1
|
Su F, Su M, Wei W, Wu J, Chen L, Sun X, Liu M, Sun S, Mao R, Bourgonje AR, Hu S. Integrating multi-omics data to reveal the host-microbiota interactome in inflammatory bowel disease. Gut Microbes 2025; 17:2476570. [PMID: 40063366 PMCID: PMC11901428 DOI: 10.1080/19490976.2025.2476570] [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: 12/17/2024] [Revised: 02/14/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Numerous studies have accelerated the knowledge expansion on the role of gut microbiota in inflammatory bowel disease (IBD). However, the precise mechanisms behind host-microbe cross-talk remain largely undefined, due to the complexity of the human intestinal ecosystem and multiple external factors. In this review, we introduce the interactome concept to systematically summarize how intestinal dysbiosis is involved in IBD pathogenesis in terms of microbial composition, functionality, genomic structure, transcriptional activity, and downstream proteins and metabolites. Meanwhile, this review also aims to present an updated overview of the relevant mechanisms, high-throughput multi-omics methodologies, different types of multi-omics cohort resources, and computational methods used to understand host-microbiota interactions in the context of IBD. Finally, we discuss the challenges pertaining to the integration of multi-omics data in order to reveal host-microbiota cross-talk and offer insights into relevant future research directions.
Collapse
Affiliation(s)
- Fengyuan Su
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Meng Su
- The First Clinical Medical School, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Wenting Wei
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jiayun Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Leyan Chen
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xiqiao Sun
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Moyan Liu
- Amsterdam UMC location Academic Medical Center, Department of Experimental Vascular Medicine, Amsterdam, The Netherlands
| | - Shiqiang Sun
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Arno R. Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shixian Hu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
2
|
Shin S, Chen S, Xie K, Duhun SA, Ortiz-Cerda T. Evaluating the anti-inflammatory and antioxidant efficacy of complementary and alternative medicines (CAM) used for management of inflammatory bowel disease: a comprehensive review. Redox Rep 2025; 30:2471737. [PMID: 40056427 DOI: 10.1080/13510002.2025.2471737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic autoimmune condition whose pathogenesis has not been fully elucidated, and current treatments are not definitive and often carry several side effects. The Complementary and Alternative Medicine (CAM) offers a new approach to conventional medicine. However, their clinical application and mechanisms remain limited.Objective: The aim of this review is to evaluate the anti-inflammatory, impact on microbiota and antioxidant efficacy of currently available CAM for IBD.Methods: The literature collection was obtained from Google Scholar, MEDLINE, PubMed and Web of Science (WOS). Studies in both human and animal models, published in English language between 2018 and 2024, were selected. Sixty-seven studies were included in the current review after inclusion and exclusion screening processes.Results: Mostly, studies showed significant anti-inflammatory, gut microbiota restoring, antioxidant effects of polyphenols, polysaccharides, emodin, short-chain fatty acids (SCFA; including butyrate, propionate and acetate), and probiotics although some contrasting results were noted. Current evidence shows that polyphenols exhibit the most consistent result in alleviating IBD pathophysiology, primarily due to their significant SCFA-elevating effect.Discussion: Future studies may focus on human studies, narrowing down on individual factors which may change natural product's metabolism. Further research studies are also essential to obtain therapeutic recommendations.
Collapse
Affiliation(s)
- Sia Shin
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Siqi Chen
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Kangzhe Xie
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Suehad Abou Duhun
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Tamara Ortiz-Cerda
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Departamento de Citología e Histología Normal y Patológica, Facultad de medicina, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
3
|
Yan R, Zhang L, Chen Y, Zheng Y, Xu P, Xu Z. Therapeutic potential of gut microbiota modulation in epilepsy: A focus on short-chain fatty acids. Neurobiol Dis 2025; 209:106880. [PMID: 40118219 DOI: 10.1016/j.nbd.2025.106880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025] Open
Abstract
According to the criteria established by the International League Against Epilepsy (ILAE), epilepsy is defined as a disorder characterized by at least two unprovoked seizures occurring more than 24 h apart. Its pathogenesis is closely related to various physiological and pathological factors. Advances in high-throughput metagenomic sequencing have increasingly highlighted the role of gut microbiota dysbiosis in epilepsy. Short-chain fatty acids (SCFAs), the major metabolites of the gut microbiota and key regulators of the gut-brain axis, support physiological homeostasis through multiple mechanisms. Recent studies have indicated that SCFAs not only regulate seizures by maintaining intestinal barrier integrity and modulating intestinal immune responses, but also affect the structure and function of the blood-brain barrier (BBB) and regulate neuroinflammation. This review, based on current literatures, explores the relationship between SCFAs and epilepsy, emphasizing how SCFAs affect epilepsy by modulating the intestinal barrier and BBB. In-depth studies on SCFAs may reveal their therapeutic potential and inform the development of gut microbiota-targeted epilepsy treatments.
Collapse
Affiliation(s)
- Rong Yan
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Linhai Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ya Chen
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yongsu Zheng
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ping Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Key Laboratory of Brain Function and Brain Disease Prevention and Treatment of Guizhou Province, Zunyi, China.
| |
Collapse
|
4
|
Weng S, Zheng J, Lin Y, Fang H, Ko CY. Therapeutic effects of amisulpride in male schizophrenics: Role of short-chain fatty acids and gene expression changes. Physiol Behav 2025; 294:114864. [PMID: 40020893 DOI: 10.1016/j.physbeh.2025.114864] [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: 10/02/2024] [Revised: 02/16/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Schizophrenia (SCZ) is a complex disorder characterized by acute symptom exacerbations. Amisulpride, an antipsychotic, has shown effects beyond its primary neurochemical actions, suggesting an influence on the gut microbiome, cytokine modulation, and short-chain fatty acid (SCFA) metabolism. This study aims to investigate these broader effects by examining changes in serum SCFA levels and gene expression profiles in peripheral blood mononuclear cells (PBMCs) following amisulpride treatment. Patients with SCZ undergoing a four-week amisulpride regimen were enrolled. Serum SCFA levels were quantified by gas chromatography, and gene expression profiling was performed in PBMCs using real-time quantitative polymerase chain reaction to assess treatment-associated changes. Results revealed that treatment with amisulpride resulted in a significant increase in serum acetate levels. Gene expression analysis revealed upregulation of G-protein coupled receptor 109a (GPR109a), histone deacetylase 1 (HDAC1), G-protein coupled receptor 43 (GPR43), Toll-like receptor 2 (TLR2), soluble CD14 (sCD14), and N-methyl-d-aspartate receptor (NMDAR), while Toll-like receptor 4 (TLR4) and pregnane X receptor (PXR) were downregulated. These findings suggest that amisulpride may modulate acetate metabolism and immune signaling pathways in SCZ, potentially contributing to anti-inflammatory effects and neuroimmune regulation. The observed increase in acetate, a key microbial metabolite, and the altered expression of immune-related genes suggest a possible link between metabolic shifts and immunomodulatory responses in SCZ pathophysiology. However, direct evidence linking these changes to gut-brain axis mechanisms remains insufficient. Further research is needed to elucidate the therapeutic implications of these metabolic and immunological alterations and their potential role in symptom modulation.
Collapse
Affiliation(s)
- Saizheng Weng
- Department of Psychiatry, Neuropsychiatric Hospital Affiliated to Fujian Medical University, 350008, PR China.
| | - Jinchi Zheng
- The third hospital of Quanzhou, Quanzhou 362000, PR China.
| | - Yichuan Lin
- The third hospital of Quanzhou, Quanzhou 362000, PR China.
| | - Hongqiao Fang
- The third hospital of Quanzhou, Quanzhou 362000, PR China.
| | - Chih-Yuan Ko
- Department of Clinical Nutrition, the Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, PR China.
| |
Collapse
|
5
|
Xiong L, Huang YX, Mao L, Xu Y, Deng YQ. Targeting gut microbiota and its associated metabolites as a potential strategy for promoting would healing in diabetes. World J Diabetes 2025; 16:98788. [DOI: 10.4239/wjd.v16.i5.98788] [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: 07/05/2024] [Revised: 01/03/2025] [Accepted: 03/05/2025] [Indexed: 04/25/2025] Open
Abstract
Impaired healing of diabetic wounds is one of the most important complications of diabetes, often leading to lower limb amputations and incurring significant economic and psychosocial costs. Unfortunately, there are currently no effective prevention or treatment strategies available. Recent research has reported that an imbalance in the gut microbiota, known as dysbiosis, was linked to the onset of type 2 diabetes, as well as the development and progression of diabetic complications. Indeed, the gut microbiota has emerged as a promising therapeutic approach for treating type 2 diabetes and related diseases. However, there is few of literatures specifically discussing the relationship between gut microbiota and diabetic wounds. This review aims to explore the potential role of the gut microbiota, especially probiotics, and its associated byproducts such as short chain fatty acids, bile acids, hydrogen sulfide, and tryptophan metabolites on wound healing to provide fresh insights and novel perspectives for the treatment of chronic wounds in diabetes.
Collapse
Affiliation(s)
- Ling Xiong
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Ya-Xin Huang
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Lan Mao
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yong-Qiong Deng
- Department of Dermatology & STD, Chengdu Integrated TCM & Western Medicine Hospital, Chengdu 610000, Sichuan Province, China
- Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| |
Collapse
|
6
|
Lai Y, Xiong P. Analysis of gut microbiota and depression and anxiety: Mendelian randomization from three datasets. Gen Hosp Psychiatry 2025; 94:206-218. [PMID: 40154232 DOI: 10.1016/j.genhosppsych.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 03/14/2025] [Accepted: 03/14/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Emerging evidence supports gut microbiota's association with mental distress, particularly depression and anxiety, the microbiota-gut-brain axis was the believed to be the underlying mechanism. This study investigated the causal relationships between specific gut microbiota and depression and anxiety disorders using large-scale genome-wide association study (GWAS) data. METHODS A two-sample bidirectional Mendelian randomization (MR) analysis was conducted to explore the causal effects of 211 microbial taxa on depression and anxiety across three large GWAS databases: FinnGen, Pan-UKBB, and PGC. Sensitive analyses were followed to validate the robustness of results. Random-effect meta-analysis was further performed to enhance the statistical power. RESULTS The MR analysis revealed that the Bifidobacteriales (IVW: OR 0.90, 95 %CI 0.83 to 0.98) and Bifidobacteriaceae (IVW: OR 0.90, 95 %CI 0.83 to 0.98) had a protective effect against depression. Clostridiales (cML-MA: OR 0.88, 95 %CI 0.81 to 0.95) and Parasutterella (cML-MA: OR 0.75, 95 %CI 0.64 to 0.88) showed negative associations with depression. Increased abundance of Oxalobacteraceae (cML-MA: OR 1.78, 95 %CI 1.24 to 2.56), Deltaproteobacteria (cML-MA: OR 2.17, 95 %CI 1.38 to 3.40), and Desulfovibrionales (cML-MA: OR 2.22, 95 %CI 1.41 to 3.49) was associated with a higher risk of depression. For anxiety, protective effects were found for Actinobacteria (phylum: IVW: OR 0.83, 95 %CI 0.76 to 0.87; class: IVW: OR 0.84, 95 %CI 0.75 to 0.93), Bifidobacteriales (IVW: OR 0.80, 95 %CI 0.75 to 0.85), Bifidobacteriaceae (IVW: OR 0.80, 95 %CI 0.75 to 0.85) and Bifidobacterium [g] (IVW: OR 0.79, 95 %CI 0.74 to 0.84). Lactobacillaceae [f] (cML-MA: OR 1.18, 95 %CI 1.08 to 1.28), Clostridia [c] (cML-MA: OR 1.15, 95 %CI 0.1.06 to 1.26) and Clostridiales [o] (IVW: OR 1.15, 95 %CI 1.05 to 1.27) were associated with increased anxiety risk. Meta-analysis results indicated significant associations, particularly the protective effects of Actinobacteria (OR 0.90, 95 % CI, 0.83 to 0.98) and Clostridiaceae1 (OR 0.91, 95 % CI, 0.83 to 0.99) on depression and several taxa on anxiety. No significant instrumental variables for depression or anxiety on gut microbiota were identified. CONCLUSIONS Our findings highlight specific gut microbiota that are associated with depression and anxiety, underscoring the causal relationships between these intestinal microbes and psychiatric disorders. These results suggest potential strategies for mitigating disease symptoms and improving quality of life through microbiome-targeted therapies. Further studies, including randomized controlled trials and investigations into sex-specific effects, are essential to validate and expand upon these findings.
Collapse
Affiliation(s)
- Yaoyong Lai
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Peng Xiong
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
7
|
Situ Y, Zhang P, Zhang C, Jiang A, Zhang N, Zhu L, Mou W, Liu Z, Wong HZH, Zhang J, Cheng Q, Lin A, Luo P. The metabolic dialogue between intratumoural microbes and cancer: implications for immunotherapy. EBioMedicine 2025; 115:105708. [PMID: 40267755 DOI: 10.1016/j.ebiom.2025.105708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/25/2025] Open
Abstract
The tumour microenvironment (TME) exerts a profound influence on cancer progression and treatment outcomes. Recent investigations have elucidated the crucial role of intratumoural microbiota and their metabolites in shaping the TME and modulating anti-tumour immunity. This review critically assesses the influence of intratumoural microbial metabolites on the TME and cancer immunotherapy. We systematically analyse how microbial-derived glucose, amino acid, and lipid metabolites modulate immune cell function, cytokine secretion, and tumour growth. The roles of specific metabolites, including lactate, short-chain fatty acids, bile acids, and tryptophan derivatives, are comprehensively examined in regulating immune responses and tumour progression. Furthermore, we investigate the potential of these metabolites to augment the efficacy of cancer immunotherapies, with particular emphasis on immune checkpoint inhibitors. By delineating the mechanisms through which microbial metabolites influence the TME, this review provides insights into novel microbiome-based therapeutic strategies, thereby highlighting a promising frontier in personalised cancer medicine.
Collapse
Affiliation(s)
- Yingheng Situ
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University); Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, China
| | - Lingxuan Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Weiming Mou
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hank Z H Wong
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China.
| | - Anqi Lin
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University); Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Peng Luo
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University); Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| |
Collapse
|
8
|
Xie X, Gu Y, Liu Y, Shen M, Ji J, Gao J, Li J. An inulin-type fructan from Codonopsis pilosula ameliorates cyclophosphamide-induced immunosuppression and intestinal barrier injury in mice. Int J Biol Macromol 2025; 310:143312. [PMID: 40250123 DOI: 10.1016/j.ijbiomac.2025.143312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/08/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
In the present study, an inulin-type fructan (ITF) with the degree polymerization (DP) of 21 was isolated from Codonopsis pilosula roots and its structure was characterized by FT-IR, MALDI-TOF-MS and NMR. The immunomodulatory and intestinal protective effects of ITF were investigated on immunosuppressive mice. Male BALB/c mice were pretreated with cyclophosphamide (Cy) for 3 days to establish an immunosuppressive model followed by ITF treatment. The results demonstrated that compared with the model group, ITF administration significantly increased immune organ index (P<0.05), alleviated intestinal villus damage, stimulated serum cytokine secretion including Ig G, IL-4, IL-6, IL-2, TNF-α, and INF-γ (P<0.05), upregulated the expression of Occludin and Claudin-1 (P<0.05), and increased CD4+ and CD8+ T cells of ileum in Cy-induced mice (P<0.05). Furthermore, ITF restored the intestinal microbiota dysbiosis caused by Cy by increasing the abundance of Muribaculaceae, Blautia, Odoribacter, Lactobacillus and decreasing the abundance of Lachnospiraceae_NK4A136_group (P<0.05). Meanwhile, ITF increased the production of short-chain fatty acids (SCFAs) including acetic acid, propionic acid and butyric acid (P<0.05). These results indicated that ITF can ameliorate cyclophosphamide-induced immunosuppression and intestinal barrier injury, and restore gut microbiota dysbiosis. This study provided important evidences for the immunomodulatory and intestinal protective effects of the ITF from C. pilosula.
Collapse
Affiliation(s)
- Xingfang Xie
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Yao Gu
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Yi Liu
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Mingyue Shen
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Jiaojiao Ji
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Jianping Gao
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China
| | - Jiankuan Li
- School of Pharmaceutical Science, Shanxi Medical University, Jinzhong 030600, China.
| |
Collapse
|
9
|
Ichikawa A, Takayama T, Kojima C, Fujie S, Iemitsu M, Inoue K. Conversion Reaction of Stable-Isotope Oxygen Labeling of Carboxylic Acids for Accurate Screening LC-MS/MS Assay: Application of Behavioral Changes of Short-Chain Fatty Acids in Sports Athletes under Exercise Loading. Anal Chem 2025; 97:7765-7771. [PMID: 40183608 DOI: 10.1021/acs.analchem.4c05872] [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: 04/05/2025]
Abstract
Short-chain fatty acids (SCFAs) have attracted considerable interest as potential biomarkers, therapeutic targets, and nutritional factors in athletic training. SCFAs are typically produced by the intestinal microbiome and exhibit various structural forms, including linear- and branched-chain types. In particular, branched-chain SCFAs have been associated with muscle metabolism during exercise loading. Consequently, accurate and efficient analytical methods are essential for identifying these biomarkers. Liquid chromatography-tandem mass spectrometry is a suitable and accurate technique for SCFA analysis; however, stable isotope calibrations are required for all analytes. Because of technological limitations, the available species are restricted to certain types of SCFAs. To address this issue, this study performed a simple conversion reaction involving the incorporation of 18O into the carboxyl group. Specifically, oxygen atoms in the carboxyl groups were substituted with 18O sourced from commercially available H218O. An SCFA mixture standard solution was successfully labeled under optimized conditions, and the SIL purity and amount were sufficient for isotope dilution (95.2-96.9%, 250 assays using 10 μL of H218O). Moreover, no reversion to 16O was observed during storage or analysis. Analytical validation was performed in human serum using the substituted isotopic standard mixture, achieving good accuracy (90-110%) and precision (<10% relative standard deviation) across three concentration levels. Finally, changes in SCFA patterns were examined in athletes during exercise loading.
Collapse
Affiliation(s)
- Aoi Ichikawa
- Laboratory of Clinical and Analytical Chemistry, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan
| | - Takahiro Takayama
- Laboratory of Clinical and Analytical Chemistry, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan
| | - Chihiro Kojima
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan
| | - Shumpei Fujie
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan
| | - Motoyuki Iemitsu
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan
- Institute of Advanced Research for Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan
| | - Koichi Inoue
- Laboratory of Clinical and Analytical Chemistry, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan
- Institute of Advanced Research for Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan
| |
Collapse
|
10
|
Xiao Z, Xie J, Zhao X, Chen X, Lu Y, Xu Y, Wu M, An L, Li Q. Role of Pyroptosis in inflammatory bowel disease. Int Immunopharmacol 2025; 155:114619. [PMID: 40209313 DOI: 10.1016/j.intimp.2025.114619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
Inflammatory bowel disease (IBD) is a serious chronic condition marked by persistent and recurrent intestinal ulcers. Although the exact cause of IBD remains unclear, it is generally accepted that a complex interaction among dietary factors, gut microbiota, and immune responses in genetically predisposed individuals contributes to its development. Pyroptosis, an inflammatory form of programmed cell death activated by inflammasomes, is marked by the rupture of cell membranes and the subsequent release of inflammatory mediators. Emerging evidence indicates that pyroptosis plays a crucial role in the pathogenesis of IBD. Moderate pyroptosis activation can enhance intestinal immune defenses, while excessive inflammasome activation can trigger an inflammatory cascade, resulting in increased damage to intestinal tissues. This article reviews the molecular mechanisms underlying pyroptosis and highlights its role in the onset and progression of IBD. Furthermore, We explore recent advancements in IBD treatment, focusing on small molecule compounds that specifically target and inhibit pyroptosis.
Collapse
Affiliation(s)
- Zhiyi Xiao
- The Clinical Medical College, Guizhou Medical University, Guiyang 550004, China
| | - Jiling Xie
- The Clinical Medical College, Guizhou Medical University, Guiyang 550004, China
| | - Xun Zhao
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Xiangjun Chen
- The Clinical Medical College, Guizhou Medical University, Guiyang 550004, China
| | - Yihong Lu
- The Clinical Medical College, Guizhou Medical University, Guiyang 550004, China
| | - Yuanzhao Xu
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Manqing Wu
- Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Lingyue An
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China.
| | - Qing Li
- Department of Gastroenterology and Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China.
| |
Collapse
|
11
|
Mohammed MA, Hay NHA, Mohammed MT, Mahmoud HS, Ahmed MY, Abdelmenem A, Abdelrahim DS. The effect of adipose-derived mesenchymal stem cells against high fructose diet induced liver dysfunction and dysbiosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4525-4537. [PMID: 39500806 PMCID: PMC11978704 DOI: 10.1007/s00210-024-03518-5] [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: 06/03/2024] [Accepted: 10/05/2024] [Indexed: 04/10/2025]
Abstract
High fructose diet (HFrD) has been approved to be involved in the pathogenesis of insulin resistance. Mesenchymal stem cells have a vital role in the treatment of various diseases including metabolic disturbances. We investigated the effect of Adipose-derived mesenchymal stem cells (ADMSCs) against HFrD-induced metabolic disorders and the molecular mechanisms for this effect. Rats were divided into 3 groups; control, HFrD, and combined HFrD with ADMSCs. We assessed liver functions, gut microbiota activity, oxidative stress, adiponectin, and IL10 levels. Also, we measured SREBP-1, IRS-1 expression using Western blot, and Malat1 expression using rt-PCR. ADMSCs antagonized metabolic abnormalities induced by HFrD in the form of improvement of liver functions and alleviation of oxidative stress. In addition, ADMSCs ameliorated gut microbiota activity besides the elevation of adiponectin and IL10 levels. ADMSCs attenuated insulin resistance through upregulation of IRS1 and downregulation of SREBP-1 and Malat1. ADMSCs can protect against HFrD-induced metabolic hazards.
Collapse
Affiliation(s)
| | - Nesma Hussein Abel Hay
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Maha Tarek Mohammed
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hoda Sayed Mahmoud
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Manar Yehia Ahmed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed Abdelmenem
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Dina Sayed Abdelrahim
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Department of Pharmacology, Faculty of Medicine, Modern University for Technology and Information, Cairo, Egypt
| |
Collapse
|
12
|
Saadh MJ, Ahmed HH, Kareem RA, Sanghvi G, Ganesan S, Agarwal M, Kaur P, Taher WM, Alwan M, Jawad MJ, Hamad AK. Short-chain fatty acids in Huntington's disease: Mechanisms of action and their therapeutic implications. Pharmacol Biochem Behav 2025; 249:173972. [PMID: 39983928 DOI: 10.1016/j.pbb.2025.173972] [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: 12/03/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder characterized by motor dysfunction, cognitive decline, and emotional instability, primarily resulting from the abnormal accumulation of mutant huntingtin protein. Growing research highlights the role of intestinal microbiota and their metabolites, particularly short-chain fatty acids (SCFAs), in modulating HD progression. SCFAs, including acetate, propionate, and butyrate, are produced by gut bacteria through dietary fiber fermentation and are recognized for their neuroprotective properties. Evidence suggests that SCFAs regulate neuroinflammation, neuronal communication, and metabolic functions within the central nervous system (CNS). In HD, these compounds may support neuronal health, reduce oxidative stress, and enhance blood-brain barrier (BBB) integrity. Their mechanisms of action involve binding to G-protein-coupled receptors (GPCRs) and modulating gene expression through epigenetic pathways, underscoring their therapeutic potential. This analysis examines the significance of SCFAs in HD, emphasizing the gut-brain axis and the benefits of dietary interventions aimed at modifying gut microbiota composition and promoting SCFA production. Further research into these pathways may pave the way for novel HD management strategies and improved therapeutic outcomes.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mohit Agarwal
- Department of Pharmaceutical Chemistry, NIMS Institute of Pharmacy, NIMS University, Rajasthan, Jaipur,302131, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | | | | | | |
Collapse
|
13
|
Zhao T, Wang C, Liu Y, Li B, Shao M, Zhao W, Zhou C. The role of polysaccharides in immune regulation through gut microbiota: mechanisms and implications. Front Immunol 2025; 16:1555414. [PMID: 40230839 PMCID: PMC11994737 DOI: 10.3389/fimmu.2025.1555414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 03/05/2025] [Indexed: 04/16/2025] Open
Abstract
Polysaccharides, as complex carbohydrates, play a pivotal role in immune modulation and interactions with the gut microbiota. The diverse array of dietary polysaccharides influences gut microbial ecology, impacting immune responses, metabolism, and overall well-being. Despite their recognized benefits, there is limited understanding of the precise mechanisms by which polysaccharides modulate the immune system through the gut microbiota. A comprehensive search of Web of Science, PubMed, Google Scholar, and Embase up to May 2024 was conducted to identify relevant studies. This study employs a systematic approach to explore the interplay between polysaccharides and the gut microbiota, focusing on cytokine-mediated and short-chain fatty acid (SCFA)-mediated pathways. The findings underscore the significant role of polysaccharides in shaping the composition and function of the gut microbiota, thereby influencing immune regulation and metabolic processes. However, further research is necessary to elucidate the detailed molecular mechanisms and translate these findings into clinical applications.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Congyue Wang
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Yuhan Liu
- Department of Medical Oncology, Anshan Cancer Hospital, Anshan, China
| | - Bo Li
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Mingjia Shao
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Wuyang Zhao
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Chuang Zhou
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| |
Collapse
|
14
|
Shu JZ, Huang YH, He XH, Liu FY, Liang QQ, Yong XT, Xie YF. Gut microbiota differences, metabolite changes, and disease intervention during metabolic - dysfunction - related fatty liver progression. World J Hepatol 2025; 17:103854. [PMID: 40177201 PMCID: PMC11959672 DOI: 10.4254/wjh.v17.i3.103854] [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: 12/12/2024] [Revised: 01/17/2025] [Accepted: 02/12/2025] [Indexed: 03/26/2025] Open
Abstract
In the current era, metabolic dysfunction-associated steatotic liver disease (MASLD) has gradually developed into a major type of chronic liver disease that is widespread globally. Numerous studies have shown that the gut microbiota plays a crucial and indispensable role in the progression of MASLD. Currently, the gut microbiota has become one of the important entry points for the research of this disease. Therefore, the aim of this review is to elaborate on the further associations between the gut microbiota and MASLD, including the changes and differences in the microbiota between the healthy liver and the diseased liver. Meanwhile, considering that metabolic dysfunction-associated fatty liver and metabolic dysfunction-associated steatohepatitis are abnormal pathological states in the development of the disease and that the liver exhibits different degrees of fibrosis (such as mild fibrosis and severe fibrosis) during the disease progression, we also conduct a comparison of the microbiota in these states and use them as markers of disease progression. It reveals the changes in the production and action mechanisms of short-chain fatty acids and bile acids brought about by changes in the gut microbiota, and the impact of lipopolysaccharide from Gram-negative bacteria on the disease. In addition, the regulation of the gut microbiota in disease and the production and inhibition of related disease factors by the use of probiotics (including new-generation probiotics) will be explored, which will help to monitor the disease progression of patients with different gut microbiota compositions in the future and carry out personalized targeted therapies for the gut microbiota. This will achieve important progress in preventing and combating this disease.
Collapse
Affiliation(s)
- Jian-Zhong Shu
- Department of Encephalopathy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400015, China
- School of Life Health Information Science and Engineering, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
- College of Integrated Traditional Chinese and Western Medicine, Chongqing University of Traditional Chinese Medicine, Chongqing 402760, China
| | - Yu-Han Huang
- School of Life Health Information Science and Engineering, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| | - Xiao-Hong He
- School of Life Health Information Science and Engineering, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| | - Feng-Ying Liu
- School of Life Health Information Science and Engineering, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| | - Qian-Qian Liang
- School of Life Health Information Science and Engineering, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| | - Xue-Tong Yong
- School of Life Health Information Science and Engineering, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| | - Yong-Fang Xie
- School of Life Health Information Science and Engineering, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
- Institute of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing 400065, China.
| |
Collapse
|
15
|
Simon P, Török É, Szalontai K, Kari B, Neuperger P, Zavala N, Kanizsai I, Puskás LG, Török S, Szebeni GJ. Nutritional Support of Chronic Obstructive Pulmonary Disease. Nutrients 2025; 17:1149. [PMID: 40218907 PMCID: PMC11990120 DOI: 10.3390/nu17071149] [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: 02/05/2025] [Revised: 03/14/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Background: COPD is a heterogenous disease of the respiratory tract caused by diverse genetic factors along with environmental and lifestyle-related effects such as industrial dust inhalation and, most frequently, cigarette smoking. These factors lead to airflow obstruction and chronic respiratory symptoms. Additionally, the increased risk of infections exacerbates airway inflammation in COPD patients. As a consequence of the complex pathomechanisms and difficulty in treatment, COPD is among the leading causes of mortality both in the western countries and in the developing world. Results: The management of COPD is still a challenge for the clinicians; however, alternative interventions such as smoking cessation and lifestyle changes from a sedentary life to moderate physical activity with special attention to the diet may ameliorate patients' health. Here, we reviewed the effects of different dietary components and supplements on the conditions of COPD. Conclusions: COPD patients are continuously exposed to heavy metals, which are commonly present in cigarette smoke and polluted air. Meanwhile, they often experience significant nutrient deficiencies, which affect the detoxification of these toxic metals. This in turn can further disrupt nutritional balance by interfering with the absorption, metabolism, and utilization of essential micronutrients. Therefore, awareness and deliberate efforts should be made to check levels of micronutrients, with special attention to ensuring adequate levels of antioxidants, vitamin D, vitamin K2, magnesium, and iron, as these may be particularly important in reducing the risk of COPD development and limiting disease severity.
Collapse
Grants
- 2023-1.1.1-PIACI_FÓKUSZ-2024-00036 National Research, Development, and Innovation Office (NKFI), Hungary
- 2020-1.1.6-JÖVŐ-2021-00003 National Research, Development, and Innovation Office (NKFI), Hungary
- 2022-1.2.6-TÉT-IPARI-TR-2022-00023 National Research, Development, and Innovation Office (NKFI), Hungary
- 142877 FK22 National Research, Development, and Innovation Office (NKFI), Hungary.
- BO/00582/22/8 János Bolyai Research Scholarship of the Hungarian Academy of Sciences
Collapse
Affiliation(s)
- Péter Simon
- National Korányi Institute of Pulmonology, 1121 Budapest, Hungary;
| | - Éva Török
- Gastroenterology Center Buda, 1117 Budapest, Hungary;
| | - Klára Szalontai
- Department of Pulmonology, Szent-Györgyi Albert Medical Center, University of Szeged, 6772 Deszk, Hungary;
| | - Beáta Kari
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
| | - Patrícia Neuperger
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
| | - Norma Zavala
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
| | | | - László G. Puskás
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
- Anthelos Ltd., 6726 Szeged, Hungary
| | - Szilvia Török
- National Korányi Institute of Pulmonology, 1121 Budapest, Hungary;
| | - Gabor J. Szebeni
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
- Department of Internal Medicine, Hematology Centre, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary
| |
Collapse
|
16
|
Firoozi D, Masoumi SJ, Mohammad-Kazem Hosseini Asl S, Fararouei M, Jamshidi S. Effects of Short Chain Fatty Acid-Butyrate Supplementation on the Disease Severity, Inflammation, and Psychological Factors in Patients With Active Ulcerative Colitis: A Double-Blind Randomized Controlled Trial. J Nutr Metab 2025; 2025:3165876. [PMID: 40123849 PMCID: PMC11930386 DOI: 10.1155/jnme/3165876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 02/13/2025] [Indexed: 03/25/2025] Open
Abstract
Background: Depression and anxiety are common in UC patients due to gut microbiota dysbiosis and increased proinflammatory markers. Butyrate, a short-chain fatty acid, participates in the regulation of gut microbiota and inflammation and has neuroprotective effects in neurodegenerative disease. Therefore, we assessed the effects of sodium butyrate supplementation on the disease severity, inflammation, and psychological factors in active UC patients. Methods: This study was a randomized, parallel, double-blind controlled trial. Participants in the intervention (n = 18) and control (n = 18) groups received 600 mg/kg of sodium butyrate or rice starch as a placebo with their main meal, respectively, for 12 weeks. The partial Mayo score was used to evaluate disease severity, while the Westergren method was employed to assess the erythrocyte sedimentation rate (ESR). NLR and PLR were determined using an automated analyzer (XS-500i, Sysmex). Moreover, the psychological factors were assessed by the hospital anxiety depression scale (HADS) and the general health questionnaire (GHQ). Results: In comparison with placebo, sodium-butyrate supplementation significantly decreased the ESR level (-6.66 ± 1.56 vs. 3.00 ± 2.11, p=0.01), NLR (-0.24 ± 0.1 vs. 0.33 ± 0.23, p=0.02), Mayo score (-2.33 ± 0.41 vs. 0.22 ± 0.40, p < 0.001), HADS anxiety score (-2.77 ± 0.64 vs. 0.94 ± 0.63, p=0.001), HADS depression score (-2.38 ± 0.47 vs. 0.61 ± 0.33, p < 0.001), and GHQ total score (-12.11 ± 1.48 vs. 3.55 ± 1.39, p < 0.001). Conclusion: Butyrate could serve as an effective adjuvant treatment for reducing disease severity and alleviating psychological symptoms. This trial was registered on the Research Ethics Committee of Shiraz University of Medical Sciences, with the reference number IR.SUMS.SCHEANUT.REC.1400.037. Trial Registration: Iranian Registry of Clinical Trials: IRCT20211214053401N1.
Collapse
Affiliation(s)
- Donya Firoozi
- Student Research Committee, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Science, Shiraz, Iran
| | - Seyed Jalil Masoumi
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Science, Shiraz, Iran
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammad Fararouei
- Department of Epidemiology, School of Public Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sanaz Jamshidi
- Center for Cohort Study of Shiraz University of Medical Sciences Employees' Health, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
17
|
Attiq A. Early-life antibiotic exposures: Paving the pathway for dysbiosis-induced disorders. Eur J Pharmacol 2025; 991:177298. [PMID: 39864578 DOI: 10.1016/j.ejphar.2025.177298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/06/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Microbiota encompasses a diverse array of microorganisms inhabiting specific ecological niches. Gut microbiota significantly influences physiological processes, including gastrointestinal motor function, neuroendocrine signalling, and immune regulation. They play a crucial role in modulating the central nervous system and bolstering body defence mechanisms by influencing the proliferation and differentiation of innate and adaptive immune cells. Given the potential consequences of antibiotic therapy on gut microbiota equilibrium, there is a need for prudent antibiotic use to mitigate associated risks. Observational studies have linked increased antibiotic usage to various pathogenic conditions, including obesity, inflammatory bowel disease, anxiety-like effects, asthma, and pulmonary carcinogenesis. Addressing dysbiosis incidence requires proactive measures, including prophylactic use of β-lactamase drugs (SYN-004, SYN-006, and SYN-007), hydrolysing the β-lactam in the proximal GIT for maintaining intestinal flora homeostasis. Prebiotic and probiotic supplementations are crucial in restoring intestinal flora equilibrium by competing with pathogenic bacteria for nutritional resources and adhesion sites, reducing luminal pH, neutralising toxins, and producing antimicrobial agents. Faecal microbiota transplantation (FMT) shows promise in restoring gut microbiota composition. Rational antibiotic use is essential to preserve microflora and improve patient compliance with antibiotic regimens by mitigating associated side effects. Given the significant implications on gut microbiota composition, concerted intervention strategies must be pursued to rectify and reverse the occurrence of antibiotic-induced dysbiosis. Here, antibiotics-induced microbiota dysbiosis mechanisms and their systemic implications are reviewed. Moreover, proposed interventions to mitigate the impact on gut microflora are also discussed herein.
Collapse
Affiliation(s)
- Ali Attiq
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| |
Collapse
|
18
|
Liu Y, Li F, Wang J, Yang R. Exploring effects of gut microbiota on tertiary lymphoid structure formation for tumor immunotherapy. Front Immunol 2025; 15:1518779. [PMID: 40124706 PMCID: PMC11925796 DOI: 10.3389/fimmu.2024.1518779] [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: 10/28/2024] [Accepted: 12/20/2024] [Indexed: 03/25/2025] Open
Abstract
Anti-tumor immunity, including innate and adaptive immunity is critical in inhibiting tumorigenesis and development of tumor. The adaptive immunity needs specific lymph organs such as tertiary lymphoid structures (TLSs), which are highly correlated with improved survival outcomes in many cancers. In recent years, with increasing attention on the TLS in tumor microenvironment, TLSs have emerged as a novel target for anti-tumor therapy. Excitingly, studies have shown the contribution of TLSs to the adaptive immune responses. However, it is unclear how TLSs to form and how to more effectively defense against tumor through TLS formation. Recent studies have shown that the inflammation plays a critical role in TLS formation. Interestingly, studies have also found that gut microbiota can regulate the occurrence and development of inflammation. Therefore, we here summarize the potential effects of gut microbiota- mediated inflammation or immunosuppression on the TLS formation in tumor environments. Meanwhile, this review also explores how to manipulate mature TLS formation through regulating gut microbiota/metabolites or gut microbiota associated signal pathways for anti-tumor immunity, which potentially lead to a next-generation cancer immunotherapy.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Fan Li
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Juanjuan Wang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
| |
Collapse
|
19
|
Liu MJ, Zhang Y, Zhu K, Li WW, Liu C, Jiang S, Shang EX, Duan JA. Xiexin Tang restores gut barrier function by regulating the differentiation of CD4 + T cells via GPRs and HDACs in T2DM rats. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025:1-15. [PMID: 40029093 DOI: 10.1080/10286020.2025.2459603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 03/05/2025]
Abstract
This study aimed to explore the potential mechanism of Xiexin Tang in improving type 2 diabetes mellitus from the perspective of intestinal barrier function. The results indicated that Xiexin Tang could notably promote the expression of GPRs while suppressing the expression of HDACs in colon epithelial cells, then significantly elevate the levels of TGF-β1 and IL-18 to regulate the differentiation of T cells and further maintain the intestinal immune homeostasis. Meanwhile, it could markedly inhibit the inflammatory signaling pathway to improve intestinal barrier function, relieving type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Mei-Juan Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing210023, China
| | - Yun Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing210023, China
| | - Ke Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing210023, China
| | - Wen-Wen Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing210023, China
| | - Chen Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing210023, China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing210023, China
| | - Er-Xin Shang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing210023, China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing210023, China
| |
Collapse
|
20
|
Barra NG, Fang H, Bhatwa A, Schmidt AM, Syed SA, Steinberg GR, Morrison KM, Surette MG, Wade MG, Holloway AC, Schertzer JD. Food supply toxicants and additives alter the gut microbiota and risk of metabolic disease. Am J Physiol Endocrinol Metab 2025; 328:E337-E353. [PMID: 39871724 DOI: 10.1152/ajpendo.00364.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/09/2024] [Accepted: 01/13/2025] [Indexed: 01/29/2025]
Abstract
Metabolic disease is rising along with both global industrialization and the use of new commercial, agricultural, and industrial chemicals and food additives. Exposure to these compounds may contribute to aspects of metabolic diseases such as obesity, diabetes, and fatty liver disease. Ingesting compounds in the food supply is a key route of human exposure, resulting in the interaction between toxicants or additives and the intestinal microbiota. Toxicants can influence the composition and function of the gut microbiota, and these microbes can metabolize and transform toxicants and food additives. Microbe-toxicant interactions in the intestine can alter host mucosal barrier function, immunity, and metabolism, which may contribute to the risk or severity of metabolic disease development. Targeting the connection between toxicants, food, and immunity in the gut using strategies such as fermentable fiber (i.e., inulin) may mitigate some of the effects of these compounds on host metabolism. Understanding causative factors in the microbe-host relationship that promote toxicant-induced dysmetabolism is an important goal. This review highlights the role of common toxicants (i.e., persistent organic pollutants, pesticides, and fungicides) and food additives (emulsifiers and artificial sweeteners) found in our food supply that alter the gut microbiota and promote metabolic disease development.
Collapse
Affiliation(s)
- Nicole G Barra
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Han Fang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Arshpreet Bhatwa
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Angela M Schmidt
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Saad A Syed
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Gregory R Steinberg
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Katherine M Morrison
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Michael G Wade
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Alison C Holloway
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
21
|
Chenghan M, Wanxin L, Bangcheng Z, Yao H, Qinxi L, Ting Z, Xiaojie L, Kun Z, Yingqian Z, Zhihui Z. Short-chain fatty acids mediate gut microbiota-brain communication and protect the blood-brain barrier integrity. Ann N Y Acad Sci 2025; 1545:116-131. [PMID: 39998158 DOI: 10.1111/nyas.15299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
The human gut, with a complex community of microbes, is essential for maintaining overall health. This gut microbiota engages in two-way communication with the central nervous system, collectively known as the gut microbiota-brain axis. Alterations in gut microbiota have been associated with various neurological disorders, and disruptions to the blood-brain barrier (BBB) may be crucial, though the exact mechanisms remain unknown. In the current study, we investigated the impacts of short-chain fatty acids (SCFAs) on the integrity of the BBB, which was compromised by orally administered antibiotics in rhesus monkeys and C57BL/6n mice. Our results showed that SCFA supplementation notably enhanced BBB integrity in rhesus monkeys with gut dysbiosis. Similar outcomes were observed in mice with gut dysbiosis, accompanied by decreased cortical claudin-5 mRNA levels. In particular, propionate, but not acetate or butyrate, could reverse the antibiotic-induced BBB permeability increase in mice. Additionally, in vitro studies demonstrated that propionate boosted the expression of tight junction proteins in brain endothelial cells. These results suggest that the propionate can maintain BBB integrity through a free fatty acid receptor 2-dependent mechanism. This study offers new insights into the gut-brain axis and underscores potential therapeutic targets for interventions based on gut microbiota.
Collapse
Affiliation(s)
- Mei Chenghan
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurological Disease Modeling and Translational Research, Institute of Neurological Diseases, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China
- Guizhou Academy of Testing and Analysis, Guizhou Academy of Sciences, Guiyang, China
| | - Li Wanxin
- Department of Pharmacy, Shaanxi Provincial Cancer Hospital, Xi'an, China
| | | | - He Yao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurological Disease Modeling and Translational Research, Institute of Neurological Diseases, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China
| | - Li Qinxi
- Sichuan Junhui Biotechnology Co., Ltd, Chengdu, China
| | - Zhang Ting
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurological Disease Modeling and Translational Research, Institute of Neurological Diseases, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China
| | - Li Xiaojie
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurological Disease Modeling and Translational Research, Institute of Neurological Diseases, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China
| | - Zhang Kun
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Zhang Yingqian
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurological Disease Modeling and Translational Research, Institute of Neurological Diseases, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Junhui Biotechnology Co., Ltd, Chengdu, China
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhong Zhihui
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurological Disease Modeling and Translational Research, Institute of Neurological Diseases, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Sandu R, Singh J. A comprehensive review on calcitonin gene-related peptide in the management of gastrointestinal disorders. Inflammopharmacology 2025; 33:1043-1059. [PMID: 39934537 DOI: 10.1007/s10787-025-01657-6] [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: 12/06/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025]
Abstract
The prevalence of gastrointestinal disorders caused by alcohol, Helicobacter pylori, non-steroidal anti-inflammatory drugs, chronic stress and sedentary lifestyle is on the rise. Calcitonin gene-related peptide (CGRP), a 37-amino acid neuropeptide, has emerged as a protective factor against various gastrointestinal issues. Despite its known benefits, the dual role of CGRP in gastrointestinal damage remains unclear. Discovered 30 years ago through alternative RNA processing of the calcitonin gene, CGRP is known to be a potent vasodilator involved in crucial defensive mechanisms for both physiological and pathological conditions. Promising evidences from preclinical research have attracted the interest of scientists for the exploration of CGRP as a therapeutic neuropeptide. Numerous evidences suggest that this neuropeptide is secreted by the neurons under the influence of endogenous as well as exogenous stimuli. CGRP repairs the gastric mucosal barrier and maintain mucosal integrity by suppressing NF-κB activation, thereby reducing tumour necrosis factor-alpha expression. In addition, recent studies suggest that CGRP modulates immune responses and enhances epithelial cell proliferation, further contributing to its cytoprotective effects. Consequently, CGRP and the CGRP secretagogues represent promising novel targets for clinical applications. This review aims to elucidate the role of CGRP and CGRP secretagogues in the management of gastrointestinal disorders, highlighting its potential as a therapeutic agent in the context of evidence-based modern gastroenterology.
Collapse
Affiliation(s)
- Rajesh Sandu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062, Punjab, India
| | - Jagtar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062, Punjab, India.
| |
Collapse
|
23
|
Liu Y, Yan D, Chen R, Zhang Y, Wang C, Qian G. Recent insights and advances in gut microbiota's influence on host antiviral immunity. Front Microbiol 2025; 16:1536778. [PMID: 40083779 PMCID: PMC11903723 DOI: 10.3389/fmicb.2025.1536778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
A diverse array of microbial organisms colonizes the human body, collectively known as symbiotic microbial communities. Among the various pathogen infections that hosts encounter, viral infections represent one of the most significant public health challenges worldwide. The gut microbiota is considered an important biological barrier against viral infections and may serve as a promising target for adjuvant antiviral therapy. However, the potential impact of symbiotic microbiota on viral infection remains relatively understudied. In this review, we discuss the specific regulatory mechanisms of gut microbiota in antiviral immunity, highlighting recent advances in how gut microbiota regulate the host immune response, produce immune-related molecules, and enhance the host's defense against viruses. Finally, we also discuss the antiviral potential of oral probiotics.
Collapse
Affiliation(s)
- Ying Liu
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Danying Yan
- Department of Infectious Diseases, The First Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Ran Chen
- Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yingying Zhang
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Chuwen Wang
- Department of Infectious Diseases, The First Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Guoqing Qian
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- Department of Infectious Diseases, The First Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
24
|
Fachi JL, de Oliveira S, Trsan T, Penati S, Gilfillan S, Cao S, Ribeiro Castro P, Fernandes MF, Hyrc KL, Liu X, Rodrigues PF, Bhattarai B, Layden BT, Vinolo MAR, Colonna M. Fiber- and acetate-mediated modulation of MHC-II expression on intestinal epithelium protects from Clostridioides difficile infection. Cell Host Microbe 2025; 33:235-251.e7. [PMID: 39826540 PMCID: PMC11974464 DOI: 10.1016/j.chom.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/18/2024] [Accepted: 12/27/2024] [Indexed: 01/22/2025]
Abstract
Here, we explore the relationship between dietary fibers, colonic epithelium major histocompatibility complex class II (MHC-II) expression, and immune cell interactions in regulating susceptibility to Clostridioides difficile infection (CDI). We find that a low-fiber diet increases MHC-II expression in the colonic epithelium, which, in turn, worsens CDI by promoting the development of pathogenic CD4+ intraepithelial lymphocytes (IELs). The influence of dietary fibers on MHC-II expression is mediated by its metabolic product, acetate, and its receptor, free fatty acid receptor 2 (FFAR2). While acetate activation of FFAR2 on epithelial cells helps resist CDI, it does not directly regulate MHC-II expression. Instead, MHC-II is regulated by FFAR2 in type 3 innate lymphoid cells (ILC3s). Acetate enhances interleukin-22 (IL-22) production by ILC3s, which then suppresses MHC-II expression on the colonic epithelium. In conclusion, a low-fiber diet reduces acetate-induced IL-22 production by ILC3s, leading to increased MHC-II on the colonic epithelium. This change affects recovery from CDI by expanding the population of pathogenic CD4+ IELs.
Collapse
Affiliation(s)
- José L Fachi
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA.
| | - Sarah de Oliveira
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | - Tihana Trsan
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA
| | - Silvia Penati
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA
| | - Susan Gilfillan
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA
| | - Siyan Cao
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA; Division of Gastroenterology, Department of Medicine, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA
| | - Pollyana Ribeiro Castro
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | - Mariane Font Fernandes
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | - Krzysztof L Hyrc
- Alafi Neuroimaging Laboratory, The Hope Center of Neurological Disorders, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA
| | - Xiuli Liu
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA
| | - Patrick Fernandes Rodrigues
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA
| | - Bishan Bhattarai
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA
| | - Brian T Layden
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Marco Aurélio R Vinolo
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110, USA.
| |
Collapse
|
25
|
Ren K, Yong C, Jin Y, Rong S, Xue K, Cao B, Wei H. Unraveling the microbial mysteries: gut microbiota's role in ulcerative colitis. Front Nutr 2025; 12:1519974. [PMID: 39996003 PMCID: PMC11847676 DOI: 10.3389/fnut.2025.1519974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by persistent inflammation of the colon. Recent research has highlighted the significant role of gut microbiota in the pathogenesis and treatment of UC. This review aims to provide a comprehensive overview of the current understanding of the relationship between gut microbiota and UC. We discuss the involvement of gut microbiota in the onset of UC, including the dysbiosis observed in patients and its potential mechanisms. Additionally, the role of extra-intestinal microbiota in UC pathogenesis is explored, which has been less studied but is gaining attention. The influence of gut microbiota on the efficacy of biological immunotherapy for UC is also examined, highlighting how microbial composition can influence treatment outcomes. Furthermore, we review microbiota transplantation, and their potential benefits in UC management. Finally, we consider the combined use of antibiotics and biological agents in UC treatment, discussing their synergistic effects and potential drawbacks. This review underscores the importance of gut microbiota in UC and suggests that targeting microbial communities could offer new avenues for effective treatment.
Collapse
Affiliation(s)
- Keyu Ren
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chunming Yong
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanchun Jin
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shanwei Rong
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kuijin Xue
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Cao
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongyun Wei
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
26
|
Song Z, Qiao Z, Liu J, Han L, Chen X, Wang Y. Sea buckthorn berries alleviate ulcerative colitis via regulating gut Faecalibaculum rodentium-mediated butyrate biosynthesis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156490. [PMID: 40010029 DOI: 10.1016/j.phymed.2025.156490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND The gut microbiota is firmly associated with the progression of ulcerative colitis (UC). Beneficial microbial metabolites, such as butyrate, exert vital roles in maintaining intestinal homeostasis. The Sea buckthorn berry is a traditional Chinese medicine homologous to food and medicine which is widely applied in the prevention and treatment of UC in clinic practice. Recent studies have exhibited the potential function of Sea buckthorn on regulating the gut microbiota, however, the mechanism underlying its anti-colitis effects and the key gut microbes mediating its efficacy are still unclear. PURPOSE This study is intended to explore the pharmacological mechanism of the efficacy of Sea buckthorn berries extract (SBE) in alleviating UC from the perspective of the gut microbial regulation. METHODS The effect of SBE on UC was evaluated on dextran sulfate sodium (DSS)-induced murine model by assessing the body weight change, colon length, disease activity index (DAI), histopathological staining and the transcriptional expression of genes associated with inflammation and mucosal integrity. The dependence of the gut microbiota in the therapeutical effects of SBE on UC was confirmed by pseudo-germ-free mice and the co-housing experiment. The differential gut microbes altered by SBE were discovered by 16S rRNA sequencing and qPCR. The levels of short chain fatty acids (SCFAs) in bacterial medium and colonic contents were determined by GC-MS/MS. Bacterial colonization was conducted to estimate the effects of the bacteria on UC and to verify the involvement of the functional bacteria in the efficacy of SBE. A butyrate receptor G protein-coupled receptor (GPR)109A antagonist mepenzolate bromide (MPN) was used to validate the important role of butyrate and GPR109A in the anti-colitis effects of SBE and functional bacteria on UC. Two-way ANOVA was employed for multiple curve comparison and One-way ANOVA and Brown-Forsythe ANOVA test were used for multiple group comparison. Statistical significance was defined as p< 0.05. RESULTS SBE treatment significantly alleviated DSS-induced UC and its therapeutical effects was impaired in pseudo-germ-free mice. Moreover, the co-housing experiment exhibited that SBE-altered microbiota could effectively ameliorate UC. Further research demonstrated that Faecalibaculum rodentium was obviously increased by SBE and could be transferable by co-housing. Moreover, butyrate, a product of F. rodentium, dramatically decreased in UC mice while could be recovered by SBE administration. Abolishment of F. rodentium using vancomycin deprived the efficacy of SBE, but this could be reversed by recolonization of F. rodentium. Finally, blockage of the butyrate's receptor GPR109A weakened the effects of F. rodentium, indicating the indispensability of butyrate and GPR109A in the anti-colitis effects of F. rodentium. CONCLUSION SBE has potent therapeutical efficacy on UC including relieving inflammation and enhancing intestinal epithelial integrity, which is mediated by the gut F. rodentium-regulated butyrate-GPR109A axis.
Collapse
Affiliation(s)
- Zhe Song
- China Pharmaceutical University Center for Analysis and Testing, China Pharmaceutical University, Nanjing, PR China.
| | - Zhou Qiao
- China Pharmaceutical University Center for Analysis and Testing, China Pharmaceutical University, Nanjing, PR China.
| | - Jia Liu
- The Animal Experimental Center, China Pharmaceutical University, Nanjing, PR China.
| | - Lingfei Han
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, PR China.
| | - Xi Chen
- China Pharmaceutical University Center for Analysis and Testing, China Pharmaceutical University, Nanjing, PR China.
| | - Yun Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, PR China.
| |
Collapse
|
27
|
Li C, Yao J, Yang C, Yu S, Yang Z, Wang L, Li S, He N. Gut microbiota-derived short chain fatty acids act as mediators of the gut-liver-brain axis. Metab Brain Dis 2025; 40:122. [PMID: 39921774 DOI: 10.1007/s11011-025-01554-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
The gut microbiota plays a crucial role in the communication between the gut, liver, and brain through the production of short chain fatty acids (SCFAs). SCFAs serve as key mediators in the Gut-Liver-Brain Axis, influencing various physiological processes and contributing to overall health. SCFAs are produced by bacterial fermentation of dietary fiber in the gut, and they exert systemic effects by signaling through various pathways. In the Gut-Liver axis, SCFAs regulate liver metabolism through peroxisome proliferator-activated receptor-γ (PPAR-γ), AMP-activated protein kinase (AMPK) and other pathways, promotes fat oxidation, modulate inflammation through mTOR pathway, and impact metabolic health. In the Gut-Brain axis, SCFAs influence brain function, behavior, and may have implications for neurological disorders, in which G-protein coupled receptors (GPCRs) play an essential role, along with other pathways such as hypothalamic-pituitary-adrenal (HPA) pathway. Understanding the mechanisms by which SCFAs mediate communication between the gut, liver, and brain is crucial for elucidating the complex interplay of the Gut-Liver-Brain Axis. This review aims to provide insight into the role of gut microbiota-derived SCFAs as mediators of the Gut-Liver-Brain Axis and their potential therapeutic implications. Further research in this area will be instrumental in developing novel strategies to target the Gut-Liver-Brain Axis for the prevention and treatment of various health conditions.
Collapse
Affiliation(s)
- Cunyin Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
- Department of Obstetrics and Gynecology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, 266000, China
| | - Jingtong Yao
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
| | - Chang Yang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
| | - Shengnan Yu
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
- Affiliated Hospital of Inner Mongolia University for Nationalities, TongLiao, 028005, China
| | - Zizhen Yang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
| | - Lijing Wang
- Department of Obstetrics and Gynecology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, 266000, China.
- Department of Obstetrics, Qingdao Municipal Hospital, Qingdao, 266000, China.
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
- Department of Ultrasound, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
28
|
Yu W, Sun S, Yan Y, Zhou H, Liu Z, Fu Q. The role of short-chain fatty acid in metabolic syndrome and its complications: focusing on immunity and inflammation. Front Immunol 2025; 16:1519925. [PMID: 39991152 PMCID: PMC11842938 DOI: 10.3389/fimmu.2025.1519925] [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: 10/30/2024] [Accepted: 01/09/2025] [Indexed: 02/25/2025] Open
Abstract
Metabolic syndrome (Mets) is an important contributor to morbidity and mortality in cardiovascular, liver, neurological, and reproductive diseases. Short-chain fatty acid (SCFA), an organismal energy donor, has recently been demonstrated in an increasing number of studies to be an important molecule in ameliorating immuno-inflammation, an important causative factor of Mets, and to improve lipid distribution, blood glucose, and body weight levels in animal models of Mets. This study reviews recent research advances on SCFA in Mets from an immune-inflammatory perspective, including complications dominated by chronic inflammation, as well as the fact that these findings also contribute to the understanding of the specific mechanisms by which gut flora metabolites contribute to metabolic processes in humans. This review proposes an emerging role for SCFA in the inflammatory Mets, followed by the identification of major ambiguities to further understand the anti-inflammatory potential of this substance in Mets. In addition, this study proposes novel strategies to modulate SCFA for the treatment of Mets that may help to mitigate the prognosis of Mets and its complications.
Collapse
Affiliation(s)
- Wenqian Yu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Siyuan Sun
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yutong Yan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Hong Zhou
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Ziyi Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Qiang Fu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
29
|
Wang AYL, Aviña AE, Liu YY, Chang YC, Kao HK. Transcription Factor Blimp-1: A Central Regulator of Oxidative Stress and Metabolic Reprogramming in Chronic Inflammatory Diseases. Antioxidants (Basel) 2025; 14:183. [PMID: 40002370 PMCID: PMC11851694 DOI: 10.3390/antiox14020183] [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: 10/31/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
B-lymphocyte-induced maturation protein 1 (Blimp-1) is a transcription factor that, among other functions, modulates metabolism and helps to regulate antioxidant pathways, which is important in the context of chronic inflammatory diseases like diabetes, cardiovascular disease, and autoimmune disease. In immune cell function, Blimp-1 has a modulatory role in the orchestration of metabolic reprogramming and as a promoter of anti-inflammatory cytokines, including IL-10, responsible for modulating oxidative stress and immune homeostasis. Moreover, Blimp-1 also modulates key metabolic aspects, such as glycolysis and fatty acid oxidation, which regulate reactive oxygen species levels, as well as tissue protection. This review depicts Blimp-1 as an important regulator of antioxidant defenses and anti-inflammation and suggests that the protein could serve as a therapeutic target in chronic inflammatory and metabolic dysregulation conditions. The modulation of Blimp-1 in diseases such as diabetic coronary heart disease and atherosclerosis could alleviate oxidative stress, augment the protection of tissues, and improve disease outcomes. The therapeutic potential for the development of new treatments for these chronic conditions lies in the synergy between the regulation of Blimp-1 and antioxidant therapies, which are future directions that may be pursued. This review emphasizes Blimp-1's emerging importance as a novel regulator in the pathogenesis of inflammatory diseases, providing new opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Aline Yen Ling Wang
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (A.E.A.); (Y.-Y.L.)
| | - Ana Elena Aviña
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (A.E.A.); (Y.-Y.L.)
- International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yen-Yu Liu
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (A.E.A.); (Y.-Y.L.)
| | - Yun-Ching Chang
- Department of Health Industry Technology Management, Chung Shan Medical University, Taichung 402, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Huang-Kai Kao
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
30
|
Huang W, Jiang T, He J, Ruan J, Wu B, Tao R, Xu P, Wang Y, Chen R, Wang H, Yang Q, Zhang K, Jin L, Sun D, You J. Modulation of Intestinal Flora: a Novel Immunotherapeutic Approach for Enhancing Thyroid Cancer Treatment. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10471-z. [PMID: 39890752 DOI: 10.1007/s12602-025-10471-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Over the past 3 years, there has been a growing interest in clinical research regarding the potential involvement of intestinal flora in thyroid cancer (TC). This review delves into the intricate connection between intestinal flora and TC, focusing on the particular intestinal flora that is directly linked to the disease and identifying which may be able to predict potential microbial markers of TC. In order to shed light on the inflammatory pathways connected to the onset of TC, we investigated the impact of intestinal flora on immune modulation and the connection between chronic inflammation when investigating the role of intestinal flora in the pathogenesis of TC. Furthermore, the potential role of intestinal flora metabolites in the regulation of thyroid function was clarified by exploring the effects of short-chain fatty acids and lipopolysaccharide on thyroid hormone synthesis and metabolism. Based on these findings, we further explore the effects of probiotics, prebiotics, postbiotics, vitamins, and trace elements.
Collapse
Affiliation(s)
- Weiqiang Huang
- Department of General Surgery, The First People's Hospital of Jiashan, Jiashan Hospital Afliated of Jiaxing University, Jiaxing, 314100, China
| | - Tao Jiang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jiaxuan He
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jing Ruan
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Baihui Wu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Runchao Tao
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Peiye Xu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Yongpan Wang
- Department of General Surgery, The First People's Hospital of Jiashan, Jiashan Hospital Afliated of Jiaxing University, Jiaxing, 314100, China
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, SAR 999077, China
| | - Hanbing Wang
- The University of Hong Kong School of Biomedical Sciences, Hong Kong, 999077, SAR, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Kun Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Jinfeng You
- Department of Obstetrics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China.
| |
Collapse
|
31
|
Aja E, Zeng A, Gray W, Connelley K, Chaganti A, Jacobs JP. Health Effects and Therapeutic Potential of the Gut Microbe Akkermansia muciniphila. Nutrients 2025; 17:562. [PMID: 39940420 PMCID: PMC11820462 DOI: 10.3390/nu17030562] [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: 12/31/2024] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
Akkermansia muciniphila is a bacterium commonly found in the human gastrointestinal tract that has received considerable interest as a potential probiotic for the improvement of gut health and overall metabolic function. A. muciniphila is enriched in the mucus layer of the intestinal lining, where it degrades mucin and plays a significant role in gut barrier maintenance and immune regulation. A higher abundance of A. muciniphila has been observed in the gut of healthy individuals relative to those with metabolic disorders, and multiple metabolic benefits, including improved glucose management, reduced body fat, and reduced inflammation have been linked to A. muciniphila. Current research on A. muciniphila primarily relies on mouse models, with limited human interventional studies available. While these animal studies offer valuable insights into the potential roles of A. muciniphila in health and disease, further clinical investigations in humans are needed to fully understand its impact. Here, we explore the current scope of A. muciniphila research and its potential as a therapeutic agent to improve gut and metabolic health while also emphasizing the need to optimize techniques to further improve studies of this organism.
Collapse
Affiliation(s)
- Ezinne Aja
- Goodman-Luskin Microbiome Center, University of California, Los Angeles, CA 90095, USA;
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, CA 90095, USA; (A.Z.); (W.G.); (K.C.); (A.C.)
| | - Amber Zeng
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, CA 90095, USA; (A.Z.); (W.G.); (K.C.); (A.C.)
| | - Weston Gray
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, CA 90095, USA; (A.Z.); (W.G.); (K.C.); (A.C.)
| | - Kaden Connelley
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, CA 90095, USA; (A.Z.); (W.G.); (K.C.); (A.C.)
| | - Anil Chaganti
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, CA 90095, USA; (A.Z.); (W.G.); (K.C.); (A.C.)
| | - Jonathan P. Jacobs
- Goodman-Luskin Microbiome Center, University of California, Los Angeles, CA 90095, USA;
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, CA 90095, USA; (A.Z.); (W.G.); (K.C.); (A.C.)
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| |
Collapse
|
32
|
Kern L, Mastandrea I, Melekhova A, Elinav E. Mechanisms by which microbiome-derived metabolites exert their impacts on neurodegeneration. Cell Chem Biol 2025; 32:25-45. [PMID: 39326420 DOI: 10.1016/j.chembiol.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/18/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024]
Abstract
Recent developments in microbiome research suggest that the gut microbiome may remotely modulate central and peripheral neuronal processes, ranging from early brain development to age-related changes. Dysbiotic microbiome configurations have been increasingly associated with neurological disorders, such as neurodegeneration, but causal understanding of these associations remains limited. Most mechanisms explaining how the microbiome may induce such remote neuronal effects involve microbially modulated metabolites that influx into the 'sterile' host. Some metabolites are able to cross the blood-brain barrier (BBB) to reach the central nervous system, where they can impact a variety of cells and processes. Alternatively, metabolites may directly signal to peripheral nerves to act as neurotransmitters or exert modulatory functions, or impact immune responses, which, in turn, modulate neuronal function and associated disease propensity. Herein, we review the current knowledge highlighting microbiome-modulated metabolite impacts on neuronal disease, while discussing unknowns, controversies and prospects impacting this rapidly evolving research field.
Collapse
Affiliation(s)
- Lara Kern
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Ignacio Mastandrea
- Microbiome & Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anna Melekhova
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Microbiome & Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
33
|
Jones K, de Brito CB, Byndloss MX. Metabolic tug-of-war: Microbial metabolism shapes colonization resistance against enteric pathogens. Cell Chem Biol 2025; 32:46-60. [PMID: 39824157 DOI: 10.1016/j.chembiol.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 11/06/2024] [Accepted: 12/11/2024] [Indexed: 01/20/2025]
Abstract
A widely recognized benefit of gut microbiota is that it provides colonization resistance against enteric pathogens. The gut microbiota and their products can protect the host from invading microbes directly via microbe-pathogen interactions and indirectly by host-microbiota interactions, which regulate immune system function. In contrast, enteric pathogens have evolved mechanisms to utilize microbiota-derived metabolites to overcome colonization resistance and increase their pathogenic potential. This review will focus on recent studies of metabolism-mediated mechanisms of colonization resistance and virulence strategies enteric pathogens use to overcome them, along with how induction of inflammation by pathogenic bacteria changes the landscape of the gut and enables alternative metabolic pathways. We will focus on how intestinal pathogens counteract the protective effects of microbiota-derived metabolites to illustrate the growing appreciation of how metabolic factors may serve as crucial virulence determinants and overcome colonization resistance.
Collapse
Affiliation(s)
- Katerina Jones
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Camila Bernardo de Brito
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mariana Xavier Byndloss
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Howard Hughes Medical Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
34
|
Ren Y, Chen G, Hong Y, Wang Q, Lan B, Huang Z. Novel Insight into the Modulatory Effect of Traditional Chinese Medicine on Cerebral Ischemia-Reperfusion Injury by Targeting Gut Microbiota: A Review. Drug Des Devel Ther 2025; 19:185-200. [PMID: 39810832 PMCID: PMC11731027 DOI: 10.2147/dddt.s500505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025] Open
Abstract
Cerebral ischemia-reperfusion injury (CIRI) is clinically characterized by high rates of morbidity, disability, mortality, and recurrence as well as high economic burden. The clinical manifestations of CIRI are often accompanied by gastrointestinal symptoms such as intestinal bacterial dysbiosis and gastrointestinal bleeding. Gut microbiota plays an important role in the pathogenesis of CIRI, and its potential biological effects have received extensive attention. The gut microbiota not only affects intestinal barrier function but also regulates gastrointestinal immunity and host homeostasis. Traditional Chinese medicine (TCM), a multi-component and multi-targeted drug, has shown remarkable effects and few adverse reactions in the prevention and treatment of CIRI. Notably, the effect of TCM on CIRI by regulating gut microbiota and maintaining gastrointestinal homeostasis has gradually become a hot topic. This review summarizes the functional role of the gut microbiota in the development and progression of CIRI and the therapeutic effects of TCM on CIRI by improving gut microbiota dysbiosis, affecting gut microbiota metabolism, and maintaining host immunity. The active ingredients of TCM used for the treatment of CIRI in relevant studies were saponins, triterpenoids, phenolics, and alkaloids. In addition, the clinical effects of TCM used to treat CIRI were briefly discussed. This review established the clinical significance and development prospects of TCM-based CIRI treatments and provided the necessary theoretical support for the further development of TCM resources for the treatment of CIRI.
Collapse
Affiliation(s)
- Yisong Ren
- Department of Critical Care Medicine, Chengdu Pidu District Hospital of Traditional Chinese Medicine, Chengdu, Sichuan Province, 611731, People’s Republic of China
| | - Gang Chen
- Department of Critical Care Medicine, Chengdu Pidu District Hospital of Traditional Chinese Medicine, Chengdu, Sichuan Province, 611731, People’s Republic of China
| | - Ying Hong
- Department of Critical Care Medicine, Chengdu Pidu District Hospital of Traditional Chinese Medicine, Chengdu, Sichuan Province, 611731, People’s Republic of China
| | - Qianying Wang
- Department of Critical Care Medicine, Chengdu Pidu District Hospital of Traditional Chinese Medicine, Chengdu, Sichuan Province, 611731, People’s Republic of China
| | - Bo Lan
- Department of Critical Care Medicine, Chengdu Pidu District Hospital of Traditional Chinese Medicine, Chengdu, Sichuan Province, 611731, People’s Republic of China
| | - Zhaozhao Huang
- Department of Critical Care Medicine, Chengdu Pidu District Hospital of Traditional Chinese Medicine, Chengdu, Sichuan Province, 611731, People’s Republic of China
| |
Collapse
|
35
|
Yu D, Shen J, Li L, Long Q, Xie S, Zhou M, Tian Q, Cai Y. Investigating the biological significance of the TCM principle "promoting urination to regulate bowel movements" through the influence of the intestinal microbiota and their metabolites on the renal-intestinal axis. Front Cell Infect Microbiol 2025; 14:1523708. [PMID: 39867340 PMCID: PMC11757259 DOI: 10.3389/fcimb.2024.1523708] [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: 11/06/2024] [Accepted: 12/20/2024] [Indexed: 01/28/2025] Open
Abstract
Treatment methods in traditional Chinese medicine (TCM) are foundational to their theoretical, methodological, formulaic, and pharmacological systems, significantly contributing to syndrome differentiation and therapy. The principle of "promoting urination to regulate bowel movements" is a common therapeutic approach in TCM. The core concept is "promoting the dispersion and drainage of water dampness, regulating urination to relieve diarrhea," yet its scientific underpinning remains unclear. Modern medical treatment for watery diarrhea primarily focuses on electrolyte replenishment, as diuretics may lead to dehydration and other side effects. Some reports suggest that this TCM approach lacks scientific validity. Microecology, an area associated with the origins of TCM, is closely related to the development, diagnosis, and treatment of diarrhea. The renal-intestinal axis offers a molecular biological basis for examining associated pathological mechanisms, advancing therapeutic targets such as "treating the intestine to address kidney issues" and highlighting the interactions within the "renal-intestinal microbiota-liquid metabolism" framework, thus providing an endogenous mechanism to support "treating the intestine through the kidney." An increasing number of studies have shown that the intestinal microbiota and its metabolites, as unique mediators, are involved in the physiological and pathological changes of the body. Therefore, this study explores the relationship between fluid metabolism and diarrhea from the perspective of the intestinal microbiota and its metabolites, aiming to elucidate the biological mechanisms underlying the "promoting urination to regulate bowel movements" therapeutic approach and to clarify the scientific basis for treating diarrhea via the renal-intestinal axis. This research provides new insights for the study of TCM microbiology.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ying Cai
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
36
|
Zhao X, Lin T, Jiang W, Lin Y, Xiao L, Tian Y, Ma K, Zhang C, Ji F, Mahsa GC, Rui X, Li W. Lactobacillus helveticus LZ-R-5 Ameliorates DSS-Induced Colitis in Mice by Modulating Gut Microbiota and Enhancing Intestinal Barrier Function. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:464-477. [PMID: 39688942 DOI: 10.1021/acs.jafc.4c07895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Lactobacillus helveticus LZ-R-5 (R-5), a strain with high epithelial adhesion and bioactive exopolysaccharide production, was isolated from Tibetan kefir grains. This study investigated its potential to alleviate intestinal inflammation using a DSS-induced colitis model in BALB/c mice. We integrated microbial diversity and serological analyses to assess changes in gut flora and cytokines following the R-5 treatment. Pathological assessments showed that R-5 reduced crypt distortion in the proximal colon and mitigated hepatic immune challenges by enhancing gut barrier function. The increased relative expression of TGF-β1 and the downregulation of NLRP3-related inflammatory factors were conducive to preventing organ damage in the thymus and spleen of mice with colitis. Additionally, R-5 stimulated GPR43 expression and improved epithelial nutrition, promoting mucin production to prevent enterotoxin leakage. It also modulated the gut microbiota by suppressing Bacteroides and Erysipelatoclostridium, leading to a microbiota composition more akin to that of normal flora.
Collapse
Affiliation(s)
- Xiaogan Zhao
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Tao Lin
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
- Quality Standards and Testing Technology Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, Yunnan 650205, PR China
| | - Wenkai Jiang
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Yihan Lin
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjing 301617, PR China
| | - Luyao Xiao
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Yufang Tian
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Kai Ma
- Jiangsu New-Bio Biotechnology Co., Ltd, Jiangyin, Jiangsu 214400, PR China
- Jiangsu Biodep Biotechnology Co., Ltd, Jiangyin, Jiangsu 214400, PR China
| | - Changliang Zhang
- Jiangsu New-Bio Biotechnology Co., Ltd, Jiangyin, Jiangsu 214400, PR China
- Jiangsu Biodep Biotechnology Co., Ltd, Jiangyin, Jiangsu 214400, PR China
| | - Feng Ji
- Jiangsu New-Bio Biotechnology Co., Ltd, Jiangyin, Jiangsu 214400, PR China
- Jiangsu Biodep Biotechnology Co., Ltd, Jiangyin, Jiangsu 214400, PR China
| | - Ghahvechi Chaeipeima Mahsa
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Xin Rui
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Wei Li
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| |
Collapse
|
37
|
Zhang H, Zhao T, Gu J, Tang F, Zhu L. Gut microbiota and inflammasome-mediated pyroptosis: a bibliometric analysis from 2014 to 2023. Front Microbiol 2025; 15:1413490. [PMID: 39834371 PMCID: PMC11743621 DOI: 10.3389/fmicb.2024.1413490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Background The role of gut microbiota in inflammatory disease development and progression has been recognized more recently. Inflammasome-mediated pyroptosis in involved in these diseases. This complex relationship between gut microbiota and inflammasome-mediated pyroptosis provides an important field of research. Bibliometric analysis provides a comprehensive understanding of this relationship, offering valuable insights into emerging research trends. Materials and methods Leveraging data spanning from 2014 to 2023 sourced from the Web of Science Core Collection, our analysis was conducted using advanced tools such as SCImago Graphica, VOSviewer, and CiteSpace software. Visualizations were created using GraphPad Prism software. We explored the nuanced aspects of research hotspots, collaborative networks, and developing trends in this field. Results A global bibliometric analysis identified 520 relevant studies spanning 41 countries and 887 institutions. Over the past decade, publication trends have shown consistent growth, with China and the United States leading the research output. Southern Medical University and Nanjing Medical University in China emerged as leading institutions in this filed. Prominent contributors include Jia Sun, Yuan Zhang, Wei Chen, Jing Wang, and Hongtao Liu from China, alongside Eicke Latz from Germany. High-impact journals such as Frontiers in Immunology and Nature Communications have been pivotal in disseminating research in this domain. Keyword analysis highlighted a primary focus on gut microbiota, NLRP3 inflammasome, pyroptosis pathways, and inflammatory diseases, themes that persist in recent studies. Furthermore, burst keyword analysis identified "butyrate" as the sole term currently experiencing a marked increase in research interest. Conclusion Research has been deeply focused on the gut microbiota and inflammasome triggered pyroptosis in years. Over the past decade, the exploration of how gut microbiota and NLRP3 or NLRP6 inflammasome-mediated pyroptosis has been an area of interest. Future investigations in this filed may primarily revolve around understanding the correlation between butyrate and NLRP3 inflammasome induced pyroptosis in relation to conditions. However, an in-depth analysis, through studies is crucial to uncover and elucidate the complex mechanisms linking these elements.
Collapse
Affiliation(s)
- Hang Zhang
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- The Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi, China
| | - Tian Zhao
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- The Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi, China
| | - Juan Gu
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China
- Department of Pharmacy, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Fushan Tang
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- The Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi, China
| | - Lei Zhu
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- The Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi, China
| |
Collapse
|
38
|
Li T, Wang P, Zhi Z, Guo T, Zhou J, Zhang H, Cao C, Cai Y, Li Y, Zhang J. Free-caged rearing modes regulate chicken intestinal metabolism by influencing gut microbial homeostasis. Poult Sci 2025; 104:104381. [PMID: 39693966 PMCID: PMC11720617 DOI: 10.1016/j.psj.2024.104381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 12/20/2024] Open
Abstract
Free-caged rearing modes, which prioritize animal welfare, are believed to enhance the quality of animal products. The impact of rearing modes on meat quality may play a key role in the superior quality of local chicken breeds. This study analyzed the cecal contents of free-range and caged black-bone chickens at different ages using metagenomic and metabolomic sequencing. We identified 32 metabolites and 367 microbial species significantly affected by the rearing mode. Linear discriminant analysis Effect Size (LefSe) highlighted five key microorganisms, Gemmiger formicilis, Bacteria unclassified, Bacteroides sp. ET225, Massilistercora timonensis, and Bacteroidales unclassified, that showed distinct abundance patterns across all age points. Among them, Bacteroides sp. ET225 and Massilistercora timonensis were positively associated with certain phospholipids and plant-derived metabolites, while negatively correlated with others like demissidine and acylcarnitine. Functional analysis revealed that rearing modes impact gut metabolites involved in gut metabolism as well as broader processes such as signal transduction, protein digestion, and autophagy. This study offers new insights into how rearing modes influence gut microbiota and metabolites, shedding light on the study of rearing mode-mediated muscle development and fat deposition.
Collapse
Affiliation(s)
- Tao Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Peng Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Zhuo Zhi
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Tong Guo
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Jie Zhou
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Huiya Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Chang Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yingjie Cai
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yuxiao Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Jianqin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
39
|
Lai Y, Huang X, Sun H, Hui Q, Hu S. Research Progress in the Relationship between Intestinal Flora and Diabetes Mellitus. Endocr Metab Immune Disord Drug Targets 2025; 25:281-290. [PMID: 38956918 DOI: 10.2174/0118715303308965240624054156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024]
Abstract
Diabetes mellitus is a common chronic metabolic disease characterized by a high incidence and disability rate. Intestinal flora refers to the microbial community that lives in the intestines and plays a crucial role in maintaining intestinal health and the human immune system. In recent years, an increasing body of research has revealed a close relationship between intestinal flora and diabetes. The pathophysiological mechanisms between them have also been constantly uncovered, and the regulation of intestinal flora has shown promising efficacy in the adjuvant treatment of diabetes. This study mainly summarized the characteristics and mechanisms of intestinal flora in patients with diabetes in recent years, as well as the methods of regulating intestinal flora to prevent and treat diabetes, and prospected the future research direction. This will offer a theoretical basis for the clinical adjuvant treatment of diabetes with intestinal flora and the development of new drugs.
Collapse
Affiliation(s)
- Yingji Lai
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xianfeng Huang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongwei Sun
- Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qi Hui
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shanshan Hu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
40
|
Sullivan JP, Jones MK. The Multifaceted Impact of Bioactive Lipids on Gut Health and Disease. Int J Mol Sci 2024; 25:13638. [PMID: 39769399 PMCID: PMC11728145 DOI: 10.3390/ijms252413638] [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: 12/02/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Bioactive lipids have a multifaceted role in health and disease and are recognized to play an important part in gut immunity and disease conditions such as inflammatory bowel disease and colon cancer. Advancements in lipidomics, enabled by mass spectrometry and chromatographic techniques, have enhanced our understanding of lipid diversity and functionality. Bioactive lipids, including short-chain fatty acids, saturated fatty acids, omega-3 fatty acids, and sphingolipids, exhibit diverse effects on inflammation and immune regulation. Short-chain fatty acids like butyrate demonstrate anti-inflammatory properties, enhancing regulatory T cell function, gut barrier integrity, and epigenetic regulation, making them promising therapeutic targets for inflammatory bowel disease and colon cancer. Conversely, saturated fatty acids promote inflammation by disrupting gut homeostasis, triggering oxidative stress, and impairing immune regulation. Omega-3 lipids counteract these effects, reducing inflammation and supporting immune balance. Sphingolipids exhibit complex roles, modulating immune cell trafficking and inflammation. They can exert protective effects or exacerbate colitis depending on their source and context. Additionally, eicosanoids can also prevent pathology through prostaglandin defense against damage to epithelial barriers. This review underscores the importance of dietary lipids in shaping gut health and immunity and also highlights the potential use of lipids as therapeutic strategies for managing inflammatory conditions and cancer.
Collapse
Affiliation(s)
| | - Melissa K. Jones
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL 32611, USA;
| |
Collapse
|
41
|
Kozai H, Watanabe C, Kosaka R, Aoki T, Hamada H, Kawashima M, Kono T, Akagi K, Kremenik MJ, Yano H, Tanaka M, Oyanagi E. Nopalea cochenillifera Regulates the Immune Response and Gut Microbiota in Mice. Nutrients 2024; 16:4376. [PMID: 39770998 PMCID: PMC11677944 DOI: 10.3390/nu16244376] [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: 10/23/2024] [Revised: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Cactus contains dietary fiber and minerals and is expected to have preventive effects against diabetes, arteriosclerosis, and other diseases. Additionally, cactus intake induces the production of short-chain fatty acids derived from the gut microbiota, which might influence immune functions. In this study, we examined the effects of a cactus (Nopalea cochenillifera: NC)-supplemented diet on lipopolysaccharide (LPS)-induced immune responses and intestinal barrier function. METHODS Male C3H/HeN mice were randomly divided into three groups-no fiber (NF), cellulose-containing fiber (Cellu), and cactus-added (NC) diets-for 6 weeks. The TNF-α and IL-10 responses to LPS, antibody titers, and intestinal barrier function, as well as the fecal microbiota, were analyzed. RESULTS The plasma TNF-α but not the IL-10 concentrations were significantly higher in the NC group than in the NF and Cellu groups. Furthermore, the plasma IgG antibody titers were significantly higher in the NC group than in the other groups. The NC group showed higher mucin content and IgA antibody titers in their feces compared with the Cellu group. The succinate and lactate contents, which induce a reduction in TNF-α secretion by macrophages, in the cecum of the NC group were significantly lower than those in the Cellu and NF groups. In contrast, the butyrate content was significantly higher in the cecum of the NC group compared to that of the Cellu group, with a significantly higher relative abundance of butyrate-producing bacteria. CONCLUSIONS Taken together, we found that cactus intake regulates innate and adaptive immune function via the gut microbiota in mice. Therefore, cactus supplementation might serve as a strategy to develop novel functional foods with dietary fiber.
Collapse
Affiliation(s)
- Hana Kozai
- Graduate School of Bioscience and Biotechnology, Chubu University, Kasugai 487-8501, Japan; (H.K.); (R.K.); (M.T.)
| | - Chihiro Watanabe
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki 701-0193, Japan; (C.W.); (H.H.); (M.K.); (T.K.); (K.A.); (H.Y.)
| | - Rina Kosaka
- Graduate School of Bioscience and Biotechnology, Chubu University, Kasugai 487-8501, Japan; (H.K.); (R.K.); (M.T.)
| | - Takafumi Aoki
- Department of Clinical Nutrition, Kawasaki University of Medical Welfare, Kurashiki 701-0193, Japan;
| | - Hiroki Hamada
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki 701-0193, Japan; (C.W.); (H.H.); (M.K.); (T.K.); (K.A.); (H.Y.)
| | - Masato Kawashima
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki 701-0193, Japan; (C.W.); (H.H.); (M.K.); (T.K.); (K.A.); (H.Y.)
| | - Takumi Kono
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki 701-0193, Japan; (C.W.); (H.H.); (M.K.); (T.K.); (K.A.); (H.Y.)
| | - Kosuke Akagi
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki 701-0193, Japan; (C.W.); (H.H.); (M.K.); (T.K.); (K.A.); (H.Y.)
| | - Michael J. Kremenik
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki 701-0193, Japan; (C.W.); (H.H.); (M.K.); (T.K.); (K.A.); (H.Y.)
| | - Hiromi Yano
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki 701-0193, Japan; (C.W.); (H.H.); (M.K.); (T.K.); (K.A.); (H.Y.)
| | - Mamoru Tanaka
- Graduate School of Bioscience and Biotechnology, Chubu University, Kasugai 487-8501, Japan; (H.K.); (R.K.); (M.T.)
| | - Eri Oyanagi
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki 701-0193, Japan; (C.W.); (H.H.); (M.K.); (T.K.); (K.A.); (H.Y.)
| |
Collapse
|
42
|
Wang S, Yin J, Liu Z, Liu X, Tian G, Xin X, Qin Y, Feng X. Metabolic disorders, inter-organ crosstalk, and inflammation in the progression of metabolic dysfunction-associated steatotic liver disease. Life Sci 2024; 359:123211. [PMID: 39491769 DOI: 10.1016/j.lfs.2024.123211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/20/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a global health concern, affecting over 30 % of adults. It is a principal driver in the development of cirrhosis and hepatocellular carcinoma. The complex pathogenesis of MASLD involves an excessive accumulation of lipids, subsequently disrupting lipid metabolism and prompting inflammation within the liver. This review synthesizes the recent research progress in understanding the mechanisms contributing to MASLD progression, with particular emphasis on metabolic disorders and interorgan crosstalk. We highlight the molecular mechanisms linked to these factors and explore their potential as novel targets for pharmacological intervention. The insights gleaned from this article have important implications for both the prevention and therapeutic management of MASLD.
Collapse
Affiliation(s)
- Shendong Wang
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Zhaojun Liu
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Xin Liu
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Ge Tian
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, China
| | - Xijian Xin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Yiming Qin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China.
| |
Collapse
|
43
|
Tian S, Goand UK, Paudel D, Le GV, Tiwari AK, Prabhu KS, Singh V. Processed Dietary Fiber Partially Hydrolyzed Guar Gum Increases Susceptibility to Colitis and Colon Tumorigenesis in Mice. RESEARCH SQUARE 2024:rs.3.rs-5522559. [PMID: 39711544 PMCID: PMC11661293 DOI: 10.21203/rs.3.rs-5522559/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The vital role of naturally occurring dietary fibers (DFs) in maintaining intestinal health has fueled the incorporation of isolated DFs into processed foods. A select group of soluble DFs, such as partially hydrolyzed guar gum (Phgg), are being promoted as dietary supplements to meet recommended DF intake. However, the potential effects of regular consumption of these processed DFs on gastrointestinal health remain largely unknown. The present study assessed the impact of Phgg on the development of intestinal inflammation and colitis-associated colon carcinogenesis (CAC). Wild-type C57BL/6 mice were fed isocaloric diets containing either 7.5% Phgg and 2.5% cellulose (Phgg group) or 10% cellulose (control) for four weeks. To induce colitis, a subgroup of mice from each group was switched to 1.4% dextran sulfate sodium (DSS) in drinking water for seven days. CAC was induced in another subgroup through a single dose of azoxymethane (AOM, 7.5 mg/kg i.p.) followed by three DSS/water cycles. To our surprise, Phgg feeding exacerbated DSS-induced colitis, as evidenced by body weight loss, disrupted colonic crypt architecture, and increased pro-inflammatory markers accompanied by a decrease in anti-inflammatory markers. Additionally, Phgg feeding led to increased colonic expression of genes promoting cell proliferation. Accordingly, extensive colon tumorigenesis was observed in Phgg-fed mice in the AOM/DSS model, whereas the control group exhibited no visible tumors. To investigate whether reducing Phgg has a distinct effect on colitis and CAC development, mice were fed a low-Phgg diet (2.5% Phgg). The low-Phgg group also exhibited increased colitis and tumorigenesis compared to the control, although the severity was markedly lower than in the regular Phgg (7.5%) group, suggesting a dose-dependent effect of Phgg in colitis and CAC development. Our study reveals that Phgg supplementation exacerbates colitis and promotes colon tumorigenesis, warranting further investigation into the potential gastrointestinal health risks associated with processed Phgg consumption.
Collapse
|
44
|
Tang M, Wu Y, Liang J, Yang S, Huang Z, Hu J, Yang Q, Liu F, Li S. Gut microbiota has important roles in the obstructive sleep apnea-induced inflammation and consequent neurocognitive impairment. Front Microbiol 2024; 15:1457348. [PMID: 39712898 PMCID: PMC11659646 DOI: 10.3389/fmicb.2024.1457348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
Obstructive sleep apnea (OSA) is a state of sleep disorder, characterized by repetitive episodes of apnea and chronic intermittent hypoxia. OSA has an extremely high prevalence worldwide and represents a serious challenge to public health, yet its severity is frequently underestimated. It is now well established that neurocognitive dysfunction, manifested as deficits in attention, memory, and executive functions, is a common complication observed in patients with OSA, whereas the specific pathogenesis remains poorly understood, despite the likelihood of involvement of inflammation. Here, we provide an overview of the current state of the art, demonstrating the intimacy of OSA with inflammation and cognitive impairment. Subsequently, we present the recent findings on the investigation of gut microbiota alteration in the OSA conditions, based on both patients-based clinical studies and animal models of OSA. We present an insightful discussion on the role of changes in the abundance of specific gut microbial members, including short-chain fatty acid (SCFA)-producers and/or microbes with pathogenic potential, in the pathogenesis of inflammation and further cognitive dysfunction. The transplantation of fecal microbiota from the mouse model of OSA can elicit inflammation and neurobehavioral disorders in naïve mice, thereby validating the causal relationship to inflammation and cognitive abnormality. This work calls for greater attention on OSA and the associated inflammation, which require timely and effective therapy to protect the brain from irreversible damage. This work also suggests that modification of the gut microbiota using prebiotics, probiotics or fecal microbiota transplantation may represent a potential adjuvant therapy for OSA.
Collapse
Affiliation(s)
- Mingxing Tang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Yongliang Wu
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Junyi Liang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Shuai Yang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Zuofeng Huang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Jing Hu
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Qiong Yang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Fei Liu
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Shuo Li
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| |
Collapse
|
45
|
Xu X, Gao Y, Xiao Y, Yu Y, Huang J, Su W, Li N, Xu C, Gao S, Wang X. Characteristics of the gut microbiota and the effect of Bifidobacterium in very early-onset inflammatory bowel disease patients with IL10RA mutations. Front Microbiol 2024; 15:1479779. [PMID: 39687875 PMCID: PMC11647010 DOI: 10.3389/fmicb.2024.1479779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
Very early-onset inflammatory bowel disease (VEO-IBD) is a distinct subtype of inflammatory bowel disease (IBD) characterized by onset before the age of 6 years, and patients often exhibit more severe clinical features. Interleukin 10 receptor alpha (IL10RA) is a hotspot mutation in the Chinese population and is associated with a poor prognosis closely linked to the onset of IBD. However, limited knowledge exists regarding how the IL10RA mutation influences the host microbiota and its role in disease development. We employed 16S rRNA sequencing to conduct a comprehensive assessment of microbial changes in different types of IBD, employed database to thoroughly examine the influence of Bifidobacterium in IBD and to demonstrate a potential positive effect exerted by Bifidobacterium breve M16V (M16V) through a mouse model. The study demonstrated a significant reduction in the abundance and diversity of the gut microbiota among children with IL10RA mutations compared to those with late-onset pediatric IBD and nonmutated VEO-IBD. Furthermore, the analysis identified genera capable of distinguishing between various types of IBD, with the genus Bifidobacterium emerging as a potential standalone diagnostic indicator and Bifidobacterium may also be involved in related pathways that influence the progression of IBD, such as the biosynthesis of amino acids and inflammation-related pathways. This study corroborated the efficacy of Bifidobacterium in alleviating intestinal inflammation. The impact of IL10RA mutations on VEO-IBD may be mediated by alterations in microbes. M16V demonstrates efficacy in alleviating colitis and holds promise as a novel microbial therapy.
Collapse
Affiliation(s)
- Xu Xu
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanqi Gao
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Xiao
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Yu
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiebin Huang
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Su
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Na Li
- Department of Tropical Diseases, The Second Affiliated Hospital of Hainan Medical University, Key Laboratory of Tropical Translational Medicine of Ministry of Education, NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Chundi Xu
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shenshen Gao
- Clinical Research and Development Center of Shanghai Municipal Hospitals, Shanghai Shenkang Hospital Development Center, Shanghai, China
| | - Xinqiong Wang
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
46
|
Bauset C, Carda-Diéguez M, Cejudo-Garcés A, Buetas E, Seco-Cervera M, Macias-Ceja DC, Navarro-Vicente F, Esplugues JV, Calatayud S, Mira Á, Ortiz-Masiá D, Barrachina MD, Cosín-Roger J. A disturbed metabolite-GPCR axis is associated with microbial dysbiosis in IBD patients: Potential role of GPR109A in macrophages. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167489. [PMID: 39233260 DOI: 10.1016/j.bbadis.2024.167489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/20/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
Inflammatory Bowel Disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract characterized by disrupted immune function. Indeed, gut microbiota dysbiosis and metabolomic profile alterations, are hallmarks of IBD. In this scenario, metabolite-sensing G-protein coupled receptors (GPCRs), involved in several biological processes, have emerged as pivotal players in the pathophysiology of IBD. The aim of this study was to characterize the axis microbiota-metabolite-GPCR in intestinal surgical resections from IBD patients. Results showed that UC patients had a lower microbiota richness and bacterial load, with a higher proportion of the genus Cellulosimicrobium and a reduced proportion of Escherichia, whereas CD patients showed a decreased abundance of Enterococcus. Furthermore, metabolomic analysis revealed alterations in carboxylic acids, fatty acids, and amino acids in UC and CD samples. These patients also exhibited upregulated expression of most metabolite-sensing GPCRs analysed, which positively correlated with pro-inflammatory and pro-fibrotic markers. The role of GPR109A was studied in depth and increased expression of this receptor was detected in epithelial cells and cells from lamina propria, including CD68+ macrophages, in IBD patients. The treatment with β-hydroxybutyrate increased gene expression of GPR109A, CD86, IL1B and NOS2 in U937-derived macrophages. Besides, when GPR109A was transiently silenced, the mRNA expression and secretion of IL-1β, IL-6 and TNF-α were impaired in M1 macrophages. Finally, the secretome from siGPR109A M1 macrophages reduced the gene and protein expression of COL1A1 and COL3A1 in intestinal fibroblasts. A better understanding of metabolite-sensing GPCRs, such as GPR109A, could establish their potential as therapeutic targets for managing IBD.
Collapse
Affiliation(s)
- Cristina Bauset
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | | | - Andrea Cejudo-Garcés
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Elena Buetas
- Genomics & Health Department, FISABIO Foundation, Valencia, Spain
| | | | | | | | - Juan Vicente Esplugues
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), Madrid, Spain
| | - Sara Calatayud
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), Madrid, Spain
| | - Álex Mira
- Genomics & Health Department, FISABIO Foundation, Valencia, Spain; CIBER Center for Epidemiology and Public Health, Madrid, Spain
| | - Dolores Ortiz-Masiá
- CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), Madrid, Spain; Departamento de Medicina, Facultad de Medicina, Universidad de Valencia, Valencia, Spain.
| | - María Dolores Barrachina
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), Madrid, Spain.
| | - Jesús Cosín-Roger
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), Madrid, Spain
| |
Collapse
|
47
|
Ferrer M, Buey B, Grasa L, Mesonero JE, Latorre E. Protective role of short-chain fatty acids on intestinal oxidative stress induced by TNF-α. Cell Stress Chaperones 2024; 29:769-776. [PMID: 39547594 DOI: 10.1016/j.cstres.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] [Received: 09/09/2024] [Revised: 10/31/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024] Open
Abstract
Inflammatory bowel diseases (IBDs) are driven by an exaggerated inflammatory response, which leads to a marked increase in oxidative stress. This, in turn, exacerbates the inflammatory process and causes significant cellular and tissue damage. Intestinal dysbiosis, a common observation in IBD patients, alters the production of bacterial metabolites, including short-chain fatty acids (SCFAs), which are key by-products of dietary fiber fermentation. While the role of SCFAs in intestinal physiology is still being elucidated, this study aimed to investigate their effects on intestinal oxidative stress, particularly under inflammatory conditions induced by the proinflammatory mediator tumor necrosis factor alpha (TNF-α). The Caco-2/TC7 cell line was employed as an in vitro model of the intestinal epithelium, and the cells were treated with a range of SCFAs, including acetate, propionate, and butyrate. The levels of protein and lipid oxidation were quantified, as well as the activity of antioxidant enzymes. Our findings demonstrate that microbiota-derived SCFAs can effectively mitigate TNF-α-induced oxidative stress by modulating antioxidant enzyme activity. The proinflammatory mediator TNF-α induces lipid peroxidation by inhibiting catalase and glutathione peroxidase activities. SCFAs are able to upregulate antioxidant enzyme activity to restore lipid oxidative levels. These results underscore the critical role of the gut microbiota in maintaining intestinal homeostasis and highlight the therapeutic potential of SCFAs in managing oxidative stress-related pathologies.
Collapse
Affiliation(s)
- Miguel Ferrer
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
| | - Berta Buey
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - Laura Grasa
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón-IA2, Universidad de Zaragoza-CITA, Zaragoza, Spain
| | - Jose Emilio Mesonero
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón-IA2, Universidad de Zaragoza-CITA, Zaragoza, Spain
| | - Eva Latorre
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain; Instituto Agroalimentario de Aragón-IA2, Universidad de Zaragoza-CITA, Zaragoza, Spain.
| |
Collapse
|
48
|
Thapa R, Magar AT, Shrestha J, Panth N, Idrees S, Sadaf T, Bashyal S, Elwakil BH, Sugandhi VV, Rojekar S, Nikhate R, Gupta G, Singh SK, Dua K, Hansbro PM, Paudel KR. Influence of gut and lung dysbiosis on lung cancer progression and their modulation as promising therapeutic targets: a comprehensive review. MedComm (Beijing) 2024; 5:e70018. [PMID: 39584048 PMCID: PMC11586092 DOI: 10.1002/mco2.70018] [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: 07/06/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
Lung cancer (LC) continues to pose the highest mortality and exhibits a common prevalence among all types of cancer. The genetic interaction between human eukaryotes and microbial cells plays a vital role in orchestrating every physiological activity of the host. The dynamic crosstalk between gut and lung microbiomes and the gut-lung axis communication network has been widely accepted as promising factors influencing LC progression. The advent of the 16s rDNA sequencing technique has opened new horizons for elucidating the lung microbiome and its potential pathophysiological role in LC and other infectious lung diseases using a molecular approach. Numerous studies have reported the direct involvement of the host microbiome in lung tumorigenesis processes and their impact on current treatment strategies such as radiotherapy, chemotherapy, or immunotherapy. The genetic and metabolomic cross-interaction, microbiome-dependent host immune modulation, and the close association between microbiota composition and treatment outcomes strongly suggest that designing microbiome-based treatment strategies and investigating new molecules targeting the common holobiome could offer potential alternatives to develop effective therapeutic principles for LC treatment. This review aims to highlight the interaction between the host and microbiome in LC progression and the possibility of manipulating altered microbiome ecology as therapeutic targets.
Collapse
Affiliation(s)
- Rajan Thapa
- Department of Pharmacy, Universal college of medical sciencesTribhuvan UniversityBhairahawaRupendehiNepal
| | - Anjana Thapa Magar
- Department of MedicineKathmandu Medical College Teaching Hospital, SinamangalKathmanduNepal
| | - Jesus Shrestha
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Nisha Panth
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Sobia Idrees
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Tayyaba Sadaf
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Saroj Bashyal
- Department of Pharmacy, Manmohan Memorial Institute of Health SciencesTribhuvan University, SoalteemodeKathmanduNepal
| | - Bassma H. Elwakil
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences TechnologyPharos University in AlexandriaAlexandriaEgypt
| | - Vrashabh V. Sugandhi
- Department of pharmaceutical sciences, College of Pharmacy & Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Satish Rojekar
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ram Nikhate
- Department of PharmaceuticsDattakala Shikshan Sanstha, Dattakala college of pharmacy (Affiliated to Savitribai Phule Pune universityPuneMaharashtraIndia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiIndia
- Centre of Medical and Bio‐allied Health Sciences ResearchAjman UniversityAjmanUAE
| | - Sachin Kumar Singh
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraIndia
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Philip M Hansbro
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| |
Collapse
|
49
|
Yang C, Li X, Hu M, Li T, Jiang L, Zhang Y. Gut Microbiota as Predictive Biomarker for Chronic Heart Failure in Patients with Different Nutritional Risk. J Cardiovasc Transl Res 2024; 17:1240-1257. [PMID: 38913293 DOI: 10.1007/s12265-024-10529-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 05/21/2024] [Indexed: 06/25/2024]
Abstract
To examine the relationship between gut microbiota and disease development in chronic heart failure patients with different nutritional risk. The study analyzed stool samples from 62 CHF patients and 21 healthy peoples using 16S rRNA gene sequencing. CHF patients were separated into risk (n = 30) and non-risk group (n = 32) based on NRS2002 scores. Analysis methods used were LEfSe, random forest regression model, ROC curves, BugBase, PICRUSt2, metagenomeSeq. Risk group includes 11 cases of HFrEF, 6 cases of HFpEF, and 13 cases of HFmrEF. LefSe analysis confirmed that the risk group had higher levels of Enterobacter and Escherichia-Shigella. Correlation analysis revealed a negative correlation between prealbumin and Escherichia-Shigella. The presence of Enterobacter and Escherichia-Shigella worsens intestinal inflammation in CHF patients, impacting lysine metabolism by influencing its degradation metabolic function. This interference further disrupts albumin and prealbumin synthesis, leading to malnutrition in CHF patients and ultimately worsening the disease.
Collapse
Affiliation(s)
- Chen Yang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
| | - Xiaopeng Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
| | - Miaomiao Hu
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Dian Diagnostics Group Co., Ltd., Hangzhou, 310030, Zhejiang Province, China
| | - Ting Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
| | - Li Jiang
- Department of Cardiovascular Medicine, Dongcheng branch of the First Affiliated Hospital of Anhui Medical University (Feidong County People's Hospital), 218 Jixi Road, Hefei, 230000, Anhui Province, China
| | - Yong Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China.
- Department of Cardiovascular Medicine, Dongcheng branch of the First Affiliated Hospital of Anhui Medical University (Feidong County People's Hospital), 218 Jixi Road, Hefei, 230000, Anhui Province, China.
| |
Collapse
|
50
|
Zhang Y, Fan J. Impact of gut microbiota on metabolic syndrome and its comprising traits: a two-sample mendelian randomization study. Diabetol Metab Syndr 2024; 16:279. [PMID: 39578862 PMCID: PMC11585155 DOI: 10.1186/s13098-024-01520-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND The prevalence of metabolic syndrome is on the rise globally. Understanding the etiology and discovering potential treatment target have become a priority. Observational data have linked gut microbiota with metabolic syndrome and its comprising traits. However, whether these relations underlie causal effects remains unclear. METHODS Using Inver Variance Weighted (IVW) as primary analysis method, we performed two-sample Mendelian Randomization (MR) analyses to explore the causal relationship between gut microbiota and metabolic syndrome with its comprising traits. Methods including MR-Egger regression, MR Pleiotropy RESidual Sum and Outlier (MR-PRESSO), Weighted Mode, and Weighted Median were chosen for additional MR analysis to test the robustness of MR results. Cochran's IVW Q test and leave-one-out IVW analysis tested the heterogeneity among instrumental variables (IVs). Steiger filtering was utilized to exclude all IVs with reverse causality. Genome-wide association study (GWAS) data used in this study were all from the largest respective GWAS studies available. RESULTS Out of 1172 tests, a total of 16 associations with evidence of causality were identified after sensitivity analyses, but only 3 remained after multiple testing correction. Class Melainabacteria (β = 0.02, adjusted P = 0.01) with affiliated order Gastranaerophilales (β = 0.02, adjusted P = 1.20*10- 3) and genus Eubacterium hallii (β = 0.03, adjusted P = 0.03) showed a positive effect on abdominal obesity. All effect sizes were small (abs(β) < 0.1). All causal relationships identified were unidirectional. CONCLUSIONS Given the study's limitations, we found little evidence supporting a large causal effect, i.e. absolute effect size > 0.1, of gut microbial taxa abundance on metabolic syndrome and its comprising traits. This study also suggests that previously reported associations between gut microbiota and metabolic syndrome with its comprising traits may not necessarily lead to causal relations. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Yaodong Zhang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jinhai Fan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|