1
|
Zhang B, Chen L, Kang M, Ai L, Tao Y. Gegen Qinlian Decoction improves Alzheimer's disease through TLR4/NF-κB/NLRP3 pathway. Tissue Cell 2025; 95:102818. [PMID: 40056656 DOI: 10.1016/j.tice.2025.102818] [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: 07/30/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025]
Abstract
OBJECTIVE Alzheimer's disease (AD) is a neurodegenerative disease that leads to dementia, but effective treatments are lacking. This study aims to evaluate the therapeutic effects of Gegen Qinlian Decoction (GGQLD) on AD and investigate the underlying mechanisms. METHODS Using network pharmacology and bioinformatics, we identified 376 active ingredients of GGQLD and 427 drug targets. Among these, 7 potential targets (CASP1, MKI67, NFKB1, TLR4, NLRP3, IL1B, and AKT1) were identified as intersecting targets of both GGQLD and AD. Functional enrichment analysis revealed that GGQLD regulates pyroptosis-related pathways. In vivo, GGQLD was administered to AD rat models to assess its effects on spatial learning, memory, and brain tissue injury. RESULTS GGQLD significantly reduced latency time by 40 % and increased platform crossings by 60 % in AD rats, demonstrating improved spatial learning and memory abilities. It also reduced hippocampal tissue damage and abnormal Aβ deposition. Mechanistically, GGQLD downregulated pyroptosis-related targets (TLR4, NF-κB, NLRP3, IL-1β, and Caspase-1), which were significantly upregulated in AD. ROC analysis demonstrated strong diagnostic significance for these genes, with AUC values exceeding 0.70. Functional enrichment and KEGG analysis further indicated that GGQLD exerts its therapeutic effects through multiple pathways, particularly the NOD-like receptor pathway, Necroptosis, and NF-kappa B pathway. CONCLUSIONS This study demonstrates that GGQLD improves spatial learning, reduces brain tissue damage, and alleviates inflammation in AD through the regulation of pyroptosis-related pathways, providing evidence for its potential as a therapeutic agent for AD.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China
| | - Liudan Chen
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China
| | - Mengru Kang
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China
| | - Liang Ai
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China
| | - Yangu Tao
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China.
| |
Collapse
|
2
|
Wu X, Cao T, Ye J, Shi R, Bao X, Ge Y, Li D, Hao S, Liu F, Liu X. Supplementation of 2'-Fucosyllactose during the Growth Period Improves Neurodevelopmental Disorders in Offspring Mice Induced by Maternal Immune Activation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40350763 DOI: 10.1021/acs.jafc.5c01184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Autism spectrum disorder is a serious neurodevelopmental disorder whose early onset significantly affects an individual's social interactions and cognitive function. Recent research suggests that modulating the gut microbiota could be a potential intervention strategy for autism spectrum disorder symptoms. 2'-Fucosyllactose has been identified as a regulator of gut microbiota homeostasis, however, its effectiveness in addressing autism spectrum disorder remains unclear. In this study, the effects of daily supplementation of 2'-FL in 3-week-old male offspring mice for 5 weeks were examined. The results showed that 2'-fucosyllactose significantly improved autism spectrum disorder-like behavioral deficits. Furthermore, supplementation with 2'-fucosyllactose restored intestinal barrier integrity and increased relative abundance of beneficial gut bacteria, particularly Akkermansia and Bifidobacterium that are closely related to bile acid metabolism. Notably, 2'-fucosyllactose treatment elevated the content of bile acids and upregulated the expression of bile acid receptors in the brain. Co-housing experiments further confirmed the crucial role of gut microbiota in mediating the beneficial effects of 2'-fucosyllactose. Overall, this study suggests that 2'-fucosyllactose could alleviate maternal immune activation-induced behavioral deficits and neuroinflammation through the regulation of the gut-brain axis, offering potential therapeutic value.
Collapse
Affiliation(s)
- Xiaoning Wu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Tengzheng Cao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jin Ye
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Renjie Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xiaowei Bao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yunshu Ge
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Dongning Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Shijin Hao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Feitong Liu
- H&H Research, China Research and Innovation Center, H&H Group, Guangzhou 510700, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| |
Collapse
|
3
|
Xu W, Liu A, Gong Z, Xiao W. L-theanine prevents ulcerative colitis by regulating the CD4+ T cell immune response through the gut microbiota and its metabolites. J Nutr Biochem 2025; 139:109845. [PMID: 39922394 DOI: 10.1016/j.jnutbio.2025.109845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 12/29/2024] [Accepted: 01/20/2025] [Indexed: 02/10/2025]
Abstract
The disturbance of gut microbiota and its metabolites are considered to be the causes of ulcerative colitis (UC), which leads to immune abnormalities. Diet is the most important regulator of gut microbiota; therefore, it has a beneficial impact on UC. A novel food ingredient, l-theanine, alters the gut microbiota, thereby regulating gut immunity. However, whether l-theanine prevents UC by altering the gut microbiota, as well as the underlying mechanisms, remains unknown. Here, l-theanine was used to optimize the gut microbiota and its metabolites. Furthermore, to explore the mechanism by which l-theanine prevents UC, an l-theanine fecal microbiota solution was used to prevent dextran sulfate sodium-induced UC via fecal microbiota transplantation. Improvements in the colonic structure, colon histology scores, immune factors (IL-10), and inflammatory factors (IL-1β) demonstrated the preventive effect of l-theanine on UC. The 16S rDNA and metabolomic results showed that tryptophan-, short chain fatty acid-, and bile acid-related microbiota, such as Muribaculaceae, Lachnospiraceae, Alloprevotella, and Prevotellaceae were the dominant. Flow cytometry results showed that l-theanine decreased helper T (Th)1 and Th17 immune responses, and increased Th2 and T-regulatory immune responses via regulation of antigen-presenting cell responses, such as dendritic cells and macrophages. Therefore, l-theanine regulated the immune response of colon CD4 + T cells to dendritic cell and macrophage antigen presentation via tryptophan-, short chain fatty acid-, and bile acid-related microbiota, thereby preventing dextran sulfate sodium-induced UC.
Collapse
Affiliation(s)
- Wei Xu
- Key Lab of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, China; National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan, China
| | - Aoxiang Liu
- Key Lab of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, China; National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan, China
| | - Zhihua Gong
- Key Lab of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, China; National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan, China; Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan, China
| | - Wenjun Xiao
- Key Lab of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, China; National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan, China; Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, Hunan, China.
| |
Collapse
|
4
|
Lu H, Zhang G, Wang K, Liu K, Gao Y, Chen J, Li Y, Yan J. The Role of Lactiplantibacillus plantarum CGMCC9513 in Alleviating Colitis by Synergistic Enhancement of the Intestinal Barrier Through Modulating Gut Microbiota and Activating the Aryl Hydrocarbon Receptor. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10551-0. [PMID: 40301232 DOI: 10.1007/s12602-025-10551-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2025] [Indexed: 05/01/2025]
Abstract
Ulcerative colitis (UC) has become a global health issue. This study evaluated whether administering Lactiplantibacillus plantarum CGMCC9513 to dextran sulfate sodium (DSS)-induced colitis mice could alleviate colitis by modulating gut microbiota imbalance and activating the aryl hydrocarbon receptor (AhR) to enhance intestinal barrier function. The anti-inflammatory effect and AhR activation ability of L. plantarum CGMCC9513 were evaluated with lipopolysaccharide (LPS)-induced cell inflammation model; 25 male BALB/c mice were divided into blank group (CNG), model group (DSS), L. plantarum CGMCC9513-treated group (LPDT), and L. plantarum CGMCC9513 control group (LP). The mice were pre-administered L. plantarum CGMCC9513 for 14 days and continued to receive it during DSS induction. Symptoms during induction, goblet cell count, expression of MUC2 and Occludin proteins, and changes in gut microbiota were observed. Subsequently, the expression of cytokines interleukin-10 (IL-10), tumor necrosis factor (TNF-α), interleukin-1β (IL-1β) and AhR activation status was determined. The study found that L. plantarum CGMCC9513 could alleviate cell inflammation induced by LPS and activate AhR in vitro. For colitis mice, it could reduce colonic mucosal damage and enhance intestinal barrier function. Regarding gut microbiota changes, L. plantarum CGMCC9513 mainly downregulated Bacteroides, Blautia, Escherichia-Shigella, and Lachnospiraceae_ NK4A136_group and upregulated Firmicutes, Lactobacillus. It reduces the risk of bacterial translocation and increases beneficial gut bacteria. L. plantarum CGMCC9513 reduced the expression of pro-inflammatory cytokines TNF-α and IL-1β while increasing the expression of anti-inflammatory cytokine IL-10. Meanwhile, increased expression of AhR and Cytochrome P450 1A1 (CYP1A1) proteins indicated AhR activation by L. plantarum CGMCC9513. In conclusion, L. plantarum CGMCC9513 can synergistically enhance intestinal barrier alleviation in colitis mice by modulating gut microbiota imbalance and activating AhR.
Collapse
Affiliation(s)
- Hongyu Lu
- Medical School of Guangxi University, Nanning, 530004, China
| | - Guoqing Zhang
- Medical School of Guangxi University, Nanning, 530004, China
| | - Kaidi Wang
- Medical School of Guangxi University, Nanning, 530004, China
| | - Kefei Liu
- Tianjin Shengji Group., Co., Ltd, Huayuan Industrial Zone, No. 2, Hai Tai Development 2 Road, Tianjin, 300384, China
| | - Yingrui Gao
- Tianjin Shengji Group., Co., Ltd, Huayuan Industrial Zone, No. 2, Hai Tai Development 2 Road, Tianjin, 300384, China
| | - Jinyan Chen
- Tianjin Shengji Group., Co., Ltd, Huayuan Industrial Zone, No. 2, Hai Tai Development 2 Road, Tianjin, 300384, China
| | - Yixiang Li
- Medical School of Guangxi University, Nanning, 530004, China.
| | - Jianhua Yan
- Medical School of Guangxi University, Nanning, 530004, China.
| |
Collapse
|
5
|
Liu Y, Zhang JT, Sun M, Song J, Sun HM, Wang MY, Wang CM, Liu W. Targeting ferroptosis in the treatment of ulcerative colitis by traditional Chinese medicine: A novel therapeutic strategies. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156539. [PMID: 39987602 DOI: 10.1016/j.phymed.2025.156539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/04/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND The incidence of ulcerative colitis (UC) has been rising rapidly in recent years, and there is currently no effective method to prevent its recurrence. Owing to its long treatment duration, difficulty in treatment, prolonged remission, and high costs, it has attracted global attention. Exploring safe, effective, and sustainable treatment regimens has become an urgent global issue. Traditional Chinese medicine (TCM) has unique advantages such as low cost, low drug resistance, and fewer side effects, and has accumulated rich experience in the treatment of UC. PURPOSE Ferroptosis, as a new form of non-apoptotic cell death, is characterized by iron homeostatic imbalance and lipid peroxidation in the redox system. Studies have shown that inhibited ferroptosis in intestinal epithelial cells can protect the intestinal mucosa. Targeted intervention in ferroptosis may be a new direction for the treatment of UC. METHODS We conducted a systematic literature search with Google Scholar, PubMed, Web of Science, ScienceDirect and X-mol databases have been utilized to retrieve relevant literature up to October 2024, using keywords included ferroptosis, Inflammatory bowel disease (IBD), UC, Crohn's disease and TCM, Chinese traditional prescription, Chinese medicine extract and active ingredients. The existing literature was comprehensively studied and sorted out. RESULTS Currently, UC is mainly treated with drugs, including corticosteroids, amino salicylates, biologics, and immunomodulators, but drug resistance and adverse reactions are common. Increasing evidence suggests that TCM may treat UC by interfering with ferroptosis. Scholars have confirmed that TCM can inhibit ferroptosis, and recent studies have shown that TCM can not only inhibit iron dependent lipid peroxidation in intestinal cells but also enhance the antioxidant and anti-inflammatory abilities of intestinal mucosa, thus playing a role in the treatment of UC. This review explores the relevance of TCM intervention in ferroptosis and the treatment of UC, discusses the possible mechanisms of ferroptosis in UC, and aims to provide a basis for the diagnosis and treatment of UC. CONCLUSION It is revealed that TCM targeted ferroptosis has a good application prospect in the treatment of UC, providing a theoretical basis for elucidating the pathogenesis of UC and the study of TCM targeting ferroptosis regulating lipid metabolism in the treatment of UC, and providing a new perspective for the treatment of IBD in the future.
Collapse
Affiliation(s)
- Ying Liu
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Jing-Tian Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Meng Sun
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Jian Song
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Hai-Ming Sun
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Meng-Yang Wang
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Chun-Mei Wang
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Wei Liu
- College of Pharmacy, Beihua University, jilin 132013, China.
| |
Collapse
|
6
|
Zhao L, Jiang T, Zhang Y, Shen Z. Epimedium polysaccharides ameliorate ulcerative colitis by inhibiting oxidative stress and regulating autophagy. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:2655-2670. [PMID: 39540346 DOI: 10.1002/jsfa.14037] [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: 05/09/2024] [Revised: 10/09/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Epimedium polysaccharide (EPS) is a bioactive compound derived from the traditional Chinese herb Epimedium brevicornum. The objective of this study was to investigate the protective effects of EPS on ulcerative colitis (UC) and to elucidate the underlying mechanisms involved. RESULTS The findings showed that EPS treatment mitigated UC symptoms, including weight loss, anal bleeding, elevated disease activity index (DAI), and colon shortening. Hematoxylin and eosin (H&E) and Alcian blue-periodic acid-Schiff (AB-PAS) staining demonstrated that EPS alleviated histopathological damage and improved the integrity of the colonic mucosa. Mechanistically, EPS was found to substantially decrease inflammation by inhibiting the Toll-like receptor 4/nuclear factor-κB (TLR4/NF-κB) signaling pathway and to alleviate oxidative stress through modulation of the Kelch-like ECH-associated protein 1/nuclear factor erythroid-derived 2-like 2 (Keap1/Nrf2) pathway. Notably, EPS failed to exert protective effects against dextran sulfate sodium (DSS)-induced UC in Nrf2-knockout (Nrf2-/-) mice. Additionally, Western blotting and immunohistochemical analysis demonstrated that EPS facilitated autophagy via the adenosine monophosphate-dependent protein kinase/mammalian target of rapamycin (AMPK/mTOR) pathway. In vitro experiments revealed that EPS effectively suppressed lipopolysaccharide (LPS)-mediated cellular damage and oxidative stress by regulating Keap1/Nrf2 pathway. Transcriptomic analysis of LPS-treated Caco-2 cells following EPS treatment revealed a significant up-regulation of Nrf2 expression. CONCLUSION In conclusion, the findings of this study suggest that EPS exerts protective effects against DSS-induced UC through the inhibition of the TLR4/NF-κB signaling pathway, regulation of the Keap1/Nrf2 pathway, and promotion of autophagy via the AMPK/mTOR pathway. Consequently, EPS may represent a potential therapeutic target for the treatment of UC. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Linxian Zhao
- Department of Gastrointestinal, Colorectal, and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Tao Jiang
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuxin Zhang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhen Shen
- Department of Gastrointestinal, Colorectal, and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
7
|
Zheng J, Sun T, Qin T, Wu Y, Zhang W, Qiu Y, Chen J. Tryptophan Attenuates Chronic Restraint Stress-Induced Intestinal Injury Through Modulation of Intestinal Barrier Integrity and Gut Microbiota Homeostasis. Nutrients 2025; 17:975. [PMID: 40290020 PMCID: PMC11944976 DOI: 10.3390/nu17060975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Chronic stress is associated with detrimental effects on physical health, such as chronic restraint stress (CRS), which can damage the intestinal tract. Although tryptophan has many benefits in maintaining intestinal health, the underlying mechanism of its protective effects against stress-induced intestinal injury remains unclear. Methods: In this study, we constructed a CRS model by using a behavioral restraint device in which mice were restrained for 6 h per day over 14 days and investigated the effects, as well as the potential mechanism of a high-tryptophan diet (0.4% tryptophan), on CRS-induced intestinal injury using scanning electron microscopy, 16S rRNA sequencing, and LC-MS. Results: A 0.4% tryptophan diet (fed ad libitum for 24 days) attenuated CRS-induced pathologies, including weight loss, elevated corticosterone, intestinal barrier injury, increased permeability, and epithelial apoptosis. Tryptophan modulated the gut microbiota composition in CRS-induced mice, increasing the abundance of Bacteroidota and decreasing the abundance of Firmicutes, as well as enhancing metabolic function through pathways identified by KEGG analysis. Additionally, tryptophan restored the levels of short-chain fatty acids (SCFAs), including acetic, propionic, isobutyric, butyric, and valeric acids. Correlation analyses showed interactions between tryptophan, intestinal permeability, SCFAs, and gut microbiota. Conclusions: Tryptophan supplementation attenuates CRS-induced intestinal injury by modulating intestinal barrier integrity and gut microbiota homeostasis, and the beneficial effects are largely associated with the SCFA-mediated regulation of intestinal permeability and microbiota-associated energy metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | - Yefeng Qiu
- Academy of Military Medical Sciences, Beijing 100193, China; (J.Z.); (T.S.); (T.Q.); (Y.W.); (W.Z.)
| | - Jingqing Chen
- Academy of Military Medical Sciences, Beijing 100193, China; (J.Z.); (T.S.); (T.Q.); (Y.W.); (W.Z.)
| |
Collapse
|
8
|
Zhao Y, Cui D, Xiao Y, Han X, Jiang M, Gong Y. Clinical Analysis and Network Pharmacology in Revealing the Mechanism of Daifu Decoction on the Relapse of UC. Drug Des Devel Ther 2025; 19:1629-1653. [PMID: 40070532 PMCID: PMC11895689 DOI: 10.2147/dddt.s497944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Background Daifu Decoction (DFD), a patented herbal prescription used to prevent and treat ulcerative colitis (UC). This study aimed to reveal the effect of DFD on the relapse of UC and its mechanism via integrated retrospective clinical analysis, network pharmacology and in vivo and in vitro experimental validation. Methods First, the clinical data of UC patients treated with DFD were reviewed from a real-world study (RWS), and the relapse at 24 weeks after drug withdrawal was recorded to evaluate the relapse rate. Next, the chemical components of DFD were identified via ultra performance liquid chromatography‒mass spectrometry (UPLC‒MS), and the differentially expressed genes (DEGs) between UC patients in the active and remission stages were screened as disease targets related to the relapse of UC from the Gene Expression Omnibus (GEO) database. The core components, targets and key signalling pathways of DFD for preventing the relapse of UC were discussed via network pharmacology. Finally, the above results were verified via molecular docking and in vivo and in vitro experiments. Results A total of 475 UC patients were included, and the relapse rate of UC treated with DFD was 23.9%. Additionally, the 221 components identified by UPLC-MS and 398 DEGs related to the relapse of UC enriched the main pathway of the relapse of UC was IL-17 signaling pathway and the inflammatory-related targets, such as IL6, PTGS2, MMP7, MMP3, MMP1. Moreover, molecular docking revealed that the core components of DFD were able to bind to inflammation-related targets, and in vivo and in vitro experiments demonstrated that DFD could inhibit the IL-17 pathway, increase the level of claudin-1, and control inflammation to prevent UC relapse. Conclusion DFD can effectively prevent the relapse of UC which may be related to inhibiting the activation of IL-17 signalling pathway.
Collapse
Affiliation(s)
- Yangyang Zhao
- Department of Traditional Chinese Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning, People’s Republic of China
| | - Danyang Cui
- Department of Traditional Chinese Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning, People’s Republic of China
| | - Yanan Xiao
- Department of Traditional Chinese Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning, People’s Republic of China
| | - Xu Han
- Institute of Basic Research in Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing, People’s Republic of China
| | - Miao Jiang
- Institute of Basic Research in Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing, People’s Republic of China
| | - Yang Gong
- Department of Traditional Chinese Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning, People’s Republic of China
| |
Collapse
|
9
|
Shan C, Liu T, Miao F, Guo G. Macadamia oil alleviates dextran sulfate sodium-induced ulcerative colitis in mice via activating the Nrf2/Ho-1 pathway. Food Sci Biotechnol 2025; 34:1027-1036. [PMID: 39974866 PMCID: PMC11832958 DOI: 10.1007/s10068-024-01719-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/08/2024] [Accepted: 09/18/2024] [Indexed: 02/21/2025] Open
Abstract
Macadamia nut oil (MO) fatty acids are mainly composed of oleic acid and palmitoleic acid, which have a variety of health benefits. This study established an ulcerative colitis (UC) mouse model using dextran sulfate sodium (DSS), and the ameliorative effects of MO on UC were investigated. The results revealed that MO supplementation mitigated weight loss and colon shortening, increased goblet cell counts, and alleviated histopathologic changes in UC mice. MO significantly increased the intestinal antioxidant levels in UC mice. Moreover, Nrf2 and Ho-1 mRNA and protein expression levels were significantly upregulated in UC mice following treatment with low- and high-dose MO. In contrast, expression levels of Keap1 were significantly downregulated. Lastly, MO inhibited the inflammatory factors (TNF-α, IL-6 and IL-1β) expression in UC mice. These results indicate that MO could enhance colonic antioxidant levels, induce apoptosis, and activate the Nrf2/Ho-1 pathway, thereby ameliorating the pathological injuries associated with UC.
Collapse
Affiliation(s)
- Chunlan Shan
- College of Animal Science, Guizhou University, Guiyang, 550025 P. R. China
| | - Ting Liu
- College of Animal Science, Guizhou University, Guiyang, 550025 P. R. China
| | - Fujun Miao
- Yunnan Academy of Forestry and Grassland, Kunming, 650204 China
| | - Gangjun Guo
- Yunnan Institute of Tropical Crops, Jinghong, 666100 P. R. China
| |
Collapse
|
10
|
Bai X, Liu B, Fan D, Lu Y, Zhao X. Modulating the gut microbiota: A novel perspective in colorectal cancer treatment. Cancer Lett 2025; 612:217459. [PMID: 39805389 DOI: 10.1016/j.canlet.2025.217459] [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: 10/08/2024] [Revised: 01/07/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Colorectal cancer (CRC), the second leading cause of cancer-related deaths worldwide, is intricately linked to the dysregulation of the gut microbiota. Manipulating the gut microbiota has emerged as a novel strategy for the prevention and treatment of CRC. Natural products, a pivotal source in new drug discovery, have shown promise in recent research as regulators of the gut microbiota, offering potential applications in the prevention and treatment of CRC. In this work, commencing with a focus on the gut microbiota, we first elucidate the latest research on the intricate relationship between the gut microbiota and CRC. Additionally, we explore the impact of the gut microbiota on immunotherapy and chemotherapy treatments for CRC. Subsequently, we review the latest research findings on the regulation of the gut microbiota for CRC prevention through various mechanisms by natural products. These mechanisms include promoting the growth of beneficial bacteria, eradicating harmful bacteria, and enhancing the synthesis of beneficial metabolites. Furthermore, we summarize the advancements in research on natural products that alleviate chemotherapy toxicity and enhance the efficacy of immunotherapy by modulating the gut microbiota. Ultimately, we aspire to leverage advancements in nanomedicine and multiomics technologies to gain a deeper understanding of the mechanisms by which natural products regulate the gut microbiota. This work leverages gut microbiota as a focal point, aiming to offer new perspectives for developing novel natural products for colorectal cancer prevention and treatment.
Collapse
Affiliation(s)
- Xue Bai
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Boyang Liu
- Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710000, China
| | - Daiming Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yuanyuan Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Xiaodi Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
11
|
Deng H, Zhao Y, He Y, Teng H, Chen L. Unveiling the Dark Side of Flavonoid: Rutin Provokes Hepatotoxicity in Low-Dose 2-Amino-3-methylimidazo [4,5- f] Quinoline-Exposed Mice via Regulating Gut Microbiota and Liver Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:4253-4269. [PMID: 39862407 DOI: 10.1021/acs.jafc.4c07330] [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: 01/27/2025]
Abstract
2-Amino-3-methylimidazole [4,5-f] quinoline (IQ) is a kind of heterocyclic amine (HCAs) with high carcinogenicity in hot processed meat. Rutin (Ru) is a flavonoid compound with anti-inflammatory and antioxidant properties. However, whether Ru is scatheless under IQ-stimulated potential unhealthy conditions, especially liver function, in vivo, is unknown. In this study, we explored the effects and underlying mechanism of Ru on liver injury induced by a low dose of IQ in mice. Results showed that Ru supplement led to liver injury upon low-dose IQ alone administration, as shown by histological analysis, inflammatory, and serum biochemical indexes. Additionally, nontargeted metabolomics analysis revealed that coexposure of Ru and IQ disrupted liver metabolic balance, leading to significant changes in metabolites and metabolic pathways, hinting at a possible relationship with intestinal microbiota. Furthermore, the 16S rRNA sequencing data indicated that a combination of Ru and IQ caused gut microbiota dysbiosis and decreased the level of short-chain fatty acids (SCFAs). Correlation analysis between gut microbiota, SCFAs, liver metabolites, and liver damage markers highlighted the crucial role of the gut-liver axis in IQ and Ru coexposure-induced liver injury in vivo. In general, this study offers a valuable perspective on flavones and HCA compounds in the realms of food safety and human health.
Collapse
Affiliation(s)
- Hongting Deng
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Provincial Engineering Technology Research Center of Seafood, Zhanjiang, Guangdong 524000, People's Republic of China
- Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, Guangdong 524000, People's Republic of China
| | - Yanan Zhao
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Provincial Engineering Technology Research Center of Seafood, Zhanjiang, Guangdong 524000, People's Republic of China
- Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, Guangdong 524000, People's Republic of China
| | - Yuanju He
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Provincial Engineering Technology Research Center of Seafood, Zhanjiang, Guangdong 524000, People's Republic of China
- Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, Guangdong 524000, People's Republic of China
| | - Hui Teng
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Provincial Engineering Technology Research Center of Seafood, Zhanjiang, Guangdong 524000, People's Republic of China
- Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, Guangdong 524000, People's Republic of China
| | - Lei Chen
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang, Guangdong 524000, People's Republic of China
- Guangdong Provincial Engineering Technology Research Center of Seafood, Zhanjiang, Guangdong 524000, People's Republic of China
- Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, Guangdong 524000, People's Republic of China
- Shenzhen Institute of Guangdong Ocean University, Shenzhen 518108, China
| |
Collapse
|
12
|
Liang C, Chen S, Liu C, Wang L, Cui H, Du K, Wei W, He J, Li J, Chang Y. Active components unveiling and pharmacodynamic research on Valeriana jatamansi jones for ameliorating ulcerative colitis based on pharmacokinetics and network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119299. [PMID: 39743185 DOI: 10.1016/j.jep.2024.119299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Valeriana jatamansi Jones (V. jatamansi), a traditional Chinese medicine, is widely used in the treatment of gastrointestinal disorders, such as ulcerative colitis (UC). However, the active components of V. jatamansi with protective effect on the intestinal barrier remains elusive. AIM OF THE STUDY This investigation was conducted to confirm the efficacy of V. jatamansi ethanol extract for the treatment of UC and to identify the active compounds in ethanol extract of V. jatamansi that have a protective effect against intestinal barrier damage. MATERIALS AND METHODS The role of V. jatamansi extract was assessed on dextrose sodium sulfate (DSS) induced colitis in vivo. The wound healing, apoptosis and epithelial barrier permeability of intestinal epithelial cells (NCM460 cells) were evaluated in vitro. UHPLC-MS method was used to determine the pharmacokinetic characteristics of V. jatamansi extract after oral administration. The effect of absorbed compounds on the intestinal barrier was assessed on NCM460 cells. Fuzzy matter-element analysis was used to explore the main compounds, and the results were verified by FITC-dextran assay. Network pharmacology was used to predict the potential mechanisms and western blotting was used to validate the results. RESULTS The ethanol extract ofV. jatamansi relieves symptoms, inflammatory response and intestinal barrier damage in DSS-induced UC. It effectively alleviated DSS-induced epithelial barrier damage in NCM460 cells. Pharmacokinetic analyses indicated that the five components (chlorogenic acid, hesperidin, valerosidate, isochlorogenic acid B and cryptochlorogenic acid) were quickly absorbed into the blood after oral administration of ethanol extract. The fuzzy matter-element analysis and FITC-dextran assay demonstrated that valerosidate and chlorogenic acid played an important role in the protection of the intestinal barrier damage. Network pharmacological analyses and western blotting analyses showed that the ethanol extract of and absorbed components significantly inhibited the Bcl-2/Bax/Caspase-3 signaling pathway, and suppressed DSS-induced apoptosis in NCM460 cells. CONCLUSION The ethanol extract of V. jatamansi ameliorates intestinal barrier injury in UC mice through multi-components. Valerosidate and chlorogenic acid were identified as anti-colitis compounds. These provided new insights into finding the active components for the treatment of UC and contribute to the clinical application of V. jatamansi.
Collapse
Affiliation(s)
- Chunxiao Liang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Phytochemistry and Pharmaceutical Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shujing Chen
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Phytochemistry and Pharmaceutical Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Changqing Liu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lirong Wang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Huan Cui
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Kunze Du
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Phytochemistry and Pharmaceutical Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wei Wei
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Phytochemistry and Pharmaceutical Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Jun He
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Phytochemistry and Pharmaceutical Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jin Li
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yanxu Chang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Phytochemistry and Pharmaceutical Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
13
|
Zhang Y, Zhu M, Dai Y, Gao L, Cheng L. Research Progress in Ulcerative Colitis: The Role of Traditional Chinese Medicine on Gut Microbiota and Signaling Pathways. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2277-2336. [PMID: 39756829 DOI: 10.1142/s0192415x24500885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Ulcerative colitis (UC), one among other refractory diseases worldwide, has shown an increasing trend of progression to colorectal cancer in recent years. In the treatment of UC, traditional Chinese medicine has demonstrated good efficacy, with a high cure rate, fewer adverse effects, great improvement in the quality of patient survival, and reduction in the tendency of cancerous transformation. It shows promise as a complementary and alternative therapy. This review aims to evaluate and discuss the current research on UC, signaling pathways, and gut microbiota. We also summarized the mechanisms of action of various Chinese medicines (active ingredients or extracts) and herbal formulas, through signaling pathways and gut microbiota, with the expectation that they can provide references and evidence for treating UC and preventing inflammation-associated colorectal cancer by traditional Chinese medicine. We illustrate that multiple signaling pathways, such as TLR4, STAT3, PI3K/Akt, NF-[Formula: see text]B, and Keap1/Nrf2, can be inhibited by Chinese herbal treatments through the combined regulation of signaling pathways and gut microbiota, which can act individually or synergistically to inhibit intestinal inflammatory cell infiltration, attenuate gut oxidative responses, and repair the intestinal barrier.
Collapse
Affiliation(s)
- Yuyi Zhang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Mingfang Zhu
- Graduate School, Zunyi Medical University Zunyi, P. R. China
| | - Yueying Dai
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Longying Gao
- Department of Anorectal, The First Affiliated Hospital of Heilongjiang, University of Chinese Medicine Harbin, P. R. China
| | - Limin Cheng
- Department of Anorectal, The First Affiliated Hospital of Heilongjiang, University of Chinese Medicine Harbin, P. R. China
| |
Collapse
|
14
|
Xu Y, Cai Q, Zhao C, Zhang W, Xu X, Lin H, Lin Y, Chen D, Lin S, Jia P, Wang M, Zhang L, Lin W. Gegen Qinlian Decoction Attenuates Colitis-Associated Colorectal Cancer via Suppressing TLR4 Signaling Pathway Based on Network Pharmacology and In Vivo/In Vitro Experimental Validation. Pharmaceuticals (Basel) 2024; 18:12. [PMID: 39861077 PMCID: PMC11768880 DOI: 10.3390/ph18010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/29/2024] [Accepted: 12/13/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Gegen Qinlian Decoction (GQD), is used for intestinal disorders like ulcerative colitis, irritable bowel syndrome, and colorectal cancer. But the precise mechanisms underlying its anti-inflammatory and anti-tumor effects are not fully elucidated. Methods: Use network pharmacology to identify targets and pathways of GQD. In vivo (azoxymethane/dextran sodium sulfate (AOM/DSS)-induced colitis-associated colorectal cancer (CAC) mouse model) and in vitro (lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages) experiments were conducted to explore GQD's anti-inflammatory and anti-tumor effects. We monitored mouse body weight and disease activity index (DAI), and evaluated colon cancer tissues using hematoxylin and eosin staining. Expression of Ki67 and F4/80 was determined by immunohistochemistry analysis. The protein levels of TLR4 signaling pathway were assessed by western blotting analysis. Enzyme-linked immunosorbent assay measured IL-1β, IL-6, and TNF-α levels. Immunofluorescence (IF) staining visualized NF-κB and IRF3 translocation. Results: There were 18, 9, 24 and 77 active ingredients in the four herbs of GQD, respectively, targeting 435, 156, 485 and 691 genes. Through data platform analysis, it was concluded that there were 1104 target genes of GQD and 2022 target genes of CAC. Moreover, there were 99 intersecting genes between GQD and CAC. The core targets of GQD contained NFKB1, IL1B, IL6, TLR4, and TNF, and GQD reduced inflammation by inhibiting the TLR4 signaling pathway. In vivo experiment, GQD increased mouse body weight, lowered DAI scores, while also alleviating histopathological changes in the colon and decreasing the expressions of Ki67 and F4/80 in the AOM/DSS-induced mice. GQD reduced IL-1β, IL-6, and TNF-α levels in the serum and downregulated TLR4, MyD88, and phosphorylation of IκBα, P65, and IRF3 in the colon tissue from AOM/DSS-induced mice. In vitro, GQD suppressed pro-inflammatory cytokines and TLR4 signaling pathway in the LPS-induced RAW264.7 cells, and combined with TAK242, it further reduced the phosphorylation of IκBα, P65. Conclusions: GQD mitigated CAC by inhibiting the TLR4 signaling pathway, offering a potential therapeutic approach for CAC management.
Collapse
Affiliation(s)
- Yaoyao Xu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Y.X.); (Q.C.); (C.Z.); (W.Z.); (X.X.); (H.L.); (Y.L.); (S.L.); (P.J.); (M.W.)
| | - Qiaoyan Cai
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Y.X.); (Q.C.); (C.Z.); (W.Z.); (X.X.); (H.L.); (Y.L.); (S.L.); (P.J.); (M.W.)
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Chunyu Zhao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Y.X.); (Q.C.); (C.Z.); (W.Z.); (X.X.); (H.L.); (Y.L.); (S.L.); (P.J.); (M.W.)
| | - Weixiang Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Y.X.); (Q.C.); (C.Z.); (W.Z.); (X.X.); (H.L.); (Y.L.); (S.L.); (P.J.); (M.W.)
| | - Xinting Xu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Y.X.); (Q.C.); (C.Z.); (W.Z.); (X.X.); (H.L.); (Y.L.); (S.L.); (P.J.); (M.W.)
| | - Haowei Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Y.X.); (Q.C.); (C.Z.); (W.Z.); (X.X.); (H.L.); (Y.L.); (S.L.); (P.J.); (M.W.)
| | - Yuxing Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Y.X.); (Q.C.); (C.Z.); (W.Z.); (X.X.); (H.L.); (Y.L.); (S.L.); (P.J.); (M.W.)
| | - Daxin Chen
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China;
| | - Shan Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Y.X.); (Q.C.); (C.Z.); (W.Z.); (X.X.); (H.L.); (Y.L.); (S.L.); (P.J.); (M.W.)
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Peizhi Jia
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Y.X.); (Q.C.); (C.Z.); (W.Z.); (X.X.); (H.L.); (Y.L.); (S.L.); (P.J.); (M.W.)
| | - Meiling Wang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Y.X.); (Q.C.); (C.Z.); (W.Z.); (X.X.); (H.L.); (Y.L.); (S.L.); (P.J.); (M.W.)
| | - Ling Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Y.X.); (Q.C.); (C.Z.); (W.Z.); (X.X.); (H.L.); (Y.L.); (S.L.); (P.J.); (M.W.)
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Wei Lin
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China;
| |
Collapse
|
15
|
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
|
16
|
Zhou C, Peng B, Zhang M, Yang Y, Yi Z, Wu Y. Ganjiang Huangqin Huanglian Renshen Decoction protects against ulcerative colitis by modulating inflammation, oxidative stress, and gut microbiota. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156172. [PMID: 39471735 DOI: 10.1016/j.phymed.2024.156172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/22/2024] [Accepted: 10/20/2024] [Indexed: 11/01/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is a disease that is difficult to treat and has been associated with high rates of recurrence. Moreover, the current medications for UC induce serious side effects following prolonged use. Ganjiang Huangqin Huanglian Renshen Decoction (GJHQHLRSD), has been traditionally used to treat UC. However, its protective mechanisms have not been fully studied. PURPOSE In this study the mechanisms by which GJHQHLRSD treats UC was investigated. METHODS The GJHQHLRSD and GJHQHLRSD drug-containing serum (GJHQHLRSD-DS) were characterized using LC-MS/MS. The therapeutic effect of GJHQHLRSD on dextran sodium sulfate (DSS)-induced UC was explored by assessing various parameters including intestinal flora 16S rRNA, intestinal barrier function, oxidative stress (OS) response, inflammatory cytokines, colonic histopathological injury, colon length, disease activity index (DAI) and body weight. RESULTS Treatment with GJHQHLRSD increased body weight, ameliorated colon length shortening and edema, reduced the DAI score, improved the pathological injury, down-regulated the levels of IL-1β, IL-6, IL-8, TNF-α, LPS, LDH, TLR4, and NLRP3, and up-regulated the ZO-1 and Occludin levels in UC mice. It also decreased intestinal oxidative stress in UC mice and improved mitogenic activity by modulating mitochondrial ultrastructure as well as the expression level of PINK1, LC3-II/Ⅰ, Beclin-1, p62, and Parkin proteins. In addition, we found that the effects of GJHQHLRSD on UC mice were inhibited by 3-MA.GJHQHLRSD treatment reduced the imbalance of intestinal flora in UC mice, by regulating the inflammation and oxidative stress. CONCLUSION These findings suggested that GJHQHLRSD effectively attenuated inflammatory responses, inhibited the TLR4/NF-κB/NLRP3 signalling, oxidative stress, and modulated the gut microbiota, and alleviated the DSS-induced UC symptoms, making it a promising and innovative therapeutic option for the treatment of UC.
Collapse
Affiliation(s)
- Ce Zhou
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China.
| | - Bo Peng
- Department of Radiology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China
| | - Mingxing Zhang
- Department of Radiology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China
| | - Yang Yang
- Department of Radiology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China
| | - Zelin Yi
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Yinghua Wu
- Department of Radiology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China.
| |
Collapse
|
17
|
Yuan L, Li W, Hu S, Wang Y, Wang S, Tian H, Sun X, Yang X, Hu M, Zhang Y. Protective effects of ginsenosides on ulcerative colitis: a meta-analysis and systematic review to reveal the mechanisms of action. Inflammopharmacology 2024; 32:3079-3098. [PMID: 38977646 DOI: 10.1007/s10787-024-01516-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/07/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory disease of the colon. Ginsenoside may be an ideal agent for UC treatment. However, its efficacy and safety are unknown. We aim to conduct a systematic evaluation to assess the effects and potential mechanisms of ginsenosides in animal models of UC. METHODS Six electronic databases will be searched (PubMed, Embase, Web of Science, China Knowledge Network (CNKI), China Science and Technology Journal Database (CQVIP), and Wanfang Data Knowledge). SYRCLE list will be used to assess the quality of literature, and STATA 15.1 for data analysis. Time-dose effects analysis will be used to reveal the time-dosage response relations between ginsenosides and UC. RESULTS Ultimately, fifteen studies involving 300 animals were included. Preliminary evidence was shown that ginsenosides could reduce Disease Activity Index (DAI) scores, weight loss, histological colitis score (HCS), spleen weight, Malondialdehyde (MDA), Myeloperoxidase (MPO) activity, interleukin-1β (IL-1β), interleukin 6 (IL-6), tumor necrosis factor α (TNF-α) and increase colon length (CL), myeloperoxidase (GSH), interleukin 4 (IL-4), interleukin 10 (IL-10), Zonula Occludens-1 (ZO-1) and occludin. Results of time-dose interval analysis indicated that ginsenosides at a dosage of 5-200 mg/kg with an intervention time of 7-28 days were relatively effective. CONCLUSIONS Preclinical evidence suggests that ginsenoside is a novel treatment for UC. And the mechanisms of ginsenosides in treating UC may involve anti-inflammatory, antioxidant, barrier protection, intestinal flora regulation, and immune regulation. Although, due to the high heterogeneity, further large-scale and high-quality preclinical studies are needed to examine the protection of ginsenosides against UC.
Collapse
Affiliation(s)
- Lingling Yuan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuangyuan Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yingyi Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shaofeng Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huai'e Tian
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuhui Sun
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuli Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengyun Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
18
|
Li X, He J, Gao X, Zheng G, Chen C, Chen Y, Xing Z, Wang T, Tang J, Guo Y, He Y. GPX4 restricts ferroptosis of NKp46 +ILC3s to control intestinal inflammation. Cell Death Dis 2024; 15:687. [PMID: 39300068 PMCID: PMC11413021 DOI: 10.1038/s41419-024-07060-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/31/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
Group 3 innate lymphoid cells (ILC3s) are essential for both pathogen defense and tissue homeostasis in the intestine. Dysfunction of ILC3s could lead to increased susceptibility to intestinal inflammation. However, the precise mechanisms governing the maintenance of intestinal ILC3s are yet to be fully elucidated. Here, we demonstrated that ferroptosis is vital for regulating the survival of intestinal ILC3. Ferroptosis-related genes, including GPX4, a key regulator of ferroptosis, were found to be upregulated in intestinal mucosal ILC3s from ulcerative colitis patients. Deletion of GPX4 resulted in a decrease in NKp46+ILC3 cell numbers, impaired production of IL-22 and IL-17A, and exacerbated intestinal inflammation in a T cell-independent manner. Our mechanistic studies revealed that GPX4-mediated ferroptosis in NKp46+ILC3 cells was regulated by the LCN2-p38-ATF4-xCT signaling pathway. Mice lacking LCN2 in ILC3s or administration of a p38 pathway inhibitor exhibited similar phenotypes of ILC3 and colitis to those observed in GPX4 conditional knock-out mice. These observations provide novel insights into therapeutic strategies for intestinal inflammation by modulating ILC3 ferroptosis.
Collapse
Affiliation(s)
- Xinyao Li
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences); Department of Immunology, School of Basic Medical Sciences; Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
- Department of Immunology; Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Junyu He
- Department of Immunology; Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiang Gao
- Department of Gastroenterology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guilang Zheng
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University; Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chunling Chen
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University; Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yimin Chen
- Department of Immunology; Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhe Xing
- Department of Immunology; Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tianci Wang
- Department of Immunology; Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jian Tang
- Department of Gastroenterology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuxiong Guo
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University; Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Yumei He
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences); Department of Immunology, School of Basic Medical Sciences; Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China.
- Department of Immunology; Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
19
|
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
|
20
|
Cai X, Zhang Q, Wang J, Miao Y, Sun Y, Xia Z, Zhang L, Yu Q, Jiang Z. Novel Dual PPAR δ/γ Partial Agonist Induces Hepatic Lipid Accumulation through Direct Binding and Inhibition of AKT1 Phosphorylation, Mediating CD36 Upregulation. Chem Res Toxicol 2024; 37:1574-1587. [PMID: 39235066 DOI: 10.1021/acs.chemrestox.4c00268] [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: 09/06/2024]
Abstract
ZLY06 is a dual agonist of peroxisome proliferator-activated receptor (PPAR) δ/γ, showing potential therapeutic effects on metabolic syndrome. However, our research has revealed that ZLY06 exhibits hepatotoxicity in normal C57BL/6J mice, though the precise mechanism remains unclear. This study aims to investigate the manifestations and mechanisms of ZLY06-induced hepatotoxicity. We administered ZLY06 via oral gavage to C57BL/6J mice (once daily for six weeks) and monitored various indicators to preliminarily explore its hepatotoxicity. Additionally, we further investigate the specific mechanisms of ZLY06-induced hepatotoxicity using PPAR inhibitors (GW9662 and GSK0660) and the Protein kinase B (AKT) activator (SC79). Results showed that ZLY06 led to increased serum ALP, ALT and AST, as well as elevated liver index and hepatic lipid levels. There was upregulation in the gene and protein expression of lipid metabolism-related molecules Acc, Scd1, Cd36, Fabp1 and Fabp2 in hepatocytes, with Cd36 showing the most significant change. Furthermore, cotreatment with SC79 significantly reduced ZLY06-induced hepatotoxicity in AML12 cells, evidenced by decreased intracellular TG levels and downregulation of CD36 expression. Specific knockdown of CD36 also mitigated ZLY06-induced hepatotoxicity. The study found that ZLY06 may bind to AKT1, inhibiting its phosphorylation activation, with the downregulation of p-AKT1 preceding the upregulation of CD36. In summary, ZLY06 mediates the upregulation of CD36 by potentially binding to and inhibiting the phosphorylation of AKT1, leading to hepatic lipid metabolism disorder and inducing liver toxicity.
Collapse
Affiliation(s)
- Xiaotong Cai
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Qin Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jiwei Wang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yingying Miao
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yuqing Sun
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Ziyin Xia
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Luyong Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qinwei Yu
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenzhou Jiang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Center for Pharmacodynamics Research and Evaluation, Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
21
|
Yue Y, Ke Y, Zheng J, Wang Z, Liu H, Liu S. Microbiota-derived tryptophan metabolism and AMPK/mTOR pathway mediate antidepressant-like effect of Shugan Hewei Decoction. Front Pharmacol 2024; 15:1466336. [PMID: 39351096 PMCID: PMC11439769 DOI: 10.3389/fphar.2024.1466336] [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: 07/17/2024] [Accepted: 08/30/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Depression is a common psychological disorder, accompanied by a disturbance of the gut microbiota and its metabolites. Recently, microbiota-derived tryptophan metabolism and AMPK/mTOR pathway were found to be strongly linked to the development of depression. Shugan Hewei Decoction (SHD) is a classical anti-depression traditional Chinese medicine formula. Although, we have shown that SHD exerted antidepressant effects via cecal microbiota and cecum NLRP3 inflammasome, the specific mechanism of SHD on metabolism driven by gut microbiota is unknown. In this study, we focus on the tryptophan metabolism and AMPK/mTOR pathway to elucidate the multifaceted mechanisms of SHD. Methods Male rats were established to the chronic unpredictable stress (CUS)/social isolation for 6 weeks, and SHD-L (7.34 g/kg/d), SHD-H (14.68 g/kg/d), Fructooligosaccharide (FOS) (3.15 g/kg/d) were given by intragastric administration once daily during the last 2 weeks. Behavioral experiments were carried out to evaluate the model. The colonic content was taken out for shotgun metagenomic sequencing combined with the untargeted metabolomics, the targeted tryptophan metabolomics. ELISA was used to detect the levels of zonula occludens 1 (ZO-1), Occludin in colon, as well as lipopolysaccharide (LPS), diamine oxidase (DAO), D-lactate (DLA) in serum. The expressions of mRNA and proteins of adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway of autophagy were examined using RT-qPCR and Western blot in colon. Results SHD modulated gut microbiota function and biological pathways, which were related to tryptophan metabolism. In addition, SHD could regulate microbiota-derived tryptophan production (such as reduction of 3-HK, 3-HAA etc., increment of ILA, IAA etc.), which metabolites belong to kynurenine (KYN) and indole derivatives. Further, SHD reduced intestinal permeability and enhanced the intestinal barrier function. Moreover, SHD could upregulate the levels of AMPK, microtubule associated protein light chain 3 (LC3), autophagy related protein 5 (ATG5) and Beclin1, downregulate the levels of mTOR, p62, promoted autophagy in colon. Spearman's analysis illustrated the close correlation between tryptophan metabolites and intestinal barrier, AMPK/mTOR pathway. Conclusion SHD may exert antidepressant-like effects by regulating microbiota-derived tryptophan metabolism, and triggering the AMPK/mTOR pathway of autophagy, enhancing the intestinal barrier function.
Collapse
Affiliation(s)
- Yingying Yue
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
| | - Youlan Ke
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
| | - Junping Zheng
- Hubei Shizhen Laboratory, Wuhan, China
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Zicheng Wang
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
| | - Hongtao Liu
- Hubei Shizhen Laboratory, Wuhan, China
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Songlin Liu
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
| |
Collapse
|
22
|
Xu W, Liu AX, Liu KH, Zhang S, Gong ZH, Xiao WJ. l-Theanine Alleviates Ulcerative Colitis by Regulating Colon Immunity via the Gut Microbiota in an MHC-II-Dependent Manner. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:19852-19868. [PMID: 39197065 DOI: 10.1021/acs.jafc.4c04379] [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: 08/30/2024]
Abstract
Alterations to the gut microbiota are associated with ulcerative colitis (UC), whereas restoration of normobiosis can effectively alleviate UC. l-Theanine has been shown to reshape the gut microbiota and regulate gut immunity. To investigate the mechanisms by which l-theanine alleviates UC, we used l-theanine and l-theanine fecal microbiota solution to treat UC mice. In this study, we used l-theanine and l-theanine fecal microbiota solution to treat UC mice to explore the mechanism by which l-theanine alleviates UC. By reducing inflammation in the colon, we demonstrated that l-theanine alleviates symptoms of UC. Meanwhile, l-theanine can improve the abundance of microbiota related to short-chain fatty acid, bile acid, and tryptophan production. Single-cell sequencing results indicated that l-theanine-mediated suppression of UC was associated with immune cell changes, especially regarding macrophages and T and B cells, and validated the immune cell responses to the gut microbiota. Further, flow cytometry results showed that the ability of dendritic cells, macrophages, and monocytes to present microbiota antigens to colonic T cells in an MHC-II-dependent manner was reduced after treating normal mouse fecal donors with l-theanine. These results demonstrate that l-theanine modulates colon adaptive and innate immunity by regulating the gut microbiota in an MHC-II-dependent manner, thereby alleviating UC.
Collapse
Affiliation(s)
- Wei Xu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, Hunan, China
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - Ao-Xiang Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, Hunan, China
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - Ke-Hong Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, Hunan, China
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - Sheng Zhang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, Hunan, China
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, Hunan, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha 410128, Hunan, China
| | - Zhi-Hua Gong
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, Hunan, China
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, Hunan, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha 410128, Hunan, China
| | - Wen-Jun Xiao
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, Hunan, China
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, Hunan, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha 410128, Hunan, China
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, Hunan, China
| |
Collapse
|
23
|
Li J, Zhang C, Xu Y, Yang L. Efficacy and safety of berberine plus 5-ASA for ulcerative colitis: A systematic review and meta-analysis. PLoS One 2024; 19:e0309144. [PMID: 39241013 PMCID: PMC11379390 DOI: 10.1371/journal.pone.0309144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 08/05/2024] [Indexed: 09/08/2024] Open
Abstract
PURPOSE This study aimed to assess the efficacy and safety of berberine(BBR) plus 5-aminosalicylic acid (5-ASA) for treating ulcerative colitis (UC). METHODS A comprehensive search was conducted in electronic databases, including Medline/PubMed, Sinomed, Embase, CNKI, Wanfang, and VIP, through January 2024 to identify all randomized controlled trials (RCTs) that administered BBR conjunction in standard therapy(5-ASA) for to support the treatment of UC. The data were synthesized using a meta-analysis approach with RevMan 5.4.1. The primary endpoint was the clinical efficacy rate. In contrast, the secondary endpoints included the Baron score, disease activity index (DAI) score, symptom relief latency, inflammatory markers, immunological indicators, and adverse events. RESULTS In this analysis, 10 RCTs comprising 952 patients with UC were examined. BBR considerably improved the clinical efficacy rate (RR = 1.22, 95% CI [1.15, 1.30], P < 0.00001), attenuated the Baron score (SMD = -1.72, 95% CI [-2.30, -1.13], P < 0.00001) and reduced the DAI score (SMD = -2.93, 95% CI [-4.42, -1.43], P < 0.00001). Additionally, it ameliorated clinical symptoms (SMD = -2.74, 95% CI [-3.45, 2.02], P < 0.00001), diminished inflammatory responses (SMD = -1.59, 95% CI [-2.14, 1.04], P < 0.00001), and modulated immune reactions (SMD = 1.06,95% CI [0.24,1.87], P <0.00001). Nonetheless, the impact of BBR on reducing adverse reactions was not statistically significant (RR = 0.75, 95% CI [0.42, 1.33], P > 0.05). CONCLUSION BBR demonstrates substantial efficacy in treating UC without causing severe adverse reactions and may serve as a viable complementary therapy. However, its clinical application warrants confirmation by additional high-quality, low-bias RCTs.
Collapse
Affiliation(s)
- Jilei Li
- Department of Oncology Diseases, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Chenchen Zhang
- Graduate School Department, Beijing University of Chinese Medicine, Beijing, China
| | - Yanchao Xu
- Department of Oncology Diseases, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Lili Yang
- Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
24
|
Huang S, Ye Q, Wang A, Chen Y. Paeoniae Decoction restores intestinal barrier dysfunction by promoting the interaction between ILC3 and gut flora. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155873. [PMID: 39024673 DOI: 10.1016/j.phymed.2024.155873] [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: 03/02/2024] [Revised: 06/18/2024] [Accepted: 07/09/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Intestinal barrier dysfunction is a significant contributor to the recurrence and refractory of ulcerative colitis (UC). Promoting the interaction between group 3 innate lymphoid cells (ILC3s) and gut flora is a valuable strategy for mucosal repair. Paeoniae decoction (PD) is a compound commonly used in clinical treatment of UC, but its exact mechanism remains unclear. PURPOSE We aimed to investigate the protective effect of PD on intestinal mucosal injury induced by dextran sulfate sodium (DSS) in chronic colitis, as well as to elucidate its potential mechanism. METHODS C57BL/6 mice were induced with chronic colitis by 2 % DSS and divided into four groups: control group, model group, PD low dose (4 g/kg), and high dose (8 g/kg) group. The effectiveness of PD in treating chronic colitis mice was evaluated based on changes in body weight, colon length, colon pathological tissue scores, and the mRNA levels of inflammatory factors IL-6 and IL-1β. The expressions of intestinal epithelial tight junction proteins (ZO-1 and Occludin), IL-22, and MUC2 were observed using immunofluorescence and RT-PCR. Additionally, the proportion of ILC3 and natural cytotoxicity receptor (NCR)+ ILC3 in the colon were detected using flow cytometry. Furthermore, UHPLC-QE-MS was utilized to identify chemical components of PD and network pharmacology was employed to predict potential pathways for PD intervention in UC. Subsequently, MNK-3 cells (ILC3 in vitro cell line) and NCM460 cells were used to verify the network pharmacology results. Finally, the effects of PD on UC gut flora have been explored using in vitro fermentation and 16S rDNA techniques. RESULTS The results showed that PD significantly restored body weight and colon length in mice with chronic colitis, while also reducing colon inflammatory cell infiltration and the expression of IL-6 and IL-1β. Additionally, PD notably promoted the expression of MUC2, ZO-1, Occludin, and IL-22, as well as increasing the ratio of ILC3 and NCR+ILC3. UHPLC-QE-MS analysis identified 443 components of PD, and network pharmacology suggested that PD could target the aryl hydrocarbon receptor (AHR) signaling pathway, which was confirmed by MNK-3 cells and in vitro fermentation experiments. Furthermore, MNK-3-conditioned medium (CM) increased the expression of ZO-1 and Occludin in NCM460 cells. In addition, 16S rDNA results indicated that PD promoted the abundance of Lactobacillales, thus contributing to mucosal damage repair by activating the AHR signal in ILC3s. CONCLUSION In summary, our study demonstrates that PD repairs intestinal mucosal damage in chronic colitis by regulating the interaction of gut flora with ILC3, and the specific mechanism is related to the activation of AHR signaling pathway.
Collapse
Affiliation(s)
- Shaowei Huang
- Integrative Microecology Clinical Center, Shenzhen Key Laboratory of Gastrointestinal Microbiota and Disease, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Technology Research Center of Gut Microbiota Transplantation, Shenzhen Hospital, Southern Medical University, Shenzhen, PR China
| | - Qiujuan Ye
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Anjiang Wang
- Integrative Microecology Clinical Center, Shenzhen Key Laboratory of Gastrointestinal Microbiota and Disease, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Technology Research Center of Gut Microbiota Transplantation, Shenzhen Hospital, Southern Medical University, Shenzhen, PR China.
| | - Ye Chen
- Integrative Microecology Clinical Center, Shenzhen Key Laboratory of Gastrointestinal Microbiota and Disease, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Technology Research Center of Gut Microbiota Transplantation, Shenzhen Hospital, Southern Medical University, Shenzhen, PR China; Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China.
| |
Collapse
|
25
|
Kim S, Seo SU, Kweon MN. Gut microbiota-derived metabolites tune host homeostasis fate. Semin Immunopathol 2024; 46:2. [PMID: 38990345 PMCID: PMC11239740 DOI: 10.1007/s00281-024-01012-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/15/2024] [Indexed: 07/12/2024]
Abstract
The gut microbiota, housing trillions of microorganisms within the gastrointestinal tract, has emerged as a critical regulator of host health and homeostasis. Through complex metabolic interactions, these microorganisms produce a diverse range of metabolites that substantially impact various physiological processes within the host. This review aims to delve into the intricate relationships of gut microbiota-derived metabolites and their influence on the host homeostasis. We will explore how these metabolites affect crucial aspects of host physiology, including metabolism, mucosal integrity, and communication among gut tissues. Moreover, we will spotlight the potential therapeutic applications of targeting these metabolites to restore and sustain host equilibrium. Understanding the intricate interplay between gut microbiota and their metabolites is crucial for developing innovative strategies to promote wellbeing and improve outcomes of chronic diseases.
Collapse
Affiliation(s)
- Seungil Kim
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine / Asan Medical Center, Seoul, Republic of Korea
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine / Asan Medical Center, Seoul, Republic of Korea.
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
26
|
Zhou Y, Zhang D, Cheng H, Wu J, Liu J, Feng W, Peng C. Repairing gut barrier by traditional Chinese medicine: roles of gut microbiota. Front Cell Infect Microbiol 2024; 14:1389925. [PMID: 39027133 PMCID: PMC11254640 DOI: 10.3389/fcimb.2024.1389925] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024] Open
Abstract
Gut barrier is not only part of the digestive organ but also an important immunological organ for the hosts. The disruption of gut barrier can lead to various diseases such as obesity and colitis. In recent years, traditional Chinese medicine (TCM) has gained much attention for its rich clinical experiences enriched in thousands of years. After orally taken, TCM can interplay with gut microbiota. On one hand, TCM can modulate the composition and function of gut microbiota. On the other hand, gut microbiota can transform TCM compounds. The gut microbiota metabolites produced during the actions of these interplays exert noticeable pharmacological effects on the host especially gut barrier. Recently, a large number of studies have investigated the repairing and fortifying effects of TCM on gut barriers from the perspective of gut microbiota and its metabolites. However, no review has summarized the mechanism behand this beneficiary effects of TCM. In this review, we first briefly introduce the unique structure and specific function of gut barrier. Then, we summarize the interactions and relationship amidst gut microbiota, gut microbiota metabolites and TCM. Further, we summarize the regulative effects and mechanisms of TCM on gut barrier including physical barrier, chemical barrier, immunological barrier, and microbial barrier. At last, we discuss the effects of TCM on diseases that are associated gut barrier destruction such as ulcerative colitis and type 2 diabetes. Our review can provide insights into TCM, gut barrier and gut microbiota.
Collapse
Affiliation(s)
- Yaochuan Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dandan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hao Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinlu Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Juan Liu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wuwen Feng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
27
|
Hu Y, Tang J, Xie Y, Xu W, Zhu W, Xia L, Fang J, Yu D, Liu J, Zheng Z, Zhou Q, Shou Q, Zhang W. Gegen Qinlian decoction ameliorates TNBS-induced ulcerative colitis by regulating Th2/Th1 and Tregs/Th17 cells balance, inhibiting NLRP3 inflammasome activation and reshaping gut microbiota. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:117956. [PMID: 38428658 DOI: 10.1016/j.jep.2024.117956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chinese herbal medicine Gegen Qinlian Decoction (GQD) has been clinically shown to be an effective treatment of ulcerative colitis (UC) in China. However, the underlying mechanism of GQD's anti-ulcerative colitis properties and its effect on gut microbiota still deserve further exploration. AIM OF THE STUDY This study observed the regulatory effects of GQD on Th2/Th1 and Tregs/Th17 cells balance, the NOD-like receptor family pyrin domain containing 3 (NLRP3) infammasome and gut microbiota in TNBS-induced UC in BALB/c mice. MATERIALS AND METHODS 61 main chemical compounds in the GQD were determined by UPLC-Q-TOF/MS. The UC BALB/c model was established by intrarectal administration of trinitrobenzene sulfonic acid (TNBS), and GQD was orally administered at low and high dosages of 2.96 and 11.83 g/kg/day, respectively. The anti-inflammatory effects of GQD for ulcerative colitis were evaluated by survival rate, body weight, disease activity index (DAI) score, colonic weight and index, spleen index, hematoxylin-eosin (HE) staining and histopathological scores. Flow cytometry was used to detect the percentage of CD4, Th1, Th2, Th17 and Tregs cells. The levels of Th1-/Th2-/Th17-/Tregs-related inflammatory cytokines and additional proinflammatory cytokines (IL-1β, IL-18) were detected by CBA, ELISA, and RT-PCR. The expressions of GATA3, T-bet, NLRP3, Caspase-1, IL-Iβ, Occludin and Zonula occludens-1 (ZO-1) on colon tissues were detected by Western blot and RT-PCR. Transcriptome sequencing was performed using colon tissue and 16S rRNA gene sequencing was performed on intestinal contents. Fecal microbiota transplantation (FMT) was employed to assess the contribution of intestinal microbiota and its correlation with CD4 T cells and the NLRP3 inflammasome. RESULTS GQD increased the survival rate of TNBS-induced UC in BALB/c mice, and significantly improved their body weight, DAI score, colonic weight and index, spleen index, and histological characteristics. The intestinal barrier dysfunction was repaired after GQD administration through promoting the expression of tight junction proteins (Occludin and ZO-1). GQD restored the balance of Th2/Th1 and Tregs/Th17 cells immune response of colitis mice, primarily inhibiting the increase in Th2/Th1 ratio and their transcription factor production (GATA3 and T-bet). Morever, GQD changed the secretion of Th1-/Th2-/Th17-/Tregs-related cytokines (IL-2, IL-12, IL-5, IL-13, IL-6, IL-10, and IL-17A) and reduced the expressions of IL-1β, IL-18. Transcriptome results suggested that GQD could also remodel the immune inflammatory response of colitis by inhibiting NOD-like receptor signaling pathway, and Western blot, immunohistochemistry and RT-PCR further revealed that GQD exerted anti-inflammatory effects by inhibiting the NLRP3 inflammasome, such as down-regulating the expression of NLRP3, Caspase-1 and IL-1β. More interestingly, GQD regulated gut microbiota dysbiosis, suppressed the overgrowth of conditional pathogenic gut bacteria like Helicobacter, Proteobacteria, and Mucispirillum, while the probiotic gut microbiota, such as Lactobacillus, Muribaculaceae, Ruminiclostridium_6, Akkermansia, and Ruminococcaceae_unclassified were increased. We further confirmed that GQD-treated gut microbiota was sufficient to relieve TNBS-induced colitis by FMT, involving the modulation of Th2/Th1 and Tregs/Th17 balance, inhibition of NLRP3 inflammasome activation, and enhancement of colonic barrier function. CONCLUSIONS GQD might alleviate TNBS-induced UC via regulating Th2/Th1 and Tregs/Th17 cells Balance, inhibiting NLRP3 inflammasome and reshaping gut microbiota, which may provide a novel strategy for patients with colitis.
Collapse
Affiliation(s)
- Yingnan Hu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jingyi Tang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yongfeng Xie
- Department of Burn Plastic Surgery, Huai'an Hospital Affiliated to Xuzhou Medical University, Jiangsu, 223001, China
| | - Wenjun Xu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Weihan Zhu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Linying Xia
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Jintao Fang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Dian Yu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jingjing Liu
- Department of General Surgery, Haining City Central Hospital, Jiaxing, 314408, China
| | - Zhipeng Zheng
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China
| | - Qiujing Zhou
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China
| | - Qiyang Shou
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| | - Wei Zhang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China; The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| |
Collapse
|
28
|
Zhu H, Yang X, Zhao Y. Recent Advances in Current Uptake Situation, Metabolic and Nutritional Characteristics, Health, and Safety of Dietary Tryptophan. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:6787-6802. [PMID: 38512048 DOI: 10.1021/acs.jafc.3c06419] [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: 03/22/2024]
Abstract
Tryptophan (Trp) is an essential amino acid which is unable to be synthesized in the body. Main sources of Trp are uptake of foods such as oats and bananas. In this review, we describe the status of current dietary consumption, metabolic pathways and nutritional characteristics of Trp, as well as its ingestion and downstream metabolites for maintaining body health and safety. This review also summarizes the recent advances in Trp metabolism, particularly the 5-HT, KYN, and AhR activation pathways, revealing that its endogenous host metabolites are not only differentially affected in the body but also are closely linked to health. More attention should be paid to targeting its specific metabolic pathways and utilizing food molecules and probiotics for manipulating Trp metabolism. However, the complexity of microbiota-host interactions requires further exploration to precisely refine targets for innovating the gut microbiota-targeted diagnostic approaches and informing subsequent studies and targeted treatments of diseases.
Collapse
Affiliation(s)
- Haoyan Zhu
- Key Laboratory of the Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yan Zhao
- Key Laboratory of the Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| |
Collapse
|
29
|
Huang X, Chen X, Wan G, Yang D, Zhu D, Jia L, Zheng J. Mechanism of intestinal microbiota disturbance promoting the occurrence and development of esophageal squamous cell carcinoma--based on microbiomics and metabolomics. BMC Cancer 2024; 24:245. [PMID: 38388357 PMCID: PMC10885407 DOI: 10.1186/s12885-024-11982-8] [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: 09/04/2023] [Accepted: 02/07/2024] [Indexed: 02/24/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a high-risk malignant tumor that has been reported in China. Some studies indicate that gut microbiota disorders can affect the occurrence and development of ESCC, but the underlying mechanism remains unclear. In this study, we aimed to explore the possible underlying mechanisms using microbiomics and metabolomics. Fifty ESCC patients and fifty healthy controls were selected as the study subjects according to sex and age, and fecal samples were collected. 16S rDNA sequencing and LC‒MS were used for microbiomics and nontargeted metabolomics analyses. We found significant differences in the composition of the gut microbiota and metabolites between the ESCC patients and control individuals (P < 0.05). ESCC patients exhibited increased abundances of Fusobacteriaceae and Lactobacillus, increased levels of GibberellinA34 and decreased levels of 12-hydroxydodecanoic acid; these metabolites could be diagnostic and predictive markers of ESCC. An increase in the abundance of Enterobacteriaceae and Lactobacillus significantly reduced the content of L-aspartate and pantothenic acid, which may be involved in the occurrence and development of ESCC by downregulating the expression of proteins in the pantothenate and coenzyme A biosynthesis pathways. An imbalance in the intestinal flora may decrease the number of eosinophils in peripheral blood, resulting in the activation of an inflammatory response and immune dysfunction, leading to ESCC deterioration. We hypothesize that this imbalance in the gut microbiota can cause an imbalance in intestinal metabolites, which can activate carcinogenic metabolic pathways, affect inflammation and immune function, and play a role in the occurrence and development of ESCC.
Collapse
Affiliation(s)
- Xingqiang Huang
- The First Clinical College, Changzhi Medical College, 046000, Shanxi, China
| | - Xueyi Chen
- The First Clinical College, Changzhi Medical College, 046000, Shanxi, China
| | - Guowei Wan
- The First Clinical College, Changzhi Medical College, 046000, Shanxi, China
| | - Dandan Yang
- The First Clinical College, Changzhi Medical College, 046000, Shanxi, China
| | - Dongqiang Zhu
- The First Clinical College, Changzhi Medical College, 046000, Shanxi, China
| | - Linqian Jia
- The First Clinical College, Changzhi Medical College, 046000, Shanxi, China
| | - Jinping Zheng
- The First Clinical College, Changzhi Medical College, 046000, Shanxi, China.
| |
Collapse
|
30
|
Wang X, Xie X, Li Y, Xie X, Huang S, Pan S, Zou Y, Pan Z, Wang Q, Chen J, Zhou L, Luo X. Quercetin ameliorates ulcerative colitis by activating aryl hydrocarbon receptor to improve intestinal barrier integrity. Phytother Res 2024; 38:253-264. [PMID: 37873559 DOI: 10.1002/ptr.8027] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 10/25/2023]
Abstract
Ulcerative colitis (UC) pathogenesis is largely associated with intestinal epithelial barrier dysfunction. A therapeutic approach to UC involves the repair of damaged intestinal barrier. Our study aimed to investigate whether aryl hydrocarbon receptor (AhR) mediated the intestinal barrier repair effects of quercetin to ameliorate UC. 3% dextran sulfate sodium was used to induce colitic mice, and quercetin (25, 50, and 100 mg/kg) was administered orally for 10 days to assess the therapeutic effects. In vitro, Caco-2 cells were used to explore the effect of quercetin on tight junction protein expression and AhR activation. The results showed that quercetin alleviated colitic mice by restoring tight junctions (TJs) integrity via an AhR-dependent manner (p < 0.05). In vitro, quercetin dose-dependently elevated the expressions of TJs protein ZO-1 and Claudin1, and activated AhR by enhancing the expression of CYP1A1 and facilitating AhR nuclear translocation in Caco-2 cells (p < 0.05). While AhR antagonist CH223191 reversed the therapeutic effects of quercetin (p < 0.05) and blocked quercetin-induced AhR activation and enhancement of TJs protein (p < 0.05). In conclusion, quercetin repaired intestinal barrier dysfunction by activating AhR-mediated enhancement of TJs to alleviate UC. Our research offered new perspectives on how quercetin enhanced intestinal barrier function.
Collapse
Affiliation(s)
- Xiaojing Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuting Xie
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanyang Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xueqian Xie
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shaowei Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Simin Pan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanling Zou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zengfeng Pan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qing Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinyan Chen
- Basic Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lian Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xia Luo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
31
|
Hou JJ, Ma AH, Qin YH. Activation of the aryl hydrocarbon receptor in inflammatory bowel disease: insights from gut microbiota. Front Cell Infect Microbiol 2023; 13:1279172. [PMID: 37942478 PMCID: PMC10628454 DOI: 10.3389/fcimb.2023.1279172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory intestinal disease that affects more than 3.5 million people, with rising prevalence. It deeply affects patients' daily life, increasing the burden on patients, families, and society. Presently, the etiology of IBD remains incompletely clarified, while emerging evidence has demonstrated that altered gut microbiota and decreased aryl hydrocarbon receptor (AHR) activity are closely associated with IBD. Furthermore, microbial metabolites are capable of AHR activation as AHR ligands, while the AHR, in turn, affects the microbiota through various pathways. In light of the complex connection among gut microbiota, the AHR, and IBD, it is urgent to review the latest research progress in this field. In this review, we describe the role of gut microbiota and AHR activation in IBD and discussed the crosstalk between gut microbiota and the AHR in the context of IBD. Taken as a whole, we propose new therapeutic strategies targeting the AHR-microbiota axis for IBD, even for other related diseases caused by AHR-microbiota dysbiosis.
Collapse
Affiliation(s)
| | | | - Yue-Hua Qin
- Department of Gastroenterology, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
32
|
Zong Y, Meng J, Mao T, Han Q, Zhang P, Shi L. Repairing the intestinal mucosal barrier of traditional Chinese medicine for ulcerative colitis: a review. Front Pharmacol 2023; 14:1273407. [PMID: 37942490 PMCID: PMC10628444 DOI: 10.3389/fphar.2023.1273407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/10/2023] [Indexed: 11/10/2023] Open
Abstract
Damage to the intestinal mucosal barrier play an important role in the pathogenesis of ulcerative colitis (UC). Discovering the key regulators and repairing the disturbed barrier are crucial for preventing and treating UC. Traditional Chinese medicine (TCM) has been proved to be effective on treating UC and has exhibited its role in repairing the intestinal mucosal barrier. We summarized the evidence of TCM against UC by protecting and repairing the physical barrier, chemical barrier, immune barrier, and biological barrier. Mechanisms of increasing intestinal epithelial cells, tight junction proteins, and mucins, promoting intestinal stem cell proliferation, restoring the abundance of the intestinal microbiota, and modulating the innate and adaptive immunity in gut, were all involved in. Some upstream proteins and signaling pathways have been elucidated. Based on the existing problems, we suggested future studies paying attention to patients' samples and animal models of UC and TCM syndromes, conducting rescue experiments, exploring more upstream regulators, and adopting new technical methods. We hope this review can provide a theoretical basis and novel ideas for clarifying the mechanisms of TCM against UC via repairing the intestinal mucosal barrier.
Collapse
Affiliation(s)
- Yichen Zong
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Meng
- Department of Gastroenterology and Hepatology, Beijing University of Chinese Medicine Affiliated Dongfang Hospital, Beijing, China
| | - Tangyou Mao
- Department of Gastroenterology and Hepatology, Beijing University of Chinese Medicine Affiliated Dongfang Hospital, Beijing, China
| | - Qiang Han
- Department of Traditional Chinese Medicine, Health Service Center of Beiyuan Community, Beijing, China
| | - Peng Zhang
- Department of Gastroenterology and Hepatology, Beijing University of Chinese Medicine Affiliated Dongfang Hospital, Beijing, China
| | - Lei Shi
- Department of Gastroenterology and Hepatology, Beijing University of Chinese Medicine Affiliated Dongfang Hospital, Beijing, China
| |
Collapse
|
33
|
Wang X, Quan J, Xiu C, Wang J, Zhang J. Gegen Qinlian decoction (GQD) inhibits ulcerative colitis by modulating ferroptosis-dependent pathway in mice and organoids. Chin Med 2023; 18:110. [PMID: 37649073 PMCID: PMC10466729 DOI: 10.1186/s13020-023-00819-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/10/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Gegen Qinlian decoction (GQD) is a classic prescription for treating ulcerative colitis (UC) in traditional Chinese medicine. However, the therapeutic mechanism has not been fully clarified. PURPOSE In the present study, we aimed to evaluate the role of ferroptosis-mediated IEC death in UC treated mice with GQD by using DSS-induced a colitis mouse model and RSL3-induced ferroptosis in intestinal organoids. METHODS The effects of GQD on DSS-treated colitis were examined via daily body weight, DAI, colon length, HE staining, PAS staining, ZO-1 and Occludin immunohistochemical staining. Ferroptosis was determined by analysis of iron load, MDA, GSH, mitochondrial morphology, and expression of ferroptosis-associated proteins (GPX4, SLC7A11 and ACSL4). RESULTS In vivo, GQD administration reduced body weight loss and DAI scores, increased colon length, and improved intestinal histological characteristics and epithelial barrier dysfunction. GQD administration obviously improved the levels of ferroptosis markers (iron load, MDA, GSH, and mitochondrial morphology) and the expression of ferroptosis-associated proteins (GPX4, SLC7A11 and ACSL4). Consistent with in vivo results, GQD administration partially reversed the levels of mtROS, Fe2+ and MDA in intestinal organoids induced by RSL3, and notably improved morphological destruction, histological damage and epithelial barrier dysfunction in organoids. CONCLUSIONS In this study, we demonstrated that ferroptosis was triggered in DSS-induced experimental colitis and that GQD adiministration could protect against colonic damage and intestinal epithelial barrier dysfunction by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Xue Wang
- Beijing Key Laboratory of Research of Chinese Medicine on Preventional and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jianye Quan
- Beijing Key Laboratory of Research of Chinese Medicine on Preventional and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chengkui Xiu
- Beijing Key Laboratory of Research of Chinese Medicine on Preventional and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jiali Wang
- Beijing Key Laboratory of Research of Chinese Medicine on Preventional and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jiaqi Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, No.1 Xiyuan Playground, Haidian District, Beijing, 100091, China.
| |
Collapse
|
34
|
Kargbo RB. Microbiome: The Next Frontier in Psychedelic Renaissance. J Xenobiot 2023; 13:386-401. [PMID: 37606422 PMCID: PMC10443327 DOI: 10.3390/jox13030025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/11/2023] [Accepted: 07/21/2023] [Indexed: 08/23/2023] Open
Abstract
The psychedelic renaissance has reignited interest in the therapeutic potential of psychedelics for mental health and well-being. An emerging area of interest is the potential modulation of psychedelic effects by the gut microbiome-the ecosystem of microorganisms in our digestive tract. This review explores the intersection of the gut microbiome and psychedelic therapy, underlining potential implications for personalized medicine and mental health. We delve into the current understanding of the gut-brain axis, its influence on mood, cognition, and behavior, and how the microbiome may affect the metabolism and bioavailability of psychedelic substances. We also discuss the role of microbiome variations in shaping individual responses to psychedelics, along with potential risks and benefits. Moreover, we consider the prospect of microbiome-targeted interventions as a fresh approach to boost or modulate psychedelic therapy's effectiveness. By integrating insights from the fields of psychopharmacology, microbiology, and neuroscience, our objective is to advance knowledge about the intricate relationship between the microbiome and psychedelic substances, thereby paving the way for novel strategies to optimize mental health outcomes amid the ongoing psychedelic renaissance.
Collapse
Affiliation(s)
- Robert B Kargbo
- Usona Institute, 2800 Woods Hollow Rd., Madison, WI 53711-5300, USA
| |
Collapse
|
35
|
Miao Y, Fan X, Wei L, Wang B, Diao F, Fu J, Zhuang P, Zhang Y. Lizhong decoction ameliorates pulmonary infection secondary to severe traumatic brain injury in rats by regulating the intestinal physical barrier and immune response. JOURNAL OF ETHNOPHARMACOLOGY 2023; 311:116346. [PMID: 36898448 DOI: 10.1016/j.jep.2023.116346] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The pathogenesis of pulmonary infection secondary to severe traumatic brain injury (sTBI) is closely related to damage to the intestinal barrier. Lizhong decoction (LZD) is a prominent traditional Chinese medicine (TCM) that is widely used in clinical treatment to regulate gastrointestinal movement and enhance resistance. Nevertheless, the role and mechanism of LZD in lung infection secondary to sTBI have yet to be elucidated. AIM OF THE STUDY Here, we evaluate the therapeutic effect of LZD on pulmonary infection secondary to sTBI in rats and discuss potential regulatory mechanisms. MATERIALS AND METHODS The chemical constituents of LZD were analyzed by ultra-high performance liquid chromatography-Q Exactive-tandem mass spectrometry(UPLC-QE-MS/MS). The efficacy of LZD on rats with lung infection secondary to sTBI was examined by changes in brain morphology, coma time, brain water content, mNSS score, colony counts, 16S rRNA/RNaseP/MRP30 kDa(16S/RPP30), myeloperoxidase (MPO) content and pathology of lung tissue. The concentration of fluorescein isothiocyanate(FITC)-dextran in serum and the contents of secretory immunoglobulin A (SIgA) in colon tissue were detected by enzyme-linked immunosorbent assay (ELISA). Subsequently, Alcian Blue Periodic acid Schiff (AB-PAS) was used to detect colonic goblet cells. Immunofluorescence (IF) was used to detect the expression of tight junction proteins. The proportions of CD3+ cell, CD4+CD8+ T cells, CD45+ cell and CD103+ cells in the colon were analyzed by flow cytometry (FC). In addition, colon transcriptomics were analyzed by Illumina mRNA-Seq sequencing. Real-time quantitative polymerase chain reaction (qRT‒PCR) was used to verify the genes associated with LZD alleviation of intestinal barrier function. RESULTS Twenty-nine chemical constituents of LZD were revealed with UPLC-QE-MS/MS analysis. Administration of LZD significantly reduced colony counts, 16S/RPP30 and MPO content in lung infection secondary to sTBI rats. In addition, LZD also reduced the serum FITC-glucan content and the SIgA content of the colon. Additionally, LZD significantly increased the number of colonic goblet cells and the expression of tight junction proteins. Furthermore, LZD significantly decreased the proportion of CD3+ cell, CD4+CD8+ T cells,CD45+ and CD103+ cells in colon tissue. Transcriptomic analysis identified 22 upregulated genes and 56 downregulated genes in sTBI compared to the sham group. The levels of seven genes were recovered after LZD treatment. qRT‒PCR successfully validated two genes (Jchain and IL-6) at the mRNA level. CONCLUSION LZD can improves sTBI secondary lung infection by regulating the intestinal physical barrier and immune response. Thees results suggested that LZD may be a prospective treatment for pulmonary infection secondary to sTBI.
Collapse
Affiliation(s)
- Yulu Miao
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuejin Fan
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Luge Wei
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bin Wang
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fengyin Diao
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiafeng Fu
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Pengwei Zhuang
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Yanjun Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Department of Integrated Rehabilitation, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|