1
|
Wang L, Lian YJ, Dong JS, Liu MK, Liu HL, Cao ZM, Wang QN, Lyu WL, Bai YN. Traditional Chinese medicine for chronic atrophic gastritis: Efficacy, mechanisms and targets. World J Gastroenterol 2025; 31:102053. [DOI: 10.3748/wjg.v31.i9.102053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/06/2024] [Accepted: 01/21/2025] [Indexed: 02/18/2025] Open
Abstract
Chronic atrophic gastritis (CAG) is an important stage of precancerous lesions of gastric cancer. Effective treatment and regulation of CAG are essential to prevent its progression to malignancy. Traditional Chinese medicine (TCM) has shown multi-targeted efficacy in CAG treatment, with advantages in enhancing gastric mucosal barrier defense, improving microcirculation, modulating inflammatory and immune responses, and promoting lesion healing, etc. Clinical studies and meta-analyses indicate that TCM provides significant benefits, with specific Chinese herbal compounds and monomers demonstrating protective effects on the gastric mucosa through mechanisms including anti-inflammation, anti-oxidation, and regulation of cellular proliferation and apoptosis, etc. Finally, it is pointed out that the efficacy of TCM in the treatment of CAG requires standardized research and unified standards, and constantly clarifies and improves the evaluation criteria of each dimension of gastric mucosal barrier function.
Collapse
Affiliation(s)
- Li Wang
- Department of Gastroenterology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yan-Jie Lian
- Division of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Jin-Sheng Dong
- Department of Gastroenterology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ming-Kun Liu
- Department of Gastroenterology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Hong-Liang Liu
- Department of Gastroenterology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Zheng-Min Cao
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Qing-Nan Wang
- Department of Dermatology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Wen-Liang Lyu
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yu-Ning Bai
- Department of Gastroenterology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
2
|
He Y, Wang X, Chen LS, Chang L, He TT, Zhang AZ, Li HT, Wei SZ, Jing MY, Zhao YL. Effects of Rutaecarpine on Chronic Atrophic Gastritis Through Nucleotide-binding Oligomerization Domain-like Receptors and Inflammasomes. WORLD JOURNAL OF TRADITIONAL CHINESE MEDICINE 2024; 10:303-315. [DOI: 10.4103/wjtcm.wjtcm_55_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/04/2023] [Indexed: 01/06/2025] Open
Abstract
Objective:
Chronic atrophic gastritis (CAG) is a complex and burdensome disease. However, side effects and compliance issues cannot be ignored due to the long treatment cycle. Numerous studies have confirmed the effectiveness of rutaecarpine (RUT) for treating digestive dysfunction. However, the potential mechanism of action of RUT in the context of CAG treatment remains unclear. This study aimed to explore the therapeutic effects and mechanisms of RUT in 1-methyl-3-nitro-1-nitrosoguanidine-induced CAG using network pharmacology, metabolomics, and traditional pharmacological approaches.
Materials and Methods:
Pathological tests and ELISA assays were used to observe the therapeutic effects of RUT treatment on CAG. Differential metabolites were identified using ultra-high-performance liquid chromatography-tandem mass spectrometry, and metabolism-related target genes were enriched. The same target genes were identified between RUT and CAG diseases. The intersectional target genes were uploaded to Cytoscape for enrichment, and the nucleotide-binding oligomerization domain (NOD)-like receptor signaling pathway was selected to validate the mechanisms of the study. Finally, cell pyroptosis status was evaluated using the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay, and the expressions of associated proteins of the NOD-like receptor signaling pathway were assessed by Western blotting and immunohistochemistry.
Results:
RUT alleviated gastric mucosal damage and significantly downregulated indicators associated with inflammation and gastric atrophy. A total of 29 intersection target genes was identified, and core pathways were obtained. The NOD-like receptor signaling pathway and pyroptosis status were selected to validate the mechanisms of RUT treatment in CAG rats. The expression of NOD-related proteins and downstream factors was downregulated in the RUT group.
Conclusions:
RUT exerts a pharmacological effect on relieving gastric damage in CAG rats by inhibiting NOD-like receptors and inflammasomes.
Collapse
Affiliation(s)
- Yong He
- Department of Pharmacy, Chongqing Rongchang Hospital of TCM, Chongqing, China
| | - Xin Wang
- Department of Pharmacy, Chongqing Rongchang Hospital of TCM, Chongqing, China
| | - Li-Sheng Chen
- Department of Pharmacy, Chongqing Rongchang Hospital of TCM, Chongqing, China
| | - Lei Chang
- Department of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Ting-Ting He
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Ao-Zhe Zhang
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Hao-Tian Li
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Shi-Zhang Wei
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Man-Yi Jing
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Yan-Ling Zhao
- Department of Pharmacy, Chongqing Rongchang Hospital of TCM, Chongqing, China
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
3
|
Zhang N, Chen P, Liang X, Sun J, Liu Q, Guan S, Wang Q. Luteolin targets the AGE-RAGE signaling to mitigate inflammation and ferroptosis in chronic atrophic gastritis. Aging (Albany NY) 2024; 16:10918-10930. [PMID: 38917486 PMCID: PMC11272119 DOI: 10.18632/aging.205969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024]
Abstract
Chronic atrophic gastritis (CAG) is a chronic inflammatory disease and precancerous lesion in stomach cancer. Abnormal activation cellular ferroptosis further damages gastric tissue, which is susceptible to inflammation. Luteolin has powerful anti-inflammatory and regulatory potential for cellular ferroptosis. We aimed to clarify the involvement of luteolin in inflammation and ferroptosis during CAG. Luteolin targets were searched to identify intersecting genes in the chronic atrophic gastritis disease database. The AGE-RAGE pathway is a potential target of luteolin for the treatment of chronic atrophic gastritis and a binding site between luteolin and RAGE was predicted through a computer simulation of molecular docking. We established a CAG rat model using N-methyl-N-nitro-N-nitroguanidine. The therapeutic effect of luteolin on CAG was detected using western blotting, qPCR, hematoxylin and eosin staining, lipid oxidation (MDA), and Fe2+ assays. Luteolin inhibited the AGE-RAGE signaling pathway and reduced the inflammatory response in gastric tissues. Additionally, luteolin downregulated the concentration of (MDA) and Fe2+, and CAG downregulated the expression levels of ACSL4 and NOX1 and upregulated the expression levels of FIH1 and GPX4 ferroptosis-related proteins, thus inhibiting the ferroptosis of gastric tissue cells, which had a therapeutic effect on CAG.
Collapse
Affiliation(s)
- Nailin Zhang
- Clinical Research Base Office, Hebei Provincial Hospital of Chinese Medicine, Hebei, China
- Key Laboratory of Integrated Chinese and Western Medicine for Gastroenterology Research, Hebei Provincial Hospital of Chinese Medicine, Hebei, China
| | - Pingping Chen
- Department of Pharmacology, Hebei University of Chinese Medicine, Hebei, China
| | - Xiaoyan Liang
- Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhui Sun
- Hebei Key Laboratory of Turbidity Toxin Syndrome, Hebei University of Chinese Medicine, Hebei, China
| | - Qiquan Liu
- Department of Spleen and Stomach Diseases, Hebei Provincial Hospital of Chinese Medicine, Hebei, China
| | - Shengjiang Guan
- Pharmaceutical Department, Hebei Provincial Hospital of Chinese Medicine, Hebei, China
| | - Qiao Wang
- Pharmacological Analysis Teaching and Research Department, Hebei Medical University, Hebei, China
| |
Collapse
|
4
|
Weng J, Wu XF, Shao P, Liu XP, Wang CX. Medicine for chronic atrophic gastritis: a systematic review, meta- and network pharmacology analysis. Ann Med 2024; 55:2299352. [PMID: 38170849 PMCID: PMC10769149 DOI: 10.1080/07853890.2023.2299352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
PURPOSE The aim of this study is to determine the effectiveness and reliability of adding traditional Chinese medicine (TCM) in the clinical intervention and explore mechanisms of action for chronic atrophic gastritis (CAG) through meta- and network pharmacology analysis (NPAs). METHODS A predefined search strategy was used to retrieve literature from PubMed, Embase database, Cochrane Library, China National Knowledge Infrastructure (CNKI), Chinese BioMedical Literature Database (CBM), Wan Fang Data and China Science and Technology Journal Database (VIP). After applying inclusion and exclusion criteria, a total of 12 randomized controlled trials (RCTs) were included for meta-analysis to provide clinical evidence of the intervention effects. A network meta-analysis using Bayesian networks was conducted to observe the relative effects of different intervention measures and possible ranking of effects. The composition of the TCM formulation in the experimental group was analysed, and association rule mining was performed to identify hub herbal medicines. Target genes for CAG were searched in GeneCards, Online Mendelian Inheritance in Man, PharmGKB, Therapeutic Target Database and DrugBank. A regulatory network was constructed to connect the target genes with active ingredients of the hub herbal medicines. Enrichment analyses were performed using the Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) to examine the central targets from a comprehensive viewpoint. Protein-protein interaction networks (PPINs) were constructed to identify hub genes and conduct molecular docking with differentially expressed genes (DEGs) and corresponding active molecules. RESULTS A total of 1140 participants from 12 RCTs were included in the statistical analysis, confirming that the experimental group receiving the addition of TCM intervention had better clinical efficacy. Seven hub TCMs (Paeonia lactiflora, Atractylodes macrocephala, Pinellia ternata, Citrus reticulata, Codonopsis pilosula, Salvia miltiorrhiza and Coptis chinensis) were identified through association rule analysis of all included TCMs. Thirteen hub genes (CDKN1A, CASP3, STAT1, TP53, JUN, MAPK1, STAT3, MAPK3, MYC, HIF1A, FOS, MAPK14 and AKT1) were obtained from 90 gene PPINs. Differential gene expression analysis between the disease and normal gastric tissue identified MAPK1 and MAPK3 as the significant genes. Molecular docking analysis revealed that naringenin, luteolin and quercetin were the main active compounds with good binding activities to the two hub targets. GO analysis demonstrated the function of the targets in protein binding, while KEGG analysis indicated their involvement in important pathways related to cancer. CONCLUSIONS The results of a meta-analysis of 12 RCTs indicate that TCM intervention can improve the clinical treatment efficacy of CAG. NPAs identified seven hub TCM and 13 target genes associated with their actions, while bioinformatics analysis identified two DEGs between normal and CAG gastric tissues. Finally, molecular docking was employed to reveal the mechanism of action of the active molecules in TCM on the DEGs. These findings not only reveal the mechanisms of action of the active components of the TCMs, but also provide support for the development of new drugs, ultimately blocking the progression from chronic gastritis to gastric cancer.
Collapse
Affiliation(s)
- Jiao Weng
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xiu-fang Wu
- The Second Hospital Affiliated with Shenyang Medical University, Shenyang, China
| | - Peng Shao
- The Second Hospital Affiliated with Shenyang Medical University, Shenyang, China
| | - Xing-pu Liu
- The Second Hospital Affiliated with Shenyang Medical University, Shenyang, China
| | - Cai-xia Wang
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
5
|
He Y, Liu HH, Zhou XL, He TT, Zhang AZ, Wang X, Wei SZ, Li HT, Chen LS, Chang L, Zhao YL, Jing MY. Rutaecarpine Ameliorates Murine N-Methyl-N'-Nitro-N-Nitrosoguanidine-Induced Chronic Atrophic Gastritis by Sonic Hedgehog Pathway. Molecules 2023; 28:6294. [PMID: 37687125 PMCID: PMC10489734 DOI: 10.3390/molecules28176294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/25/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
CAG is a burdensome and progressive disease. Numerous studies have shown the effectiveness of RUT in digestive system diseases. The therapeutic effects of RUT on MNNG-induced CAG and the potential mechanisms were probed. MNNG administration was employed to establish a CAG model. The HE and ELISA methods were applied to detect the treatment effects. WB, qRT-PCR, immunohistochemistry, TUNEL, and GES-1 cell flow cytometry approaches were employed to probe the mechanisms. The CAG model was successfully established. The ELISA and HE staining data showed that the RUT treatment effects on CAG rats were reflected by the amelioration of histological damage. The qRT-PCR and WB analyses indicated that the protective effect of RUT is related to the upregulation of the SHH pathway and downregulation of the downstream of apoptosis to improve gastric cellular survival. Our data suggest that RUT induces a gastroprotective effect by upregulating the SHH signaling pathway and stimulating anti-apoptosis downstream.
Collapse
Affiliation(s)
- Yong He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.H.); (X.W.)
| | - Hong-Hong Liu
- Department of Pharmacy, Chinese PLA General Hospital, Beijing 100039, China
| | - Xue-Lin Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100039, China
| | - Ting-Ting He
- Department of Pharmacy, Chinese PLA General Hospital, Beijing 100039, China
| | - Ao-Zhe Zhang
- Department of Pharmacy, Chinese PLA General Hospital, Beijing 100039, China
| | - Xin Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.H.); (X.W.)
| | - Shi-Zhang Wei
- Department of Pharmacy, Chinese PLA General Hospital, Beijing 100039, China
| | - Hao-Tian Li
- Department of Pharmacy, Chinese PLA General Hospital, Beijing 100039, China
| | - Li-Sheng Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.H.); (X.W.)
| | - Lei Chang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yan-Ling Zhao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.H.); (X.W.)
- Department of Pharmacy, Chinese PLA General Hospital, Beijing 100039, China
| | - Man-Yi Jing
- Department of Pharmacy, Chinese PLA General Hospital, Beijing 100039, China
| |
Collapse
|