1
|
Gao H, Sun M, Li A, Gu Q, Kang D, Feng Z, Li X, Wang X, Chen L, Yang H, Cong Y, Liu Z. Microbiota-derived IPA alleviates intestinal mucosal inflammation through upregulating Th1/Th17 cell apoptosis in inflammatory bowel disease. Gut Microbes 2025; 17:2467235. [PMID: 39956891 PMCID: PMC11834480 DOI: 10.1080/19490976.2025.2467235] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/09/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025] Open
Abstract
The gut microbiota-derived metabolite indole-3-propionic acid (IPA) plays an important role in maintaining intestinal mucosal homeostasis, while the molecular mechanisms underlying IPA regulation on mucosal CD4+ T cell functions in inflammatory bowel disease (IBD) remain elusive. Here we investigated the roles of IPA in modulating mucosal CD4+ T cells and its therapeutic potential in treatment of human IBD. Leveraging metabolomics and microbial community analyses, we observed that the levels of IPA-producing microbiota (e.g. Peptostreptococcus, Clostridium, and Fournierella) and IPA were decreased, while the IPA-consuming microbiota (e.g. Parabacteroides, Erysipelatoclostridium, and Lachnoclostridium) were increased in the feces of IBD patients than those in healthy donors. Dextran sulfate sodium (DSS)-induced acute colitis and CD45RBhighCD4+ T cell transfer-induced chronic colitis models were then established in mice and treated orally with IPA to study its role in intestinal mucosal inflammation in vivo. We found that oral administration of IPA attenuated mucosal inflammation in both acute and chronic colitis models in mice, as characterized by increased body weight, and reduced levels of pro-inflammatory cytokines (e.g. TNF-α, IFN-γ, and IL-17A) and histological scores in the colon. We further utilized RNA sequencing, molecular docking simulations, and surface plasmon resonance analyses and identified that IPA exerts its biological effects by interacting with heat shock protein 70 (HSP70), leading to inducing Th1/Th17 cell apoptosis. Consistently, ectopic expression of HSP70 in CD4+ T cells conferred resistance to IPA-induced Th1/Th17 cell apoptosis. Therefore, these findings identify a previously unrecognized pathway by which IPA modulates intestinal inflammation and provide a promising avenue for the treatment of IBD.
Collapse
Affiliation(s)
- Han Gao
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Mingming Sun
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Ai Li
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Qiaoyan Gu
- Department of Gastroenterology, Yanan University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dengfeng Kang
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Zhongsheng Feng
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Xiaoyu Li
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Xuehong Wang
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liang Chen
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Hong Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yingzi Cong
- Division of Gastroenterology and Hepatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Human Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Zhanju Liu
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| |
Collapse
|
2
|
Tan X, Gao B, Xu Y, Zhao Q, Jiang J, Sun D, Zhang Y, Zhou S, Fan JB, Zhang M, Zhao K. Atractylodes macrocephala-derived extracellular vesicles-like particles enhance the recovery of ulcerative colitis by remodeling intestinal microecological balance. J Nanobiotechnology 2025; 23:433. [PMID: 40490795 DOI: 10.1186/s12951-025-03506-8] [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: 03/05/2025] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
Current treatment of ulcerative colitis (UC) remains challenging, with the mainstay of therapy being 5-aminosalicylic acid-based drugs, which have limited and inconsistent results. Atractylodes macrocephala (AM) is a traditional Chinese medicine commonly used in the clinical treatment of various inflammatory diseases. Herein, we demonstrate that AM-derived extracellular vesicle-like particles (AMEVLP) can effectively modulate the gut microbiota, thereby significantly improving the treatment efficiency of UC. This is achieved by enhancing the alpha diversity of the gut microbiota and re-establishing beneficial types, which in turn alter tryptophan metabolism, leading to an increase in indole derivatives within the gut. This process also protects the gut barrier and exerts anti-inflammatory effects. The mechanism behind these anti-inflammatory effects is closely associated with the Th17 cell differentiation signaling pathway. It is believed that the AMEVLP enable them to efficiently remodel gut microbiota, providing an avenue for the treatment of various inflammatory diseases. Significantly, preliminary clinical trials have shown that AMEVLP can substantially slow the progression of the disease in UC patients.
Collapse
Affiliation(s)
- Xuejun Tan
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, The Third Affiliated Hospital of Guangzhou, University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378, China
- Guangdong Engineering Research Center of Chinese Herbal Vesicles, Guangzhou University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378, China
| | - Bowen Gao
- School of Basic Medical Sciences, Xi'an Jiaotong University, Shaanxi, 710061, Xi'an, China
| | - Yukun Xu
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378, China
| | - Qing Zhao
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, The Third Affiliated Hospital of Guangzhou, University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378, China
- Guangdong Engineering Research Center of Chinese Herbal Vesicles, Guangzhou University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378, China
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378, China
| | - Jiazan Jiang
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, The Third Affiliated Hospital of Guangzhou, University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378, China
| | - Dexuan Sun
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, The Third Affiliated Hospital of Guangzhou, University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378, China
| | - Yirong Zhang
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, The Third Affiliated Hospital of Guangzhou, University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378, China
| | - Sirui Zhou
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378, China
| | - Jun-Bing Fan
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China, 510515.
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Shaanxi, 710061, Xi'an, China.
| | - Kewei Zhao
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, The Third Affiliated Hospital of Guangzhou, University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378, China.
- Guangdong Engineering Research Center of Chinese Herbal Vesicles, Guangzhou University of Chinese Medicine, No.261 and 263, Longxi Avenue, Guangzhou, 510378, China.
| |
Collapse
|
3
|
Gao Z, Yu X, Su W, Huang P, Li Z, Lin Y, Chen LL, Cao Y, Liu Y, Chen J, Yang D, Cao G. Atractylenolide-1 Alleviates Ulcerative Colitis via Restraining RhoA/ROCK/MLC Pathway-Mediated Intestinal Barrier Dysfunction. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12690-12701. [PMID: 40364748 DOI: 10.1021/acs.jafc.4c11976] [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: 05/15/2025]
Abstract
Using Atractylenolide-1 (AT-1) is a confident strategy for the treatment of ulcerative colitis (UC) due to its natural origin and notable pharmacological activity. The study investigated the therapeutic effect of AT-1 in dextran sodium sulfate (DSS)-induced mice and Caco-2 cells while also exploring the underlying molecular mechanisms. In this study, AT-1 treatment could reduce weight loss and colon shortening and significantly reduce disease activity index (DAI), spleen index, and histopathological scores in UC mice. And AT-1 was observed to restore cell necrosis and monolayer damage and restored F-actin-mediated tight junction (TJ) protein redistribution to alleviate mucosal injury in UC mice and Caco-2 cells. Moreover, AT-1 regulated alanine, aspartic acid, and glutamate metabolism; increased the content of related metabolites; and promoted cell proliferation to restore damaged mucous membranes in UC mice. The results of molecular docking and molecular dynamics simulation showed that the binding of AT-1 to RhoA had a stable conformation, and it was speculated that RhoA was the main target of AT-1. Further investigations revealed that the interference of RhoA disrupted the regulatory pathway of AT-1. Thus, AT-1 could inhibit the reduction of TJ proteins, alter DSS-mediated cytoskeletal migration, promote amino acid metabolism, and subsequently reduce the permeability of the colon epithelium, thereby restoring mucosal barrier dysfunction features.
Collapse
Affiliation(s)
- Zengxiang Gao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Xuecheng Yu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Wenlong Su
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Peng Huang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Zhenhui Li
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Yunya Lin
- Hubei Shizhen Laboratory,Wuhan 430065, People's Republic of China
| | - Lin-Lin Chen
- Hubei Shizhen Laboratory,Wuhan 430065, People's Republic of China
- Key Laboratory of Traditional Chinese Medicine Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine,Wuhan 430065, People's Republic of China
| | - Yan Cao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
- Hubei Shizhen Laboratory,Wuhan 430065, People's Republic of China
| | - Yanju Liu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
- Hubei Shizhen Laboratory,Wuhan 430065, People's Republic of China
| | - Jianbei Chen
- Hubei Shizhen Laboratory,Wuhan 430065, People's Republic of China
| | - Desen Yang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
- Hubei Shizhen Laboratory,Wuhan 430065, People's Republic of China
| | - Guosheng Cao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
- Hubei Shizhen Laboratory,Wuhan 430065, People's Republic of China
| |
Collapse
|
4
|
Liao Z, Liu X, Li L, Li S, Xing X, Zheng X, Song W, Gui P, Liu Q, Rong G, Shao Y, Zou M, Liao H, Wu X. Mechanism of the Proprietary Chinese Medicine "JiuLiWan" to Treat Ulcerative Colitis Revealed by Network Pharmacology, Molecular Docking, and Experimental Verification In Vitro. ACS OMEGA 2025; 10:19598-19613. [PMID: 40415848 PMCID: PMC12096223 DOI: 10.1021/acsomega.5c00261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/14/2025] [Accepted: 03/25/2025] [Indexed: 05/27/2025]
Abstract
JiuLiWan (JLW), as a classic traditional Chinese medicine formula, has been clinically used against ulcerative colitis (UC). However, the exact mechanism of its therapeutic effect remains unclear. This study aims to explore and validate the main components and pharmacological mechanism of JLW in the treatment of UC through network pharmacology, molecular docking, and cell experiments. Network pharmacology analyses indicated a total of 107 main components and 286 core targets of JLW against UC. Pathway enrichment analysis demonstrated the involvement of PI3K-AKT, MAPK, Ras, Rap1, TNF, T cell receptor, HIF-1, C-type lectin receptor, VEGF, and Th17 cell differentiation signal pathways in the efficacy of the formula. The molecular docking results indicated that the prominent components (ailanthone (AIL), butylidenephthalide, honokiol, dehydrocostuslactone, ganoderic acid A, atractylenolide I, neokurarinol, glycyrrhetinic acid, palmatine, tangeretin, and bruceine A) could bind to core targets AKT1, P53, STAT3, c-JUN, and ERK1. Subsequently, AIL was used as a representative compound to conduct cell experiments to verify its role and mechanism in anti-inflammation and immunomodulation. Interestingly, AIL could switch Jurkat T cells into a quiescence state without activating the inflammatory and immune status. However, AIL could significantly decrease the levels of interleukin-2 (IL-2) and interferon-gamma (IFN-γ), as well as the expression of surface activation markers CD69 and CD25, in PMA/ionomycin-activated Jurkat T cells by suppressing the RAF/ERK/STAT3 signaling pathway and increasing the phosphorylation of p53. This study combines network pharmacology prediction with experimental verification in vitro to demonstrate the mechanism of JLW in treating UC and provides an effective, safe, and inexpensive strategy for UC treatment.
Collapse
Affiliation(s)
- Zhifang Liao
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Xiao Liu
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Linxuan Li
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
- The Key Laboratory
of Sepsis Translational Medicine, The Second
Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
- Interdisciplinary
Science Research Center of Western Guangdong, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
| | - Sikai Li
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Xingxing Xing
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Xiwen Zheng
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Wenyu Song
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Pin Gui
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Qi Liu
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Guanghong Rong
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Yiming Shao
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
- The Key Laboratory
of Sepsis Translational Medicine, The Second
Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
| | - Mingzhi Zou
- The Key Laboratory
of Sepsis Translational Medicine, The Second
Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
- Interdisciplinary
Science Research Center of Western Guangdong, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
| | - Hongbo Liao
- Guangdong
Provincial Key Laboratory of Research and Development of Natural Drugs,
School of Pharmacy, Guangdong Medical University, Zhanjiang, Guangdong Province524023, P.R. of
China
| | - Xin Wu
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
- The Key Laboratory
of Sepsis Translational Medicine, The Second
Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
| |
Collapse
|
5
|
Liu Y, Ren Q, Qin H, Huang M, Mao J, Xi B, Zhang S. Comparative study on the anti-alcoholic liver disease efficiency of the ethanol- and water-soluble polysaccharides from Baijiu vinasses. Int J Biol Macromol 2025; 299:140087. [PMID: 39842569 DOI: 10.1016/j.ijbiomac.2025.140087] [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: 09/20/2024] [Revised: 01/10/2025] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
Ethanol- and water-soluble polysaccharides were extracted from Baijiu vinasses (EP and WP), respectively. EP was dominantly composed by arabinose, glucose and xylose with molar ratio of 8.81: 76.82: 6.9. While, WP was dominantly composed by galactose, glucose and mannose with molar ratio of 8.32: 56.05: 25.19. The molecular weights and reducing sugar contents in EP and WP were 6.2 kDa vs. 16.1 kDa and 24.52 ± 0.97 % vs. 19.77 ± 0.75 %, respectively. Alterations in activation of the Nrf2/HO-1 signalling pathway and increases in the abundance of Lachnospiraceae and Akkermansia and their associated metabolisms could be the general mechanism by which Baijiu vinasses (BV) polysaccharides alleviated alcohol-induced liver disease (ALD) in mice. Due to the different physicochemical characteristics, the ALD alleviation efficiency was different. EP exhibited higher efficiency in oxidative stress suppressing and lipid alternation by activating the peroxisome proliferators-activated receptors (PPAR) signalling pathway. WP exhibited higher efficiency in liver damage repairing with the controlment in tryptophan metabolism pathway. This study exhibited the potential biofunction of BV polysaccharides in ALD alleviation and could promote the BV upcycling.
Collapse
Affiliation(s)
- Yizhou Liu
- Luzhou Laojiao Co, Ltd, Luzhou, Sichuan 646000, China; National Engineering Research Center of Solid-State Brewing, Luzhou, Sichuan 646000, China; School of Environmental Science and Engineering, Tianjin University, Tianjin 300350, China
| | - Qingxi Ren
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hui Qin
- Luzhou Laojiao Co, Ltd, Luzhou, Sichuan 646000, China; National Engineering Research Center of Solid-State Brewing, Luzhou, Sichuan 646000, China
| | - Mengyang Huang
- Luzhou Laojiao Co, Ltd, Luzhou, Sichuan 646000, China; National Engineering Research Center of Solid-State Brewing, Luzhou, Sichuan 646000, China
| | - Jian Mao
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| | - Beidou Xi
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300350, China; State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China.
| | - Suyi Zhang
- Luzhou Laojiao Co, Ltd, Luzhou, Sichuan 646000, China; National Engineering Research Center of Solid-State Brewing, Luzhou, Sichuan 646000, China.
| |
Collapse
|
6
|
Yuan M, Wan Y, Wang Y, Li S, Tang J, Liang X, Tan X, Yi S, Wei X, Li X, Guo L, Guo Y. Ursodeoxycholic acid grafted chitosan oligosaccharide self-assembled micelles with enhanced oral absorption and antidiabetic effect of oleanolic acid. Food Chem 2025; 470:142708. [PMID: 39752745 DOI: 10.1016/j.foodchem.2024.142708] [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: 07/18/2024] [Revised: 11/26/2024] [Accepted: 12/28/2024] [Indexed: 01/29/2025]
Abstract
Oleanolic acid (OA) is a food-derived bioactive component with antidiabetic activity, but its water solubility and oral bioavailability are notably restricted. In this study, to overcome these limitations, ursodeoxycholic acid-modified chitosan oligosaccharide (UCOS) was synthesized to encapsulate OA in self-assembled nanomicelles (UCOS-OA). The encapsulation efficiency and drug loading of UCOS-OA were 86 % and 11 %, respectively. UCOS-OA exhibited enhanced gastrointestinal stability and prolonged intestinal retention time when compared with free OA, resulting in a 10.6-fold increase in oral bioavailability. The enhanced antidiabetic activity of UCOS-OA was confirmed in the type 2 diabetes mellitus mice model, as it significantly improved glycolipid metabolism disorders and mitigated liver injury. Furthermore, UCOS-OA ameliorated the dysbiosis of gut microbiota and fecal metabolites. In conclusion, UCOS serves as an effective polymeric carrier for encapsulating OA, thereby improving its bioavailability and antidiabetic activity. This work provides valuable insights for the advancement of oral delivery systems for bioactive compounds.
Collapse
Affiliation(s)
- Minghao Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Yan Wan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Yulu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Sihui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Jiamei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Xue Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Xin Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Sirui Yi
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Xiaohang Wei
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Xiaohong Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Yiping Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| |
Collapse
|
7
|
Luo J, Yang Q, Jiang W, Liu Y, Hu Q, Peng X. The interaction between Angelica sinensis polysaccharide ASP-2pb and specific gut bacteria alleviates rheumatoid arthritis in rats. Int J Biol Macromol 2025; 301:140473. [PMID: 39889994 DOI: 10.1016/j.ijbiomac.2025.140473] [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: 10/18/2024] [Revised: 01/07/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Angelica sinensis polysaccharide (ASP) alleviated Rheumatoid arthritis (RA), but whether the relief was attributed to ASP itself or its microbial metabolites remained unclear. We characterized the main fraction of ASP (ASP-2pb) as a polysaccharide with molecular weight of 92.02 kDa. It contained approximately 48 repeating units of →6)-β-D-Galp-(1 → 3)-4-OMe-β-D-Galp-(1 → 4)-α-D-GalpA-(1 → 6)-β-D-Galp-(1 → 3)-4-OMe-β-D-Galp-(1→3)-β-D-Galp-(1 → 3)-β-D-Galp-(1 → 3)-β-D-Galp-(1 → with branches of Araf and Galp. Using ASP-2pb as intervention, the symptoms of RA in rats including joint swelling and inflammation were alleviated. Pseudo-germ-free animal test confirmed the necessity of specific gut bacteria during this alleviation. Bacteria such as Candidatus_Saccharimonas, Lactobacillus, Bifidobacterium, Faecalibaculum, Parvibacter, Ruminococcus_torques_group, Fournierella and Alloprevotella ought to be the key bacteria. Metabolites generated by these gut bacteria such as myristoleic acid, cuminaldehyde, 4-deoxypyridoxine and galactosylhydroxylysine, should be the key to RA remission. Therefore, specific metabolites were the consequence of the interaction between ASP-2pb and specific intestinal bacteria, and were responsible for the RA improvement.
Collapse
Affiliation(s)
- Jianming Luo
- Department of Food Science and Engineering, Jinan University, 601 Huangpu Avenue, Guangzhou, Guangdong 510632, China.
| | - Qianyi Yang
- Department of Food Science and Engineering, Jinan University, 601 Huangpu Avenue, Guangzhou, Guangdong 510632, China
| | - Wenwen Jiang
- Department of Food Science and Engineering, Jinan University, 601 Huangpu Avenue, Guangzhou, Guangdong 510632, China
| | - Yanghanxiu Liu
- Department of Food Science and Engineering, Jinan University, 601 Huangpu Avenue, Guangzhou, Guangdong 510632, China
| | - Qing Hu
- Department of Food Science and Engineering, Jinan University, 601 Huangpu Avenue, Guangzhou, Guangdong 510632, China
| | - Xichun Peng
- Department of Food Science and Engineering, Jinan University, 601 Huangpu Avenue, Guangzhou, Guangdong 510632, China.
| |
Collapse
|
8
|
Liao W, Cao L, Jiang X, Che L, Fang Z, Xu S, Lin Y, Zhuo Y, Hua L, Li J, Liu G, Sun M, Wu D, Wang H, Feng B. Intestinal overexpression of Pla2g10 alters the composition, diversity and function of gut microbiota in mice. Front Cell Infect Microbiol 2025; 15:1535204. [PMID: 40160470 PMCID: PMC11949945 DOI: 10.3389/fcimb.2025.1535204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
The intestinal microbiota is important for the health of the host and recent studies have shown that some genes of the host regulated the composition of the intestinal microbiota. Group 10 phospholipase A2 (PLA2G10) is a member of the lipolytic enzyme family PLA2, which hydrolyze the ester bond at the sn-2 position of phospholipids to produce free fatty acids and lysophospholipids. PLA2G10 is secreted into the intestinal lumen, but its impact on the gut microbiota remains unclear. In this study, we generated intestine-specific Pla2g10 knock-in mice, and used 16S RNA sequencing to compare their gut microbiota with that of their wild-type (WT) littermates. Results showed that gut-specific Pla2g10 knock-in induced both PLA2G10 mRNA and protein levels in the colon. Moreover, intestinal Pla2g10 overexpression reduced the α-diversity of the gut microbiota relative to that of WT mice. The abundance of Bacteroidetes was lower in the Pla2g10 knock-in mice than that in the control mice, while the ratio of Firmicutes/Bacteroidetes was higher. Furthermore, the abundance of the genus Allobaculum was reduced, whereas the abundance of beneficial bacteria genera, including Enterorhabdus, Dubosiella, and Lactobacillus, was increased by host intestinal Pla2g10 overexpression. In summary, intestinal Pla2g10 overexpression increased the proportions of beneficial bacterial in the colonic chyme of mice, providing a potential therapeutic target for future improvement of the gut microbiota.
Collapse
Affiliation(s)
- Wenhao Liao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lei Cao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xuemei Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lianqiang Che
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhengfeng Fang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Shengyu Xu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yan Lin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yong Zhuo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lun Hua
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jian Li
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Guangmang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mengmeng Sun
- College of Science, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - De Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hairui Wang
- Chengdu Research Base of Giant Panda Breeding, Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu, China
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Park C, Kim M, Park JW, Kim J, Bu Y, Ko SJ. Effect of Bojanggunbi-tang and its primary constituent herbs on the gastrointestinal tract: a scoping review. Front Pharmacol 2025; 16:1543194. [PMID: 40144663 PMCID: PMC11938064 DOI: 10.3389/fphar.2025.1543194] [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: 12/11/2024] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
Background Bojanggunbi-tang (BGT), a herbal prescription used in traditional Korean medicine, has been used to treat various gastrointestinal (GI) diseases. Methods Studies on BGT published until May 2024 were retrieved from the electronic databases of Medline, CENTRAL, Embase, AMED, CNKI, CiNii, Kmbase, KISS, NDSL, and OASIS using GI-related terms. All study types, regardless of the research method or language, were eligible for inclusion. Additional articles on Lonicera japonica, Atractylodes macrocephala, and Alisma canaliculatum, which are key components of BGT, were retrieved from the databases of Medline, CENTRAL, Embase, and Web of Science using GI-specific terms. The basic information, research models, administration methods, evaluation methods, and treatment outcomes of the selected studies were examined subsequently. Results Fourteen studies, comprising nine animal studies, one cell-based study, and four human studies, were included in the final analysis. BGT was found to exhibit anti-inflammatory effects, promote restoration of the gastrointestinal mucosa, and regulate GI motility. Analysis of the key herbal components L. japonica, A. macrocephala, and A. canaliculatum revealed that they inhibit inflammatory cytokines and oxidative substances, regulate serotonin and cholinergic pathways, and modulate intestinal microbiota. Conclusion This scoping review confirmed the therapeutic potential and mechanisms of action of BGT and its main components, L. japonica, A. macrocephala, and A. canaliculatum, thereby indicating its ability to enhance GI health. Further studies, including randomized clinical trials, must be conducted in the future to confirm these findings. Scoping review registration The study was registered in OSF, an international scoping review database: https://doi.org/10.17605/OSF.IO/ATU4S.
Collapse
Affiliation(s)
- Chaehyun Park
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Korean Internal Medicine, Kyung Hee University College of Korean Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Minjeong Kim
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Korean Internal Medicine, Kyung Hee University College of Korean Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Jae-Woo Park
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Korean Internal Medicine, Kyung Hee University College of Korean Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
- Department of Digestive Diseases, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jinsung Kim
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Digestive Diseases, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Division of Digestive Diseases, Department of Korean Internal Medicine, Kyung Hee University Korean Medicine Hospital, Seoul, Republic of Korea
| | - Youngmin Bu
- Department of Herbal Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Seok-Jae Ko
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Korean Internal Medicine, Kyung Hee University College of Korean Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
- Department of Digestive Diseases, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
10
|
Yang L, Fang Z, Zhu J, Li X, Yang B, Liu H, Lou F. The potential of Sijunzi decoction in the fight against gastrointestinal disorders: a review. Front Pharmacol 2025; 16:1464498. [PMID: 40103588 PMCID: PMC11913818 DOI: 10.3389/fphar.2025.1464498] [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: 07/14/2024] [Accepted: 02/03/2025] [Indexed: 03/20/2025] Open
Abstract
Sijunzi Decoction (SJZD) is a traditional Chinese medicine formula widely used in the treatment of gastrointestinal disorders. Clinical studies have substantiated the efficacy of SJZD in managing conditions such as functional dyspepsia, chronic gastritis, gastric cancer, irritable bowel syndrome, colorectal cancer, and ulcerative colitis. Despite its proven effectiveness, the precise mechanisms by which SJZD operates remain incompletely understood. In this study, we undertake a systematic review of both the clinical applications and the mechanistic underpinnings of SJZD in the context of gastrointestinal disease treatment. Research indicates that SJZD functions through a spectrum of mechanisms including the regulation of intestinal flora, alleviation of inflammation, modulation of immune responses, and facilitation of mucosal repair in the treatment of gastrointestinal ailments. This comprehensive analysis aims to provide a clearer understanding of how SJZD benefits patients with gastrointestinal disorders.
Collapse
Affiliation(s)
- Liangjun Yang
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province (Zhejiang Academy of Traditional Chinese Medicine), Hangzhou, China
| | - Zheng Fang
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province (Zhejiang Academy of Traditional Chinese Medicine), Hangzhou, China
| | - Jiajie Zhu
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province (Zhejiang Academy of Traditional Chinese Medicine), Hangzhou, China
| | - Xiaofang Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bo Yang
- Department of Pharmacy, Tongde Hospital of Zhejiang Province (Zhejiang Academy of Traditional Chinese Medicine), Hangzhou, China
| | - Haiyan Liu
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province (Zhejiang Academy of Traditional Chinese Medicine), Hangzhou, China
| | - Feiyan Lou
- Endoscopy Center, Tongde Hospital of Zhejiang Province (Zhejiang Academy of Traditional Chinese Medicine), Hangzhou, China
| |
Collapse
|
11
|
Luo W, Zhang K, Wang Y, Ye M, Zhang Y, Xu W, Chen L, Li H. The Rhizome of Atractylodes macrocephala Koidz.: A Comprehensive Review on the Traditional Uses, Phytochemistry and Pharmacology. Chem Biodivers 2025; 22:e202401879. [PMID: 39473269 DOI: 10.1002/cbdv.202401879] [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: 08/03/2024] [Accepted: 10/28/2024] [Indexed: 03/16/2025]
Abstract
Atractylodes macrocephala Koidz. (A. macrocephala) is a perennial herb of the genus Atractylodes. The rhizome of A. macrocephala (AMR) is its medicinal part. It primarily grows in Southeast Asia and function to invigorate the spleen and qi, drying dampness and removing water. It has long been used for cancer treatment, relieving inflammation, and improving gastrointestinal function, highlighting its remarkable medicinal value. This paper focuses on recent advancements in the traditional uses, phytochemistry, and pharmacology of AMR from 2018 to the present, while exploring its therapeutic and scientific potential. In recent years, more than 120 compounds have been identified in AMR. The primary active components have been identified as sesquiterpenoids, polysaccharides and polyacetylenes. Modern pharmacological studies have demonstrated that AMR has anti-inflammatory, anti-tumor, immunity enhancement, gastrointestinal function improvement, and other pharmacological effects. It is mainly employed in the clinical treatment of tumors and gastrointestinal diseases, showing promising developmental potential. Its mechanism may be related to reducing oxidative stress, inhibiting the expression of inflammatory mediators and factors, and alleviating apoptosis through related signaling pathways. It is hoped that this review can provide a theoretical reference and scientific basis for further systematic research and extensive clinical application of AMR.
Collapse
Affiliation(s)
- Weihong Luo
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Kexin Zhang
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Yali Wang
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Miao Ye
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Yuqin Zhang
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Wei Xu
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Lixia Chen
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Wuya College of Innovation, School of Pharmacy, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Hua Li
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Wuya College of Innovation, School of Pharmacy, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| |
Collapse
|
12
|
Lin Q, Chen Y, Yu B, Chen Z, Zhou H, Su J, Yu J, Yan M, Chen S, Lv G. Atractylodes macrocephala Rhizoma alleviates blood hyperviscosity induced by high-fat, high-sugar, and high-salt diet by inhibiting gut-liver inflammation and fibrinogen synthesis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119034. [PMID: 39505223 DOI: 10.1016/j.jep.2024.119034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/20/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Unhealthy dietary patterns and lifestyle changes have been linked to increased blood viscosity, which is recognized as an important pathogenic factor in cardiovascular and cerebrovascular diseases. The underlying mechanism may involve chronic inflammation resulting from intestinal barrier disruption induced by unhealthy diets. The rhizome of Atractylodes macrocephala Koidz. (Called Baizhu in China), is a well-used "spleen-reinforcing" traditional Chinese medicinal herb used for thousands of years. Previous research has demonstrated its multiple gastrointestinal health benefits and its ability to regulate metabolic disorders. However, the effects of Baizhu on blood hyperviscosity induced by long-term unhealthy diets remain unclear. AIM OF THE STUDY This study aimed to investigate the effects of the aqueous extract of Baizhu on blood hyperviscosity induced by unhealthy diet and to explore the possible mechanisms. MATERIALS AND METHODS The blood hyperviscosity model in SD rats was established utilizing a high-fat, high-sugar, and high-salt diet (HFSSD). Subsequently, the rats underwent a twelve-week intervention with varying doses of Baizhu and a positive control. To evaluate the efficacy of Baizhu on blood hyperviscosity in model rats, we measured behavioral index, hemorheological parameters, inflammatory cytokines, hematology, adhesion molecules, as well as biochemical indicators in serum and liver. We also assessed the pathological states of the colon and liver. Furthermore, Western blotting, ELISA, IHC, and qRT-PCR were used to determine the effect of Baizhu on the IL-6/STAT3/ESRRG signaling pathway and FIB synthesis. RESULTS The intervention of Baizhu showed evident attenuating effects on blood viscosity and microcirculation disorders, and exhibit the capacity to moderately modulate parameters including grip, autonomous activities, vertigo time, TC, TG, LDL-c, inflammatory factors, adhesion factors, hematological indicators, etc. At the same time, it reduces liver lipid droplet deposition, restores intestinal integrity, and lowers LPS level in the serum. Subsequent experimental results showed that Baizhu downregulated the expression of TLR4 and NF-κB in colon tissue, as well as the expression of IL-6, TLR4, p-JAK2, p-STAT3, and ESRRG in liver tissue. Finally, we also found that Baizhu could regulate the levels of FIB in plasma and liver. CONCLUSION Baizhu protects HFSSD-induced rats from blood hyperviscosity, likely through repairing the intestinal barrier and inhibiting LPS/TLR4-associated liver inflammatory activation, thus suppressing FIB synthesis through the downregulation of IL-6/STAT3/ESRRG pathway.
Collapse
Affiliation(s)
- Qiwei Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou, 310053, China
| | - Yigong Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou, 310053, China
| | - Bingqing Yu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou, 310053, China
| | - Ziyan Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou, 310053, China
| | - Hengpu Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou, 310053, China
| | - Jie Su
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou, 310053, China
| | - Jingjing Yu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou, 310053, China
| | - Meiqiu Yan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou, 310053, China.
| | - Suhong Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou, 310053, China.
| | - Guiyuan Lv
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou, 310053, China.
| |
Collapse
|
13
|
Beauchemin ET, Hunter C, Maurice CF. Dextran sodium sulfate-induced colitis alters the proportion and composition of replicating gut bacteria. mSphere 2025; 10:e0082524. [PMID: 39723822 PMCID: PMC11774032 DOI: 10.1128/msphere.00825-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/15/2024] [Indexed: 12/28/2024] Open
Abstract
The bacteria living in the human gut are essential for host health. Though the composition and metabolism of these bacteria are well described in both healthy hosts and those with intestinal disease, less is known about the metabolic activity of the gut bacteria prior to, and during, disease development-especially regarding gut bacterial replication. Here, we use a recently developed single-cell technique alongside existing metagenomics-based tools to identify, track, and quantify replicating gut bacteria both ex vivo and in situ in the dextran sodium sulfate (DSS) mouse model of colitis. We show that the proportion of replicating gut bacteria decreases when mice have the highest levels of inflammation and returns to baseline levels as mice begin recovering. In addition, we report significant alterations in the composition of the replicating gut bacterial community ex vivo during colitis development. On the taxa level, we observe significant changes in the abundance of taxa such as the mucus-degrading Akkermansia and the poorly described Erysipelatoclostridium genus. We further demonstrate that many taxa exhibit variable replication rates in situ during colitis, including Akkermansia muciniphila. Lastly, we show that colitis development is positively correlated with increases in the presence and abundance of bacteria in situ which are predicted to be fast replicators. This could suggest that taxa with the potential to replicate quickly may have an advantage during intestinal inflammation. These data support the need for additional research using activity-based approaches to further characterize the gut bacterial response to intestinal inflammation and its consequences for both the host and the gut microbial community.IMPORTANCEIt is well known that the bacteria living inside the gut are important for human health. Indeed, the type of bacteria that are present and their metabolism are different in healthy people versus those with intestinal disease. However, less is known about how these gut bacteria are replicating, especially as someone begins to develop intestinal disease. This is particularly important as it is thought that metabolically active gut bacteria may be more relevant to health. Here, we begin to address this gap using several complementary approaches to characterize the replicating gut bacteria in a mouse model of intestinal inflammation. We reveal which gut bacteria are replicating, and how quickly, as mice develop and recover from inflammation. This work can serve as a model for future research to identify how actively growing gut bacteria may be impacting health, or why these particular bacteria tend to thrive during intestinal inflammation.
Collapse
Affiliation(s)
- Eve T. Beauchemin
- Department of Microbiology & Immunology, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Claire Hunter
- Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, England, United Kingdom
| | - Corinne F. Maurice
- Department of Microbiology & Immunology, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
- McGill Centre for Microbiome Research, Montreal, Quebec, Canada
| |
Collapse
|
14
|
Zhang L, Wang K, Huang L, Deng B, Chen C, Zhao K, Wang W. Ganoderic Acid A Alleviates Severe Acute Pancreatitis by Modulating Gut Homeostasis and Inhibiting TLR4-NLRP3 Signaling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:1563-1579. [PMID: 39811933 PMCID: PMC11740897 DOI: 10.1021/acs.jafc.4c07635] [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: 08/19/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025]
Abstract
Background Severe acute pancreatitis (SAP) manifests as a critical state marked by acute abdominal symptoms, often associated with intestinal barrier dysfunction, exacerbating SAP retroactively. Ganoderic acid A (GAA) demonstrates anti-inflammatory properties in various inflammatory disorders. Nonetheless, its potential therapeutic impact on SAP and the underlying mechanisms remain unexplored. Methods In both wild-type and TLR4-/- mice, experimental SAP was induced using caerulein plus lipopolysaccharide. Caerulein injections were administered intraperitoneally following 7 days of intragastric GAA administration. Additionally, the potential mechanisms by which GAA ameliorates SAP were further investigated using fecal microbiota transplantation and TLR4-overexpressing IEC-6 cells. Results We observed that GAA treatment significantly ameliorated serum levels of amylase, lipase, and pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) in SAP mice. Pretreatment with GAA mitigated pathological injuries and reduced M1 macrophage and neutrophil infiltration in pancreatic or ileal tissues. Additionally, GAA treatment down-regulated TLR4-MAPK/NF-κB signaling and NLRP3 inflammasome activation in the pancreatic and ileal tissues of SAP mice. The results further revealed that the gavage of GAA decreased bacterial translocation (Escherichia coli and EUB338), repaired intestinal barrier dysfunction (ZO-1, occludin, DAO, and FITC), increased lysozyme and MUC2 expression, and raised the levels of short-chain fatty acids. Analysis of the gut microbiome showed that the beneficial effects of GAA treatment were associated with improvements in pancreatitis-associated gut microbiota dysbiosis, characterized by notable increases in α-diversity and the abundance of probiotics such as Akkermansia, GCA-900066575, and Parvibacter. Fecal transplantation experiments further confirmed that GAA exerts protective effects by modulating intestinal flora. The protective role of GAA in intestinal and pancreatic injuries is mediated by the inhibition of TLR4 signaling, as further evidenced in TLR4-deficient mice and TLR4-overexpressed IEC-6 cells. The results of docking indicated that GAA interacts with TLR4 via a hydrophobic interaction. Conclusions The study demonstrates that GAA significantly alleviates SAP through its anti-inflammatory and antioxidant capacities, as well as by restoring intestinal homeostasis, thereby providing insights into novel treatments for SAP.
Collapse
Affiliation(s)
- Lilong Zhang
- Department
of General Surgery, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Hubei
Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- General
Surgery Laboratory, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Central
Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Kunpeng Wang
- Department
of General Surgery, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Hubei
Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- General
Surgery Laboratory, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Central
Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Li Huang
- Department
of Gastroenterology, Renmin Hospital of
Wuhan University, Wuhan, Hubei 430060, China
| | - Beiying Deng
- Department
of Gastroenterology, Renmin Hospital of
Wuhan University, Wuhan, Hubei 430060, China
| | - Chen Chen
- Department
of General Surgery, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Hubei
Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- General
Surgery Laboratory, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Central
Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Kailiang Zhao
- Department
of General Surgery, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Hubei
Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- General
Surgery Laboratory, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Central
Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Weixing Wang
- Department
of General Surgery, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Hubei
Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- General
Surgery Laboratory, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Central
Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| |
Collapse
|
15
|
Yuan Q, Liu W, Hao W, Chen Y, Xiao Y, Li H, Shui M, Wu DT, Wang S. Glycosidic linkages of fungus polysaccharides influence the anti-inflammatory activity in mice. J Adv Res 2025; 67:161-172. [PMID: 38309691 PMCID: PMC11725157 DOI: 10.1016/j.jare.2024.01.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 02/05/2024] Open
Abstract
INTRODUCTION Over decades, the source-function relationships of bioactive polysaccharides have been progressively investigated, however, it is still unclear how a defined structure may conduce to the bioactivities of polysaccharides. OBJECTIVES To explore the structure-function relationship of fungus polysaccharides, we employed a dextran sulfate sodium (DSS)-induced colitis mouse model to compare the anti-inflammatory activity of two fungus polysaccharides from Dictyophora indusiata (DIP) and Tremella fuciformis (TFP), which exhibit distinct glycosidic linkages. METHODS The structures of DIP and TFP were characterized through molecular weight detection, molecular morphology analysis, methylation analysis, and NMR analysis. Subsequently, we employed a DSS-induced colitis model to assess the anti-inflammatory efficacy of DIP and TFP. The colitis symptoms, histological morphology, intestinal inflammatory cytokines, and the composition and function of gut microbiota before and after polysaccharides treatment in colitis mice were also investigated. RESULTS DIP, l,3-β-D-glucan with 1,4-β and 1,6-β-D-Glcp as branched chains, exhibited superior therapeutic effect than that of TFP consisted of a linear 1,3-α-D-mannose backbone with D-xylose and L-fucose in the side chains. Both DIP and TFP relieved DSS-induced colitis in a gut microbiota-dependent manner. Furthermore, metagenomics showed that DIP and TFP could partially reverse the bacterial function in colitis mice. Glycoside Hydrolase 1 (GH1) and GH3 were identified as being involved in hydrolyzing the glucose linkages in DIP, while GH92 and GH29 were predicted to be active in cleaving the α-1,3-linked mannose linkages and the glycosidic bonds of L-fucose residues in TFP. CONCLUSION Our findings highlight the pivotal role of glycosidic linkages in anti-inflammatory activities of fungus polysaccharides and would promote the design and discovery of polysaccharides with designated activity to be used as functional foods and/or therapeutics.
Collapse
Affiliation(s)
- Qin Yuan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China; Macao Centre for Research and Development in Chinese Medicine, University of Macau, Macau, China
| | - Wen Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China; Macao Centre for Research and Development in Chinese Medicine, University of Macau, Macau, China
| | - Wei Hao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China; Macao Centre for Research and Development in Chinese Medicine, University of Macau, Macau, China
| | - Yi Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China; Macao Centre for Research and Development in Chinese Medicine, University of Macau, Macau, China
| | - Yaqin Xiao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Hongyi Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Mingju Shui
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Ding-Tao Wu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu 610106, China; Institute for Advanced Study, Chengdu University, Chengdu 610106, China.
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China; Macao Centre for Research and Development in Chinese Medicine, University of Macau, Macau, China.
| |
Collapse
|
16
|
Bai SH, Chandnani A, Cao S. Bile Acids in Inflammatory Bowel Disease: From Pathophysiology to Treatment. Biomedicines 2024; 12:2910. [PMID: 39767816 PMCID: PMC11673883 DOI: 10.3390/biomedicines12122910] [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: 11/06/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic condition that affects about 7 million people worldwide, and new therapies are needed. Understanding the complex roles that bile acids (BAs) play in IBD may lead to the development of novel IBD treatments independent of direct immunosuppression. This review discusses the latest discoveries in the roles BAs play in IBD pathogenesis and explores how these discoveries offer promising new therapeutic targets to treat IBD and improve patient outcomes. Several therapies discussed include specific BA receptor (BAR) agonists, dietary therapies, supplements, probiotics, and mesenchymal stem cell therapies that have all been shown to decrease IBD disease activity.
Collapse
Affiliation(s)
| | | | - Siyan Cao
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.H.B.); (A.C.)
| |
Collapse
|
17
|
Niu Y, Zhao T, Liu Z, Li D, Wen D, Li B, Huang X. Brassica rapa L. crude polysaccharide meditated synbiotic fermented whey beverage ameliorates hypobaric hypoxia induced intestinal damage. Food Funct 2024; 15:11975-11989. [PMID: 39555987 DOI: 10.1039/d4fo04667f] [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: 11/19/2024]
Abstract
Hypobaric hypoxia causes oxidative stress and inflammatory responses and disrupts the gut microbiome and metabolome. In this study, we developed a synbiotic fermented whey beverage, combining kefir and Brassica rapa L. crude polysaccharides, to explore its protective effects against high-altitude induced injury in mice. The beverage, formulated with 0.8% (w/v) polysaccharides and kefir inoculation, demonstrated robust fermentation parameters and antioxidative capacity. When applied to a hypobaric hypoxia mouse model, the synbiotic fermented whey significantly reduced oxidation and protected the intestinal barrier by lowering inflammation, protecting the intestinal structure, increasing goblet cell counts, and reducing apoptosis. It also modulated the gut microbiota, enriching beneficial taxa as Intestinimonas and Butyricicoccaceae, while reducing harmful ones like Marvinbryantia and Proteus, and enhancing short-chain fatty acid (SCFA) production. Notably, the beverage increased berberine and nicotinic acid levels, activating the adenosine monophosphate-activated protein kinase (AMPK) signalling pathway and influencing nicotinate and nicotinamide metabolites linked to the suppression of Marvinbryantia, thereby alleviating intestinal inflammation and barrier damage. These effects contributed to the alleviation of hypoxia-induced intestinal damage in mice. This study highlights the potential of synbiotics and whey fermentation in novel nutritional interventions in high altitude environments.
Collapse
Affiliation(s)
- Yuanlin Niu
- School of Public Health, Lanzhou University, Lanzhou, China.
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Xizang, China
| | - Tingting Zhao
- School of Public Health, Lanzhou University, Lanzhou, China.
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Xizang, China
| | - Zhenjiang Liu
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Xizang, China
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Diantong Li
- School of Public Health, Lanzhou University, Lanzhou, China.
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Xizang, China
| | - Dongxu Wen
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Xizang, China
| | - Bin Li
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Xizang, China
| | - Xiaodan Huang
- School of Public Health, Lanzhou University, Lanzhou, China.
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Xizang, China
| |
Collapse
|
18
|
Liu Y, Huang K, Zhang Y, Li S, Song H, Guan X. Oat anthranilamides regulates high-fat diet-induced intestinal inflammation by the TLR4/NF-κb signalling pathway and gut microbiota. Int J Food Sci Nutr 2024; 75:786-799. [PMID: 39285614 DOI: 10.1080/09637486.2024.2401130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/22/2024] [Accepted: 09/01/2024] [Indexed: 11/26/2024]
Abstract
Oat anthranilamides have demonstrated antioxidant and anti-inflammatory effects; however, the precise mechanism of action remains unclear. This study investigated the impact of oat anthranilamide B (AVN B) on high-fat diet (HFD)-induced intestinal inflammation in mice and its underlying mechanisms. The results indicated that AVN B supplementation mitigated weight gain and reduced inflammatory and oxidative stress markers in serum, liver, and intestines. It improved intestinal barrier dysfunction by upregulating the expression levels of Occludin and MUC2 while simultaneously reducing intestinal inflammation by inhibiting the TLR4/NF-κB signalling pathway. Additionally, AVN B treatment improved gut microbiota composition. It increased the abundance of beneficial flora and the production of short-chain fatty acids (SCFAs), especially propionate and butyrate, associated with reduced production of pro-inflammatory factors and enhanced intestinal protection. The findings provide scientific evidence for the potential of AVN B as an anti-inflammatory agent.
Collapse
Affiliation(s)
- Yongyong Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, P.R. China
| | - Kai Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, P.R. China
| | - Yu Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, P.R. China
| | - Sen Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, P.R. China
| | - Hongdong Song
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, P.R. China
| | - Xiao Guan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, P.R. China
| |
Collapse
|
19
|
Wang L, Qin N, Shi L, Liu R, Zhu T. Gut Microbiota and Tryptophan Metabolism in Pathogenesis of Ischemic Stroke: A Potential Role for Food Homologous Plants. Mol Nutr Food Res 2024; 68:e2400639. [PMID: 39551995 DOI: 10.1002/mnfr.202400639] [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: 08/10/2024] [Revised: 10/15/2024] [Indexed: 11/19/2024]
Abstract
SCOPE The intestinal flora is involved in the maintenance of human health and the development of diseases, and is closely related to the brain. As an essential amino acid, tryptophan (TRP) participates in a variety of physiological functions in the body and affects the growth and health of the human body. TRP catabolites produced by the gut microbiota are important signaling molecules for microbial communities and host-microbe interactions, and play an important role in maintaining health and disease pathogenesis. METHODS AND RESULTS The review first demonstrates the evidence of TRP metabolism in stroke and the relationship between gut microbiota and TRP metabolism. Furthermore, the review reveals that food homologous plants (FHP) bioactive compounds have been shown to regulate various metabolic pathways of the gut microbiota, including the biosynthesis of valine, leucine, isoleucine, and vitamin B6 metabolism. The most notable metabolic alteration is in TRP metabolism. CONCLUSION The interaction between gut microbiota and TRP metabolism offers a plausible explanation for the notable bioactivities of FHP in the treatment of ischemic stroke (IS). This review enhances the comprehension of the underlying mechanisms associated with the bioactivity of FHP on IS.
Collapse
Affiliation(s)
- Lei Wang
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Na Qin
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
| | - Liuliu Shi
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, PR China
| | - Rujuan Liu
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, PR China
| | - Ting Zhu
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
| |
Collapse
|
20
|
Huang S, Zhang M, Li X, Pei J, Zhou Z, Lei P, Wang M, Zhang P, Yu H, Fan G, Han L, Yu H, Wang Y, Jiang M. Formulation, characterization, and evaluation of curcumin-loaded ginger-derived nanovesicles for anti-colitis activity. J Pharm Anal 2024; 14:101014. [PMID: 39834559 PMCID: PMC11743112 DOI: 10.1016/j.jpha.2024.101014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 01/05/2025] Open
Abstract
Plant-derived nanovesicles have gained attention given their similarity to mammalian exosomes and advantages such as low cost, sustainability, and tissue targeting. Thus, they hold promise for disease treatment and drug delivery. In this study, we proposed a time-efficient method, PEG 8000 combined with sucrose density gradient centrifugation to prepare ginger-derived nanovesicles (GDNVs). Subsequently, curcumin (CUR) was loaded onto GDNV by ultrasonic incubation. The optimum conditions for ginger-derived nanovesicles loaded with curcumin (CG) were ultrasound time of 3 min, a carrier-to-drug ratio (GDNV:CUR) of 1:1. The study achieved a high loading capacity (94.027% ± 0.094%) and encapsulation efficiency (89.300% ± 0.344%). Finally, the drugs' in vivo distribution and anti-colitis activity were investigated in mice. CG was primarily distributed in the colon after oral administration. Compared to CUR and GDNV, CG was superior in improving disease activity, colon length, liver and spleen coefficients, myeloperoxidase activity, and biochemical factor levels in ulcerative colitis (UC) mice. In addition, CG plays a protective role against UC by modulating serum metabolite levels and gut flora. In summary, our study demonstrated that GDNV can be used for CUR delivery with enhanced therapeutic potential.
Collapse
Affiliation(s)
- Shengjie Huang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Min Zhang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Xiaoge Li
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jierong Pei
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Zhirong Zhou
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Peng Lei
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Meng Wang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Peng Zhang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Heshui Yu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Guanwei Fan
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lifeng Han
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Haiyang Yu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuefei Wang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Miaomiao Jiang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| |
Collapse
|
21
|
Fan S, Zhou Y, Zhu N, Meng Q, Zhao Y, Xu J, Tang Y, Dai S, Yuan X. Exogenous Application of dsRNA-Inducing Silencing of the Fusarium oxysporum Tup1 Gene and Reducing Its Virulence. Int J Mol Sci 2024; 25:10286. [PMID: 39408614 PMCID: PMC11476490 DOI: 10.3390/ijms251910286] [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: 08/05/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Fusarium oxysporum is a widespread soil-borne fungal pathogen that can infect various plants, causing wilt and root rot diseases. The root rot disease of Atractylodes macrocephala caused by F. oxysporum is among the most serious diseases associated with continuous cropping, significantly hindering its sustainable development. In this study, we aimed to investigate the effect of exogenous application of double-stranded RNA (dsRNA) on silencing the F. oxysporum Tup1 gene to reduce its virulence and to evaluate its potential application in controlling root rot disease in A. macrocephala. The Tup1 gene was amplified from the F. oxysporum genome, and different lengths of Tup1-dsRNA were designed and synthesized. The uptake of dsRNA by the fungus was verified using Tup1-dsRNA labeled with fluorescein, and in vitro dsRNA treatment experiments were conducted to assess its impact on the growth and virulence of F. oxysporum. Additionally, Tup1-dsRNA was applied to the roots of A. macrocephala to evaluate its effectiveness in controlling root rot disease. The experimental results showed that F. oxysporum could effectively uptake exogenously applied Tup1-dsRNA, significantly reducing Tup1 gene expression. All lengths of Tup1-dsRNA inhibited fungal growth and caused morphological changes in the fungal hyphae. Further plant experiments and Reverse Transcription Quantitative Polymerase Chain Reaction (RT-qPCR) analysis indicated that Tup1-dsRNA treatment significantly reduced the incidence of root rot disease in A. macrocephala, which was supported by the reduction in peroxidase (POD) and catalase (CAT) enzyme activities, malondialdehyde (MDA) content, and proline (Pro) levels in treated root tissues. This study demonstrated that exogenous dsRNA could reduce the virulence of F. oxysporum by silencing the Tup1 gene and effectively mitigate the root rot disease it causes in A. macrocephala. The successful application of Tup1-dsRNA provided strong evidence for the potential of RNA interference (RNAi) technology in plant disease control. Future research could further optimize the design and application of dsRNA to enhance its practical value in agriculture.
Collapse
Affiliation(s)
- Sen Fan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310051, China; (S.F.); (Y.Z.); (N.Z.); (Q.M.); (Y.Z.); (J.X.); (Y.T.); (S.D.)
| | - Yanguang Zhou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310051, China; (S.F.); (Y.Z.); (N.Z.); (Q.M.); (Y.Z.); (J.X.); (Y.T.); (S.D.)
| | - Na Zhu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310051, China; (S.F.); (Y.Z.); (N.Z.); (Q.M.); (Y.Z.); (J.X.); (Y.T.); (S.D.)
| | - Qingling Meng
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310051, China; (S.F.); (Y.Z.); (N.Z.); (Q.M.); (Y.Z.); (J.X.); (Y.T.); (S.D.)
| | - Yujin Zhao
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310051, China; (S.F.); (Y.Z.); (N.Z.); (Q.M.); (Y.Z.); (J.X.); (Y.T.); (S.D.)
| | - Jingyan Xu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310051, China; (S.F.); (Y.Z.); (N.Z.); (Q.M.); (Y.Z.); (J.X.); (Y.T.); (S.D.)
| | - Yunjia Tang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310051, China; (S.F.); (Y.Z.); (N.Z.); (Q.M.); (Y.Z.); (J.X.); (Y.T.); (S.D.)
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
| | - Shijie Dai
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310051, China; (S.F.); (Y.Z.); (N.Z.); (Q.M.); (Y.Z.); (J.X.); (Y.T.); (S.D.)
| | - Xiaofeng Yuan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310051, China; (S.F.); (Y.Z.); (N.Z.); (Q.M.); (Y.Z.); (J.X.); (Y.T.); (S.D.)
| |
Collapse
|
22
|
Liu R, Luo Y, Ma J, Zhang Q, Sheng Y, Li J, Li H, Zhao T. Traditional Chinese medicine for functional gastrointestinal disorders and inflammatory bowel disease: narrative review of the evidence and potential mechanisms involving the brain-gut axis. Front Pharmacol 2024; 15:1444922. [PMID: 39355776 PMCID: PMC11443704 DOI: 10.3389/fphar.2024.1444922] [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: 06/06/2024] [Accepted: 08/23/2024] [Indexed: 10/03/2024] Open
Abstract
Functional gastrointestinal disorders (FGIDs) and inflammatory bowel disease (IBD) are common clinical disorders characterized by recurrent diarrhea and abdominal pain. Although their pathogenesis has not been fully clarified, disruptions in intestinal motility and immune function are widely accepted as contributing factors to both conditions, and the brain-gut axis plays a key role in these processes. Traditional Chinese Medicine (TCM) employs a holistic approach to treatment, considers spleen and stomach impairments and liver abnormality the main pathogenesis of these two diseases, and offers a unique therapeutic strategy that targets these interconnected pathways. Clinical evidence shows the great potential of TCM in treating FGIDs and IBD. This study presents a systematic description of the pathological mechanisms of FGIDs and IBD in the context of the brain-gut axis, discusses clinical and preclinical studies on TCM and acupuncture for the treatment of these diseases, and summarizes TCM targets and pathways for the treatment of FGIDs and IBD, integrating ancient wisdom with contemporary biomedical insights. The alleviating effects of TCM on FGID and IBD symptoms are mainly mediated through the modulation of intestinal immunity and inflammation, sensory transmission, neuroendocrine-immune network, and microbiota and their metabolism through brain-gut axis mechanisms. TCM may be a promising treatment option in controlling FGIDs and IBD; however, further high-quality research is required. This review provides a reference for an in-depth exploration of the interventional effects and mechanisms of TCM in FGIDs and IBD, underscoring TCM's potential to recalibrate the dysregulated brain-gut axis in FGIDs and IBD.
Collapse
Affiliation(s)
- RuiXuan Liu
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - YunTian Luo
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - JinYing Ma
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qi Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yudong Sheng
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiashan Li
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongjiao Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - TianYi Zhao
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
23
|
Zhang L, Zhu W, Zhang Z. Combined analysis of cecal microbiota and metabolomics reveals the intervention mechanism of Dayuan Yin in acute lung injury. Front Pharmacol 2024; 15:1436017. [PMID: 39318776 PMCID: PMC11420052 DOI: 10.3389/fphar.2024.1436017] [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: 05/21/2024] [Accepted: 07/10/2024] [Indexed: 09/26/2024] Open
Abstract
The ancient Chinese medicinal formula, Dayuan Yin (DYY), has a long history of use in treating respiratory ailments and is shown to be effective in treating acute infectious diseases. This study aims to explore how DYY may impact intestinal flora and metabolites induced by acute lung injury (ALI). ALI rats were induced with lipopolysaccharide (LPS) to serve as models for assessing the anti-ALI efficacy of DYY through multiple lung injury indices. Changes in intestinal microflora were assessed via 16SrRNA gene sequencing, while cecum contents were analyzed using non-targeted metabonomics. Differential metabolites were identified through data analysis, and correlations between metabolites, microbiota, and inflammatory markers were examined using Pearson's correlation analysis. DYY demonstrated a significant improvement in LPS-induced lung injury and altered the composition of intestinal microorganisms, and especially reduced the potential harmful bacteria and enriched the beneficial bacteria. At the gate level, DYY exhibited a significant impact on the abundance of Bacteroidota and Firmicutes in ALI rats, as well as on the regulation of genera such as Ruminococcus, Lactobacillus, and Romboutsia. Additionally, cecal metabonomics analysis revealed that DYY effectively modulated the abnormal expression of 12 key metabolic biomarkers in ALI rats, thereby promoting intestinal homeostasis through pathways such as purine metabolism. Furthermore, Pearson's analysis indicated a strong correlation between the dysregulation of intestinal microbiota, differential metabolites, and inflammation. These findings preliminarily confirm that ALI is closely related to cecal microbial and metabolic disorders, and DYY can play a protective role by regulating this imbalance, which provides a new understanding of the multi-system linkage mechanism of DYY improving ALI.
Collapse
Affiliation(s)
- Lei Zhang
- Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China
| | - Wei Zhu
- Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China
| | - Zepeng Zhang
- Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China
| |
Collapse
|
24
|
Tang L, Liu Y, Tao H, Feng W, Ren C. Network pharmacology integrated with molecular docking and molecular dynamics simulations to explore the mechanism of Tongxie Yaofang in the treatment of ulcerative colitis. Medicine (Baltimore) 2024; 103:e39569. [PMID: 39252247 PMCID: PMC11383260 DOI: 10.1097/md.0000000000039569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Tongxie Yaofang (TXYF), a classical traditional Chinese medicine, is commonly used in China to treat ulcerative colitis (UC). The aim of this study was to integrate network pharmacology with molecular docking and molecular dynamics simulations to explore the mechanism of Tongxie Yaofang in the treatment of UC. The traditional Chinese medicine systems pharmacology database was used to retrieve the relevant chemical compositions of the herbs contained in TXYF. The DisGeNET, GeneCards, Online Mendelian Inheritance in Man, and Therapeutic Target Database databases were used to retrieve UC-related targets. To construct protein-protein interaction networks and screen for key targets, gene ontology and Kyoto Encyclopedia of Genes and Genomes analyses of the key targets of TXYF in the treatment of UC were performed using R 4.3.2 software. AutoDock Tools 1.5.7 was used for molecular docking. Molecular dynamics simulations of protein complexes and complexes of proteins with small-molecule ligands and eutectic ligands were carried out with Gromacs 2022 software. Network pharmacology analysis revealed that TXYF could act on UC through multiple targets and pathways. It may exert therapeutic effects mainly through the AGE/RAGE, TOLL, JAK/STAT, and Th17 signaling pathways. The possible targets of TXYF in the treatment of UC could be AKT1, BCL2, EGFR, HMOX1, HSP90AA1, and TGFβ1. Molecular docking analysis revealed that AKT1 had the highest binding energy (-10.55 kcal/mol). Molecular dynamics simulations revealed that the complexes formed by the AKT1 protein and the chemical compounds MOL001910 and MOL00035 had good stability and high binding strength. AKT1 may be the most critical target of TXYF in treating UC, and the key chemical components of TXYF in treating UC may include β-sitosterol (MOL000358) and 11alpha,12alpha-epoxy-3beta-23-dihydroxy-30-norolean-20-en-28,12beta-olide (MOL00 1910). This study revealed that TXYF may exert therapeutic effects on UC through multiple targets, multiple biological functions, and multiple signaling pathways. This study provides a new insight into the pharmacological mechanism of TXYF in treating UC.
Collapse
Affiliation(s)
- Lili Tang
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yuedong Liu
- The Third Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Hongwu Tao
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Wenzhe Feng
- Affiliated Hospital of Shaanxi University of Chinese Medicine, Shenyang, China
| | - Cong Ren
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
25
|
Li A, Liu F, Si W, Wang Y, Wang D, Yuan Z, Li L, Kiani FA, Jiang X. Pesticide butachlor exposure perturbs gut microbial homeostasis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 281:116646. [PMID: 38954906 DOI: 10.1016/j.ecoenv.2024.116646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024]
Abstract
Agricultural production relies heavily on the use of pesticides, which may accumulate in soil and water, posing a significant threat to the global ecological environment and biological health. Butachlor is a commonly used herbicide and environmental pollutant, which has been linked to liver and kidney damage, as well as neurological abnormalities. However, the potential impact of butachlor exposure on the gut microbiota remains understudied. Thus, our aim was to investigate the potential negative effects of butachlor exposure on host health and gut microbiota. Our results demonstrated that butachlor exposure significantly reduced the host antioxidant capacity, as evidenced by decreased levels of T-AOC, SOD, and GSH-Px, and increased levels of MDA. Serum biochemical analysis also revealed a significant increase in AST and ALT levels during butachlor exposure. Microbial analysis showed that butachlor exposure significantly reduced the abundance and diversity of gut microbiota. Furthermore, butachlor exposure also significantly altered the gut microbial composition. In conclusion, our findings indicate that butachlor exposure can have detrimental health effects, including dysregulation of antioxidant enzymes, abnormalities in transaminases, and hepatointestinal damage. Furthermore, it disrupts the gut microbial homeostasis by altering microbial composition and reducing diversity and abundance. In the context of the increasingly serious use of pesticides, this study will help provide impetus for standardizing the application of pesticides and reducing environmental pollution.
Collapse
Affiliation(s)
- Aoyun Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Fang Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Wenyu Si
- Xinxiang County Agriculture and Rural Affairs Bureau, Xinxiang 453799, China
| | - Yan Wang
- Institute of Animal Husbandry and Veterinary, Tibet Autonomous Region Academy of Agricultural Sciences, Tibet, Lhasa 850009, China
| | - Dongjing Wang
- Institute of Animal Husbandry and Veterinary, Tibet Autonomous Region Academy of Agricultural Sciences, Tibet, Lhasa 850009, China
| | - Zhenjie Yuan
- Institute of Animal Husbandry and Veterinary, Tibet Autonomous Region Academy of Agricultural Sciences, Tibet, Lhasa 850009, China
| | - Liangliang Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Faisal Ayub Kiani
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiong Jiang
- Hubei Three Gorges Polytechnic, Yichang 443000, China.
| |
Collapse
|
26
|
Li B, Jiang XF, Dong YJ, Zhang YP, He XLS, Zhou CL, Ding YY, Wang N, Wang YB, Cheng WQ, Jiang NH, Su J, Lv GY, Chen SH. The effects of Atractylodes macrocephala extract BZEP self-microemulsion based on gut-liver axis HDL/LPS signaling pathway to ameliorate metabolic dysfunction-associated fatty liver disease in rats. Biomed Pharmacother 2024; 175:116519. [PMID: 38663104 DOI: 10.1016/j.biopha.2024.116519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 06/03/2024] Open
Abstract
OBJECTIVES To elucidate the therapeutic effects and mechanisms of Atractylodes macrocephala extract crystallize (BZEP) and BZEP self-microemulsion (BZEPWR) on metabolic dysfunction-associated fatty liver disease (MAFLD) induced by "high sugar, high fat, and excessive alcohol consumption" based on the gut-liver axis HDL/LPS signaling pathway. METHODS In this study, BZEP and BZEPWR were obtained via isolation, purification, and microemulsification. Furthermore, an anthropomorphic MAFLD rat model of "high sugar, high fat, and excessive alcohol consumption" was established. The therapeutic effects of BZEPWR and BZEP on the model rats were evaluated in terms of liver function, lipid metabolism (especially HDL-C), serum antioxidant indexes, and liver and intestinal pathophysiology. To determine the lipoproteins in the serum sample, the amplitudes of a plurality of NMR spectra were derived via deconvolution of the composite methyl signal envelope to yield HDL-C subclass concentrations. The changes in intestinal flora were detected via 16 S rRNA gene sequencing. In addition, the gut-liver axis HDL/LPS signaling pathway was validated using immunohistochemistry, immunofluorescence, and western blot. RESULTS The findings established that BZEPWR and BZEP improved animal signs, serum levels of liver enzymes (ALT and AST), lipid metabolism (TC, TG, HDL-C, and LDL-C), and antioxidant indexes (GSH, SOD, and ROS). In addition, pathological damage to the liver, colon, and ileum was ameliorated, and the intestinal barrier function of the model rats was restored. At the genus level, BZEPWR and BZEP exerted positive effects on beneficial bacteria, such as Lactobacillus and norank_f__Muribaculaceae, and inhibitory effects on harmful bacteria, such as unclassified_f__Lachnospiraceae and Blautia. Twenty HDL-C subspecies were detected, and their levels were differentially increased in both BZEPWR and BZEP groups, with BZEPWR exhibiting a stronger elevating effect on specific HDL-C subspecies. Also, the gut-liver axis HDL/LPS signaling pathway was studied, which indicated that BZEPWR and BZEP significantly increased the expressions of ABCA1, LXR, occludin, and claudin-1 proteins in the gut and serum levels of HDL-C. Concomitantly, the levels of LPS in the serum and TLR4, Myd88, and NF-κB proteins in the liver were decreased. CONCLUSION BZEPWR and BZEP exert restorative and reversal effects on the pathophysiological damage to the gut-liver axis in MAFLD rats, and the therapeutic mechanism may be related to the regulation of the intestinal flora and the HDL/LPS signaling pathway.
Collapse
Affiliation(s)
- Bo Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, Zhejiang 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, Zhejiang 313200, China
| | - Xiao-Feng Jiang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, Zhejiang 310014, China
| | - Ying-Jie Dong
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, Zhejiang 310014, China; College of Pharmaceutical Science, Zhejiang Chinese Medical University, No. 548, Binwen Road, Binjiang District, Hangzhou, Zhejiang 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, Zhejiang 313200, China
| | - Yi-Piao Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, Zhejiang 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, Zhejiang 313200, China
| | - Xing-Li-Shang He
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, Zhejiang 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, Zhejiang 313200, China
| | - Cheng-Liang Zhou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, Zhejiang 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, Zhejiang 313200, China
| | - Yan-Yan Ding
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, Zhejiang 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, Zhejiang 313200, China
| | - Ning Wang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, Zhejiang 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, Zhejiang 313200, China
| | - Yi-Bin Wang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, Zhejiang 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, Zhejiang 313200, China
| | - Wan-Qi Cheng
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, Zhejiang 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, Zhejiang 313200, China
| | - Ning-Hua Jiang
- The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, China.
| | - Jie Su
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, No. 548, Binwen Road, Binjiang District, Hangzhou, Zhejiang 310014, China.
| | - Gui-Yuan Lv
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, No. 548, Binwen Road, Binjiang District, Hangzhou, Zhejiang 310014, China.
| | - Su-Hong Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, Zhejiang 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, Zhejiang 313200, China.
| |
Collapse
|
27
|
Jing R, Zhang L, Li R, Yang Z, Song J, Wang Q, Cao N, Han G, Yin H. Milk-derived extracellular vesicles functionalized with anti-tumour necrosis factor-α nanobody and anti-microbial peptide alleviate ulcerative colitis in mice. J Extracell Vesicles 2024; 13:e12462. [PMID: 38840457 PMCID: PMC11154809 DOI: 10.1002/jev2.12462] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/16/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
Ulcerative colitis (UC) manifests clinically with chronic intestinal inflammation and microflora dysbiosis. Although biologics can effectively control inflammation, efficient delivery to the colon and colon epithelial cells remains challenging. Milk-derived extracellular vesicles (EV) show promise as an oral delivery tool, however, the ability to load biologics into EV presents challenges to therapeutic applications. Here, we demonstrate that fusing cell-penetrating peptide (TAT) to green fluorescent protein (GFP) enabled biologics loading into EV and protected against degradation in the gastrointestinal environment in vitro and in vivo after oral delivery. Oral administration of EV loaded with anti-tumour necrosis factor-α (TNF-α) nanobody (VHHm3F) (EVVHH) via TAT significantly reduced tissue TNF-α levels and alleviated pathologies in mice with acute UC, compared to VHH alone. In mice with chronic UC, simultaneously introducing VHH and an antimicrobial peptide LL37 into EV (EVLV), then administering orally improved intestinal barrier, inflammation and microbiota balance, resulted in relief of UC-induced depression and anxiety. Collectively, we demonstrated that oral delivery of EVLV effectively alleviated UC in mice and TAT efficiently loaded biologics into EV to confer protection from degradation in the gastrointestinal tract. This therapeutic strategy is promising for UC and is a simple and generalizable approach towards drug-loaded orally-administrable EV treatment for other diseases.
Collapse
Affiliation(s)
- Renwei Jing
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Leijie Zhang
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Ruibin Li
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Zhongqiu Yang
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Jun Song
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Qian Wang
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Nan Cao
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Gang Han
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - HaiFang Yin
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
- Department of Clinical LaboratoryTianjin Medical University General HospitalTianjinChina
| |
Collapse
|
28
|
Li R, Yang P, Liu B, Ye Z, Zhang P, Li M, Gong Y, Huang Y, Yang L, Li M. Lycium barbarum polysaccharide remodels colon inflammatory microenvironment and improves gut health. Heliyon 2024; 10:e30594. [PMID: 38774318 PMCID: PMC11107222 DOI: 10.1016/j.heliyon.2024.e30594] [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: 07/17/2023] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/24/2024] Open
Abstract
Aim Disturbed intestinal microbiota has been implicated in the inflammatory microenvironment of the colon, which usually results in ulcerative colitis (UC). Given the limitations of these drugs, it is important to explore alternative means of protecting the gut health from UC. This study aimed to investigate the potential of polysaccharides as beneficial nutrients in the regulation of the gut microbiota, which determines the inflammatory microenvironment of the colon. Materials and methods Mice were treated with dextran sulfate sodium (DSS) to evaluate the effects and mechanisms of Lycium barbarum polysaccharide (LBP) in remodeling the inflammatory microenvironment and improving gut health. Body weight and disease activity indices were monitored daily. Hematoxylin and eosin staining was used to analyze colon dynamics. The levels of inflammatory indicators and expression of MUC-2, claudin-1, ZO-1, and G-protein-coupled receptor 5 (TGR5) were determined using assay kits and immunohistochemistry, respectively. 16S rRNA high-throughput sequencing of the intestinal microbiota and liquid chromatography-tandem mass spectrometry for related bile acids were used. Results LBP significantly improved the colonic tissue structure by upregulating MUC-2, claudin-1, and ZO-1 protein expression. The bacterial genus Dubosiella was dominant in healthy mice, but significantly decreased in mice treated with DSS. LBP rehabilitated Dubosiella in the sick guts of DSS mice to a level close to that of healthy mice. The levels of other beneficial bacterial genera Akkermansia and Bifidobacterium were also increased, whereas those of the harmful bacterial genera Turicibacter, Clostridium_sensu_stricto_1, Escherichia-Shigella, and Faecalibaculum decreased. The activity of beneficial bacteria promoted the bile acids lithocholic and deoxycholic acids in mice with UC, which improved the gut barrier function through the upregulation of TGR5. Conclusion The inflammatory microenvironment in the gut is determined by the balance of the gut microbiota. LBP showed great potential as a beneficial nutrient for rehabilitating Dubosiella which is dominant in the gut of healthy mice. Nutrient-related LBP may play an important role in gut health management.
Collapse
Affiliation(s)
- Rong Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Ping Yang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Bowen Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Ziru Ye
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Puyue Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Mingjian Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yanju Gong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yong Huang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Lan Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Min Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| |
Collapse
|
29
|
Guo H, Xie W, Ji Z, Wang B, Ren W, Gao W, Yuan B. Oyster Peptides Ameliorate Dextran Sulfate Sodium-Induced Ulcerative Colitis via Modulating the Gut Microbiota and Inhibiting the TLR4/NF-κB Pathway. Nutrients 2024; 16:1591. [PMID: 38892524 PMCID: PMC11175164 DOI: 10.3390/nu16111591] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease with an increasing prevalence year over year, and the medications used to treat patients with UC clinically have severe side effects. Oyster peptides (OPs) have anti-inflammatory and antioxidant properties as functional foods that can alleviate a wide range of inflammatory conditions. However, the application of oyster peptides in ulcerative colitis is not well studied. In this work, an animal model of acute colitis was established using 3% dextran sulfate sodium (DSS), and the impact of OP therapy on colitis in mice was examined. Supplementing with OPs prevented DSS-induced colitis from worsening, reduced the expression of oxidative stress and inflammatory markers, and restored the intestinal barrier damage caused by DSS-induced colitis in mice. The 16S rDNA results showed that the OP treatment improved the gut microbiota structure of the UC mice, including increasing microbial diversity, increasing beneficial bacteria, and decreasing harmful bacteria. In the UC mice, the OP therapy decreased the relative abundance of Family_XIII_AD3011_group and Prevotella_9 and increased the relative abundance of Alistipes. In conclusion, OP treatment can inhibit the TLR4/NF-κB pathway and improve the intestinal microbiota in UC mice, which in turn alleviates DSS-induced colitis, providing a reference for the treatment of clinical UC patients.
Collapse
Affiliation(s)
- Haixiang Guo
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (H.G.); (W.X.); (Z.J.); (B.W.); (W.R.)
| | - Wenyin Xie
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (H.G.); (W.X.); (Z.J.); (B.W.); (W.R.)
| | - Zhonghao Ji
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (H.G.); (W.X.); (Z.J.); (B.W.); (W.R.)
- Department of Basic Medicine, Changzhi Medical College, Changzhi 046000, China
| | - Bingbing Wang
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (H.G.); (W.X.); (Z.J.); (B.W.); (W.R.)
| | - Wenzhi Ren
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (H.G.); (W.X.); (Z.J.); (B.W.); (W.R.)
| | - Wei Gao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (H.G.); (W.X.); (Z.J.); (B.W.); (W.R.)
| | - Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (H.G.); (W.X.); (Z.J.); (B.W.); (W.R.)
| |
Collapse
|
30
|
Peng N, Wang J, Zhu H, Liu Z, Ren J, Li W, Wang Y. Protective effect of carbon dots as antioxidants on intestinal inflammation by regulating oxidative stress and gut microbiota in nematodes and mouse models. Int Immunopharmacol 2024; 131:111871. [PMID: 38492339 DOI: 10.1016/j.intimp.2024.111871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/12/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024]
Abstract
Inflammatory bowel disease (IBD) is a recurrent chronic colitis disease with increasing incidence and prevalence year by year. The single efficacy and significant side effects of traditional IBD treatment drugs have promoted the flourishing development of new drugs. Inspired by many health benefits of carbon dots (CDs) based nanomedicine in biomedical applications, a metal-free carbon dots (CP-CDs) was synthesized from citric acid and polyethylene polyamine to treat colitis. Oxidative stress tests at the cellular and nematode levels demonstrated CP-CDs have good antioxidant effects, while the toxicity of CP-CDs to cells and nematodes is low. CP-CDs were further applied to dextran sodium sulfate (DSS)-induced colitis in mice models, and it was found that CP-CDs can reduce the disease activity index (DAI) score of colon tissue and restore the intestinal barrier. Further, the anti-colitis mechanisms of CP-CDs were explored, one of which is to regulate intestinal oxidative stress in inflammatory mice, further reducing the expression of inflammatory cytokines, and thus alleviating colitis. Notably, 16S rRNA sequence analysis showed that the abundance of beneficial bacteria (Ligilactobacillus and Enterorhabdus) in the intestinal tract increased, while that of harmful bacteria (unclassified_Clostridia_UCG_014) decreased after CP-CDs treatment, indicating that CP-CDs rebalancing the gut microbiota destroyed by DSS is another important mechanism. In short, these non-toxic carbon dots not only have the potential for multi-factor combined relief of colitis but also offer an alternative therapy medicine for patients suffering from IBD.
Collapse
Affiliation(s)
- Nannan Peng
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China
| | - Jingmin Wang
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China.
| | - Haimei Zhu
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China
| | - Ziyue Liu
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China
| | - Jiayi Ren
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China
| | - Wenjing Li
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China
| | - Yongzhong Wang
- School of Life Sciences, Anhui University, Hefei 230601, Anhui, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei 230601, Anhui, China.
| |
Collapse
|
31
|
Wu R, Zhang Z, Xu Q, Liu F, Zhan Y, Wang Q, Du L, Tang X. Integration of network pharmacology and experimental verifications reveals the Bian-Se-Tong mixture can alleviate constipation in STC rats by reducing apoptosis of Cajal cells via activating PI3K-Akt signaling pathway. Heliyon 2024; 10:e28022. [PMID: 38586320 PMCID: PMC10998068 DOI: 10.1016/j.heliyon.2024.e28022] [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: 11/10/2023] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024] Open
Abstract
Bian-Se-Tong mixture (BSTM) is an optimized formulation based on the classical prescription "Zhizhu pill", which is widely used in the clinical treatment of slow-transit constipation (STC). The potential molecular mechanism of BSTM therapy for STC was investigated by network pharmacology prediction combined with animal experiments. The active components of BSTM were screened via the TCMSP platform. The GeneCards, OMIM and DrugBank databases were used to search for STC targets. With the help of the Biogenet tool, a protein interaction network between drugs and disease targets was constructed, and the intersection network of the two was extracted to obtain the key targets of BSTM in the treatment of STC. GO and KEGG enrichment analyses of key targets were carried out with Metascape. Loperamide hydrochloride was used to establish an STC rat model, and the key targets and related pathways were preliminarily verified. The important signaling pathways included the PI3K-Akt, MAPK, IL-17, cAMP, and cell cycle signaling pathways. The experimental results showed that BSTM treatment increased the body weight of STC rats and increased the fecal particle number, fecal water content and intestinal carbon ink promotion rate within 24 h. Further pathological changes in the colon of the rats were also observed. In-depth mechanistic studies have shown that BSTM can significantly reduce the apoptosis of intestinal Cajal cells, downregulate the expression of Bax and c-Caspase 3, upregulate the expression of Bcl-2 and c-kit, and promote the phosphorylation of AKT. The results showed that BSTM can significantly relieve constipation in STC rats via a mechanism related to activating the PI3K-Akt signaling pathway and improving Cajal cell apoptosis.
Collapse
Affiliation(s)
- Rong Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Zhibin Zhang
- North Sichuan Medical College, Nanchong 637000, China
| | - Qingxia Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Fang Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
- North Sichuan Medical College, Nanchong 637000, China
| | - Yu Zhan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Qiuxiao Wang
- North Sichuan Medical College, Nanchong 637000, China
| | - Lijuan Du
- Department of Anorectal, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610000, China
- Department of Anorectal, Chengdu Thrid People's Hospital, Chengdu 610000, China
| | - Xuegui Tang
- North Sichuan Medical College, Nanchong 637000, China
| |
Collapse
|
32
|
Zhao H, Mo Q, Kulyar MFEA, Guan J, Zhang X, Luo X, Li J. Metagenomic Analysis Reveals A Gut Microbiota Structure and Function Alteration between Healthy and Diarrheic Juvenile Yaks. Animals (Basel) 2024; 14:1181. [PMID: 38672329 PMCID: PMC11047321 DOI: 10.3390/ani14081181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Diarrhea-induced mortality among juvenile yaks is highly prevalent in the pastoral areas of the Qinghai-Tibet plateau. Although numerous diseases have been linked to the gut microbial community, little is known about how diarrhea affects the gut microbiota in yaks. In this work, we investigated and compared changes in the gut microbiota of juvenile yaks with diarrhea. The results demonstrated a considerable drop in the alpha diversity of the gut microbiota in diarrheic yaks, accompanied by Eysipelatoclostridium, Parabacteroides, and Escherichia-Shigella, which significantly increased during diarrhea. Furthermore, a PICRust analysis verified the elevation of the gut-microbial metabolic pathways in diarrhea groups, including glycine, serine, and threonine metabolism, alanine, aspartate, oxidative phosphorylation, glutamate metabolism, antibiotic biosynthesis, and secondary metabolite biosynthesis. Taken together, our study showed that the harmful bacteria increased, and beneficial bacteria decreased significantly in the gut microbiota of yaks with diarrhea. Moreover, these results also indicated that the dysbiosis of the gut microbiota may be a significant driving factor of diarrhea in yaks.
Collapse
Affiliation(s)
- Hongwen Zhao
- Sichuan Academy of Grassland Sciences, Chengdu 611731, China; (H.Z.); (J.G.); (X.Z.)
| | - Quan Mo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Q.M.); (M.F.-e.-A.K.)
| | | | - Jiuqiang Guan
- Sichuan Academy of Grassland Sciences, Chengdu 611731, China; (H.Z.); (J.G.); (X.Z.)
| | - Xiangfei Zhang
- Sichuan Academy of Grassland Sciences, Chengdu 611731, China; (H.Z.); (J.G.); (X.Z.)
| | - Xiaolin Luo
- Sichuan Academy of Grassland Sciences, Chengdu 611731, China; (H.Z.); (J.G.); (X.Z.)
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Q.M.); (M.F.-e.-A.K.)
| |
Collapse
|
33
|
Wang J, Wang X, Xiu W, Zhou Z, Yu S, Yang M, Zhou K, Ma Y. The sweet corn cob selenium polysaccharide alleviates type 2 diabetes via modulation of LPS/IκBα/NFκB and the intestinal microbiota. FOOD BIOSCI 2024; 58:103742. [DOI: 10.1016/j.fbio.2024.103742] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
34
|
Guo M, Fang L, Chen M, Shen J, Tan Z, He W. Dysfunction of cecal microbiota and CutC activity in mice mediating diarrhea with kidney-yang deficiency syndrome. Front Microbiol 2024; 15:1354823. [PMID: 38500584 PMCID: PMC10944907 DOI: 10.3389/fmicb.2024.1354823] [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: 12/13/2023] [Accepted: 02/22/2024] [Indexed: 03/20/2024] Open
Abstract
OBJECTIVE Previous studies have indicated that diarrhea with kidney-yang deficiency syndrome leads to a disorder of small intestine contents and mucosal microbiota. However, the relationship of TMA-lyase (CutC) activity and TMAO with diarrhea with kidney-yang deficiency syndrome remains unexplored. Therefore, this study explores the relationship between cecal microbiota and choline TMA-lyase (CutC) activity, as well as the correlation between trimethylamine oxide (TMAO), inflammatory index, and CutC activity. METHOD Twenty SPF-grade male KM mice were randomly divided into the normal group (CN) and the diarrhea model group (CD). Diarrhea mouse models were established by adenine combined with Folium sennae administration. CutC activity, TMAO, interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) levels were detected, and the cecal content microbiota was sequenced. RESULT After 14 days, diarrhea occurred in the CD group. Compared with the CN group, there was no significant change in the activity of CutC in the small intestine of the CD group, while the activity of CutC in the cecum was significantly increased, and the levels of TMAO, IL-6, and TNF-α showed a significant increase. The Chao1 index, Observed_species index, Shannon index, and Simpson index all exhibited a decreasing trend. The main changes at the bacterial genus level were Alistipes, Enterorhabdus, Desulfovibrio, Bacteroides, Candidatus_Saccharimonas, and [Ruminococcus]_torques_group. The results of LEfSe analysis, random forest analysis and ROC curve analysis revealed Paludicola, Blautia, Negativibacillus, Paraprevotella, Harryflintia, Candidatus_Soleaferrea, Anaerotruncus, Oscillibacter, Colidextribacter, [Ruminococcus]_torques_group, and Bacteroides as characteristic bacteria in the CD group. Correlation analysis showed a significant negative correlation between cecal CutC activity and Ligilactobacillus, and a significant positive correlation with Negativibacillus and Paludicola. The level of TMAO was significantly positively correlated with CutC activity and IL-6. CONCLUSION Diarrhea with kidney-yang deficiency syndrome significantly affects the physiological status, digestive enzyme activity, CutC activity, TMAO levels, and inflammatory response in mice. Additionally, there are changes in the composition and function of cecal microbiota, indicating an important impact of diarrhea with kidney-yang deficiency syndrome on the host intestinal microbiota balance. The occurrence of diarrhea with kidney-yang deficiency syndrome may be associated with dysbiosis of intestinal microbiota, increased CutC activity, elevated TMAO levels, and heightened inflammatory factor levels.
Collapse
Affiliation(s)
- Mingmin Guo
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Leyao Fang
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Meili Chen
- Changsha Hospital of Traditional Chinese Medicine, Changsha, China
| | - Junxi Shen
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Zhoujin Tan
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Wenzhi He
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
35
|
Wang D, Zeng J, Wujin C, Ullah Q, Su Z. Lactobacillus reuteri derived from horse alleviates Escherichia coli-induced diarrhea by modulating gut microbiota. Microb Pathog 2024; 188:106541. [PMID: 38224920 DOI: 10.1016/j.micpath.2024.106541] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 01/17/2024]
Abstract
Diarrhea is a prevalent health issue in farm animals and poses a significant challenge to the progress of animal husbandry. Recent evidence suggested that probiotics can alleviate diarrhea by maintaining gut microbial balance and enhancing the integrity of the intestinal barrier. However, there is a scarcity of studies investigating the efficacy of equine Lactobacillus reuteri in relieving E. coli-induced diarrhea. Hence, this study aimed to examine the potential of equine-derived Lactobacillus reuteri in alleviating E. coli diarrhea from the perspective of gut microbiota. Results demonstrated that supplementation of Lactobacillus reuteri had the potential to alleviate diarrhea induced by E. coli infection and restore the decline of tight junction genes, such as Claudin-1 and ZO-1. Additionally, Lactobacillus reuteri supplementation can restore the expression of inflammatory factors (IL-6, IL-10, TNF-α, and IFN-γ) and reduce colon inflammatory damage. Diversity analysis, based on amplicon sequencing, revealed a significant reduction in the diversity of gut microbiota during E. coli-induced diarrhea. Moreover, there were notable statistical differences in the composition and structure of gut microbiota among the different treatment groups. E. coli could induce gut microbial dysbiosis by decreasing the abundance of beneficial bacteria, including Lactobacillus, Bifidobacterium, Ligilactobacillus, Enterorhabdus, and Lachnospiraceae_UCG_001, in comparison to the control group. Conversely, supplementation with Lactobacillus reuteri could restore the abundance of beneficial bacteria and increase the diversity of the gut microbiota, thereby reshaping gut microbiota. Additionally, we also observed that supplementation with Lactobacillus reuteri alone improved the gut microbial composition and structure. In summary, the findings suggest that Lactobacillus reuteri can alleviate E. coli-induced diarrhea by preserving the integrity of the intestinal barrier and modulating the composition of the gut microbiota. These results not only contribute to understanding of the mechanism underlying the beneficial effects of Lactobacillus reuteri in relieving diarrhea, but also provide valuable insights for the development of probiotic products aimed at alleviating diarrheal diseases.
Collapse
Affiliation(s)
- Dongjing Wang
- Institute of Animal Husbandry and Veterinary, Tibet Autonomous Region Academy of Agricultural Sciences, Lhasa, Tibet, 850009, China
| | - Jiangyong Zeng
- Institute of Animal Husbandry and Veterinary, Tibet Autonomous Region Academy of Agricultural Sciences, Lhasa, Tibet, 850009, China
| | - Cuomu Wujin
- Institute of Animal Husbandry and Veterinary, Tibet Autonomous Region Academy of Agricultural Sciences, Lhasa, Tibet, 850009, China
| | - Qudrat Ullah
- Department of Theriogenology, Faculty of Veterinary and Animal Sciences, The University of Agriculture, Dera Ismail Khan, 29111, Pakistan
| | - Zhonghua Su
- Tibet Autonomous Region Animal Disease Prevention and Control Center, Lhasa, Tibet, 850009, China.
| |
Collapse
|
36
|
Wang L, Wu S, Chen T, Xiong L, Wang F, Song H, Zhou J, Wei S, Ren B, Shen X. A quinoa peptide protects impaired mucus barriers in colitis mice by inhibiting NF-κB-TRPV1 signaling and regulating the gut microbiota. Food Funct 2024; 15:1223-1236. [PMID: 38226896 DOI: 10.1039/d3fo04905a] [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: 01/17/2024]
Abstract
Inflammatory bowel diseases (IBD) are chronic inflammatory conditions that lead to the disruption of the colonic mucus barrier. Quinoa has a well-balanced profile of essential amino acids and exhibits excellent anti-inflammatory effects. We recently explored the beneficial effects and relevant mechanisms of a novel quinoa peptide TPGAFF on impaired mucus barriers in mice with chemically induced colitis. Our findings demonstrated that TPGAFF, administered in low and high doses for 28 days, effectively attenuated the pathological phenotype and reduced intestinal permeability in colitis mice. TPGAFF demonstrated its protective abilities by restoring the impaired mucus barrier, inhibiting the activation of inflammatory signaling and reducing inflammatory cytokine levels. Moreover, TPGAFF positively influenced the composition of the gut microbiota by reducing inflammation-related microbes. Additionally, TPGAFF inhibited the activation of TRPV1 nociceptor and decreased the levels of neuropeptides. Conclusively, our results indicated that oral administration of TPGAFF may be an optional approach for the treatment of mucus barrier damage.
Collapse
Affiliation(s)
- Luanfeng Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Shufeng Wu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Tong Chen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Ling Xiong
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Fang Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Jianxin Zhou
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Shixiang Wei
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| | - Bo Ren
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| | - Xinchun Shen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| |
Collapse
|
37
|
Yu Y, Liu Y, Meng Z. Role of traditional Chinese medicine in age-related macular degeneration: exploring the gut microbiota's influence. Front Pharmacol 2024; 15:1356324. [PMID: 38333011 PMCID: PMC10850396 DOI: 10.3389/fphar.2024.1356324] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
The pathogenesis of age-related macular degeneration (AMD), a degenerative retinopathy, remains unclear. Administration of anti-vascular endothelial growth factor agents, antioxidants, fundus lasers, photodynamic therapy, and transpupillary warming has proven effective in alleviating symptoms; however, these interventions cannot prevent or reverse AMD. Increasing evidence suggests that AMD risk is linked to changes in the composition, abundance, and diversity of the gut microbiota (GM). Activation of multiple signaling pathways by GM metabolites, including lipopolysaccharides, oxysterols, short-chain fatty acids (SCFAs), and bile acids (BAs), influences retinal physiology. Traditional Chinese medicine (TCM), known for its multi-component and multi-target advantages, can help treat AMD by altering GM composition and regulating the levels of certain substances, such as lipopolysaccharides, reducing oxysterols, and increasing SCFA and BA contents. This review explores the correlation between GM and AMD and interventions for the two to provide new perspectives on treating AMD with TCM.
Collapse
Affiliation(s)
- Yujia Yu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yong Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong Province Hospital of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhaoru Meng
- The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| |
Collapse
|
38
|
Lu W, Cen J, Dai Q, Tao H, Peng L. Gut microbiota does not play a mediating role in the causal association between inflammatory bowel disease and several its associated extraintestinal manifestations: a Mendelian randomization study. Front Immunol 2024; 14:1296889. [PMID: 38288127 PMCID: PMC10822939 DOI: 10.3389/fimmu.2023.1296889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/20/2023] [Indexed: 01/31/2024] Open
Abstract
Background and objectives Observational study has found inflammatory bowel disease to be associated with multiple extraintestinal manifestations. To this end, we characterized the causal association between inflammatory bowel disease and extraintestinal manifestations through a Mendelian randomization study and further explored the role of intestinal flora in inflammatory bowel disease and the extraintestinal manifestations associated with it. Materials and methods We genetically predicted the causal relationship between inflammatory bowel disease and twenty IBD-related extraintestinal manifestations (including sarcoidosis, iridocyclitis, interstitial lung disease, atopic dermatitis, ankylosing spondylitis, psoriatic arthropathies, primary sclerosing cholangitis, primary biliary cholangitis). We used the full genome-wide association study (GWAS) summary statistics on gut microbiota in 18,340 participants from 24 cohorts to explore its role in the casual relationships between IBD and IBD-related extraintestinal manifestations. Inverse variance weighting (IVW) was used as the main analytical method to assess the causal associations. We performed Cochran's Q test to examine the heterogeneity. To assess the robustness of the IVW results, we further performed sensitivity analyses including the weighted median method, MR-Egger regression, and Mendelian Randomization Pleiotropy RESidual Sum and Outlier (MR-PRESSO) test. The leave-one-out sensitivity analysis was further performed to monitor if significant associations were dominated by a single nucleotide polymorphism (SNP). Result A total of eight extraintestinal manifestations were found to be at elevated risk of development due to inflammatory bowel diseases. A total of 11 causal relationships were found between IBD and gut microbiota, four of which were stable. Between gut microbiota and these eight extraintestinal manifestations, a total of 67 nominal causal associations were identified, of which 13 associations were stable, and notably 4 associations were strongly correlated. Conclusion Through the two-sample MR analysis, we identified extraintestinal manifestations that were causally associated with inflammatory bowel disease and obtained multiple associations from inflammatory bowel disease and gut microbiota, and gut microbiota and extraintestinal manifestations in further analyses. These associations may provide useful biomarkers and potential targets for pathogenesis and treatment.
Collapse
Affiliation(s)
| | | | | | - Heqing Tao
- *Correspondence: Liang Peng, ; Heqing Tao,
| | - Liang Peng
- *Correspondence: Liang Peng, ; Heqing Tao,
| |
Collapse
|
39
|
Zhang Q, Zhang X, Wang Q, Chen S. Dioscoreae Rhizoma starch improves chronic diarrhea by regulating the gut microbiotas and fecal metabolome in rats. Food Sci Nutr 2023; 11:6271-6287. [PMID: 37823173 PMCID: PMC10563677 DOI: 10.1002/fsn3.3567] [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: 03/19/2023] [Revised: 06/11/2023] [Accepted: 07/05/2023] [Indexed: 10/13/2023] Open
Abstract
Chinese yam (Dioscorea opposite Thunb.) has been used as food and medicine to treat diarrhea for thousands of years. This article aimed to elucidate the potential mechanism of Dioscoreae Rhizoma starch in alleviating chronic diarrhea induced by rhubarb based on gut microbiotas and fecal metabolome. The administration of the Dioscoreae Rhizoma aqueous extracts, crude polysaccharides, and starch could improve diarrhea and alleviate intestinal injury in chronic diarrhea rats. The Dioscoreae Rhizoma starch displayed the most apparent effect on regulating intestinal microbiotas by increasing the abundance and diversity of microbiotas. At the genus level, there were 17 changed intestinal microbiotas in model rats, and the treatment with Dioscoreae Rhizoma starch regulated 11 microbiotas. Metabolomics analysis revealed that Dioscoreae Rhizoma starch could regulate abnormal fecal metabolites to alleviate diarrhea, and these metabolites are involved in phenylalanine, tyrosine, and tryptophan biosynthesis; tyrosine metabolism; vitamin B6 metabolism; and purine metabolism. This study will contribute to the further research and development of Dioscoreae Rhizoma starch.
Collapse
Affiliation(s)
- Qing Zhang
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
| | - Xu Zhang
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
| | - Qing Wang
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
| | - Suiqing Chen
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
| |
Collapse
|