1
|
Li Z, Chu T, Sun X, Zhuang S, Hou D, Zhang Z, Sun J, Liu Y, Li J, Bian Y. Polyphenols-rich Portulaca oleracea L. (purslane) alleviates ulcerative colitis through restiring the intestinal barrier, gut microbiota and metabolites. Food Chem 2025; 468:142391. [PMID: 39675274 DOI: 10.1016/j.foodchem.2024.142391] [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: 08/09/2024] [Revised: 11/03/2024] [Accepted: 12/04/2024] [Indexed: 12/17/2024]
Abstract
Ulcerative colitis (UC) is a recurrent intestinal disease caused by a complex of factors, and there are serious adverse effects and tolerance problems associated with the current long-term use of therapeutic drugs. The development of natural food sources and multi-targeted drugs for the treatment of UC is imminent. Portulaca oleracea L. (PO), as a vegetable, has been shown in studies to have an anti-UC effects. However, the relationship between the abundant active ingredients contained in Portulaca oleracea L. and the improvement of intestinal barrier, gut microbiota and metabolites is unclear. In the present study, Portulaca oleracea L. which was found to be rich in phenolic acid-based active ingredients, were effective in alleviating dextran sulfate sodium (DSS)-induced body weight loss, disease activity index (DAI) score and colon length in mice. It also decreased C-reactive protein (CRP) and myeloperoxidase (MPO) responses, reduced the permeation of fluorescein isothiocyanate (FITC)-dextran, lipopolysaccharide (LPS) and evans blue (EB), and improved histopathological scores. Meanwhile, in vitro and in vivo validation revealed the protective effects of purslane on the intestinal barrier indicators ZO-1, Occludin and Claudin-1, and inhibited the expression of inflammation-associated iNOS and NLRP3 proteins through the NF-κB signaling pathway. In addition, purslane increased the diversity of the intestinal flora, enhancing the proportion of the genera Butyricoccus, Dorea and Bifidobacterium and decreasing the percentage of Bacteroides, Turicibacter and Parabacteroides. Serum metabolomics analysis showed that the imbalance of 39 metabolites was significantly reversed after PO deployment. Enrichment analysis showed that Pentose phosphate pathway and Pyruvate metabolism pathway were the key pathways of PO against UC. Overall, purslane effectively improved the intestinal barrier disruption and intestinal inflammation by inhibiting the NF-κB signaling pathway, and adjusted the disorder of gut microbiota and metabolites to exert anti-UC effects.
Collapse
Affiliation(s)
- Zheng Li
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Tianjiao Chu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xin Sun
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Shen Zhuang
- College of Veterinary Medicine & Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Dianbo Hou
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zhaohan Zhang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jialu Sun
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Jing Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Yifei Bian
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
2
|
Huang STZ, Hu YH, Gao YC, Zhou DD, Chen MY, Wang L, Song JY, Zhou HH, Zhang W, Huang WH. Hypoglycemic Effect of Ginsenoside Compound K Mediated by N-Acetylserotonin Derived From Gut Microbiota. Phytother Res 2025. [PMID: 39757809 DOI: 10.1002/ptr.8385] [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: 06/12/2024] [Revised: 09/02/2024] [Accepted: 09/18/2024] [Indexed: 01/07/2025]
Abstract
Ginsenoside compound K (GCK) has been proved to have great hypoglycemic effect pertinent to gut microbiota. However, the improvement of high-fat-diet (HFD)-induced type 2 diabetes (T2D) as well as the mechanism of GCK mediated by gut microbiota is not well-known. This study aimed to investigate the hypoglycemic effects and mechanism of GCK on a HFD-induced diabetic mouse model. HFD-induced pseudo-germ free (GF) T2D mice model and fecal microbiota transplantation (FMT) experiments were performed to clarify the role of gut microbiota in the hypoglycemic effect of GCK. Differential metabolites were screened by untargeted metabolomics analysis and their functions were verified by suppling to T2D mice. The level of glucagon-like peptide-1 (GLP-1) in plasma was detected by ELISA analysis to explore the potential hypoglycemic mechanism of GCK. The results showed GCK alleviated metabolic disorders and altered gut microbiota in HFD-induced diabetic mice, which was transmitted to pseudo-GF diabetic mice via FMT experiment to reproduce the hypoglycemic effect. Non-targeted metabolites analysis on cecal content samples indicated that N-acetylserotonin (NAS) was markedly increased after GCK treatment. Moreover, gavage with NAS improved insulin sensitivity and increased the secretion of GLP-1 in HFD mice. Our study showed that GCK had hypoglycemic effect through modifying gut microbiota profiling.
Collapse
Affiliation(s)
- Su-Tian-Zi Huang
- Engineering Research Center of Applied Technology of Pharmacogenomics (Ministry of Education, China), Hunan Key Laboratory of Pharmacomicrobiomics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yu-Hui Hu
- Engineering Research Center of Applied Technology of Pharmacogenomics (Ministry of Education, China), Hunan Key Laboratory of Pharmacomicrobiomics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Yong-Chao Gao
- Engineering Research Center of Applied Technology of Pharmacogenomics (Ministry of Education, China), Hunan Key Laboratory of Pharmacomicrobiomics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Ding-Ding Zhou
- Engineering Research Center of Applied Technology of Pharmacogenomics (Ministry of Education, China), Hunan Key Laboratory of Pharmacomicrobiomics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Man-Yun Chen
- Engineering Research Center of Applied Technology of Pharmacogenomics (Ministry of Education, China), Hunan Key Laboratory of Pharmacomicrobiomics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Wang
- Engineering Research Center of Applied Technology of Pharmacogenomics (Ministry of Education, China), Hunan Key Laboratory of Pharmacomicrobiomics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Jing-Yuan Song
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hong-Hao Zhou
- Engineering Research Center of Applied Technology of Pharmacogenomics (Ministry of Education, China), Hunan Key Laboratory of Pharmacomicrobiomics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Zhang
- Engineering Research Center of Applied Technology of Pharmacogenomics (Ministry of Education, China), Hunan Key Laboratory of Pharmacomicrobiomics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Wei-Hua Huang
- Engineering Research Center of Applied Technology of Pharmacogenomics (Ministry of Education, China), Hunan Key Laboratory of Pharmacomicrobiomics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
3
|
Li W, Mu J, Ni S, Pei W, Wan L, Wu X, Zhu J, Zhang Z, Li L. Pentachlorophenol Exposure Delays the Recovery of Colitis in Association With Altered Gut Microbiota and Purine Metabolism. ENVIRONMENTAL TOXICOLOGY 2025; 40:101-110. [PMID: 39285788 DOI: 10.1002/tox.24420] [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: 01/24/2024] [Revised: 07/08/2024] [Accepted: 08/30/2024] [Indexed: 12/11/2024]
Abstract
Pentachlorophenol (PCP) was used widely as preservative and biocide and has been banned due to with various harmful effects, such as carcinogenicity and teratogenicity. However, the effects of PCP on colitis induced by dextrose sodium sulfate (DSS) remain largely unknown. Serum metabolomics and gut microbiota were investigated to elucidate the underlying mechanisms. Exposure to 20 μg/L PCP aggravated DSS-induced body weight loss, colon shortening, severe histological injuries, and upregulation of TNFα, iNOS, IL-1β, and IL-6. Serum metabolomics showed that both DSS and PCP could significantly disrupted tryptophan metabolism in normal mice. Interestingly, PCP exposure intensified the disturbance in purine metabolism but not tryptophan metabolism caused by DSS. Quantitative analysis of tryptophan and metabolites further confirmed that PCP exposure significantly increased the serum contents of serotonin, adenine, guanine, guanosine, inosine monophosphate (IMP), inosine, and hypoxanthine in DSS-treated mice. The overall gut microbial community was significantly modified by PCP and DSS treatment alone. Rikenellaceae_RC9_Gut_group, Colidextribacter, and Desulfovibrio were more abundant in colitis mice following PCP exposure. Further integrative analysis of differential bacteria and purine metabolites highlighted a significant correlation between Desulfovibrio and several purine metabolites, including guanine, guanosine, hypoxanthine, IMP, and inosine. Adenosine ribonucleotides de novo biosynthesis, inosine-5'-phosphate biosynthesis I, and urate biosynthesis/inosine 5'-phosphate degradation pathways were depleted in colitis mice upon PCP treatment. Taken together, PCP exposure delayed the recovery of colitis induced by DSS in association with altered gut microbiota and serum metabolites, which were enriched in tryptophan and purine metabolism.
Collapse
Affiliation(s)
- Wenzheng Li
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jing Mu
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Shanhong Ni
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Wenlong Pei
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Li Wan
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Xin Wu
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jun Zhu
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Zhan Zhang
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Lei Li
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
4
|
He K, An F, Zhang H, Yan D, Li T, Wu J, Wu R. Akkermansia muciniphila: A Potential Target for the Prevention of Diabetes. Foods 2024; 14:23. [PMID: 39796314 PMCID: PMC11720440 DOI: 10.3390/foods14010023] [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: 12/11/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Akkermansia muciniphila, a Gram-negative anaerobic bacterium colonizing the intestinal mucus layer, is regarded as a promising "next-generation probiotic". There is mounting evidence that diabetes and its complications are associated with disorders of A. muciniphila abundance. Thus, A. muciniphil and its components, including the outer membrane protein Amuc_1100, A. muciniphila-derived extracellular vesicles (AmEVs), and the secreted proteins P9 and Amuc_1409, are systematically summarized with respect to mechanisms of action in diabetes mellitus. Diabetes treatments that rely on altering changes in A. muciniphila abundance are also reviewed, including the identification of A. muciniphila active ingredients, and dietary and pharmacological interventions for A. mucinihila abundance. The potential and challenges of using A. muciniphila are also highlighted, and it is anticipated that this work will serve as a reference for more in-depth studies on A. muciniphila and diabetes development, as well as the creation of new therapeutic targets by colleagues domestically and internationally.
Collapse
Affiliation(s)
- Kairu He
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China; (K.H.)
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, China
| | - Feiyu An
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China; (K.H.)
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, China
| | - Henan Zhang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China; (K.H.)
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, China
| | - Danli Yan
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China; (K.H.)
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, China
| | - Tong Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China; (K.H.)
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, China
| | - Junrui Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China; (K.H.)
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, China
| | - Rina Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China; (K.H.)
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, China
| |
Collapse
|
5
|
Ren P, Wei B, Qin W, Tang Q, Wang Y, Xue C. Impact of astaxanthin on the capacity of gut microbiota to produce tryptophan catabolites. Food Funct 2024. [PMID: 39688008 DOI: 10.1039/d4fo04890c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
This study utilized in vitro colonic fermentation to examine the impact of astaxanthin on the microbial catabolism of tryptophan. Astaxanthin significantly altered the gut microbiota and raised the tryptophan catabolism metabolite levels in an in vitro human colonic fermentation system. To eliminate the influence of substrate availability, we conducted in vitro colonic fermentation of the gut microbiota of astaxanthin-domesticated mice. We observed that the capacity of astaxanthin-domesticated gut microbiota to catalyze the conversion of tryptophan to indole and derivatives was considerably augmented. Astaxanthin significantly increased the relative abundance of Akkermansia, Ruminococcus, Bacteroides and Lactobacillus and elevated the levels of indole-3-lactic acid and indole-3-propionic acid. These results demonstrated that astaxanthin regulates tryptophan metabolism by modifying gut microbiota and increasing the levels of indole metabolites, such as indole-3-lactic acid and indole-3-propionic acid. This study provides insights into the physiological activity of astaxanthin and sheds light on the potential for enhancing tryptophan metabolism through dietary manipulation of the gut microbiota.
Collapse
Affiliation(s)
- Pengfei Ren
- State Key Laboratory of Marine Food Processing &Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| | - Biqian Wei
- State Key Laboratory of Marine Food Processing &Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| | - Wanting Qin
- State Key Laboratory of Marine Food Processing &Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| | - Qingjuan Tang
- State Key Laboratory of Marine Food Processing &Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| | - Yuming Wang
- State Key Laboratory of Marine Food Processing &Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| | - Changhu Xue
- State Key Laboratory of Marine Food Processing &Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| |
Collapse
|
6
|
Han Y, He J, Li M, Peng Y, Jiang H, Zhao J, Li Y, Deng F. Unlocking the Potential of Metagenomics with the PacBio High-Fidelity Sequencing Technology. Microorganisms 2024; 12:2482. [PMID: 39770685 PMCID: PMC11728442 DOI: 10.3390/microorganisms12122482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Traditional methods for studying microbial communities have been limited due to difficulties in culturing and sequencing all microbial species. Recent advances in third-generation sequencing technologies, particularly PacBio's high-fidelity (HiFi) sequencing, have significantly advanced metagenomics by providing accurate long-read sequences. This review explores the role of HiFi sequencing in overcoming the limitations of previous sequencing methods, including high error rates and fragmented assemblies. We discuss the benefits and applications of HiFi sequencing across various environments, such as the human gut and soil, which provides broader context for further exploration. Key studies are discussed to highlight HiFi sequencing's ability to recover complete and coherent microbial genomes from complex microbiomes, showcasing its superior accuracy and continuity compared to other sequencing technologies. Additionally, we explore the potential applications of HiFi sequencing in quantitative microbial analysis, as well as the detection of single nucleotide variations (SNVs) and structural variations (SVs). PacBio HiFi sequencing is establishing a new benchmark in metagenomics, with the potential to significantly enhance our understanding of microbial ecology and drive forward advancements in both environmental and clinical applications.
Collapse
Affiliation(s)
- Yanhua Han
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Life Science and Engineering, Foshan University, Foshan 528225, China; (Y.H.); (J.H.); (M.L.); (H.J.); (Y.L.)
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Jinling He
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Life Science and Engineering, Foshan University, Foshan 528225, China; (Y.H.); (J.H.); (M.L.); (H.J.); (Y.L.)
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Minghui Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Life Science and Engineering, Foshan University, Foshan 528225, China; (Y.H.); (J.H.); (M.L.); (H.J.); (Y.L.)
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Yunjuan Peng
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.P.); (J.Z.)
| | - Hui Jiang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Life Science and Engineering, Foshan University, Foshan 528225, China; (Y.H.); (J.H.); (M.L.); (H.J.); (Y.L.)
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Jiangchao Zhao
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.P.); (J.Z.)
| | - Ying Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Life Science and Engineering, Foshan University, Foshan 528225, China; (Y.H.); (J.H.); (M.L.); (H.J.); (Y.L.)
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Feilong Deng
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Life Science and Engineering, Foshan University, Foshan 528225, China; (Y.H.); (J.H.); (M.L.); (H.J.); (Y.L.)
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| |
Collapse
|
7
|
Feng L, Guo Z, Yao W, Mu G, Zhu X. Metagenomics and Untargeted Metabolomics Analysis Revealed the Probiotic and Postbiotic Derived from Lactiplantibacillus plantarum DPUL F232 Alleviate Whey Protein-Induced Food Allergy by Reshaping Gut Microbiota and Regulating Key Metabolites. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25436-25448. [PMID: 39485064 DOI: 10.1021/acs.jafc.4c08203] [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: 11/03/2024]
Abstract
Postbiotics have emerged as a promising alternative to probiotics. However, it remains unclear whether postbiotics can exert regulatory effects on intestinal flora and metabolism as probiotics. Thus, we investigated the effects of probiotic and postbiotic in rats with whey protein-induced food allergy, which demonstrated that postbiotic intervention effectively alleviated allergy symptoms, reduced serum immunoglobulin E (IgE) and mast cell protease-1 (mMCP-1) levels, and regulated the type helper 1 cell/2 cell (Th1/Th2) balance in both serum and spleen. Metagenomic analysis revealed that postbiotics induced more significant changes in intestinal flora. Untargeted metabolomics analysis showed that both probiotics and postbiotics significantly up-regulated various differential metabolites, which were negatively correlated with immune indices, including malvidin-3-glucoside, 3,4-dihydroxymandelic acid, nicotinamide, triterpenoids, pirbuterol, and 4-hydroxybenzoic acid. This study confirms that postbiotics can alleviate food allergies and regulate intestinal flora and metabolites, which provides a valuable reference for the use of postbiotics in mitigating allergic diseases through gut microbiota and metabolite modulation.
Collapse
Affiliation(s)
- Lu Feng
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, P. R. China
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Dalian, Liaoning 116034, P. R. China
| | - Zihao Guo
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, P. R. China
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Dalian, Liaoning 116034, P. R. China
| | - Wenpu Yao
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, P. R. China
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Dalian, Liaoning 116034, P. R. China
| | - Guangqing Mu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, P. R. China
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Dalian, Liaoning 116034, P. R. China
| | - Xuemei Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, P. R. China
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Dalian, Liaoning 116034, P. R. China
| |
Collapse
|
8
|
De la Rosa González A, Guerra-Ojeda S, Camacho-Villa MA, Valls A, Alegre E, Quintero-Bernal R, Martorell P, Chenoll E, Serna-García M, Mauricio MD, Serna E. Effect of Probiotics on Gastrointestinal Health Through the Aryl Hydrocarbon Receptor Pathway: A Systematic Review. Foods 2024; 13:3479. [PMID: 39517263 PMCID: PMC11545787 DOI: 10.3390/foods13213479] [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: 10/02/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Probiotics are living microorganisms recognized for conferring health benefits on the host by modulating the gut microbiota. They interact with various signaling pathways, including the aryl hydrocarbon receptor (AhR), which plays a crucial role in maintaining intestinal homeostasis and immune function. The activation of AhR by probiotics has been associated with benefits such as improved intestinal barrier function, reduced inflammation, and modulation of immune responses. This systematic review aims to summarize current knowledge on the signaling of AhR, mediated by probiotics in physiological conditions and gastrointestinal pathologies. We conducted a comprehensive search across databases, including PubMed and Embase, up until July 2024. Out of 163 studies screened, 18 met the inclusion criteria. Our findings revealed in healthy populations that probiotic consumption increases the production of AhR ligands promoting intestinal immune tolerance. Furthermore, in populations with gastrointestinal pathologies, probiotics ameliorated symptoms through AhR activation by Trp metabolites, leading to the upregulation of the anti-inflammatory response.
Collapse
Affiliation(s)
| | - Sol Guerra-Ojeda
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (S.G.-O.); (A.V.); (E.A.); (E.S.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - María Alejandra Camacho-Villa
- HARPEER Research Group, Yumbo 760001, Colombia; (A.D.l.R.G.); (M.A.C.-V.); (R.Q.-B.)
- Pain Study Group (GED), Physical Therapy School, Universidad Industrial de Santander, Bucaramanga Santander 680002, Colombia
| | - Alicia Valls
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (S.G.-O.); (A.V.); (E.A.); (E.S.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- MODULAhR Group, Universitat de Valencia, 46010 Valencia, Spain
| | - Eva Alegre
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (S.G.-O.); (A.V.); (E.A.); (E.S.)
| | | | - Patricia Martorell
- Archer Daniels Midland (ADM), Nutrition, Health & Wellness, Biopolis S. L. Parc Cientific, University of Valencia, 46980 Paterna, Spain; (P.M.); (E.C.)
| | - Empar Chenoll
- Archer Daniels Midland (ADM), Nutrition, Health & Wellness, Biopolis S. L. Parc Cientific, University of Valencia, 46980 Paterna, Spain; (P.M.); (E.C.)
| | - Marta Serna-García
- Department of Dentistry, Faculty of Health Sciences, Universidad Europea de Valencia, 46010 Valencia, Spain;
| | - Maria D. Mauricio
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (S.G.-O.); (A.V.); (E.A.); (E.S.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- MODULAhR Group, Universitat de Valencia, 46010 Valencia, Spain
| | - Eva Serna
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (S.G.-O.); (A.V.); (E.A.); (E.S.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- MODULAhR Group, Universitat de Valencia, 46010 Valencia, Spain
| |
Collapse
|
9
|
Tang BB, Su CX, Wen N, Zhang Q, Chen JH, Liu BB, Wang YQ, Huang CQ, Hu YL. FMT and TCM to treat diarrhoeal irritable bowel syndrome with induced spleen deficiency syndrome- microbiomic and metabolomic insights. BMC Microbiol 2024; 24:433. [PMID: 39455910 PMCID: PMC11515126 DOI: 10.1186/s12866-024-03592-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Diarrheal irritable bowel syndrome (IBS-D) is a functional bowel disease with diarrhea, and can be associated with common spleen deficiency syndrome of the prevelent traditional Chinese medicine (TCM) syndrome. Fecal microbiota transplantation (FMT) could help treating IBS-D, but may provide variable effects. Our study evaluated the efficacy of TCM- shenling Baizhu decoction and FMT in treating IBS-D with spleen deficiency syndrome, with significant implications on gut microbiome and serum metabolites. METHODS The new borne rats were procured from SPF facility and separated as healthy (1 group) and IBS-D model ( 3 groups) rats were prepared articially using mother's separation and senna leaf treatment. 2 groups of IBS-D models were further treated with TCM- shenling Baizhu decoction and FMT. The efficacy was evaluated by defecation frequency, bristol stool score, and intestinal tight junction proteins (occludin-1 and claudin-1) expression. Microbiomic analysis was conducted using 16 S rRNA sequencing and bioinformatics tools. Metabolomics were detected in sera of rats by LC-MS and annotated by using KEGG database. RESULTS Significant increment in occludin-1 and claudin-1 protein expression alleviated the diarrheal severity in IBS-D rats (P < 0.05) after treatment with FMT and TCM. FMT and TCM altered the gut microbiota and regulated the tryptophan metabolism, steroid hormone biosynthesis and glycerophospholipid metabolism of IBS-D rats with spleen deficiency syndrome.The microbial abundance were changed in each case e.g., Monoglobus, Dubosiella, and Akkermansia and othe metabolic profiles. CONCLUSION FMT and TCM treatment improved the intestinal barrier function by regulating gut microbiota and improved metabolic pathways in IBS-D with spleen deficiency syndrome.
Collapse
Affiliation(s)
- Bin-Bin Tang
- Second Outpatient Department, Tongde Hospital of Zhejiang Province, Hangzhou, China
- Clinical College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Cheng-Xia Su
- Department of Gastroenterology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- First Clinical College, Hubei University of Chinese Medicine, Wuhan, China
| | - Na Wen
- Department of Gastroenterology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- First Clinical College, Hubei University of Chinese Medicine, Wuhan, China
| | - Qian Zhang
- Department of Gastroenterology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- First Clinical College, Hubei University of Chinese Medicine, Wuhan, China
| | - Jian-Hui Chen
- Department of Gastroenterology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- First Clinical College, Hubei University of Chinese Medicine, Wuhan, China
| | - Bin-Bin Liu
- Department of Gastroenterology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- First Clinical College, Hubei University of Chinese Medicine, Wuhan, China
| | - Yi-Qing Wang
- Department of Gastroenterology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- First Clinical College, Hubei University of Chinese Medicine, Wuhan, China
| | - Chao-Qun Huang
- Department of Gastroenterology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China.
- First Clinical College, Hubei University of Chinese Medicine, Wuhan, China.
| | - Yun-Lian Hu
- Department of Gastroenterology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China.
- First Clinical College, Hubei University of Chinese Medicine, Wuhan, China.
- Hubei Shizhen Laboratory, Wuhan, China.
| |
Collapse
|
10
|
Lin J, Chen D, Yan Y, Pi J, Xu J, Chen L, Zheng B. Gut microbiota: a crucial player in the combat against tuberculosis. Front Immunol 2024; 15:1442095. [PMID: 39502685 PMCID: PMC11534664 DOI: 10.3389/fimmu.2024.1442095] [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: 06/01/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
The mammalian gastrointestinal tract quickly becomes densely populated with foreign microorganisms shortly after birth, thereby establishing a lifelong presence of a microbial community. These commensal gut microbiota serve various functions, such as providing nutrients, processing ingested compounds, maintaining gut homeostasis, and shaping the intestinal structure in the host. Dysbiosis, which is characterized by an imbalance in the microbial community, is closely linked to numerous human ailments and has recently emerged as a key factor in health prognosis. Tuberculosis (TB), a highly contagious and potentially fatal disease, presents a pressing need for improved methods of prevention, diagnosis, and treatment strategies. Thus, we aim to explore the latest developments on how the host's immune defenses, inflammatory responses, metabolic pathways, and nutritional status collectively impact the host's susceptibility to or resilience against Mycobacterium tuberculosis infection. The review addresses how the fluctuations in the gut microbiota not only affect the equilibrium of these physiological processes but also indirectly influence the host's capacity to resist M. tuberculosis. This work highlights the central role of the gut microbiota in the host-microbe interactions and provides novel insights for the advancement of preventative and therapeutic approaches against tuberculosis.
Collapse
Affiliation(s)
- Jie Lin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Dongli Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yongen Yan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Junfa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Lingming Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Biying Zheng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
11
|
Lu C, Liu D, Wu Q, Zeng J, Xiong Y, Luo T. EphA2 blockage ALW-II-41-27 alleviates atherosclerosis by remodeling gut microbiota to regulate bile acid metabolism. NPJ Biofilms Microbiomes 2024; 10:108. [PMID: 39426981 PMCID: PMC11490535 DOI: 10.1038/s41522-024-00585-7] [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: 06/13/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024] Open
Abstract
Coronary artery disease (CAD), a critical condition resulting from systemic inflammation, metabolic dysfunction, and gut microbiota dysbiosis, poses a global public health challenge. ALW-II-41-27, a specific inhibitor of the EphA2 receptor, has shown anti-inflammatory prosperities. However, the impact of ALW-II-41-27 on atherosclerosis has not been elucidated. This study aimed to examine the roles of pharmacologically inhibiting EphA2 and the underlying mechanism in ameliorating atherosclerosis. ALW-II-41-27 was administered to apoE-/- mice fed a high-fat diet via intraperitoneal injection. We first discovered that ALW-II-41-27 led to a significant reduction in atherosclerotic plaques, evidenced by reduced lipid and macrophage accumulation, alongside an increase in collagen and smooth muscle cell content. ALW-II-41-27 also significantly lowered plasma and hepatic cholesterol levels, as well as the colonic inflammation. Furthermore, gut microbiota was analyzed by metagenomics and plasma metabolites by untargeted metabolomics. ALW-II-41-27-treated mice enriched Enterococcus, Akkermansia, Eggerthella and Lactobaccilus, accompanied by enhanced secondary bile acids production. To explore the causal link between ALW-II-41-27-associated gut microbiota and atherosclerosis, fecal microbiota transplantation was employed. Mice that received ALW-II-41-27-treated mouse feces exhibited the attenuated atherosclerotic plaque. In clinical, lower plasma DCA and HDCA levels were determined in CAD patients using quantitative metabolomics and exhibited a negative correlation with higher monocytes EphA2 expression. Our findings underscore the potential of ALW-II-41-27 as a novel therapeutic agent for atherosclerosis, highlighting its capacity to modulate gut microbiota composition and bile acid metabolism, thereby offering a promising avenue for CAD.
Collapse
Affiliation(s)
- Cong Lu
- Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dan Liu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiao Wu
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Zeng
- Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Xiong
- Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tiantian Luo
- Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
12
|
Mei L, Wang J, Hao Y, Zeng X, Yang Y, Wu Z, Ji Y. A comprehensive update on the immunoregulatory mechanisms of Akkermansia muciniphila: insights into active ingredients, metabolites, and nutrient-driven modulation. Crit Rev Food Sci Nutr 2024:1-18. [PMID: 39413040 DOI: 10.1080/10408398.2024.2416481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Akkermansia muciniphila (A. muciniphila) has gained recognition as a pioneering probiotic, exhibiting considerable potential to enhance immune conditions across both humans and animals. The health benefits of A. muciniphila are attributed to its various components, including outer membrane proteins (PilQ and Amuc_1100), secreted proteins (P9 and AmTARS), extracellular vesicles, and metabolites such as SCFAs, ornithine lipids, γ-aminobutyric acid, cobalamin, and inosine. The dynamic control of the mucus layer by A. muciniphila plays a crucial role in regulating intestinal mucosal immunity. Furthermore, A. muciniphila modulates immune function by interacting with macrophages, dendritic cells, T lymphocytes, and Paneth cells. Increasing the abundance of A. muciniphila in the gut through nutritional strategies represents a safe and effective means to augment immune function. Various polyphenols, oligosaccharides, and polysaccharides have been shown to elevate the levels of this bacterium, thereby contributing to favorable immunoregulatory outcomes. This paper delves into the latest research advancements related to the probiotic mechanisms of A. muciniphila and provides an overview of the current understanding of how its abundance responds to nutrients. These insights offer a theoretical foundation for the utilization of A. muciniphila in immunoregulation.
Collapse
Affiliation(s)
- Lihua Mei
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Jiaxin Wang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Yanling Hao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Yun Ji
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| |
Collapse
|
13
|
Liang Y, Li Y, Lee C, Yu Z, Chen C, Liang C. Ulcerative colitis: molecular insights and intervention therapy. MOLECULAR BIOMEDICINE 2024; 5:42. [PMID: 39384730 PMCID: PMC11464740 DOI: 10.1186/s43556-024-00207-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/13/2024] [Indexed: 10/11/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by abdominal pain, diarrhea, rectal bleeding, and weight loss. The pathogenesis and treatment of UC remain key areas of research interest. Various factors, including genetic predisposition, immune dysregulation, and alterations in the gut microbiota, are believed to contribute to the pathogenesis of UC. Current treatments for UC include 5-aminosalicylic acids, corticosteroids, immunosuppressants, and biologics. However, study reported that the one-year clinical remission rate is only around 40%. It is necessary to prompt the exploration of new treatment modalities. Biologic therapies, such as anti-TNF-α monoclonal antibody and JAK inhibitor, primarily consist of small molecules targeting specific pathways, effectively inducing and maintaining remission. Given the significant role of the gut microbiota, research into intestinal microecologics, such as probiotics and prebiotics, and fecal microbiota transplantation (FMT) shows promising potential in UC treatment. Additionally, medicinal herbs, such as chili pepper and turmeric, used in complementary therapy have shown promising results in UC management. This article reviews recent findings on the mechanisms of UC, including genetic susceptibility, immune cell dynamics and cytokine regulation, and gut microbiota alterations. It also discusses current applications of biologic therapy, herbal therapy, microecologics, and FMT, along with their prospects and challenges.
Collapse
Affiliation(s)
- Yuqing Liang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yang Li
- Department of Respiratory, Sichuan Integrative Medicine Hospital, Chengdu, 610042, China
| | - Chehao Lee
- Department of Traditional Chinese Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ziwei Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chongli Chen
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Chao Liang
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
14
|
Zhou L, Song W, Liu T, Yan T, He Z, He W, Lv J, Zhang S, Dai X, Yuan L, Shi L. Multi-omics insights into anti-colitis benefits of the synbiotic and postbiotic derived from wheat bran arabinoxylan and Limosilactobacillus reuteri. Int J Biol Macromol 2024; 278:134860. [PMID: 39163956 DOI: 10.1016/j.ijbiomac.2024.134860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/08/2024] [Accepted: 08/17/2024] [Indexed: 08/22/2024]
Abstract
Exploring nutritional therapies that manipulate tryptophan metabolism to activate AhR signaling represents a promising approach for mitigating chronic colitis. Arabinoxylan is a bioactive constituent abundant in wheat bran. Here, we comprehensively investigated anti-colitis potentials of wheat bran arabinoxylan (WBAX), its synbiotic and postbiotic derived from WBAX and Limosilactobacillus reuteri WX-94 (i.e., a probiotic strain exhibiting tryptophan metabolic activity). WBAX fueled L. reuteri and promoted microbial conversion of tryptophan to AhR ligands during in vitro fermentation in the culture medium and in the fecal microbiota from type 2 diabetes. The WBAX postbiotic outperformed WBAX and its synbiotic in augmenting efficacy of tryptophan in restoring DSS-disturbed serum immune markers, colonic tight junction proteins and gene profiles involved in amino acid metabolism and FoxO signaling. The WBAX postbiotic remodeled gut microbiota and superiorly enhanced AhR ligands (i.e., indole metabolites and bile acids), alongside with elevation in colonic AhR and IL-22. Associations between genera and metabolites modified by the postbiotic and colitis in human were verified and strong binding capacities between metabolites and colitis-related targets were demonstrated by molecular docking. Our study advances the novel perspective of WBAX in manipulating tryptophan metabolism and anti-colitis potentials of WBAX postbiotic via promoting gut microbiota-dependent AhR signaling.
Collapse
Affiliation(s)
- Lanqi Zhou
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Wei Song
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Tianqi Liu
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Tao Yan
- School of Food Science and Engineering, South China University of Technology, Guangdong 510641, China
| | - Ziyan He
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Weitai He
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Shaanxi Normal University, Xi'an 710062, China
| | - Jiayao Lv
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Shiyi Zhang
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Xiaoshuang Dai
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; Xbiome, Scientific Research Building, Room 907, Tsinghua High-Tech Park, Shenzhen, China
| | - Li Yuan
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Lin Shi
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China.
| |
Collapse
|
15
|
Gao Y, Chen Q, Yang S, Cao J, Li F, Li R, Wu Z, Wang Y, Yuan L. Indole alleviates nonalcoholic fatty liver disease in an ACE2-dependent manner. FASEB J 2024; 38:e70061. [PMID: 39305120 DOI: 10.1096/fj.202401172rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/27/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024]
Abstract
Indole is a microbial metabolite produced by the gut microbiota through the degradation of dietary tryptophan, known for its well-established anti-inflammatory and antioxidant properties. In this study, we collected fecal samples from mice fed a high-fat diet (HFD) and those on a standard diet (SD), then conducted 16S rRNA sequencing to analyze their gut microbiota. The analysis revealed distinct differences in the dominant bacterial species between the two groups, with a significant decrease in indole-producing probiotics in the HFD mice compared to the SD group. Then we administered oral indole treatment to male C57BL/6J mice with HFD-induced NAFLD and observed a significant improvement in hepatic steatosis and inflammation. Notably, indole alleviated the HFD-induced decline in serum Angiotensin-(1-7) [Ang-(1-7)] levels and Angiotensin-Converting Enzyme 2 (ACE2) expression. To further investigate the role of indole and ACE2 in NAFLD, we conducted experiments using ACE2 knockout (ACE2KO) mice that were also induced with HFD-induced NAFLD and treated with indole. Interestingly, the protective effects of indole were compromised in the absence of ACE2. In HepG2 cells, indole similarly stimulated ACE2 expression and, in an ACE2-dependent manner, reduced ROS generation, maintained mitochondrial membrane potential stability, and increased SIRT3 expression. In summary, our results highlight the formation of a biologically active gut-liver axis between the gut microbiota and the liver through the tryptophan metabolite indole, which mitigates NAFLD in an ACE2-dependent manner. Elevating dietary tryptophan and increasing indole levels may represent an effective approach for preventing and treating NAFLD.
Collapse
Affiliation(s)
- Yuanyuan Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Songtao Yang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Cao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangyu Li
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Li
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuoying Wu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Wang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Yuan
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Wang C, Xu Q, Wei C, Hu Q, Xiao Y, Jin Y. Kynurenine Attenuates Ulcerative Colitis Mediated by the Aryl Hydrocarbon Receptor. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:21000-21012. [PMID: 39271472 DOI: 10.1021/acs.jafc.4c04933] [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: 09/15/2024]
Abstract
The higher prevalence of ulcerative colitis (UC) and the side effects of its therapeutic agents contribute to finding novel treatments. This study aimed to investigate whether kynurenine (KYN), a tryptophan metabolite, has the possibility of alleviating UC and further clarifying the underlying mechanism. The effect of KYN on treating UC was evaluated by intestinal pathology, inflammatory cytokines, and tight-junction proteins in colitis mice and LPS-stimulated Caco-2 cells. Our results revealed that KYN relieved pathological symptoms of UC, improved intestinal barrier function, enhanced AhR expression, and inhibited NF-κB signaling pathway activation in the colon of colitis mice. Moreover, the improved intestinal barrier function, the decreased inflammasome production, and the inhibited activation of the NF-κB signaling pathway by KYN were dependent on AhR in Caco-2 cells. KYN could trigger AhR activation, inactivate the NF-κB signaling pathway, and inhibit NLRP3 inflammasome production, thus alleviating intestinal epithelial barrier dysfunction and reducing intestinal inflammation. In conclusion, the present study reveals that KYN ameliorates UC by improving the intestinal epithelial barrier and activating the AhR-NF-κB-NLRP3 signaling pathway, and it can be a promising therapeutic agent and dietary supplement for alleviating UC.
Collapse
Affiliation(s)
- Caihong Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Qihao Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | | | - Qinglian Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
- Xianghu Laboratory, Hangzhou 311231, China
| |
Collapse
|
17
|
Misera A, Marlicz W, Podkówka A, Łoniewski I, Skonieczna-Żydecka K. Possible application of Akkermansia muciniphila in stress management. MICROBIOME RESEARCH REPORTS 2024; 3:48. [PMID: 39741949 PMCID: PMC11684984 DOI: 10.20517/mrr.2023.81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 01/03/2025]
Abstract
Akkermansia muciniphila (A. muciniphila) is a promising candidate bacterium for stress management due to its beneficial effects on the microbiota-gut-brain axis (MGBA). As a well-known mucin-degrading bacterium in the digestive tract, A. muciniphila has demonstrated significant benefits for host physiology. Recent research highlights its potential in treating several neuropsychiatric disorders. Proposed mechanisms of action include the bacterium's outer membrane protein Amuc_1100 and potentially its extracellular vesicles (EVs), which interact with host immune receptors and influence serotonin pathways, which are crucial for emotional regulation. Despite its potential, the administration of probiotics containing A. muciniphila faces technological challenges, prompting the development of pasteurized forms recognized as safe by the European Food Safety Authority (EFSA). This review systematically examines the existing literature on the role of A. muciniphila in stress management, emphasizing the need for further research to validate its efficacy. The review follows a structured methodology, including comprehensive database searches and thematic data analysis, to provide a detailed understanding of the relationship between stress, microbiota, and A. muciniphila therapeutic potential.
Collapse
Affiliation(s)
- Agata Misera
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Szczecin 71-460, Poland
| | - Wojciech Marlicz
- Department of Gastroenterology, Pomeranian Medical University in Szczecin, Szczecin 71-252, Poland
| | - Albert Podkówka
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Szczecin 71-460, Poland
| | - Igor Łoniewski
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Szczecin 71-460, Poland
| | | |
Collapse
|
18
|
Markus V. Gut bacterial quorum sensing molecules and their association with inflammatory bowel disease: Advances and future perspectives. Biochem Biophys Res Commun 2024; 724:150243. [PMID: 38857558 DOI: 10.1016/j.bbrc.2024.150243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/15/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024]
Abstract
Inflammatory Bowel Disease (IBD) is an enduring inflammatory disease of the gastrointestinal tract (GIT). The complexity of IBD, its profound impact on patient's quality of life, and its burden on healthcare systems necessitate continuing studies to elucidate its etiology, refine care strategies, improve treatment outcomes, and identify potential targets for novel therapeutic interventions. The discovery of a connection between IBD and gut bacterial quorum sensing (QS) molecules has opened exciting opportunities for research into IBD pathophysiology. QS molecules are small chemical messengers synthesized and released by bacteria based on population density. These chemicals are sensed not only by the microbial species but also by host cells and are essential in gut homeostasis. QS molecules are now known to interact with inflammatory pathways, therefore rendering them potential therapeutic targets for IBD management. Given these intriguing developments, the most recent research findings in this area are herein reviewed. First, the global burden of IBD and the disruptions of the gut microbiota and intestinal barrier associated with the disease are assessed. Next, the general QS mechanism and signaling molecules in the gut are discussed. Then, the roles of QS molecules and their connection with IBD are elucidated. Lastly, the review proposes potential QS-based therapeutic targets for IBD, offering insights into the future research trajectory in this field.
Collapse
Affiliation(s)
- Victor Markus
- Near East University, Faculty of Medicine, Department of Medical Biochemistry, Nicosia, TRNC Mersin 10, Turkey.
| |
Collapse
|
19
|
Jiang P, Ji S, Su D, Zhao Y, Goncalves VBE, Xu G, Zhang M. The biofunction of Akkermansia muciniphila in intestinal-related diseases. MICROBIOME RESEARCH REPORTS 2024; 3:47. [PMID: 39741950 PMCID: PMC11684987 DOI: 10.20517/mrr.2024.12] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 01/03/2025]
Abstract
Intestinal homeostasis is essential for maintaining human health, and its dysfunction is related to the onset and progression of various diseases, including immune and metabolic disorders, and even tumorigenesis. Intestinal microbiota plays a critical role in intestinal homeostasis, with Akkermansia muciniphila (A. muciniphila) emerging as a key commensal bacterium utilizing mucin as its sole carbon and nitrogen source. A. muciniphila has been recognized in both experimental and clinical studies for its beneficial role in managing intestinal inflammation, tumors, functional gastrointestinal disorders, and secondary conditions such as liver and metabolic diseases. This review provides a comprehensive overview of the research history and current understanding of A. muciniphila, its association with various intestinal-related diseases, and the potential mechanisms behind its effects. This paper also explores the possibilities of leveraging the probiotic enzyme such as the active ingredients of A. muciniphila for the innovative clinical treatment of intestinal-related diseases.
Collapse
Affiliation(s)
- Ping Jiang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China
- Authors contributed equally
| | - Siqi Ji
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
- Authors contributed equally
| | - Dan Su
- FUJIFILM Diosynth Biotechnologies, Watertown, MA 02472, USA
| | - Yu Zhao
- University of Chicago, Pritzker School of Molecular Engineering, Chicago, IL 60637, USA
| | - Viriania Berta Esperanca Goncalves
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Guifang Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China
| | - Mingming Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| |
Collapse
|
20
|
Hu Z, Yao Y, Chen F, Feng L, Yuan Z, Deng J, Huang L, Yin Y, Tang X. Integrated analyses of the intestinal microbiome and transcriptome in Ningxiang piglets. Genomics 2024; 116:110919. [PMID: 39147334 DOI: 10.1016/j.ygeno.2024.110919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/03/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Ningxiang (NX) pig has been recognized as one of the most famous Chinese indigenous breeds due to its characteristics in stress resistance. However, intestinal microbial feature and gene profiling in NX piglets have not been studied. Here, we compared the intestinal microbiome and transcriptome between NX and Duroc × Landrace × Large white (DLY) piglets and found the high enrichment of several colonic Bacteroides, Prevotella and Clostridium species in NX piglets. Further functional analyses revealed their predominant function in methane, glycolysis and gluconeogenesis metabolism. Our mRNA-sequencing data unraveled the distinct colonic gene expression between these two breeds. In particular, we showed that the improved intestinal function in NX piglets may be determined by enhanced intestinal barrier gene expression and varied immune gene expression through modulating the composition of the gut microbes. Together, our study revealed the intestinal characteristics of NX piglets, providing their potential application in improving breeding strategies and developing dietary interventions.
Collapse
Affiliation(s)
- Zhenguo Hu
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Science, Changsha, Hunan 410125, China
| | - Yuezhou Yao
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Feiyue Chen
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Luya Feng
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Zian Yuan
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Junhao Deng
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Lang Huang
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Science, Changsha, Hunan 410125, China; Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, National Center of Technology Innovation for Synthetic Biology, Tianjin 300308, China.
| | - Xiongzhuo Tang
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China; Yuelushan Laboratory, Changsha, Hunan, 410128, China.
| |
Collapse
|
21
|
Li H, Du Y, Cheng K, Chen Y, Wei L, Pei Y, Wang X, Wang L, Zhang Y, Hu X, Lu Y, Zhu X. Gut microbiota-derived indole-3-acetic acid suppresses high myopia progression by promoting type I collagen synthesis. Cell Discov 2024; 10:89. [PMID: 39187483 PMCID: PMC11347609 DOI: 10.1038/s41421-024-00709-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 07/08/2024] [Indexed: 08/28/2024] Open
Abstract
High myopia (HM) is a leading cause of blindness worldwide with currently no effective interventions available. A major hurdle lies in its often isolated perception as a purely ocular morbidity, disregarding potential systemic implications. Recent evidence suggests the existence of a gut-eye axis; however, the role of gut microbiota in the pathogenesis of HM remains largely unexplored. Herein, we provide a potential crosstalk among HM's gut dysbiosis, microbial metabolites, and scleral remodeling. Utilizing 16S rRNA gene sequencing, we observed an altered gut microbiota profile in HM patients with a significant reduction in probiotic abundance compared with healthy controls. Subsequent targeted metabolic profiling revealed a notable decrease in plasma levels of the gut microbiota-derived metabolite indole-3-acetic acid (3-IAA) among HM patients, which is closely associated with the reduced probiotics, both negatively correlated with HM severity. Genetic analyses determined that gut microbiota are causally associated with myopia risk. Importantly, when mice subjected to HM modeling receive fecal microbiota transplantation from healthy donors, there is an increase in 3-IAA plasma levels and simultaneous retardation of HM progression along with better maintenance of collagen type I alpha 1 (COL1A1) expression in the sclera. Furthermore, 3-IAA gavage achieves similar effects. Mechanistic investigations confirm the transcriptional activation of COL1A1 by 3-IAA via promoting the enrichment of SP1 to its promoter. Together, our findings provide novel insights into the gut microbiota-eye axis in the pathogenesis of HM and propose new strategies for HM intervention by remodeling the gut microbiota and indole supplementation.
Collapse
Affiliation(s)
- Hao Li
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Du
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Kaiwen Cheng
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yuxi Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Ling Wei
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yujun Pei
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Xiaoyu Wang
- Mass Spectrometry Platform, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lan Wang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Ye Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Xiaoxin Hu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yi Lu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.
| | - Xiangjia Zhu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.
| |
Collapse
|
22
|
Khalili L, Park G, Nagpal R, Salazar G. The Role of Akkermansia muciniphila on Improving Gut and Metabolic Health Modulation: A Meta-Analysis of Preclinical Mouse Model Studies. Microorganisms 2024; 12:1627. [PMID: 39203469 PMCID: PMC11356609 DOI: 10.3390/microorganisms12081627] [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: 07/25/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
Akkermansia muciniphila (A. muciniphila) and its derivatives, including extracellular vesicles (EVs) and outer membrane proteins, are recognized for enhancing intestinal balance and metabolic health. However, the mechanisms of Akkermansia muciniphila's action and its effects on the microbiome are not well understood. In this study, we examined the influence of A. muciniphila and its derivatives on gastrointestinal (GI) and metabolic disorders through a meta-analysis of studies conducted on mouse models. A total of 39 eligible studies were identified through targeted searches on PubMed, Web of Science, Science Direct, and Embase until May 2024. A. muciniphila (alive or heat-killed) and its derivatives positively affected systemic and gut inflammation, liver enzyme level, glycemic response, and lipid profiles. The intervention increased the expression of tight-junction proteins in the gut, improving gut permeability in mouse models of GI and metabolic disorders. Regarding body weight, A. muciniphila and its derivatives prevented weight loss in animals with GI disorders while reducing body weight in mice with metabolic disorders. Sub-group analysis indicated that live bacteria had a more substantial effect on most analyzed biomarkers. Gut microbiome analysis using live A. muciniphila identified a co-occurrence cluster, including Desulfovibrio, Family XIII AD3011 group, and Candidatus Saccharimonas. Thus, enhancing the intestinal abundance of A. muciniphila and its gut microbial clusters may provide more robust health benefits for cardiometabolic, and age-related diseases compared with A. muciniphila alone. The mechanistic insight elucidated here will pave the way for further exploration and potential translational applications in human health.
Collapse
Affiliation(s)
- Leila Khalili
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
| | - Gwoncheol Park
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
| | - Ravinder Nagpal
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
| | - Gloria Salazar
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
- Center for Advancing Exercise and Nutrition Research on Aging (CAENRA), Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
23
|
Liu S, Zhao S, Cheng Z, Ren Y, Shi X, Mu J, Ge X, Dai Y, Li L, Zhang Z. Akkermansia muciniphila Protects Against Antibiotic-Associated Diarrhea in Mice. Probiotics Antimicrob Proteins 2024; 16:1190-1204. [PMID: 37314693 DOI: 10.1007/s12602-023-10101-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2023] [Indexed: 06/15/2023]
Abstract
Probiotics are used to prevent antibiotic-associated diarrhea (AAD) via the restoration of the gut microbiota. However, the precise effects of Akkermansia muciniphila (Akk), which is a promising probiotics, on AAD are unknown. Here, AAD models were established via the administration of lincomycin and ampicillin with or without pasteurized Akk or Amuc_1100 treatment. A diffusion test revealed that Akk was susceptible to the majority of the antibiotics, such as ampicillin. These effects were confirmed by the reduced Akk abundance in AAD model mice. Pasteurized Akk or Amuc_1100 significantly decreased the diarrhea status score and colon injury of AAD model mice. Additionally, these treatments significantly decreased the relative abundance of Citrobacter at genus level and reshaped the metabolic function of gut microbiota. Notably, pasteurized Akk or Amuc_1100 significantly changed the serum metabolome of AAD model mice. In addition, pasteurized Akk or Amuc_1100 suppressed intestinal inflammation by upregulating the expression of GPR109A and SLC5A8 and downregulating the expression of TNFα, IFNγ, IL1β, and IL6. Furthermore, they enhanced water and electrolyte absorption by upregulating AQP4, SLC26A3, and NHE3. Pasteurized Akk or Amuc_1100 also restored intestinal barrier function by ameliorating the downregulation of ZO-1, OCLN, CLDN4, and Muc2 in AAD model mice. In summary, optimizing intestinal health with pasteurized Akk or Amuc_1100 may serve as an approach for preventing AAD.
Collapse
Affiliation(s)
- Shenyin Liu
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Suying Zhao
- Department of Laboratory Medicine, The Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, 210009, People's Republic of China
| | - Zhiwei Cheng
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Yilin Ren
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Xinyi Shi
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Jing Mu
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Xiangyang Ge
- Technical Department of Sujiu Group, Suqian, 223800, People's Republic of China
| | - Yuan Dai
- Technical Department of Sujiu Group, Suqian, 223800, People's Republic of China
| | - Lei Li
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.
| | - Zhan Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.
| |
Collapse
|
24
|
Liu M, Li S, Cao S, Liu C, Han Y, Cheng J, Zhang S, Zhao J, Shi Y. Let food be your medicine - dietary fiber. Food Funct 2024; 15:7733-7756. [PMID: 38984439 DOI: 10.1039/d3fo05641d] [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: 07/11/2024]
Abstract
Dietary fiber (DF) cannot be digested and absorbed by the digestive tract, nor can it provide the energy needed to be burned for metabolic activities. Therefore, from the 1950s to the 1980s, DF received little attention in nutrition studies. With in-depth research and developments in global nutrition, people have gradually paid attention to the fact that DF occupies an essential position in the structure of nutrition, and it can ensure the healthy development of human beings. As early as 390 B.C., the ancient Greek physician Hippocrates proposed, "Let your food be your medicine, and your medicine be your food". This concept has been more systematically validated in modern scientific research, with numerous epidemiological studies showing that the dietary intake of DF-rich foods such as whole grains, root vegetables, legumes, and fruits has the potential to regulate the balance of the gut microbiota and thereby prevent diseases. However, the crosstalk between different types of DF and the gut microbiota is quite complex, and the effects on the organism vary. In this paper, we discuss research on DF and the gut microbiota and related diseases, aiming to understand the relationship between all three better and provide a reference basis for the risk reduction of related diseases.
Collapse
Affiliation(s)
- Mengqi Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Shouren Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Shixi Cao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Cong Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Yao Han
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Jiawen Cheng
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Shuhang Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Jiangchao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas, USA
| | - Yinghua Shi
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
- Henan Forage Engineering Technology Research Center, Zhengzhou, Henan, 450002, China
| |
Collapse
|
25
|
Song Z, He J, Yu W, He C, Yang M, Li P, Li Z, Jian G, Cheng S. Exploring the multifaceted therapeutic mechanism of Schisanlactone E (XTS) in APP/PS1 mouse model of Alzheimer's disease through multi-omics analysis. Front Microbiol 2024; 15:1440564. [PMID: 39044957 PMCID: PMC11263214 DOI: 10.3389/fmicb.2024.1440564] [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: 05/29/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024] Open
Abstract
Background Schisanlactone E, also known as XueTongSu (XTS), is an active compound extracted from the traditional Tujia medicine Kadsura heteroclita ("XueTong"). Recent studies highlight its anti-inflammatory and antioxidant properties, yet the mechanisms of XTS's therapeutic effects on Alzheimer's disease (AD) are unclear. This study aims to elucidate the therapeutic efficacy and mechanisms of XTS in AD. Methods Ten C57BL/6 mice were assigned to the control group (NC), and twenty APP/PS1 transgenic mice were randomly divided into the model group (M) (10 mice) and the XTS treatment group (Tre) (10 mice). After an acclimatization period of 7 days, intraperitoneal injections were administered over a 60-day treatment period. The NC and M groups received saline, while the Tre group received XTS at 2 mg/kg. Learning and memory abilities were assessed using the Morris Water Maze (MWM) test. Histopathological changes were evaluated using hematoxylin and eosin (HE) and Nissl staining, and immunofluorescence was used to assess pathological products and glial cell activation. Cytokine levels (IL-1β, IL-6, TNF-α) in the hippocampus were quantified by qPCR. 16S rDNA sequencing analyzed gut microbiota metabolic alterations, and metabolomic analysis was performed on cortical samples. The KEGG database was used to analyze the regulatory mechanisms of XTS in AD treatment. Results XTS significantly improved learning and spatial memory in APP/PS1 mice and ameliorated histopathological changes, reducing Aβ plaque aggregation and glial cell activation. XTS decreased the expression of inflammatory cytokines IL-1β, IL-6, and TNF-α. It also enhanced gut microbiota diversity, notably increasing Akkermansia species, and modulated levels of metabolites such as isosakuranetin, 5-KETE, 4-methylcatechol, and sphinganine. Pathway analysis indicated that XTS regulated carbohydrate metabolism, neuroactive ligand-receptor interactions, and alanine, aspartate, and glutamate metabolism, mitigating gut microbiota dysbiosis and metabolic disturbances. Conclusion XTS ameliorates cognitive deficits, pathological changes, and inflammatory responses in APP/PS1 mice. It significantly modulates the gut microbiota, particularly increasing Akkermansia abundance, and influences levels of key metabolites in both the gut and brain. These findings suggest that XTS exerts anti-AD effects through the microbial-gut-brain axis (MGBA).
Collapse
Affiliation(s)
- Zhenyan Song
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jiawei He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wenjing Yu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chunxiang He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Miao Yang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ping Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ze Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gonghui Jian
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Shaowu Cheng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
26
|
Wei Y, Zhao X, Xu T, Liu Z, Zuo Y, Zhang M, Zhang Y, Yin H. Soybean Bioactive Peptide Supplementation Affects the Intestinal Immune Antioxidant Function, Microbial Diversity, and Reproductive Organ Development in Roosters. Animals (Basel) 2024; 14:1954. [PMID: 38998068 PMCID: PMC11240439 DOI: 10.3390/ani14131954] [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: 06/06/2024] [Revised: 06/23/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
Soybean is an important source of high-quality vegetable protein with various health-improving properties, and its main bioactive substances are small peptides produced by in vitro enzymatic hydrolytic processes. In traditional layer breeding, the nutritional health of roosters is frequently neglected, ultimately affecting the quality and quantity of offspring. This study investigated the effects of various quantities (0%, 0.15%, 0.30%, 0.45%, and 0.60%) of soybean bioactive peptide (SBP) feed additives on immunological and antioxidant functions, gut health, and reproductive performance of roosters. SBP supplementation significantly improved male growth and reproductive performance, including growth rate, feed conversion ratio, reproductive organ development, and semen quality. SBP also increased immune and antioxidant levels, boosted the integrity of the small intestinal physiological structure and barrier function, and diversity of cecal microbes, and decreased the apoptotic ratio of small intestinal epithelial cells. The effects of SBP on various functions of males showed a quadratic trend, with the optimal concentration determined to be 0.45%.
Collapse
Affiliation(s)
- Yimeng Wei
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiyu Zhao
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Tao Xu
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhenyan Liu
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yalan Zuo
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingxue Zhang
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yao Zhang
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Huadong Yin
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
27
|
Jena PK, Wakita D, Gomez AC, Carvalho TT, Atici AE, Narayanan M, Lee Y, Fishbein MC, Cani PD, de Vos WM, Underhill DM, Devkota S, Chen S, Shimada K, Crother TR, Arditi M, Rivas MN. The intestinal microbiota contributes to the development of immune-mediated cardiovascular inflammation and vasculitis in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596258. [PMID: 38853964 PMCID: PMC11160596 DOI: 10.1101/2024.05.28.596258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Alterations in the intestinal microbiota contribute to the pathogenesis of various cardiovascular disorders, but how they affect the development of Kawasaki disease (KD), an acute pediatric vasculitis, remains unclear. We report that depleting the gut microbiota reduces the development of cardiovascular inflammation in a murine model mimicking KD vasculitis. The development of cardiovascular lesions was associated with alterations in the intestinal microbiota composition and, notably, a decreased abundance of Akkermansia muciniphila and Faecalibacterium prausnitzii. Oral supplementation with either of these live or pasteurized individual bacteria, or with short-chain fatty acids (SCFAs) produced by them, attenuated cardiovascular inflammation. Treatment with Amuc_1100, the TLR-2 signaling outer membrane protein from A. muciniphila , also decreased the severity of vascular inflammation. This study reveals an underappreciated gut microbiota-cardiovascular inflammation axis in KD vasculitis pathogenesis and identifies specific intestinal commensals that regulate vasculitis in mice by producing metabolites or via extracellular proteins acting on gut barrier function. IN BRIEF It remains unclear whether changes in the intestinal microbiota composition are involved in the development of cardiovascular lesions associated with Kawasaki disease (KD), an immune-mediated vasculitis. Jena et al. observe alterations in the intestinal microbiota composition of mice developing vasculitis, characterized by reduced A. muciniphila and F. prausnitzii . Oral supplementation with either of these bacteria, live or pasteurized, or with bacteria-produced short-chain fatty acids (SCFAs) or Amuc_1100, the TLR-2 signaling outer membrane protein of A. muciniphila , was sufficient to alleviate the development of cardiovascular lesions in mice by promoting intestinal barrier function. HIGHLIGHTS Absence or depletion of the microbiota decreases the severity of vasculitis in a murine model mimicking KD vasculitis. Supplementation of B. wadsworthia and B. fragilis promotes murine KD vasculitis. Decreased abundances of F. prausnitzii and A. muciniphila are associated with the development of cardiovascular lesions in mice. Supplementation with either live or pasteurized A. muciniphila and F. prausnitzii, or the TLR-2 signaling Amuc_1100, reduces the severity of vasculitis by promoting gut barrier function.
Collapse
|
28
|
Guo J, Li L, Cai Y, Kang Y. The development of probiotics and prebiotics therapy to ulcerative colitis: a therapy that has gained considerable momentum. Cell Commun Signal 2024; 22:268. [PMID: 38745207 PMCID: PMC11094941 DOI: 10.1186/s12964-024-01611-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 04/10/2024] [Indexed: 05/16/2024] Open
Abstract
Ulcerative colitis (UC) is increasingly common, and it is gradually become a kind of global epidemic. UC is a type of inflammatory bowel disease (IBD), and it is a lifetime recurrent disease. UC as a common disease has become a financial burden for many people and has the potential to develop into cancer if not prevented or treated. There are multiple factors such as genetic factors, host immune system disorders, and environmental factors to cause UC. A growing body of research have suggested that intestinal microbiota as an environmental factor play an important role in the occurrence and development of UC. Meanwhile, evidence to date suggests that manipulating the gut microbiome may represent effective treatment for the prevention or management of UC. In addition, the main clinical drugs to treat UC are amino salicylate and corticosteroid. These clinical drugs always have some side effects and low success rate when treating patients with UC. Therefore, there is an urgent need for safe and efficient methods to treat UC. Based on this, probiotics and prebiotics may be a valuable treatment for UC. In order to promote the wide clinical application of probiotics and prebiotics in the treatment of UC. This review aims to summarize the recent literature as an aid to better understanding how the probiotics and prebiotics contributes to UC while evaluating and prospecting the therapeutic effect of the probiotics and prebiotics in the treatment of UC based on previous publications.
Collapse
Affiliation(s)
- Jing Guo
- Department of microbiology and immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Liping Li
- Department of microbiology and immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yue Cai
- Faculty of Life science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yongbo Kang
- Department of microbiology and immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
29
|
Liu Y, Bai X, Wu H, Duan Z, Zhu C, Fu R, Fan D. Ginsenoside CK Alleviates DSS-Induced IBD in Mice by Regulating Tryptophan Metabolism and Activating Aryl Hydrocarbon Receptor via Gut Microbiota Modulation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:9867-9879. [PMID: 38602268 DOI: 10.1021/acs.jafc.4c00245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Dysbiosis of gut microbiota is believed to be associated with inflammatory bowel disease (IBD). Ginsenoside compound K (CK), the main metabolite of Panax ginseng ginsenoside, has proven effective as an anti-inflammatory agent in IBD. However, the mechanisms by which CK modulates gut microbiota to ameliorate IBD remain poorly understood. Herein, CK demonstrated the potential to suppress the release of proinflammatory cytokines by gut microbiota modulation. Notably, supplementation with CK promoted the restoration of a harmonious balance in gut microbiota, primarily by enhancing the populations of Lactobacillus and Akkermansia. Furthermore, CK considerably elevated the concentrations of tryptophan metabolites derived from Lactobacillus that could activate the aryl hydrocarbon receptor. Overall, the promising alleviative efficacy of CK primarily stemmed from the promotion of Lactobacillus growth and production of tryptophan metabolites, suggesting that CK should be regarded as a prospective prebiotic agent for IBD in the future.
Collapse
Affiliation(s)
- Yuan Liu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Xue Bai
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Huanyan Wu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Zhiguang Duan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Chenhui Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Rongzhan Fu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| |
Collapse
|
30
|
Fang W, Jin M, Qi W, Kong C, Song G, Peng W, Wang Y. Caffeic acid combined with arabinoxylan or β-glucan attenuates diet-induced obesity in mice via modulation of gut microbiota and metabolites. Int J Biol Macromol 2024; 268:131683. [PMID: 38649076 DOI: 10.1016/j.ijbiomac.2024.131683] [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: 12/06/2023] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
Polyphenols and dietary fibers in whole grains are important bioactive compounds to reduce risks for obesity. However, whether the combination of the two components exhibits a stronger anti-obesity effect remains unclear. Caffeic acid is a major phenolic acid in cereals, and arabinoxylan and β-glucan are biological macromolecules with numerous health benefits. Here, we investigated the effect of caffeic acid combined with arabinoxylan or β-glucan on glucose and lipid metabolism, gut microbiota, and metabolites in mice fed a high-fat diet (HFD). Caffeic acid combined with arabinoxylan or β-glucan significantly reduced the body weight, blood glucose, and serum free fatty acid concentrations. Caffeic acid combined with β-glucan effectively decreased serum total cholesterol levels and hepatic lipid accumulation, modulated oxidative and inflammatory stress, and improved gut barrier function. Compared with arabinoxylan, β-glucan, and caffeic acid alone, caffeic acid combined with arabinoxylan or β-glucan exhibited a better capacity to modulate gut microbiota, including increased microbial diversity, reduced Firmicutes/Bacteroidetes ratio, and increased abundance of beneficial bacteria such as Bifidobacterium. Furthermore, caffeic acid combined with β-glucan reversed HFD-induced changes in microbiota-derived metabolites involving tryptophan, purine, and bile acid metabolism. Thus, caffeic acid and β-glucan had a synergistic anti-obesity effect by regulating specific gut microbiota and metabolites.
Collapse
Affiliation(s)
- Wei Fang
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Mingyu Jin
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China
| | - Wentao Qi
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Chunli Kong
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Ge Song
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Wenting Peng
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Yong Wang
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China.
| |
Collapse
|
31
|
Liu M, Guo S, Wang L. Systematic review of metabolomic alterations in ulcerative colitis: unveiling key metabolic signatures and pathways. Therap Adv Gastroenterol 2024; 17:17562848241239580. [PMID: 38560428 PMCID: PMC10981261 DOI: 10.1177/17562848241239580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Background Despite numerous metabolomic studies on ulcerative colitis (UC), the results have been highly variable, making it challenging to identify key metabolic abnormalities in UC. Objectives This study aims to uncover key metabolites and metabolic pathways in UC by analyzing existing metabolomics data. Design A systematic review. Data sources and methods We conducted a comprehensive search in databases (PubMed, Cochrane Library, Embase, and Web of Science) and relevant study references for metabolomic research on UC up to 28 December 2022. Significant metabolite differences between UC patients and controls were identified, followed by an analysis of relevant metabolic pathways. Results This review incorporated 78 studies, identifying 2868 differentially expressed metabolites between UC patients and controls. The metabolites were predominantly from 'lipids and lipid-like molecules' and 'organic acids and derivatives' superclasses. We found 101 metabolites consistently altered in multiple datasets within the same sample type and 78 metabolites common across different sample types. Of these, 62 metabolites exhibited consistent regulatory trends across various datasets or sample types. Pathway analysis revealed 22 significantly altered metabolic pathways, with 6 pathways being recurrently enriched across different sample types. Conclusion This study elucidates key metabolic characteristics in UC, offering insights into molecular mechanisms and biomarker discovery for the disease. Future research could focus on validating these findings and exploring their clinical applications.
Collapse
Affiliation(s)
- Meiling Liu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Siyi Guo
- Chongqing Medical University, Chongqing, China
| | - Liang Wang
- Chongqing Medical University, Chongqing, China
| |
Collapse
|
32
|
Zhang R, Chen J, Liu L, Li X, Qiu C. Gut microbiota-based discriminative model for patients with ulcerative colitis: A meta-analysis and real-world study. Medicine (Baltimore) 2024; 103:e37091. [PMID: 38457570 PMCID: PMC10919464 DOI: 10.1097/md.0000000000037091] [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: 09/19/2023] [Revised: 12/23/2023] [Accepted: 01/05/2024] [Indexed: 03/10/2024] Open
Abstract
Gut microbiota directly interacts with intestinal epithelium and is a significant factor in the pathogenesis of ulcerative colitis (UC). A meta-analysis was performed to investigate gut microbiota composition of patients with UC in the United States. We also collected fecal samples from Chinese patients with UC and healthy individuals. Gut microbiota was tested using 16S ribosomal RNA gene sequencing. Meta-analysis and 16S ribosomal RNA sequencing revealed significant differences in gut bacterial composition between UC patients and healthy subjects. The Chinese UC group had the highest scores for Firmicutes, Clostridia, Clostridiales, Streptococcaceae, and Blautia, while healthy cohort had the highest scores for P-Bacteroidetes, Bacteroidia, Bacteroidales, Prevotellaceae, and Prevotella_9. A gut microbiota-based discriminative model trained on an American cohort achieved a discrimination efficiency of 0.928 when applied to identify the Chinese UC cohort, resulting in a discrimination efficiency of 0.759. Additionally, a differentiation model was created based on gut microbiota of a Chinese cohort, resulting in an area under the receiver operating characteristic curve of 0.998. Next, we applied the model established for the Chinese UC cohort to analyze the American cohort. Our findings suggest that the diagnostic efficiency ranged from 0.8794 to 0.9497. Furthermore, a combined analysis using data from both the Chinese and US cohorts resulted in a model with a diagnostic efficacy of 0.896. In summary, we found significant differences in gut bacteria between UC individuals and healthy subjects. Notably, the model from the Chinese cohort performed better at diagnosing UC patients compared to healthy subjects. These results highlight the promise of personalized and region-specific approaches using gut microbiota data for UC diagnosis.
Collapse
Affiliation(s)
- Rong Zhang
- Department of General Surgery, The Third People’s Hospital of Chengdu, Chengdu 610014, Sichuan Province, China
| | - Jing Chen
- Department of Gastroenterology, The People’s Hospital of Dujiangyan, Dujiangyan 611830, Sichuan Province, China
| | - Li Liu
- Department of Gastroenterology, The Third People’s Hospital of Chengdu, Chengdu 610014, Sichuan Province, China
| | - Xiankun Li
- Department of Pharmacy, The People’s Hospital of Dujiangyan, Dujiangyan 611830, Sichuan Province, China
| | - Changwei Qiu
- Department of Gastroenterology, The People’s Hospital of Dujiangyan, Dujiangyan 611830, Sichuan Province, China
| |
Collapse
|
33
|
Wang L, Li M, Gu Y, Shi J, Yan J, Wang X, Li B, Wang B, Zhong W, Cao H. Dietary flavonoids-microbiota crosstalk in intestinal inflammation and carcinogenesis. J Nutr Biochem 2024; 125:109494. [PMID: 37866426 DOI: 10.1016/j.jnutbio.2023.109494] [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: 09/27/2022] [Revised: 02/20/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Colorectal cancer (CRC) is currently the third leading cancer and commonly develops from chronic intestinal inflammation. A strong association was found between gut microbiota and intestinal inflammation and carcinogenic risk. Flavonoids, which are abundant in vegetables and fruits, can inhibit inflammation, regulate gut microbiota, protect gut barrier integrity, and modulate immune cell function, thereby attenuating colitis and preventing carcinogenesis. Upon digestion, about 90% of flavonoids are transported to the colon without being absorbed in the small intestine. This phenomenon increases the abundance of beneficial bacteria and enhances the production of short-chain fatty acids. The gut microbe further metabolizes these flavonoids. Interestingly, some metabolites of flavonoids play crucial roles in anti-inflammation and anti-tumor effects. This review summarizes the modulatory effect of flavonoids on gut microbiota and their metabolism by intestinal microbe under disease conditions, including inflammatory bowel disease, colitis-associated cancer (CAC), and CRC. We focus on dietary flavonoids and microbial interactions in intestinal mucosal barriers as well as intestinal immune cells. Results provide novel insights to better understand the crosstalk between dietary flavonoids and gut microbiota and support the standpoint that dietary flavonoids prevent intestinal inflammation and carcinogenesis.
Collapse
Affiliation(s)
- Lei Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China; Department of Gastroenterology and Hepatology, The Affiliated Hospital of Chengde Medical College, Hebei, China
| | - Mengfan Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yu Gu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Junli Shi
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Chengde Medical College, Hebei, China
| | - Jing Yan
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China; Department of Nutrition, the Second Affiliated Hospital, Air Force Medical University, Xi'an, China
| | - Xin Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bingqing Li
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Chengde Medical College, Hebei, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| |
Collapse
|
34
|
Ren Y, Shi X, Mu J, Liu S, Qian X, Pei W, Ni S, Zhang Z, Li L, Zhang Z. Chronic exposure to parabens promotes non-alcoholic fatty liver disease in association with the changes of the gut microbiota and lipid metabolism. Food Funct 2024; 15:1562-1574. [PMID: 38236135 DOI: 10.1039/d3fo04347a] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become a serious public health issue due to changing dietary patterns and composition. However, the relationship between NAFLD occurrence and food additives, such as preservatives, remains unknown. This study aimed to evaluate the toxicity of parabens, namely methylparaben (MeP) and ethylparaben (EtP), in relation to NAFLD occurrence in mice under different dietary conditions. Exposure to MeP and EtP exacerbated high-fat diet (HFD)-induced obesity, glucose intolerance, higher serum lipid concentrations, and fat accumulation by upregulating genes involved in lipid metabolism. Untargeted metabolomics revealed that arachidonic acid (AA) metabolism was the top enriched pathway upon MeP and EtP exposure in the presence of HFD. 11,12-Epoxyeicosatrienoic acid (11,12-EET) was the most abundant AA metabolite and was significantly reduced upon exposure to MeP or EtP. Moreover, an integrative analysis of differential fecal taxa at the genus level and serum AA metabolites revealed significant associations. In addition, MeP and EtP enhanced lipid accumulation in AML12 cells and HepG2 cells cultured with oleic acid. 11,12-EET supplementation could significantly alleviate lipid accumulation by suppressing the expression of lipid metabolism-related genes and proteins. The present study suggests that chronic exposure to MeP and EtP promoted NAFLD via gut microbiota-dependent AA metabolism. These results highlight the need for reducing oral exposure to synthetic preservatives to improve metabolic disturbance under HFD conditions.
Collapse
Affiliation(s)
- Yilin Ren
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China.
| | - Xinyi Shi
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China.
| | - Jing Mu
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China.
| | - Shenyin Liu
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China.
| | - Xin Qian
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China.
| | - Wenlong Pei
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China.
| | - Shanhong Ni
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China.
| | - Zhengduo Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China.
| | - Lei Li
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China.
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China
| | - Zhan Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China.
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China
| |
Collapse
|
35
|
Wu H, Qi S, Yang R, Pan Q, Lu Y, Yao C, He N, Huang S, Ling X. Strategies for high cell density cultivation of Akkermansia muciniphila and its potential metabolism. Microbiol Spectr 2024; 12:e0238623. [PMID: 38059626 PMCID: PMC10782997 DOI: 10.1128/spectrum.02386-23] [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: 06/20/2023] [Accepted: 11/08/2023] [Indexed: 12/08/2023] Open
Abstract
IMPORTANCE Currently, there is significant interest in Akkermansia muciniphila as a promising next-generation probiotic, making it a hot topic in scientific research. However, to achieve efficient industrial production, there is an urgent need to develop an in vitro culture method to achieve high biomass using low-cost carbon sources such as glucose. This study aims to explore the high-density fermentation strategy of A. muciniphila by optimizing the culture process. This study also employs techniques such as LC-MS and RNA-Seq to explain the possible regulatory mechanism of high-density cell growth and increased cell surface hydrophobicity facilitating cell colonization of the gut in vitro culture. Overall, this research sheds light on the potential of A. muciniphila as a probiotic and provides valuable insights for future industrial production.
Collapse
Affiliation(s)
- Haiting Wu
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Shuhua Qi
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Ruixiong Yang
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Qihua Pan
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Yinghua Lu
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Synthetic Biotechnology, Xiamen University, Xiamen, People's Republic of China
- The Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, Xiamen, People's Republic of China
| | - Chuanyi Yao
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Synthetic Biotechnology, Xiamen University, Xiamen, People's Republic of China
| | - Ning He
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Synthetic Biotechnology, Xiamen University, Xiamen, People's Republic of China
| | - Song Huang
- Department of Microbiome and Health, Bluepha Co., Ltd, Shenzhen, People's Republic of China
| | - Xueping Ling
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Synthetic Biotechnology, Xiamen University, Xiamen, People's Republic of China
| |
Collapse
|
36
|
Li S, Cai Y, Guan T, Zhang Y, Huang K, Zhang Z, Cao W, Guan X. Quinic acid alleviates high-fat diet-induced neuroinflammation by inhibiting DR3/IKK/NF-κB signaling via gut microbial tryptophan metabolites. Gut Microbes 2024; 16:2374608. [PMID: 38972055 PMCID: PMC11229714 DOI: 10.1080/19490976.2024.2374608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 06/26/2024] [Indexed: 07/09/2024] Open
Abstract
With the increasing of aging population and the consumption of high-fat diets (HFD), the incidence of Alzheimer's disease (AD) has skyrocketed. Natural antioxidants show promising potential in the prevention of AD, as oxidative stress and neuroinflammation are two hallmarks of AD pathogenesis. Here, we showed that quinic acid (QA), a polyphenol derived from millet, significantly decreased HFD-induced brain oxidative stress and neuroinflammation and the levels of Aβ and p-Tau. Examination of gut microbiota suggested the improvement of the composition of gut microbiota in HFD mice after QA treatment. Metabolomic analysis showed significant increase of gut microbial tryptophan metabolites indole-3-acetic acid (IAA) and kynurenic acid (KYNA) by QA. In addition, IAA and KYNA showed negative correlation with pro-inflammatory factors and AD indicators. Further experiments on HFD mice proved that IAA and KYNA could reproduce the effects of QA that suppress brain oxidative stress and inflammation and decrease the levels of of Aβ and p-Tau. Transcriptomics analysis of brain after IAA administration revealed the inhibition of DR3/IKK/NF-κB signaling pathway by IAA. In conclusion, this study demonstrated that QA could counteract HFD-induced brain oxidative stress and neuroinflammation by regulating inflammatory DR3/IKK/NF-κB signaling pathway via gut microbial tryptophan metabolites.
Collapse
Affiliation(s)
- Sen Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Yuwei Cai
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Tong Guan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Yu Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Kai Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Ze Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Wangqing Cao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Xiao Guan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
37
|
Meynier M, Daugey V, Mallaret G, Gervason S, Meleine M, Barbier J, Aissouni Y, Lolignier S, Bonnet M, Ardid D, De Vos WM, Van Hul M, Suenaert P, Brochot A, Cani PD, Carvalho FA. Pasteurized akkermansia muciniphila improves irritable bowel syndrome-like symptoms and related behavioral disorders in mice. Gut Microbes 2024; 16:2298026. [PMID: 38170633 PMCID: PMC10766393 DOI: 10.1080/19490976.2023.2298026] [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: 03/20/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
Gut - brain communications disorders in irritable bowel syndrome (IBS) are associated with intestinal microbiota composition, increased gut permeability, and psychosocial disturbances. Symptoms of IBS are difficult to medicate, and hence much research is being made into alternative approaches. This study assesses the potential of a treatment with pasteurized Akkermansia muciniphila for alleviating IBS-like symptoms in two mouse models of IBS with different etiologies. Two clinically relevant animal models were used to mimic IBS-like symptoms in C57BL6/J mice: the neonatal maternal separation (NMS) paradigm and the Citrobacter rodentium infection model. In both models, gut permeability, colonic sensitivity, fecal microbiota composition and colonic IL-22 expression were evaluated. The cognitive performance and emotional state of the animals were also assessed by several tests in the C. rodentium infection model. The neuromodulation ability of pasteurized A. muciniphila was assessed on primary neuronal cells from mice dorsal root ganglia using a ratiometric calcium imaging approach. The administration of pasteurized A. muciniphila significantly reduced colonic hypersensitivity in both IBS mouse models, accompanied by a reinforcement of the intestinal barrier function. Beneficial effects of pasteurized A. muciniphila treatment have also been observed on anxiety-like behavior and memory defects in the C. rodentium infection model. Finally, a neuroinhibitory effect exerted by pasteurized A. muciniphila was observed on neuronal cells stimulated with two algogenic substances such as capsaicin and inflammatory soup. Our findings demonstrate novel anti-hyperalgesic and neuroinhibitory properties of pasteurized A. muciniphila, which therefore may have beneficial effects in relieving pain and anxiety in subjects with IBS.
Collapse
Affiliation(s)
- Maëva Meynier
- NeuroDol, UMR 1107 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France
- M2iSH, UMR 1071 INSERM, UMR1382 INRAé, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Valentine Daugey
- NeuroDol, UMR 1107 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Geoffroy Mallaret
- NeuroDol, UMR 1107 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Sandie Gervason
- NeuroDol, UMR 1107 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Mathieu Meleine
- NeuroDol, UMR 1107 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Julie Barbier
- NeuroDol, UMR 1107 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Youssef Aissouni
- NeuroDol, UMR 1107 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Stéphane Lolignier
- NeuroDol, UMR 1107 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Mathilde Bonnet
- M2iSH, UMR 1071 INSERM, UMR1382 INRAé, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Denis Ardid
- NeuroDol, UMR 1107 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Willem M. De Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- The Akkermansia Company™, Mont-Saint-Guibert, Belgium
| | - Matthias Van Hul
- Metabolism and Nutrition Research group, Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- WELBIO-Walloon Excellence in Life Sciences and Biotechnology, WELBIO department, WEL Research Institute, Wavre, Belgium
| | | | | | - Patrice D. Cani
- Metabolism and Nutrition Research group, Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- WELBIO-Walloon Excellence in Life Sciences and Biotechnology, WELBIO department, WEL Research Institute, Wavre, Belgium
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Frédéric A. Carvalho
- NeuroDol, UMR 1107 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
38
|
Sosnowski K, Przybyłkowski A. Ethanol-induced changes to the gut microbiome compromise the intestinal homeostasis: a review. Gut Microbes 2024; 16:2393272. [PMID: 39224006 PMCID: PMC11376419 DOI: 10.1080/19490976.2024.2393272] [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: 03/19/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
The intestine is the largest organ in terms of surface area in the human body. It is responsible not only for absorbing nutrients but also for protection against the external world. The gut microbiota is essential in maintaining a properly functioning intestinal barrier, primarily through producing its metabolites: short-chain fatty acids, bile acids, and tryptophan derivatives. Ethanol overconsumption poses a significant threat to intestinal health. Not only does it damage the intestinal epithelium, but, maybe foremostly, it changes the gut microbiome. Those ethanol-driven changes shift its metabolome, depriving the host of the protective effect the physiological gut microbiota has. This literature review discusses the impact of ethanol consumption on the gut, the gut microbiota, and its metabolome, providing a comprehensive overview of the mechanisms through which ethanol disrupts intestinal homeostasis and discussing potential avenues for new therapeutic intervention.
Collapse
Affiliation(s)
- Konrad Sosnowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
39
|
Su W, Yang Y, Zhao X, Cheng J, Li Y, Wu S, Wu C. Potential efficacy and mechanism of eight mild-natured and bitter-flavored TCMs based on gut microbiota: A review. CHINESE HERBAL MEDICINES 2024; 16:42-55. [PMID: 38375054 PMCID: PMC10874767 DOI: 10.1016/j.chmed.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/02/2023] [Accepted: 08/04/2023] [Indexed: 02/21/2024] Open
Abstract
The mild-natured and bitter-flavored traditional Chinese medicines (MB-TCMs) are an important class of TCMs that have been widely used in clinical practice and recognized as safe long-term treatments for chronic diseases. However, as an important class of TCMs, the panorama of pharmacological effects and the mechanisms of MB-TCMs have not been systemically reviewed. Compelling studies have shown that gut microbiota can mediate the therapeutic activity of TCMs and help to elucidate the core principles of TCM medicinal theory. In this systematic review, we found that MB-TCMs commonly participated in the modulation of metabolic syndrome, intestinal inflammation, nervous system disease and cardiovascular system disease in association with promoting the growth of beneficial bacteria Bacteroides, Akkermansia, Lactobacillus, Bifidobacterium, Roseburia as well as inhibiting the proliferation of harmful bacteria Helicobacter, Enterococcus, Desulfovibrio and Escherichia-Shigella. These alterations, correspondingly, enhance the generation of protective metabolites, mainly including short-chain fatty acids (SCFAs), bile acid (BAs), 5-hydroxytryptamine (5-HT), indole and gamma-aminobutyric acid (GABA), and inhibit the generation of harmful metabolites, such as proinflammatory factors trimethylamine oxide (TAMO) and lipopolysaccharide (LPS), to further exert multiplicative effects for the maintenance of human health through several different signaling pathways. Altogether, this present review has attempted to comprehensively summarize the relationship between MB-TCMs and gut microbiota by establishing the TCMs-gut microbiota-metabolite-signaling pathway-diseases axis, which may provide new insight into the study of TCM medicinal theories and their clinical applications.
Collapse
Affiliation(s)
- Wenquan Su
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yanan Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaohui Zhao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiale Cheng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuan Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Shengxian Wu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Chongming Wu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
40
|
Sparfel L, Ratodiarivony S, Boutet-Robinet E, Ellero-Simatos S, Jolivet-Gougeon A. Akkermansia muciniphila and Alcohol-Related Liver Diseases. A Systematic Review. Mol Nutr Food Res 2024; 68:e2300510. [PMID: 38059838 DOI: 10.1002/mnfr.202300510] [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: 07/18/2023] [Revised: 10/03/2023] [Indexed: 12/08/2023]
Abstract
SCOPE Akkermansia muciniphila (A. muciniphila) are Gram negative commensal bacteria, degrading mucin in the intestinal mucosa, modulating intestinal permeability and inflammation in the digestive tract, liver, and blood. Some components can promote the relative abundance of A. muciniphila in the gut microbiota, but lower levels of A. muciniphila are more commonly found in people with obesity, diabetes, metabolic syndromes, or inflammatory digestive diseases. Over-intake of ethanol can also induce a decrease of A. muciniphila, associated with dysregulation of microbial metabolite production, impaired intestinal permeability, induction of chronic inflammation, and production of cytokines. METHODS AND RESULTS Using a PRISMA search strategy, a review is performed on the bacteriological characteristics of A. muciniphila, the factors capable of modulating its relative abundance in the digestive tract and its probiotic use in alcohol-related liver diseases (alcoholic hepatitis, cirrhosis, hepatocellular carcinoma, hepatic transplantation, partial hepatectomy). CONCLUSION Several studies have shown that supplementation with A. muciniphila can improve ethanol-related hepatic pathologies, and highlight the interest in using this bacterial species as a probiotic.
Collapse
Affiliation(s)
- Lydie Sparfel
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, F-35000, France
| | - Sandy Ratodiarivony
- Univ Rennes, Bacterial Regulatory RNAs and Medicine (BRM), UMR_S 1230, Rennes, F-35000, France
| | - Elisa Boutet-Robinet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300, Toulouse, France
| | - Sandrine Ellero-Simatos
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300, Toulouse, France
| | - Anne Jolivet-Gougeon
- Univ Rennes, Bacterial Regulatory RNAs and Medicine (BRM), UMR_S 1230, Rennes, F-35000, France
- Teaching Hospital, CHU Rennes, 2 rue Henri Le Guilloux 35033, Rennes, F-35000, France
- INSERM, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer), U1241, INSERM 1241, Rennes, F-35000, France
| |
Collapse
|
41
|
Xie Q, Hu B. Effects of gut microbiota on prostatic cancer: a two-sample Mendelian randomization study. Front Microbiol 2023; 14:1250369. [PMID: 38029073 PMCID: PMC10659115 DOI: 10.3389/fmicb.2023.1250369] [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: 06/30/2023] [Accepted: 09/26/2023] [Indexed: 12/01/2023] Open
Abstract
Aim Recent observational and small-sample case-control studies have shown a relationship between gut microbiota composition and prostatic cancer (PCa). Nevertheless, the causal association between gut microbiota and PCa is still unclear. Herein, we used the Mendelian randomization (MR) method to explore the potential causal relationship between gut microbiota and PCa. Methods In this two-sample MR study, data were extracted from the summary statistics of gut microbiota from the largest available genome-wide association study meta-analysis conducted by the MiBioGen consortium (n = 14,306) and the Dutch Microbiome Project (n = 8,208). Summary statistics for PCa were obtained from the FinnGen consortium release data (n = 95,213). Inverse variance weighted (IVW), MR-Egger, strength test (F), and MR-PRESSO were used to examine the potential causal association between gut microbiota and PCa. Cochran's Q statistics were used to quantify the heterogeneity of instrumental variables. Results IVW estimates suggested that the relative abundance of Akkermansia muciniphila (odds ratio [OR] = 0.7926, 95% confidence interval [CI]: 0.6655-0.9440) and Bacteroides salyersiae (OR = 0.9023, 95% CI: 0.8262-0.9853) were negatively associated with the odds of PCa, while that of Eubacterium biforme (OR = 1.1629, 95% CI: 1.0110-1.3376) was positively associated with the odds of PCa. In addition, we explored these relationships among patients without other cancers and similarly found that the relative abundance of Akkermansia muciniphila, Bacteroides salyersiae, and Eubacterium biforme were linked to PCa (all P < 0.05). Conclusion Gut microbiota potentially influenced the occurrence of PCa. Our findings may provide some new ideas for researching the methods of PCa prevention. In addition, further studies are needed to explore the causal association and specific underlying mechanisms between gut microbiota and PCa.
Collapse
Affiliation(s)
| | - Bin Hu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| |
Collapse
|
42
|
Wang Y, Ji X, Zhao M, Li J, Yin H, Jin J, Zhao L. Modulation of tryptophan metabolism via AHR-IL22 pathway mediates the alleviation of DSS-induced colitis by chitooligosaccharides with different degrees of polymerization. Carbohydr Polym 2023; 319:121180. [PMID: 37567716 DOI: 10.1016/j.carbpol.2023.121180] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/27/2023] [Accepted: 07/06/2023] [Indexed: 08/13/2023]
Abstract
Oral administration of chitooligosaccharides (COS) has been reported to alleviate colitis in mice. However, the mechanism of action of COS with specific polymerization degree on gut inflammation and metabolism remains unclear. This study aimed to investigate the effects of chitobiose (COS2), chitotetraose (COS4), and chitohexaose (COS6) on colitis, and to elucidate their underlying mechanisms. COS2, COS4, and COS6 were able to significantly alleviate colonic injury and inflammation levels. COS6 has the best anti-inflammatory effect. Furthermore, COS6 could down-regulate the level of indoleamine-2,3-dioxygenase1 (IDO1) and restore the levels of indole, indoleacetic-3-acid (IAA), and indole-3-carbaldehyde (I3A) in the cecum of chronic colitis mice (p < 0.05), thereby regulating tryptophan metabolism. In the aromatic hydrocarbon receptor-IL-22 (AHR-IL-22) pathway, although there were differences between chronic colitis and acute colitis mice, COS intervention could restore the AHR-IL-22 pathway to normal, promote the expression of MUC2, and repair the intestinal mucosal barrier. In conclusion, the results of this study suggested that COS had a good inhibitory effect on IDO1 under inflammation and the changes of AHR and IL-22 levels at different stages of disease development. This provides new insights into the potential use of COS as a functional food for improving intestinal inflammation and metabolism.
Collapse
Affiliation(s)
- Yu Wang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaoguo Ji
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai 200237, China; Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai 200237, China
| | - Mengyao Zhao
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai 200237, China
| | - Juan Li
- Organ Transplant Center, Shanghai Changzheng Hospital, Shanghai 200003, China
| | - Hao Yin
- Organ Transplant Center, Shanghai Changzheng Hospital, Shanghai 200003, China
| | - Jiayang Jin
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai 200237, China.
| | - Liming Zhao
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; Organ Transplant Center, Shanghai Changzheng Hospital, Shanghai 200003, China; Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai 200237, China.
| |
Collapse
|
43
|
Hou JJ, Ma AH, Qin YH. Activation of the aryl hydrocarbon receptor in inflammatory bowel disease: insights from gut microbiota. Front Cell Infect Microbiol 2023; 13:1279172. [PMID: 37942478 PMCID: PMC10628454 DOI: 10.3389/fcimb.2023.1279172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory intestinal disease that affects more than 3.5 million people, with rising prevalence. It deeply affects patients' daily life, increasing the burden on patients, families, and society. Presently, the etiology of IBD remains incompletely clarified, while emerging evidence has demonstrated that altered gut microbiota and decreased aryl hydrocarbon receptor (AHR) activity are closely associated with IBD. Furthermore, microbial metabolites are capable of AHR activation as AHR ligands, while the AHR, in turn, affects the microbiota through various pathways. In light of the complex connection among gut microbiota, the AHR, and IBD, it is urgent to review the latest research progress in this field. In this review, we describe the role of gut microbiota and AHR activation in IBD and discussed the crosstalk between gut microbiota and the AHR in the context of IBD. Taken as a whole, we propose new therapeutic strategies targeting the AHR-microbiota axis for IBD, even for other related diseases caused by AHR-microbiota dysbiosis.
Collapse
Affiliation(s)
| | | | - Yue-Hua Qin
- Department of Gastroenterology, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
44
|
Xiang Q, Yan X, Shi W, Li H, Zhou K. Early gut microbiota intervention in premature infants: Application perspectives. J Adv Res 2023; 51:59-72. [PMID: 36372205 PMCID: PMC10491976 DOI: 10.1016/j.jare.2022.11.004] [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: 08/08/2022] [Revised: 10/30/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Preterm birth is the leading cause of death in children under the age of five. One of the major factors contributing to the high risk of diseases and deaths in premature infants is the incomplete development of the intestinal immune system. The gut microbiota has been widely recognized as a critical factor in promoting the development and function of the intestinal immune system after birth. However, the gut microbiota of premature infants is at high risk of dysbiosis, which is highly associated with adverse effects on the development and education of the early life immune system. Early intervention can modulate the colonization and development of gut microbiota and has a long-term influence on the development of the intestinal immune system. AIM OF REVIEW This review aims to summarize the characterization, interconnection, and underlying mechanism of gut microbiota and intestinal innate immunity in premature infants, and to discuss the status, applicability, safety, and prospects of different intervention strategies in premature infants, thus providing an overview and outlook of the current applications and remaining gaps of early intervention strategies in premature infants. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three key concepts. Firstly, the gut microbiota of premature infants is at high risk of dysbiosis, resulting in dysfunctional intestinal immune system processes. Secondly, contributing roles of early intervention have been observed in improving the intestinal environment and promoting gut microbiota colonization, which is significant in the development and function of gut immunity in premature infants. Thirdly, different strategies of early intervention, such as probiotics, fecal microbiota transplantation, and nutrients, show different safety, applicability, and outcome in premature infants, and the underlying mechanism is complex and poorly understood.
Collapse
Affiliation(s)
- Quanhang Xiang
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Xudong Yan
- Department of Neonatal Intensive Care Unit, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Wei Shi
- Department of Obstetrics and Gynecology, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Huiping Li
- Department of Respiratory and Critical Care Medicine, the first affiliated hospital of Southern University of Science and Technology of China, Shenzhen People's Hospital, Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
45
|
Xue C, Li G, Zheng Q, Gu X, Shi Q, Su Y, Chu Q, Yuan X, Bao Z, Lu J, Li L. Tryptophan metabolism in health and disease. Cell Metab 2023; 35:1304-1326. [PMID: 37352864 DOI: 10.1016/j.cmet.2023.06.004] [Citation(s) in RCA: 181] [Impact Index Per Article: 90.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/10/2023] [Accepted: 06/05/2023] [Indexed: 06/25/2023]
Abstract
Tryptophan (Trp) metabolism primarily involves the kynurenine, 5-hydroxytryptamine, and indole pathways. A variety of bioactive compounds produced via Trp metabolism can regulate various physiological functions, including inflammation, metabolism, immune responses, and neurological function. Emerging evidence supports an intimate relationship between Trp metabolism disorder and diseases. The levels or ratios of Trp metabolites are significantly associated with many clinical features. Additionally, studies have shown that disease progression can be controlled by modulating Trp metabolism. Indoleamine-2,3-dioxygenase, Trp-2,3-dioxygenase, kynurenine-3-monooxygenase, and Trp hydroxylase are the rate-limiting enzymes that are critical for Trp metabolism. These key regulatory enzymes can be targeted for treating several diseases, including tumors. These findings provide novel insights into the treatment of diseases. In this review, we have summarized the recent research progress on the role of Trp metabolites in health and disease along with their clinical applications.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ganglei Li
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qiuxian Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xinyu Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuanshuai Su
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhengyi Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
46
|
Grant ET, Boudaud M, Muller A, Macpherson AJ, Desai MS. Maternal diet and gut microbiome composition modulate early-life immune development. EMBO Mol Med 2023; 15:e17241. [PMID: 37278126 PMCID: PMC10405054 DOI: 10.15252/emmm.202217241] [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: 11/29/2022] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 06/07/2023] Open
Abstract
In early life, the intestinal mucosa and immune system undergo a critical developmental process to contain the expanding gut microbiome while promoting tolerance toward commensals, yet the influence of maternal diet and microbial composition on offspring immune maturation remains poorly understood. We colonized germ-free mice with a consortium of 14 strains, fed them a standard fiber-rich chow or a fiber-free diet, and then longitudinally assessed offspring development during the weaning period. Unlike pups born to dams fed the fiber-rich diet, pups of fiber-deprived dams demonstrated delayed colonization with Akkermansia muciniphila, a mucin-foraging bacterium that can also use milk oligosaccharides. The pups of fiber-deprived dams exhibited an enrichment of colonic transcripts corresponding to defense response pathways and a peak in Il22 expression at weaning. Removal of A. muciniphila from the community, but maintenance on the fiber-rich diet, was associated with reduced proportions of RORγt-positive innate and adaptive immune cell subsets. Our results highlight the potent influence of maternal dietary fiber intake and discrete changes in microbial composition on the postnatal microbiome assemblage and early immune development.
Collapse
Affiliation(s)
- Erica T Grant
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
- Faculty of Science, Technology and MedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Marie Boudaud
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| | - Arnaud Muller
- Bioinformatics Platform, Data Integration and AnalysisLuxembourg Institute of HealthStrassenLuxembourg
- LuxGen, Translational Medicine Operation HubLuxembourg Institute of HealthDudelangeLuxembourg
| | - Andrew J Macpherson
- Department of Visceral Surgery and MedicineBern University Hospital, University of BernBernSwitzerland
| | - Mahesh S Desai
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| |
Collapse
|
47
|
Huang C, Hao W, Wang X, Zhou R, Lin Q. Probiotics for the treatment of ulcerative colitis: a review of experimental research from 2018 to 2022. Front Microbiol 2023; 14:1211271. [PMID: 37485519 PMCID: PMC10358780 DOI: 10.3389/fmicb.2023.1211271] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
Ulcerative colitis (UC) has become a worldwide public health problem, and the prevalence of the disease among children has been increasing. The pathogenesis of UC has not been elucidated, but dysbiosis of the gut microbiota is considered the main cause of chronic intestinal inflammation. This review focuses on the therapeutic effects of probiotics on UC and the potential mechanisms involved. In animal studies, probiotics have been shown to alleviate symptoms of UC, including weight loss, diarrhea, blood in the stool, and a shortened colon length, while also restoring intestinal microecological homeostasis, improving gut barrier function, modulating the intestinal immune response, and attenuating intestinal inflammation, thereby providing theoretical support for the development of probiotic-based microbial products as an adjunctive therapy for UC. However, the efficacy of probiotics is influenced by factors such as the bacterial strain, dose, and form. Hence, the mechanisms of action need to be investigated further. Relevant clinical trials are currently lacking, so the extension of animal experimental findings to clinical application requires a longer period of consideration for validation.
Collapse
Affiliation(s)
- Cuilan Huang
- Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi Children’s Hospital, Wuxi, China
| | - Wujuan Hao
- Department of Digestive, Affiliated Children’s Hospital of Jiangnan University, Wuxi, China
| | - Xuyang Wang
- Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi Children’s Hospital, Wuxi, China
| | - Renmin Zhou
- Department of Digestive, Affiliated Children’s Hospital of Jiangnan University, Wuxi, China
| | - Qiong Lin
- Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi Children’s Hospital, Wuxi, China
| |
Collapse
|
48
|
Zhang F, Wang D. Potential of Akkermansia muciniphila and its outer membrane proteins as therapeutic targets for neuropsychological diseases. Front Microbiol 2023; 14:1191445. [PMID: 37440890 PMCID: PMC10333588 DOI: 10.3389/fmicb.2023.1191445] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/25/2023] [Indexed: 07/15/2023] Open
Abstract
The gut microbiota varies dramatically among individuals, and changes over time within the same individual, due to diversities in genetic backgrounds, diet, nutrient supplementations and use of antibiotics. Up until now, studies on dysbiosis of microbiota have expanded to a wider range of diseases, with Akkermansia muciniphila at the cross spot of many of these diseases. A. muciniphila is a Gram-negative bacterium that produces short-chain fatty acids (SCFAs), and Amuc_1100 is one of its most highly expressed outer membrane proteins. This review aims to summarize current knowledge on correlations between A. muciniphila and involved neuropsychological diseases published in the last decade, with a focus on the potential of this bacterium and its outer membrane proteins as therapeutic targets for these diseases, on the basis of evidence accumulated from animal and clinical studies, as well as mechanisms of action from peripheral to central nervous system (CNS).
Collapse
Affiliation(s)
- Fenghua Zhang
- Department of Laboratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Dali Wang
- Center for Clinical and Translational Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
49
|
Wang S, van Schooten FJ, Jin H, Jonkers D, Godschalk R. The Involvement of Intestinal Tryptophan Metabolism in Inflammatory Bowel Disease Identified by a Meta-Analysis of the Transcriptome and a Systematic Review of the Metabolome. Nutrients 2023; 15:2886. [PMID: 37447212 DOI: 10.3390/nu15132886] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Evidence is emerging for the role of intestinal tryptophan metabolism in the development of inflammatory bowel disease (IBD). In order to identify the role of altered intestinal tryptophan metabolism in IBD pathogenesis, a meta-analysis of the transcriptome was performed to identify differentially expressed genes involved in the tryptophan metabolism pathways in intestinal biopsies of IBD as compared to non-IBD controls. Moreover, a systematic review of the metabolome was performed to identify the concurrent changes in tryptophan metabolites. Integration of the transcriptome and metabolome identified various alterations in intestinal tryptophan metabolism during active disease in IBD patients, including decreased intestinal tryptophan absorption, enhanced kynurenine pathway, increased interstitial serotonin availability, changed indole pathway, and activated aryl hydrocarbon receptor signaling. Therefore, a network of intestinal tryptophan metabolism pathways in IBD could be established, helping to assess the potential of genes and metabolites involved in these pathways as diagnostic markers and targets for IBD management.
Collapse
Affiliation(s)
- Shan Wang
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Frederik-Jan van Schooten
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Han Jin
- Department of Pathology, School for Cardiovascular Diseases (CARIM), Maastricht University Medical Center+ (MUMC+), 6200 MD Maastricht, The Netherlands
| | - Daisy Jonkers
- Department of Internal Medicine, Division Gastroenterology-Hepatology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Roger Godschalk
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
50
|
Liao J, Cao Y, Zhao J, Yu B, Wang Y, Li W, Li H, Lv S, Wen W, Cui H, Chen Y. Aqueous extract of Polygala japonica Houtt. ameliorated nonalcoholic steatohepatitis in mice through restoring the gut microbiota disorders and affecting the metabolites in feces and liver. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154937. [PMID: 37393831 DOI: 10.1016/j.phymed.2023.154937] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/23/2023] [Accepted: 06/19/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Polygala japonica Houtt. (PJ) has been demonstrated with several biological potentials such as lipid-lowering and anti-inflammatory effects. However, the effects and mechanisms of PJ on nonalcoholic steatohepatitis (NASH) remain unclear. PURPOSE The aim of this study was to evaluate the effects of PJ on NASH and illustrate the mechanism based on modulating gut microbiota and host metabolism. MATERIALS AND METHODS NASH mouse model was induced using methionine and choline deficient (MCD) diet and orally treated with PJ. The therapeutic, anti-inflammatory, and anti-oxidative effects of PJ on mice with NASH were firstly assessed. Then, the gut microbiota of mice was analyzed using 16S rRNA sequencing to assess the changes. Finally, the effects of PJ on the metabolites in liver and feces were explored by untargeted metabolomics. RESULTS The results indicated that PJ could improve hepatic steatosis, liver injury, inflammatory response, and oxidative stress in NASH mice. PJ treatment also affected the diversity of gut microbiota and changed the relative abundances of Faecalibaculum. Lactobacillus, Muribaculaceae, Dubosiella, Akkermansia, Lachnospiraceae_NK4A136_group, and Turicibacter in NASH mice. In addition, PJ treatment modulated 59 metabolites both in liver and feces. Metabolites involved in histidine, and tryptophan metabolism pathways were identified as the key metabolites according to the correlation analysis between differential gut microbiota and metabolites. CONCLUSION Our study demonstrated the therapeutic, anti-inflammatory and anti-oxidative potentials of PJ on NASH. The mechanisms of PJ treatment were related to the improvement of gut microbiota dysbiosis and the regulation of histidine and tryptophan metabolism.
Collapse
Affiliation(s)
- Jiabao Liao
- Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang, China
| | - Yongjun Cao
- Nanjing University of Chinese Medicine, Jiangsu, China
| | - Jie Zhao
- Yunnan Provincial Hospital of Traditional Chinese Medicine, Yunnan, China
| | - Bolun Yu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuming Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenting Li
- Yunnan University of Traditional Chinese Medicine, Yunnan, China
| | - Hanzhou Li
- Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Shuquan Lv
- Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Weibo Wen
- Nanjing University of Chinese Medicine, Jiangsu, China; Yunnan University of Traditional Chinese Medicine, Yunnan, China.
| | - Huantian Cui
- Yunnan University of Traditional Chinese Medicine, Yunnan, China; Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Shandong, China.
| | - Yao Chen
- Yunnan Provincial Hospital of Traditional Chinese Medicine, Yunnan, China.
| |
Collapse
|