulating the PPAR signaling pathway. In conclusion, our findings suggest that oral administration of L. vaginalis alleviates DSS induced colonic inflammation by increasing the levels of IAA. L. vaginalis, as an emerging probiotic, provides a potential therapeutic strategy for clinical UC.
Collapse
Affiliation(s)
- Zhuoya Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Tian Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Li Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Graduate School of Jiangxi University of Chinese Medicine, Nanchang 330004, PR China
| | - Jian Xie
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Furui Tang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Yimin Pi
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Yuchun Zhong
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Zhidong He
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Wenming Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Province Key Laboratory of Precision Cell Therapy, The Institute of Translational Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China.
| | - Cihua Zheng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
27
|
Liu F, Gu Z, Yi F, Liu X, Zou W, Xu Q, Yuan Y, Chen N, Tang J. Potential of Glycyrrhiza in the prevention of colitis-associated colon cancer. Fitoterapia 2025; 181:106398. [PMID: 39842555 DOI: 10.1016/j.fitote.2025.106398] [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: 10/16/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Glycyrrhiza, a legume native to the Mediterranean region, has a long history of ethnomedicinal use in China. Due to its antiviral, antibacterial, anti-inflammatory, antioxidant, antitumor, anti-ulcer, and hepatoprotective properties, Glycyrrhiza is widely utilized in the treatment of gastrointestinal disorders. THE AIM OF THE REVIEW The specific mechanisms of the main active constituents of glycyrrhiza in the treatment of inflammatory bowel disease, precancerous lesions and colorectal cancer at all stages of the colitis-associated colon cancer "Inflammation-Dysplasia-Cancer" sequence, as well as its pharmacokinetics, toxicology, formulation improvements, and application studies, are reviewed to provide new insights and perspectives on glycyrrhiza as a dietary supplement to treat and prevent colitis-associated colon cancer. MATERIALS AND METHODS Information on Glycyrrhiza was retrieved from electronic databases, including PubMed and Web of Science. RESULTS Glycyrrhiza is a well-established medicinal plant with significant potential for applications in both the food and pharmaceutical industries. Over 400 active constituents have been identified in Glycyrrhiza, including terpenoids, flavonoids, isoflavones, coumarins, and polyphenols. Numerous studies have demonstrated that Glycyrrhiza and its active compounds can inhibit the "Inflammation-Dysplasia-Cancer" progression of colitis-associated colon cancer by mitigating inflammatory bowel disease, reducing the number of intestinal precancerous lesions, and counteracting colorectal cancer. Furthermore, derivatives and nanocarriers are crucial for the effective treatment of colitis-associated colon cancer using Glycyrrhiza and its active constituents. CONCLUSION In conclusion, Glycyrrhiza is a plant with both medicinal and nutritional value, making it a potential food ingredient and dietary supplement for the treatment of colitis-associated colon cancer.
Collapse
Affiliation(s)
- Fang Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China; North Sichuan Medical College, Nanchong, China.
| | - Zhili Gu
- North Sichuan Medical College, Nanchong, China
| | - Feiyang Yi
- North Sichuan Medical College, Nanchong, China
| | - Xue Liu
- North Sichuan Medical College, Nanchong, China
| | - Wenxuan Zou
- North Sichuan Medical College, Nanchong, China
| | - Qingxia Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yun Yuan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Nianzhi Chen
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jianyuan Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
28
|
Guo M, Jiang X, Ouyang H, Zhang X, Zhang S, Wang P, Bi G, Wu T, Zhou W, Liang F, Yang X, Fan S, Fang JH, Chen P, Bi H. Parabacteroides distasonis promotes liver regeneration by increasing β-hydroxybutyric acid (BHB) production and BHB-driven STAT3 signals. Acta Pharm Sin B 2025; 15:1430-1446. [PMID: 40370533 PMCID: PMC12069244 DOI: 10.1016/j.apsb.2025.01.024] [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: 08/01/2024] [Revised: 12/04/2024] [Accepted: 12/20/2024] [Indexed: 05/16/2025] Open
Abstract
The liver regenerative capacity is crucial for patients with end-stage liver disease following partial hepatectomy (PHx). The specific bacteria and mechanisms regulating liver regeneration post-PHx remain unclear. This study demonstrated dynamic changes in the abundance of Parabacteroides distasonis (P. distasonis) post-PHx, correlating with hepatocyte proliferation. Treatment with live P. distasonis significantly promoted hepatocyte proliferation and liver regeneration after PHx. Targeted metabolomics revealed a significant positive correlation between P. distasonis and β-hydroxybutyric acid (BHB), as well as hyodeoxycholic acid and 3-hydroxyphenylacetic acid in the gut after PHx. Notably, treatment with BHB, but not hyodeoxycholic acid or 3-hydroxyphenylacetic acid, significantly promoted hepatocyte proliferation and liver regeneration in mice after PHx. Moreover, STAT3 inhibitor Stattic attenuated the promotive effects of BHB on cell proliferation and liver regeneration both in vitro and in vivo. Mechanistically, P. distasonis upregulated the expression of fatty acid oxidation-related proteins, and increased BHB levels in the liver, and then BHB activated the STAT3 signaling pathway to promote liver regeneration. This study, for the first time, identifies the involvement of P. distasonis and its associated metabolite BHB in promoting liver regeneration after PHx, providing new insights for considering P. distasonis and BHB as potential strategies for promoting hepatic regeneration.
Collapse
Affiliation(s)
- Manlan Guo
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong–Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaowen Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong–Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hui Ouyang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong–Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xianglong Zhang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shuaishuai Zhang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong–Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Peng Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong–Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Guofang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong–Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ting Wu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong–Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wenhong Zhou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong–Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Fengting Liang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong–Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiao Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong–Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen 518055, China
| | - Shicheng Fan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong–Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jian-hong Fang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong–Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Peng Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Huichang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong–Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen 518055, China
| |
Collapse
|
29
|
Charitos IA, Scacco S, Cotoia A, Castellaneta F, Castellana G, Pasqualotto F, Venneri M, Ferrulli A, Aliani M, Santacroce L, Carone M. Intestinal Microbiota Dysbiosis Role and Bacterial Translocation as a Factor for Septic Risk. Int J Mol Sci 2025; 26:2028. [PMID: 40076650 PMCID: PMC11900423 DOI: 10.3390/ijms26052028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/18/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
The human immune system is closely linked to microbiota such as a complex symbiotic relationship during the coevolution of vertebrates and microorganisms. The transfer of microorganisms from the mother's microbiota to the newborn begins before birth during gestation and is considered the initial phase of the intestinal microbiota (IM). The gut is an important site where microorganisms can establish colonies. The IM contains polymicrobial communities, which show complex interactions with diet and host immunity. The tendency towards dysbiosis of the intestinal microbiota is influenced by local but also extra-intestinal factors such as inflammatory processes, infections, or a septic state that can aggravate it. Pathogens could trigger an immune response, such as proinflammatory responses. In addition, changes in the host immune system also influence the intestinal community and structure with additional translocation of pathogenic and non-pathogenic bacteria. Finally, local intestinal inflammation has been found to be an important factor in the growth of pathogenic microorganisms, particularly in its role in sepsis. The aim of this article is to be able to detect the current knowledge of the mechanisms that can lead to dysbiosis of the intestinal microbiota and that can cause bacterial translocation with a risk of infection or septic state and vice versa.
Collapse
Affiliation(s)
- Ioannis Alexandros Charitos
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
- Doctoral School, Applied Neurosciences, University of Bari (UNIBA), 70124 Bari, Italy
| | - Salvatore Scacco
- Dipartimento di Biomedicina Traslazionale e Neuroscienze (DiBraiN), Scuola di Medicina, Università Degli Studi di Bari, Aldo Moro, 70124 Bari, Italy;
- U.O. Medicina, Ospedale Mater Dei-CBH, 70125 Bari, Italy
| | - Antonella Cotoia
- Department of Intensive Care, University Hospital of Foggia, 71121 Foggia, Italy
| | - Francesca Castellaneta
- U.O.C. Servizio di Immunoematologia e Medicina Trasfusionale—S.I.M.T. Ospedale Di Venere, 70131 Bari, Italy;
| | - Giorgio Castellana
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
| | - Federico Pasqualotto
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
- Department of Public Health and Infectious Diseases, Pulmonary Division, Sapienza University of Rome, Policlinico Umberto I Hospital, Rome, Via del Policlinico 155, 00155 Rome, Italy
| | - Maria Venneri
- Genomics and Proteomics Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (M.V.); (A.F.)
| | - Angela Ferrulli
- Genomics and Proteomics Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (M.V.); (A.F.)
| | - Maria Aliani
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
| | - Luigi Santacroce
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, The University of Bari, 70124 Bari, Italy;
| | - Mauro Carone
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
| |
Collapse
|
30
|
Rauch CE, Henningsen K, Martinez I, Young P, Mika A, Huschtscha Z, McCubbin A, Henry R, Anderson D, Costa RJS. The Effects of Prebiotic Supplementation on Markers of Exercise-Induced Gastrointestinal Syndrome in Response to Exertional Heat Stress. Int J Sport Nutr Exerc Metab 2025:1-18. [PMID: 40010361 DOI: 10.1123/ijsnem.2024-0127] [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: 06/21/2024] [Revised: 01/08/2025] [Accepted: 01/08/2025] [Indexed: 02/28/2025]
Abstract
Exercise perturbs various aspects of gastrointestinal integrity and function, which may lead to performance impeding gastrointestinal symptoms (GIS) and/or precipitate clinical issues warranting medical management. This study aimed to determine the impact of prebiotic supplementation on gastrointestinal integrity and functional status in response to exertional heat stress (EHS). Sixteen endurance athletes completed two trials of 3-hr running at 60% V˙O2max in 30 °C at baseline (T1) and following an 8-week supplementation period (T2), with 16 g/day prebiotic (PREBIOTIC) or matched placebo (PLACEBO). Blood samples were collected pre-EHS and post-EHS and in recovery for determination of stress response (cortisol), intestinal epithelial injury (intestinal fatty acid binding protein), bacterial endotoxemia (sCD14), and systemic inflammation (C-reactive protein). GIS and feeding tolerance variables were assessed throughout the EHS. Orocecal transit time was determined via a lactulose challenge given at 2.5 hr into EHS. Plasma cortisol (combined mean: +252 ng/ml), intestinal fatty acid binding protein (+800 pg/ml), and sCD14 (+487 ng/ml) concentrations increased in response to EHS in T1 (p ≤ .05), but not for C-reactive protein (+0.8 μg/ml; p > .05), in both PREBIOTIC and PLACEBO. PREBIOTIC supplementation resulted in a blunted intestinal fatty acid binding protein response on T2 (+316 pg/ml) compared with an increase (+1,001 ng/ml) in PLACEBO (p = .005). Lower sCD14 was observed at T2 (2,799 ng/ml) versus T1 (3,246 ng/ml) in PREBIOTIC only (p = .039). No intervention effects were observed for C-reactive protein. No difference within or between PREBIOTIC and PLACEBO at T1 and T2 was observed for orocecal transit time, GIS, and feeding tolerance. In conclusion, 8 weeks of prebiotic supplementation modestly attenuates EHS associated perturbations to intestinal integrity, but does not further impair gastrointestinal transit and/or exacerbate EHS associated GIS or feeding tolerance.
Collapse
Affiliation(s)
- Christopher E Rauch
- Department of Nutrition, Dietetics & Food, Monash University, Notting Hill, VIC, Australia
| | - Kayla Henningsen
- Department of Nutrition, Dietetics & Food, Monash University, Notting Hill, VIC, Australia
| | - Isabel Martinez
- Department of Nutrition, Dietetics & Food, Monash University, Notting Hill, VIC, Australia
| | - Pascale Young
- Department of Nutrition, Dietetics & Food, Monash University, Notting Hill, VIC, Australia
| | - Alice Mika
- Department of Nutrition, Dietetics & Food, Monash University, Notting Hill, VIC, Australia
| | - Zoya Huschtscha
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Science, Deakin University, Burwood, VIC, Australia
| | - Alan McCubbin
- Department of Nutrition, Dietetics & Food, Monash University, Notting Hill, VIC, Australia
| | - Rebecca Henry
- School of Public Health and Preventive Medicine, Monash University, Clayton, VIC, Australia
| | - Doville Anderson
- Monash Proteomics and Metabolomics Platform, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, Australia
| | - Ricardo J S Costa
- Department of Nutrition, Dietetics & Food, Monash University, Notting Hill, VIC, Australia
| |
Collapse
|
31
|
Kabisch S, Hajir J, Sukhobaevskaia V, Weickert MO, Pfeiffer AFH. Impact of Dietary Fiber on Inflammation in Humans. Int J Mol Sci 2025; 26:2000. [PMID: 40076626 PMCID: PMC11900212 DOI: 10.3390/ijms26052000] [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: 01/06/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Cohort studies consistently show that a high intake of cereal fiber and whole-grain products is associated with a decreased risk of type 2 diabetes (T2DM), cancer, and cardiovascular diseases. Similar findings are also reported for infectious and chronic inflammatory disorders. All these disorders are at least partially caused by inflammaging, a chronic state of inflammation associated with aging and Metabolic Syndrome. Surprisingly, insoluble (cereal) fiber intake consistently shows stronger protective associations with most long-term health outcomes than soluble fiber. Most humans consume soluble fiber mainly from sweet fruits, which usually come with high levels of sugar, counteracting the potentially beneficial effects of fiber. In both observational and interventional studies, high-fiber diets show a beneficial impact on inflammation, which can be attributed to a variety of nutrients apart from dietary fiber. These confounders need to be considered when evaluating the effects of fiber as part of complex dietary patterns. When assessing specific types of fiber, inulin and resistant starch clearly elicit anti-inflammatory short-term effects, while results for pectins, beta-glucans, or psyllium turn out to be less convincing. For insoluble fiber, promising but sparse data have been published so far. Hypotheses on putative mechanisms of anti-inflammatory fiber effects include a direct impact on immune cells (e.g., for pectin), fermentation to pleiotropic short-chain fatty acids (for fermentable fiber only), modulation of the gut microbiome towards higher levels of diversity, changes in bile acid metabolism, a differential release of gut hormones (such as the glucose-dependent insulinotropic peptide (GIP)), and an improvement of insulin resistance via the mTOR/S6K1 signaling cascade. Moreover, the contribution of phytate-mediated antioxidative and immune-modulatory means of action needs to be considered. In this review, we summarize the present knowledge on the impact of fiber-rich diets and dietary fiber on the human inflammatory system. However, given the huge heterogeneity of study designs, cohorts, interventions, and outcomes, definite conclusions on which fiber to recommend to whom cannot yet be drawn.
Collapse
Affiliation(s)
- Stefan Kabisch
- Department of Endocrinology and Metabolism, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Jasmin Hajir
- Department of Endocrinology and Metabolism, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Varvara Sukhobaevskaia
- Department of Endocrinology and Metabolism, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Martin O. Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism; The ARDEN NET Centre, ENETS CoE; University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Centre of Applied Biological & Exercise Sciences (ABES), Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
- Translational & Experimental Medicine, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology and Metabolism, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| |
Collapse
|
32
|
Blok L, Hanssen N, Nieuwdorp M, Rampanelli E. From Microbes to Metabolites: Advances in Gut Microbiome Research in Type 1 Diabetes. Metabolites 2025; 15:138. [PMID: 39997763 PMCID: PMC11857261 DOI: 10.3390/metabo15020138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Type 1 diabetes (T1D) is a severe chronic T-cell mediated autoimmune disease that attacks the insulin-producing beta cells of the pancreas. The multifactorial nature of T1D involves both genetic and environmental components, with recent research focusing on the gut microbiome as a crucial environmental factor in T1D pathogenesis. The gut microbiome and its metabolites play an important role in modulating immunity and autoimmunity. In recent years, studies have revealed significant alterations in the taxonomic and functional composition of the gut microbiome associated with the development of islet autoimmunity and T1D. These changes include reduced production of short-chain fatty acids, altered bile acid and tryptophan metabolism, and increased intestinal permeability with consequent perturbations of host (auto)immune responses. Methods/Results: In this review, we summarize and discuss recent observational, mechanistic and etiological studies investigating the gut microbiome in T1D and elucidating the intricate role of gut microbes in T1D pathogenesis. Moreover, we highlight the recent advances in intervention studies targeting the microbiota for the prevention or treatment of human T1D. Conclusions: A deeper understanding of the evolution of the gut microbiome before and after T1D onset and of the microbial signals conditioning host immunity may provide us with essential insights for exploiting the microbiome as a prognostic and therapeutic tool.
Collapse
Affiliation(s)
- Lente Blok
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Nordin Hanssen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Elena Rampanelli
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
- Amsterdam Institute for Infection and Immunity (AII), Amsterdam, The Netherlands
| |
Collapse
|
33
|
Chi J, Patterson JS, Jin Y, Kim KJ, Lalime N, Hawley D, Lewis F, Li L, Wang X, Campen MJ, Cui JY, Gu H. Metabolic Reprogramming in Gut Microbiota Exposed to Polystyrene Microplastics. Biomedicines 2025; 13:446. [PMID: 40002859 PMCID: PMC11853289 DOI: 10.3390/biomedicines13020446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/26/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Microplastics (MPs) are small plastic fragments with diameters less than 5 mm in size and are prevalent in everyday essentials and consumables. Large global plastic production has now led to a flooding of MPs in our natural environment. Due to their detrimental impacts on the planet's ecosystems and potentially our health, MPs have emerged as a significant public health concern. In this pilot study, we hypothesize that MPs exposure will negatively affect gut microbiota composition and function, in which metabolic reprogramming plays an important role. Methods: Using in vitro experiments, three bacterial strains (Escherichia coli MG1655, Nissle 1917, and Lactobacillus rhamnosus) were selected to investigate the impacts of MPs exposure. The bacterial strains were individually cultured in an anaerobic chamber and exposed to 1 µm polystyrene MPs at various concentrations (0, 10, 20, 50, 100, and 500 µg/mL) in the culture medium. Results: MPs exposure reduced the growth of all three bacterial strains in a dose-dependent manner. Liquid chromatography mass spectrometry (LC-MS)-based untargeted metabolomics revealed significant differences in multiple metabolic pathways, such as sulfur metabolism and amino sugar and nucleotide sugar metabolism. In addition, we extracted gut microbiota from C57BL/6 mice, and 16S rRNA sequencing results showed a significant upregulation of Lactobacillales and a significant reduction in Erysipelotrichales due to MPs exposure. Furthermore, targeted and untargeted metabolomics corroborated the in vitro results and revealed alterations in microbial tryptophan metabolism and energy producing pathways, such as glycolysis/gluconeogenesis and the pentose phosphate pathway. Conclusions: These findings provide evidence that MPs exposure causes comprehensive changes to healthy gut microbiota, which may also provide insights into the mechanistic effects of MPs exposure in humans.
Collapse
Affiliation(s)
- Jinhua Chi
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (J.C.); (J.S.P.); (L.L.)
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA;
| | - Jeffrey S. Patterson
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (J.C.); (J.S.P.); (L.L.)
| | - Yan Jin
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA;
| | - Kyle Joohyung Kim
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA; (K.J.K.); (J.Y.C.)
| | - Nicole Lalime
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA;
| | - Daniella Hawley
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA; (D.H.); (X.W.)
| | - Freeman Lewis
- Environmental Health Sciences, Florida International University, Miami, FL 33199, USA;
| | - Lingjun Li
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (J.C.); (J.S.P.); (L.L.)
| | - Xuan Wang
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA; (D.H.); (X.W.)
| | - Matthew J. Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences, Albuquerque, NM 87106, USA;
| | - Julia Yue Cui
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA; (K.J.K.); (J.Y.C.)
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (J.C.); (J.S.P.); (L.L.)
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA;
| |
Collapse
|
34
|
Chulenbayeva L, Issilbayeva A, Sailybayeva A, Bekbossynova M, Kozhakhmetov S, Kushugulova A. Short-Chain Fatty Acids and Their Metabolic Interactions in Heart Failure. Biomedicines 2025; 13:343. [PMID: 40002756 PMCID: PMC11853371 DOI: 10.3390/biomedicines13020343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Short-chain fatty acids (SCFAs), produced through fermentation of dietary fibers by gut bacteria, play a central role in modulating cardiovascular function and heart failure (HF) development. The progression of HF is influenced by intestinal barrier dysfunction and microbial translocation, where SCFAs serve as key mediators in the gut-heart axis. This review examines the complex metabolic interactions between SCFAs and other gut microbiota metabolites in HF, including their relationships with trimethylamine N-oxide (TMAO), aromatic amino acids (AAAs), B vitamins, and bile acids (BAs). We analyze the associations between SCFA production and clinical parameters of HF, such as left ventricular ejection fraction (LVEF), N-terminal pro-B-type natriuretic peptide (NT-proBNP), and glomerular filtration rate (GFR). Gaining insights into metabolic networks offers new potential therapeutic targets and prognostic markers for managing heart failure, although their clinical significance needs further exploration.
Collapse
Affiliation(s)
- Laura Chulenbayeva
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (A.I.); (S.K.); (A.K.)
| | - Argul Issilbayeva
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (A.I.); (S.K.); (A.K.)
| | - Aliya Sailybayeva
- Heart Center, CF “University Medical Center”, Astana 010000, Kazakhstan; (A.S.); (M.B.)
| | | | - Samat Kozhakhmetov
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (A.I.); (S.K.); (A.K.)
| | - Almagul Kushugulova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (A.I.); (S.K.); (A.K.)
- Heart Center, CF “University Medical Center”, Astana 010000, Kazakhstan; (A.S.); (M.B.)
| |
Collapse
|
35
|
Zarei P, Sedeh PA, Vaez A, Keshteli AH. Using metabolomics to investigate the relationship between the metabolomic profile of the intestinal microbiota derivatives and mental disorders in inflammatory bowel diseases: a narrative review. Res Pharm Sci 2025; 20:1-24. [PMID: 40190827 PMCID: PMC11972020 DOI: 10.4103/rps.rps_273_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/30/2024] [Accepted: 05/28/2024] [Indexed: 04/09/2025] Open
Abstract
Individuals with inflammatory bowel disease (IBD) are at a higher risk of developing mental disorders, such as anxiety and depression. The imbalance between the intestinal microbiota and its host, known as dysbiosis, is one of the factors, disrupting the balance of metabolite production and their signaling pathways, leading to disease progression. A metabolomics approach can help identify the role of gut microbiota in mental disorders associated with IBD by evaluating metabolites and their signaling comprehensively. This narrative review focuses on metabolomics studies that have comprehensively elucidated the altered gut microbial metabolites and their signaling pathways underlying mental disorders in IBD patients. The information was compiled by searching PubMed, Web of Science, Scopus, and Google Scholar from 2005 to 2023. The findings indicated that intestinal microbial dysbiosis in IBD patients leads to mental disorders such as anxiety and depression through disturbances in the metabolism of carbohydrates, sphingolipids, bile acids, neurotransmitters, neuroprotective, inflammatory factors, and amino acids. Furthermore, the reduction in the production of neuroprotective factors and the increase in inflammation observed in these patients can also contribute to the worsening of psychological symptoms. Analyzing the metabolite profile of the patients and comparing it with that of healthy individuals using advanced technologies like metabolomics, aids in the early diagnosis and prevention of mental disorders. This approach allows for the more precise identification of the microbes responsible for metabolite production, enabling the development of tailored dietary and pharmaceutical interventions or targeted manipulation of microbiota.
Collapse
Affiliation(s)
- Parvin Zarei
- Department of Bioinformatics, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Peyman Adibi Sedeh
- Isfahan Gastroenterology and Hepatology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ahmad Vaez
- Department of Bioinformatics, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Epidemiology, University of Groningen, University Medical Centre Groningen, 9713 GZ Groningen, The Netherlands
| | | |
Collapse
|
36
|
Zuo X, Zhang T, Dong X, Liu J, Liu Y. Identification of the key tryptophan metabolic characteristics of Lactiplantibacillus plantarum for aryl hydrocarbon receptor activation and ulcerative colitis alleviation. Food Res Int 2025; 203:115766. [PMID: 40022314 DOI: 10.1016/j.foodres.2025.115766] [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: 10/10/2024] [Revised: 12/17/2024] [Accepted: 01/14/2025] [Indexed: 03/03/2025]
Abstract
Lactiplantibacillus plantarum can activate aryl hydrocarbon receptor (AHR) signaling in the gut by metabolizing tryptophan, thereby counteracting inflammation. However, the tryptophan metabolic characteristics of microorganisms are strain-specific and significantly influence their AHR-activating effects. In this study, four strains with different tryptophan metabolic profiles were screened, and a dextran sulfate sodium-induced colitis model was established in C57BL/6 mice. The key tryptophan metabolic characteristics of L. plantarum involved in AHR downstream signaling activation to alleviate colitis were explored. The results showed that strain SFFI50, characterized by poor tryptophan metabolism, and strain SFFI175, which produced high levels of IAA and ICA, did not alleviate colitis. Strain SFFI118, capable of metabolizing tryptophan to produce IAA, ICA, and ILA, could slightly restore mouse body weight, DAI, and IL-22 expression. L. plantarum SFFI23 significantly restored body weight, colon length, histopathological damage, and cytokine expression in mice. Moreover, it activated the downstream signaling of AHR, specifically CYP1A1, and repairs the intestinal barrier function. Targeted metabolomic analysis revealed a significant increase in indole-3-lactic acid (ILA) in the mouse intestine. Correlation analysis revealed a significant positive correlation between high ILA production of L. plantarum, CYP1A1 expression, intestinal barrier function restoration, ILA levels in vivo, and colitis alleviation. Therefore, we inferred that high ILA production is a key tryptophan metabolic characteristic of L. plantarum which activated AHR downstream signaling (such as CYP1A1, IL-22, and STAT3) to alleviate colitis. This study provides a theoretical basis for the development of personalized dietary interventions to improve gut health.
Collapse
Affiliation(s)
- Xuemei Zuo
- Shandong Food Ferment Industry & Design Institute, QiLu University of Technology (Shandong Academy of Sciences), Jinan 250013 China
| | - Tingting Zhang
- Shanghai Engineering Research Center of Food Microbiology, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093 China
| | - Xueqian Dong
- Shandong Food Ferment Industry & Design Institute, QiLu University of Technology (Shandong Academy of Sciences), Jinan 250013 China
| | - Jianjun Liu
- Shandong Food Ferment Industry & Design Institute, QiLu University of Technology (Shandong Academy of Sciences), Jinan 250013 China
| | - Yang Liu
- Shandong Food Ferment Industry & Design Institute, QiLu University of Technology (Shandong Academy of Sciences), Jinan 250013 China.
| |
Collapse
|
37
|
Yi C, Huang S, Zhang W, Guo L, Xia T, Huang F, Yan Y, Li H, Yu B. Synergistic interactions between gut microbiota and short chain fatty acids: Pioneering therapeutic frontiers in chronic disease management. Microb Pathog 2025; 199:107231. [PMID: 39681288 DOI: 10.1016/j.micpath.2024.107231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024]
Abstract
Microorganisms in the gut play a pivotal role in human health, influencing various pathophysiological processes. Certain microorganisms are particularly essential for maintaining intestinal homeostasis, reducing inflammation, supporting nervous system function, and regulating metabolic processes. Short-chain fatty acids (SCFAs) are a subset of fatty acids produced by the gut microbiota (GM) during the fermentation of indigestible polysaccharides. The interaction between GM and SCFAs is inherently bidirectional: the GM not only shapes SCFAs composition and metabolism but SCFAs also modulate microbiota's diversity, stability, growth, proliferation, and metabolism. Recent research has shown that GM and SCFAs communicate through various pathways, mainly involving mechanisms related to inflammation and immune responses, intestinal barrier function, the gut-brain axis, and metabolic regulation. An imbalance in GM and SCFA homeostasis can lead to the development of several chronic diseases, including inflammatory bowel disease, colorectal cancer, systemic lupus erythematosus, Alzheimer's disease, and type 2 diabetes mellitus. This review explores the synergistic interactions between GM and SCFAs, and how these interactions directly or indirectly influence the onset and progression of various diseases through the regulation of the mechanisms mentioned above.
Collapse
Affiliation(s)
- Chunmei Yi
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shanshan Huang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wenlan Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lin Guo
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tong Xia
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Fayin Huang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yijing Yan
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Huhu Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Bin Yu
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
38
|
Zheng L, Wei Z, Ni X, Shang J, Liu F, Peng Y, Liu J, Li Y. Exploring the therapeutic potential of Xiangsha Liujunzi Wan in Crohn's disease: from network pharmacology approach to experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118863. [PMID: 39343107 DOI: 10.1016/j.jep.2024.118863] [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: 07/17/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiangsha Liujunzi Wan (LJZW) is a traditional Chinese medicine (TCM) formula containing a variety of traditional Chinese herb components. Its principal components are often used in the treatment of gastrointestinal diseases and contribute to the treatment of Crohn's disease (CD). AIM OF THE STUDY To explore the therapeutic potential of LJZW in CD through network pharmacology, bioinformatics, molecular docking, and experimental verification. METHODS The principal bioactive components and corresponding targets of LJZW were ascertained from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). Potential targets for CD were identified in GeneCards, OMIM, DrugBank, DisGeNET, CTD, and Gene Expression Omnibus (GEO) databases. Intersection targets of LJZW and CD were identified using a Venn diagram and visualized using Cytoscape 3.8.0 to construct a protein-protein interaction (PPI) network. Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were employed to assess the function of intersection targets. AutoDockTools and PyMOL were used for molecular docking to recognize the association between the core ingredients of LJZW and the core targets of CD. Subsequently, a series of experiments were conducted for validation. RESULTS The network pharmacology results indicated that there were 156 bioactive components and 268 corresponding targets for LJZW, 3023 primary relevant targets for CD, and 169 intersection targets for LJZW and CD. The PPI network was employed to identify five hub genes and six clusters. The GO functional analysis indicated that intersection targets are primarily correlated with oxidative stress and inflammatory responses. KEGG pathway analysis revealed that these targets were primarily associated with the phosphotylinosital 3 kinase (PI3K)-protein kinase B (AKT) and mitogen-activated protein kinase (MAPK) signaling pathways. The molecular docking results showed that the core ingredients of LJZW had good binding ability with the core targets of CD. A series of experiments demonstrated that LJZW could effectively attenuate TNBS-induced colitis symptoms, inhibit the inflammatory response, and protect intestinal barrier function by inhibiting the PI3K-AKT and MAPK signaling pathways, thus preventing and treating CD. CONCLUSION LJZW has the characteristics of multi-component, multi-target, and multi-pathway treatment, which helps to improve the treatment of CD, protect the intestinal barrier, and exert the effect of anti-inflammatory therapy by inhibiting PI3K-AKT and MAPK signaling pathways.
Collapse
Affiliation(s)
- Linlin Zheng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, PR China
| | - Ziyun Wei
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, PR China
| | - Xiao Ni
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, PR China
| | - Jianing Shang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, PR China
| | - Fu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, PR China
| | - Yuxuan Peng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, PR China
| | - Jieyu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, PR China.
| | - Yunwei Li
- Department of Anorectal Surgery, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China.
| |
Collapse
|
39
|
Rezazadegan M, Forootani B, Hoveyda Y, Rezazadegan N, Amani R. Major heavy metals and human gut microbiota composition: a systematic review with nutritional approach. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2025; 44:21. [PMID: 39871318 PMCID: PMC11773724 DOI: 10.1186/s41043-025-00750-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/10/2025] [Indexed: 01/29/2025]
Abstract
BACKGROUND The human gut microbiota has a critical role in several aspects of host homeostasis, such as immune development, metabolism, nutrition, and defense against pathogens during life. It can be sensitive to xenobiotics including drugs, diet, or even environmental pollutants, especially heavy metals (HMs). The findings of some previous studies are heterogeneous due to the inclusion of various types of study (human, and animal studies) and wide exposures (phthalate, bisphenol A, HMS, etc.), and no comprehensive systematic review has investigated the association between HMs exposure and human gut microbiota composition. Therefore, we carried out a systematic review of human observational studies to examine this association. PubMed, Scopus, ISI Web of Science, and Google Scholar were searched using Medical Subject Headings (MeSH) and non-MeSH terms. Eventually, 12 studies for arsenic (As), lead (Pb), mercury (Hg), and cadmium (Cd) were included in this study. No eligible study was found for Aluminium. SHORT CONCLUSION The findings showed exposure to HMs disturbs the composition of gut microbiota and can lead to dysbiosis. Exposure to high levels of As, Pb, and Hg increased the abundance of Collinsella as pathobionts. Evidently, it is related to leaky gut, oxidative stress, and several diseases such as inflammatory bowel disease and cancers. Probiotic treatment and nutritional strategies such as high fiber intake and following antioxidant-rich diets should be considered in terms of HMs exposure.
Collapse
Affiliation(s)
- Mahsa Rezazadegan
- Student Research Committee, Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bita Forootani
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Yeganeh Hoveyda
- Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Niloufar Rezazadegan
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Amani
- Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
40
|
Pan H, Yang S, Kulyar MF, Ma H, Li K, Zhang L, Mo Q, Li J. Lactobacillus fermentum 016 Alleviates Mice Colitis by Modulating Oxidative Stress, Gut Microbiota, and Microbial Metabolism. Nutrients 2025; 17:452. [PMID: 39940311 PMCID: PMC11820689 DOI: 10.3390/nu17030452] [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: 01/03/2025] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/14/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic and progressive inflammatory gastrointestinal disease closely associated with gut microbiota dysbiosis and metabolic homeostasis disruption. Although targeted microbial therapies are an emerging intervention strategy for inflammatory bowel disease (IBD), the mechanisms by which specific probiotics, such as Lactobacillus fermentum 016 (LF), alleviate UC remain unclear. The current study evaluated the effects of LF supplementation on gut health in a basal model using C57BL/6 mice. Subsequently, the preventive effects and mechanisms of LF supplementation on DSS-induced UC were systematically investigated. According to our findings, LF supplementation revealed immunoregulatory capabilities with significantly altered gut the composition of microbiota and metabolic activities, particularly enhancing tryptophan metabolism. In the UC model, LF supplementation effectively mitigated weight loss, increased the disease activity index (DAI), and alleviated diarrhea, rectal bleeding, and colon shortening. Moreover, it reduced colonic pathological damage and histological injury scores. LF intervention improved antioxidant markers and intestinal mucosal barrier function with the activation of the Nrf2-Keap1 signaling pathway and regulation of systemic inflammatory markers, i.e., IL-1β, IL-6, TNF-α, IFN-γ, IL-4, and IL-10. Importantly, LF supplementation reversed metabolic disturbances by significantly increasing the abundance of beneficial genera (e.g., g_Dubosiella, g_Faecalibaculum, g_Odoribacter, g_Candidatus_saccharimonas, g_Roseburia, and g_Eubacterium_xylanophilum_group) and elevating tryptophan metabolites (e.g., melatonin, kynurenic acid, 3-indoleacetic acid, 5-methoxytryptophan, and 5-hydroxyindoleacetic acid). In conclusion, Lactobacillus fermentum 016 exhibits potential for regulating gut microbiota homeostasis, enhancing tryptophan metabolism, and alleviating UC, providing critical insights for developing probiotic-based precision therapeutic strategies for IBD.
Collapse
Affiliation(s)
- Huachun Pan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (H.P.); (M.F.K.); (H.M.); (K.L.); (Q.M.)
| | - Shumin Yang
- National Key Laboratory of Agriculture Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Md. F. Kulyar
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (H.P.); (M.F.K.); (H.M.); (K.L.); (Q.M.)
| | - Hongwei Ma
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (H.P.); (M.F.K.); (H.M.); (K.L.); (Q.M.)
| | - Kewei Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (H.P.); (M.F.K.); (H.M.); (K.L.); (Q.M.)
| | - Lihong Zhang
- College of Animal Science & Technology, Gansu Agricultural University, Lanzhou 730070, China;
| | - Quan Mo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (H.P.); (M.F.K.); (H.M.); (K.L.); (Q.M.)
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (H.P.); (M.F.K.); (H.M.); (K.L.); (Q.M.)
| |
Collapse
|
41
|
Polyák H, Galla Z, Rajda C, Monostori P, Klivényi P, Vécsei L. Plasma and Visceral Organ Kynurenine Metabolites Correlate in the Multiple Sclerosis Cuprizone Animal Model. Int J Mol Sci 2025; 26:976. [PMID: 39940744 PMCID: PMC11817772 DOI: 10.3390/ijms26030976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
The cuprizone (CPZ) model of multiple sclerosis (MS) is excellent for studying the molecular differences behind the damage caused by poisoning. Metabolic differences in the kynurenine pathway (KP) of tryptophan (TRP) degradation are observed in both MS and a CPZ mouse model. Our goal was to analyze the kynurenine, serotonin, and indole pathways of TRP degradation on the periphery, in the neurodegenerative processes of inflammation. In our study, mice were fed with 0.2% CPZ toxin for 5 weeks. We examined the metabolites in the three pathways of TRP breakdown in urine, plasma, and relevant visceral organs with bioanalytical measurements. In our analyses, we found a significant increase in plasma TRP, 5-hydroxytryptophan (5-HTP), and indole-3-acetic acid (IAA) levels, while a decrease in the concentrations of 3-hydroxy-L-kynurenine (3-HK), xanthurenic acid (XA), kynurenic acid (KYNA), and quinaldic acid in the plasma of toxin-treated group was found. A marked decrease in the levels of 3-HK, XA, KYNA, quinaldic acid, and indole-3-lactic acid was also observed in the visceral organs by the end of the poisoning. Furthermore, we noticed a decrease in the urinary levels of the TRP, KYNA, and XA metabolites, while an increase in serotonin and 5-hydroxyindoleacetic acid in the CPZ group was noticed. The toxin treatment resulted in elevated tryptamine and indoxyl sulfate levels and reduced IAA concentration. Moreover, the urinary para-cresyl sulfate concentration also increased in the treated group. In the present study, we showed the differences in the three main metabolic pathways of TRP degradation in the CPZ model. We confirmed the relationship and correlation between the content of the kynurenine metabolites in the plasma and the tissues of the visceral organs. We emphasized the suppression of the KP and the activity of the serotonin and indole pathways with a particular regard to the involvement of the microbiome by the indole pathway. Consequently, this is the first study to analyze in detail the distribution of the kynurenine, serotonin, and indole pathways of TRP degradation in the periphery.
Collapse
Affiliation(s)
- Helga Polyák
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (H.P.); (C.R.); (P.K.)
- Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6720 Szeged, Hungary
| | - Zsolt Galla
- Department of Pediatrics, Albert Szent-Györgyi Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary; (Z.G.); (P.M.)
| | - Cecilia Rajda
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (H.P.); (C.R.); (P.K.)
| | - Péter Monostori
- Department of Pediatrics, Albert Szent-Györgyi Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary; (Z.G.); (P.M.)
| | - Péter Klivényi
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (H.P.); (C.R.); (P.K.)
- HUN-REN-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (H.P.); (C.R.); (P.K.)
- HUN-REN-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| |
Collapse
|
42
|
Kaltsas A, Giannakodimos I, Markou E, Adamos K, Stavropoulos M, Kratiras Z, Zachariou A, Dimitriadis F, Sofikitis N, Chrisofos M. The Role of Gut Microbiota Dysbiosis in Erectile Dysfunction: From Pathophysiology to Treatment Strategies. Microorganisms 2025; 13:250. [PMID: 40005617 PMCID: PMC11857656 DOI: 10.3390/microorganisms13020250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Erectile dysfunction (ED) is a prevalent male sexual disorder characterized by the persistent inability to achieve or maintain an erection sufficient for satisfactory sexual performance. While its etiology is multifactorial, encompassing vascular, neurological, hormonal, and psychological components, emerging evidence suggests a significant role for gut microbiota dysbiosis in its development. The gut microbiota influences various metabolic, inflammatory, and neuropsychological processes critical to erectile function. Dysbiosis can lead to systemic inflammation, endothelial dysfunction, hormonal imbalances, and altered neurotransmitter production, all of which are key factors in ED pathogenesis. This narrative review synthesizes current research on the association between gut microbiota alterations and ED, highlighting specific bacterial taxa implicated in ED through mechanisms involving inflammation, metabolic disturbances, and hormonal regulation. This review explores potential mechanisms linking gut microbiota and ED, including pro-inflammatory cytokines, gut barrier integrity disruption, metabolic disorders, psychological factors via the gut-brain axis, and hormonal regulation. Furthermore, the gut microbiota offers promising avenues for developing non-invasive biomarkers and therapeutic interventions such as probiotics, prebiotics, dietary modifications, and fecal microbiota transplantation. Future research should focus on longitudinal studies, mechanistic explorations, and clinical trials to validate these findings and translate them into clinical practice. Understanding the interplay between the gut microbiota and erectile function could unveil novel diagnostic biomarkers and pave the way for innovative treatments targeting the microbiota, ultimately improving men's sexual and overall health.
Collapse
Affiliation(s)
- Aris Kaltsas
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Ilias Giannakodimos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Eleftheria Markou
- Department of Microbiology, University Hospital of Ioannina, 45500 Ioannina, Greece;
| | - Konstantinos Adamos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Marios Stavropoulos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Zisis Kratiras
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Athanasios Zachariou
- Laboratory of Spermatology, Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.)
| | - Fotios Dimitriadis
- Department of Urology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Nikolaos Sofikitis
- Laboratory of Spermatology, Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.)
| | - Michael Chrisofos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| |
Collapse
|
43
|
Szabó Á, Galla Z, Spekker E, Szűcs M, Martos D, Takeda K, Ozaki K, Inoue H, Yamamoto S, Toldi J, Ono E, Vécsei L, Tanaka M. Oxidative and Excitatory Neurotoxic Stresses in CRISPR/Cas9-Induced Kynurenine Aminotransferase Knockout Mice: A Novel Model for Despair-Based Depression and Post-Traumatic Stress Disorder. FRONT BIOSCI-LANDMRK 2025; 30:25706. [PMID: 39862084 DOI: 10.31083/fbl25706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/24/2024] [Accepted: 11/18/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUNDS Memory and emotion are especially vulnerable to psychiatric disorders such as post-traumatic stress disorder (PTSD), which is linked to disruptions in serotonin (5-HT) metabolism. Over 90% of the 5-HT precursor tryptophan (Trp) is metabolized via the Trp-kynurenine (KYN) metabolic pathway, which generates a variety of bioactive molecules. Dysregulation of KYN metabolism, particularly low levels of kynurenic acid (KYNA), appears to be linked to neuropsychiatric disorders. The majority of KYNA is produced by the aadat (kat2) gene-encoded mitochondrial kynurenine aminotransferase (KAT) isotype 2. Little is known about the consequences of deleting the KYN enzyme gene. METHODS In CRISPR/Cas9-induced aadat knockout (kat2-/-) mice, we examined the effects on emotion, memory, motor function, Trp and its metabolite levels, enzyme activities in the plasma and urine of 8-week-old males compared to wild-type mice. RESULTS Transgenic mice showed more depressive-like behaviors in the forced swim test, but not in the tail suspension, anxiety, or memory tests. They also had fewer center field and corner entries, shorter walking distances, and fewer jumping counts in the open field test. Plasma metabolite levels are generally consistent with those of urine: antioxidant KYNs, 5-hydroxyindoleacetic acid, and indole-3-acetic acid levels were lower; enzyme activities in KATs, kynureninase, and monoamine oxidase/aldehyde dehydrogenase were lower, but kynurenine 3-monooxygenase was higher; and oxidative stress and excitotoxicity indices were higher. Transgenic mice displayed depression-like behavior in a learned helplessness model, emotional indifference, and motor deficits, coupled with a decrease in KYNA, a shift of Trp metabolism toward the KYN-3-hydroxykynurenine pathway, and a partial decrease in the gut microbial Trp-indole pathway metabolite. CONCLUSIONS This is the first evidence that deleting the aadat gene induces depression-like behaviors uniquely linked to experiences of despair, which appear to be associated with excitatory neurotoxic and oxidative stresses. This may lead to the development of a double-hit preclinical model in despair-based depression, a better understanding of these complex conditions, and more effective therapeutic strategies by elucidating the relationship between Trp metabolism and PTSD pathogenesis.
Collapse
Affiliation(s)
- Ágnes Szabó
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, H-6725 Szeged, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, H-6720 Szeged, Hungary
| | - Zsolt Galla
- Department of Pediatrics, Albert Szent-Györgyi Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary
| | - Eleonóra Spekker
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, H-6725 Szeged, Hungary
| | - Mónika Szűcs
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, Faculty of Science and Informatics, University of Szeged, H-6720 Szeged, Hungary
| | - Diána Martos
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, H-6725 Szeged, Hungary
| | - Keiko Takeda
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - Kinuyo Ozaki
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - Hiromi Inoue
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - Sayo Yamamoto
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - József Toldi
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, H-6726 Szeged, Hungary
| | - Etsuro Ono
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, H-6725 Szeged, Hungary
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, H-6725 Szeged, Hungary
| | - Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, H-6725 Szeged, Hungary
| |
Collapse
|
44
|
He Y, He Y, Cheng B. Identification of Bacterial Lipopolysaccharide-Associated Genes and Molecular Subtypes in Autism Spectrum Disorder. Pharmgenomics Pers Med 2025; 18:1-18. [PMID: 39850061 PMCID: PMC11750731 DOI: 10.2147/pgpm.s494126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/31/2024] [Indexed: 01/25/2025] Open
Abstract
Background Autism spectrum disorder (ASD) is a complex neurodevelopmental condition marked by diverse symptoms affecting social interaction, communication, and behavior. This research aims to explore bacterial lipopolysaccharide (LPS)- and immune-related (BLI) molecular subgroups in ASD to enhance understanding of the disorder. Methods We analyzed 89 control samples and 157 ASD samples from the GEO database, identifying BLI signatures using least absolute shrinkage and selection operator regression (LASSO) and logistic regression machine learning algorithms. A nomogram prediction model was developed based on these signatures, and we performed Gene Set Enrichment Analysis (GSEA), Gene Set Variation Analysis (GSVA), and immune cell infiltration analysis to assess the impact of BLI subtypes and their underlying mechanisms. Results Our findings revealed 17 differentially expressed BLI genes in children with ASD, with BLNK, MAPK8, PRKCQ, and TNFSF12 identified as potential biomarkers. The nomogram demonstrated high diagnostic accuracy for ASD. We delineated two distinct molecular subtypes (Cluster 1 and Cluster 2), with GSVA indicating that Cluster 2 showed upregulation of immune- and inflammation-related pathways. This cluster exhibited increased levels of antimicrobial agents, chemokines, cytokines, and TNF family cytokines, alongside activation of bacterial lipoprotein-related pathways. A significant correlation was found between these pathways and distinct immune cell subtypes, suggesting a potential mechanism for neuroinflammation and immune cell infiltration in ASD. Conclusion Our research highlights the role of BLI-associated genes in the immune responses of individuals with ASD, indicating their contribution to the disorder's typification. The interplay between bacterial components, genetic predisposition, and immune dysregulation offers new insights for understanding ASD and developing personalized interventions.
Collapse
Affiliation(s)
- Yuanxia He
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637000, People’s Republic of China
- Department of Pediatrics, Affiliated Hospital, North Sichuan Medical College, Nanchong, Sichuan, 637000, People’s Republic of China
| | - Yun He
- Department of Pediatrics, Affiliated Hospital, North Sichuan Medical College, Nanchong, Sichuan, 637000, People’s Republic of China
| | - Boli Cheng
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637000, People’s Republic of China
- Department of Pediatrics, Affiliated Hospital, North Sichuan Medical College, Nanchong, Sichuan, 637000, People’s Republic of China
| |
Collapse
|
45
|
Uberoi A, Murga-Garrido SM, Bhanap P, Campbell AE, Knight SAB, Wei M, Chan A, Senay T, Tegegne S, White EK, Sutter CH, Mesaros C, Sutter TR, Grice EA. Commensal-derived tryptophan metabolites fortify the skin barrier: Insights from a 50-species gnotobiotic model of human skin microbiome. Cell Chem Biol 2025; 32:111-125.e6. [PMID: 39824155 PMCID: PMC11753614 DOI: 10.1016/j.chembiol.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 11/01/2024] [Accepted: 12/12/2024] [Indexed: 01/20/2025]
Abstract
The epidermal barrier defends the body against dehydration and harmful substances. The commensal microbiota is essential for proper differentiation and repair of the epidermal barrier, an effect mediated by the aryl hydrocarbon receptor (AHR). However, the microbial mechanisms of AHR activation in skin are less understood. Tryptophan metabolites are AHR ligands that can be products of microbial metabolism. To identify microbially regulated tryptophan metabolites in vivo, we established a gnotobiotic model colonized with fifty human skin commensals and performed targeted mass spectrometry on murine skin. Indole-related metabolites were enriched in colonized skin compared to germ-free skin. In reconstructed human epidermis and in murine models of atopic-like barrier damage, these metabolites improved barrier repair and function individually and as a cocktail. These results provide a framework for the identification of microbial metabolites that mediate specific host functions, which can guide the development of microbe-based therapies for skin disorders.
Collapse
Affiliation(s)
- Aayushi Uberoi
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
| | - Sofía M Murga-Garrido
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Preeti Bhanap
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy E Campbell
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Simon A B Knight
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Monica Wei
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anya Chan
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Taylor Senay
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saba Tegegne
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellen K White
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Clementina Mesaros
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas R Sutter
- Department of Biological Sciences, University of Memphis, Memphis, TN, USA
| | - Elizabeth A Grice
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
46
|
Rodiño-Janeiro BK, Khannous-Lleiffe O, Pigrau M, Willis JR, Salvo-Romero E, Nieto A, Expósito E, Fortea M, Pardo-Camacho C, Albert-Bayo M, González-Castro AM, Guagnozzi D, Martínez C, Lobo B, Vicario M, Santos J, Gabaldón T, Alonso-Cotoner C. Acute stress triggers sex-dependent rapid alterations in the human small intestine microbiota composition. Front Microbiol 2025; 15:1441126. [PMID: 39881982 PMCID: PMC11778178 DOI: 10.3389/fmicb.2024.1441126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Background/aims Digestive disorders of gut-brain interaction (DGBI) are very common, predominant in females, and usually associated with intestinal barrier dysfunction, dysbiosis, and stress. We previously found that females have increased susceptibility to intestinal barrier dysfunction in response to acute stress. However, whether this is associated with changes in the small bowel microbiota remains unknown. We have evaluated changes in the small intestinal microbiota in response to acute stress to better understand stress-induced intestinal barrier dysfunction. Methods Jejunal biopsies were obtained at baseline and 90 min after cold pain or sham stress. Autonomic (blood pressure and heart rate), hormonal (plasma cortisol and adrenocorticotropic hormone) and psychological (Subjective Stress Rating Scale) responses to cold pain and sham stress were monitored. Microbial DNA from the biopsies was analyzed using a 16S metabarcoding approach before and after cold pain stress and sham stress. Differences in diversity and relative abundance of microbial taxa were examined. Results Cold pain stress was associated with a significant decrease in alpha diversity (P = 0.015), which was more pronounced in females, along with significant sex differences in the abundance of specific taxa and the overall microbiota composition. Microbiota alterations significantly correlated with changes in psychological responses, hormones, and gene expression in the intestinal mucosal. Cold pain stress was also associated with activation of autonomic, hormonal and psychological response, with no differences between sexes. Conclusions Acute stress elicits rapid alterations in bacterial composition in the jejunum of healthy subjects and these changes are more pronounced in females. Our results may contribute to the understanding of female predominance in DGBI.
Collapse
Affiliation(s)
- Bruno K. Rodiño-Janeiro
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Olfat Khannous-Lleiffe
- Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marc Pigrau
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Jesse R. Willis
- Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Eloísa Salvo-Romero
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Laboratory of Translational Mucosal Immunology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Adoración Nieto
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Elba Expósito
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Laboratory of Translational Mucosal Immunology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Marina Fortea
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Laboratory of Translational Mucosal Immunology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Cristina Pardo-Camacho
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Laboratory of Translational Mucosal Immunology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Mercé Albert-Bayo
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Laboratory of Translational Mucosal Immunology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Ana María González-Castro
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Laboratory of Translational Mucosal Immunology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Danila Guagnozzi
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Laboratory of Translational Mucosal Immunology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Martínez
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Renal Physiopathology Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Beatriz Lobo
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - María Vicario
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Laboratory of Translational Mucosal Immunology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Javier Santos
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Toni Gabaldón
- Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- Centro de Investigación Biomédica En Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Alonso-Cotoner
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
47
|
Vallejos OP, Bueno SM, Kalergis AM. Probiotics in inflammatory bowel disease: microbial modulation and therapeutic prospects. Trends Mol Med 2025:S1471-4914(24)00338-1. [PMID: 39814640 DOI: 10.1016/j.molmed.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder that represents a significant public health challenge worldwide. This multifactorial condition results from complex interactions among genetic, environmental, immune, and microbial factors. Some beneficial microbes, known as probiotics, have been identified as promising therapeutic agents for inflammatory conditions, such as IBD. In this review, we explore the potential of probiotics as a therapeutic strategy for managing IBD. Probiotics have shown promise due to their ability to modulate the gut microbiota, regulate histamine levels, and enhance vitamin D metabolism, thereby promoting a tolerant immune profile and reducing inflammation. While the exact mechanisms underlying these benefits remain incompletely understood, probiotics represent a novel and emerging approach for alleviating the exacerbated inflammation characteristic of this disorder.
Collapse
Affiliation(s)
- Omar P Vallejos
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Alexis M Kalergis
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
48
|
Wang X, Pan L, Niu D, Zhou J, Shen M, Zeng Z, Gong W, Yang E, Tang Y, Cheng G, Sun C. Jingfang Granules alleviates the lipid peroxidation induced ferroptosis in rheumatoid arthritis rats by regulating gut microbiota and metabolism of short chain fatty acids. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119160. [PMID: 39608616 DOI: 10.1016/j.jep.2024.119160] [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: 09/30/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation, bone and cartilage damage, musculoskeletal pain, swelling, and stiffness. Inflammation is one of the key factors that induce RA. Jingfang Granule (JFG) is a traditional Chinese medicine (TCM) with significant anti-inflammatory effects. Clinical studies have confirmed that JFG can be used to treat RA, but the mechanism is still vague. PURPOSE This study was designed to evaluate the protective function and the mechanism of JFG on rats with RA. STUDY DESIGN AND METHODS Complete Freud's Adjuvant (CFA) was used to establish a rat RA model, and JFG or Diclofenac Sodium (Dic) was orally administered. Foot swelling and hematoxylin eosin (H&E) staining were used to test the therapeutic effect of JFG on RA treatment, while ELISA kits were used to detect serum cytokines. Malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH), catalase (CAT), and reactive oxygen species (ROS) were used to evaluate oxidative stress levels. The integration of label-free proteomics, fecal short chain fatty acid (SCFA) targeted metabolomics, peripheral blood SCFA, medium and long chain fatty acid targeted metabolomics, and 16S rDNA sequencing of gut microbiota were used to screen the mechanism. Western blot technology was used to validate the results of multiple omics studies. Serum D-Lactic acid, lipopolysaccharide specific IgA antibody (LPS IgA), diamine oxidase (DAO), and colon Claudin 5 and ZO-1 were used to evaluate the intestinal barrier. RESULTS The results confirmed that JFG effectively protected rats from RA injury, which was confirmed by improved foot swelling and synovial pathology. At the same time, JFG reduced the levels of TNF-α, IL-1β, and IL-6 in serum by inhibiting the NLRP3 inflammasome signaling pathway and TLR4/NF-κB signaling pathway in synovial tissue. Multiple omics studies indicated that JFG increased the abundance of gut microbiota and regulated the number of gut bacteria, thereby increased the levels of Acetic acid, Propionic acid, and Butyric acid in the gut and serum of RA rats, which activated AMPK to regulate fatty acid metabolism and fatty acid biosynthesis, thereby inhibited lipid oxidative stress induced ferroptosis to improve tissue damage caused by RA. Meanwhile, JFG improved the intestinal barrier by upregulating the expresses of Claudin 5 and ZO-1, which was confirmed by low concentrations of D-Lactic acid, LPS-SIgA and DAO in serum. CONCLUSIONS This study confirmed that JFG improved the disturbance of fatty acid metabolism by modulating gut microbiota and the production of fecal SCFAs to activate AMPK, and then inhibited ferroptosis caused by lipid oxidative stress in synovium tissue and prevented AR injury. This study proposes for the first time to investigate the mechanism of JFG treatment for RA from the perspective of the "Gut-joint" axis, and provides a promising approach for the treatment of RA.
Collapse
Affiliation(s)
- Xiuwen Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China.
| | - Lihong Pan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Dejun Niu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Jidong Zhou
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Mengmeng Shen
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Zhen Zeng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Wenqiao Gong
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Enhua Yang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Yunfeng Tang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Guoliang Cheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Chenghong Sun
- College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, China.
| |
Collapse
|
49
|
Corcione S, Ferrocino I, Lupia T, Busca A, Bianco G, Dellacasa C, Giaccone L, Brunello L, Butera S, Costa C, Bruno B, De Rosa FG. Influence of ESBL colonization status on gut microbiota composition during allogenic hematopoietic stem cell transplantation. Sci Rep 2025; 15:1275. [PMID: 39779737 PMCID: PMC11711636 DOI: 10.1038/s41598-025-85128-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025] Open
Abstract
After allogeneic HSCT (allo-HSCT), the diversity of the intestinal microbiota significantly decreases. The changes can be rapid and are thought to be caused by chemotherapy, antibiotics, or intestinal inflammation. Most patients are exposed to prophylactic and therapeutic antibiotics during neutropenia and several patients are colonized by ESBL bacteria. We investigated the changes in gut microbiota composition in allo-HSCT, aiming at investigating if the acquisition of ESBL colonization may affect gut microbiome diversity during allo-HSCT. This was a single-center prospective pilot study. All patients consecutively admitted to the Haematological Unit of the City of Health and Science, Molinette Hospital in Turin, Italy, and undergoing allo-HSCT between August 2017 to August 2020 were enrolled in the study. Microbiome analysis on fecal samples were collected every 7 days from hospital admission to discharge and until 1 year after HSCT. 48 patients were enrolled in the study. At baseline 14 patients (29.16%) were colonized by MDR bacteria, mostly extended-spectrum beta-lactamase (ESBL)-producing gram negatives (N = 11; 78.57%). During allo-HSCT, one patient had a positive rectal swab for a carbapenemase-producing Klebsiella pneumoniae and eight patients lost the colonization during the hospital stay. Microbiota composition was compared between patients colonized by ESBL at baseline and non-colonized patients. Patients colonized by ESBL had a greater abundances of Bifidobacterium, Blautia, Clostridium, Coprococcus, L-Ruminococcus Mogibacteriaceae, Peptostreptococceae and Oscillospira, while non-colonized ESBL patients had a greater abundance of Actinomycetales, Staphylococcus and Sutterella. Moreover, microbiota composition of colonized by ESBL that retained colonization after HSCT showed an increased in abundances of Akkermansia, Dialister, Erysipelotrichaceae and Methanobrevibacter when compared with patients that become negative at rectal swabs. From a clinical perspective, the evolution of this prospective pilot study will be to investigate markers of gut barrier functions, SCFA productions and to correlate the predictivity of these parameters with risk of invasive infections and clinical outcomes in allo-HSCT population.
Collapse
Affiliation(s)
- Silvia Corcione
- Department of Medical Sciences, Infectious Diseases, University of Turin, Turin, Italy.
- Tufts Medical Center and Tufts University School of Medicine, Boston, MA, USA.
| | - Ilario Ferrocino
- Department of Agriculture, Forest and Food Science, University of Turin, Grugliasco, Italy
| | - Tommaso Lupia
- Department of Medical Sciences, Infectious Diseases, University of Turin, Turin, Italy
| | - Alessandro Busca
- Department of Oncology, Trapianto Allogenico di Cellule Staminali, Azienda Ospedaliera Universitaria (A.O.U.) Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Gabriele Bianco
- Microbiology and Virology Unit, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Chiara Dellacasa
- Department of Oncology, Trapianto Allogenico di Cellule Staminali, Azienda Ospedaliera Universitaria (A.O.U.) Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Luisa Giaccone
- Department of Oncology, Trapianto Allogenico di Cellule Staminali, Azienda Ospedaliera Universitaria (A.O.U.) Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Lucia Brunello
- Department of Oncology, Trapianto Allogenico di Cellule Staminali, Azienda Ospedaliera Universitaria (A.O.U.) Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Sara Butera
- Department of Oncology, Trapianto Allogenico di Cellule Staminali, Azienda Ospedaliera Universitaria (A.O.U.) Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Cristina Costa
- Microbiology and Virology Unit, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Benedetto Bruno
- Department of Oncology, Trapianto Allogenico di Cellule Staminali, Azienda Ospedaliera Universitaria (A.O.U.) Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | | |
Collapse
|
50
|
Jin K, Huang Y, Che H, Wu Y. Engineered Bacteria for Disease Diagnosis and Treatment Using Synthetic Biology. Microb Biotechnol 2025; 18:e70080. [PMID: 39801378 PMCID: PMC11725985 DOI: 10.1111/1751-7915.70080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/18/2024] [Accepted: 12/26/2024] [Indexed: 01/16/2025] Open
Abstract
Using synthetic biology techniques, bacteria have been engineered to serve as microrobots for diagnosing diseases and delivering treatments. These engineered bacteria can be used individually or in combination as microbial consortia. The components within these consortia complement each other, enhancing diagnostic accuracy and providing synergistic effects that improve treatment efficacy. The application of microbial therapies in cancer, intestinal diseases, and metabolic disorders underscores their significant potential. The impact of these therapies on the host's native microbiota is crucial, as engineered microbes can modulate and interact with the host's microbial environment, influencing treatment outcomes and overall health. Despite numerous advancements, challenges remain. These include ensuring the long-term survival and safety of bacteria, developing new chassis microbes and gene editing techniques for non-model strains, minimising potential toxicity, and understanding bacterial interactions with the host microbiota. This mini-review examines the current state of engineered bacteria and microbial consortia in disease diagnosis and treatment, highlighting advancements, challenges, and future directions in this promising field.
Collapse
Affiliation(s)
- Kai Jin
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| | - Yi Huang
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| | - Hailong Che
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| | - Yihan Wu
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| |
Collapse
|