1
|
Chen M, Wu Z, Zou Y, Peng C, Hao Y, Zhu Z, Shi X, Su B, Ou L, Lai Y, Jia J, Xun M, Li H, Zhu W, Feng Z, Yao M. Phellodendron chinense C.K.Schneid: An in vitro study on its anti-Helicobacter pylori effect. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118396. [PMID: 38823658 DOI: 10.1016/j.jep.2024.118396] [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: 04/15/2024] [Revised: 05/15/2024] [Accepted: 05/26/2024] [Indexed: 06/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Phellodendron chinense C.K.Schneid(P. chinense Schneid) is known in TCM as Huang Bo, is traditionally used to support gastrointestinal function and alleviate stomach-related ailments, including gastric ulcer bleeding and symptoms of gastroesophageal reflux disease. Helicobacter pylori (H. pylori) is classified by the WHO as a Group 1 carcinogen. However, the specific activity and mechanism of action of P. chinense Schneid against H. pylori infection remain unclear. It has been noted that Huangjiu processing may alter the bitter and cold properties of P. chinense Schneid, but its effect on antimicrobial activity requires further investigation. Additionally, it remains uncertain whether berberine is the sole antimicrobial active component of P. chinense Schneid. AIM OF STUDY This study aims to elucidate the anti-H. pylori infection activity of P. chinense Schneid, along with its mechanism of action and key antimicrobial active components. MATERIALS AND METHODS Phytochemical analysis was carried out by UPLC-MS/MS. HPLC was employed to quantify the berberine content of the extracts. Antimicrobial activity was assessed using the micro broth dilution method. Morphology was observed using SEM. The impact on urease activity was analyzed through in vitro urease enzyme kinetics. RT-qPCR was employed to detect the expression of virulence genes, including adhesin, flagellum, urease, and cytotoxin-related genes. The adhesion effect was evaluated by immunofluorescence staining and agar culture. RESULTS P. chinense Schneid exhibited strong antimicrobial activity against both antibiotic-sensitive and resistant H. pylori strains, with MIC ranging from 40 to 160 μg/mL. Combination with amoxicillin, metronidazole, levofloxacin, and clarithromycin did not result in antagonistic effects. P. chinense Schneid induced alterations in bacterial morphology and structure, downregulated the expression of various virulence genes, and inhibited urease enzyme activity. In co-infection systems, P. chinense Schneid significantly attenuated H. pylori adhesion and urease relative content, thereby mitigating cellular damage caused by infection. Huangjiu processing enhanced the anti-H. pylori activity of P. chinense Schneid. Besides berberine, P. chinense Schneid contained seven other components with anti-H. pylori activity, with palmatine exhibiting the strongest activity, followed by jatrorrhizine. CONCLUSIONS This study sheds light on the potential therapeutic mechanisms of P. chinense Schneid against H. pylori infection, demonstrating its capacity to disrupt bacterial structure, inhibit urease activity, suppress virulence gene transcription, inhibit adhesion, and protect host cells. The anti-H. pylori activity of P. chinense Schneid was potentiated by Huangjiu processing, and additional components beyond berberine were identified as possessing strong anti-H. pylori activity. Notably, jatrorrhizine, a core component of P. chinense Schneid, exhibited significant anti-H. pylori activity, marking a groundbreaking discovery.
Collapse
Affiliation(s)
- Meiyun Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Ziyao Wu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Yuanjing Zou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Chang Peng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Yajie Hao
- Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Zhixiang Zhu
- School of Medicine and Pharmacy (Qingdao), Ocean University of China, Qingdao, 266003, China.
| | - Xiaoyan Shi
- Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Bingmei Su
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Ling Ou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Yuqian Lai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Junwei Jia
- Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Mingjin Xun
- Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Hui Li
- Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Weixing Zhu
- Qingyuan Hospital of Traditional Chinese Medicine, Qingyuan, 511500, China.
| | - Zhong Feng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China; International Pharmaceutical Engineering Lab of Shandong Province, Feixian, 273400, China; Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Meicun Yao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
2
|
Ou L, Hao Y, Liu H, Zhu Z, Li Q, Chen Q, Wei R, Feng Z, Zhang G, Yao M. Chebulinic acid isolated from aqueous extracts of Terminalia chebula Retz inhibits Helicobacter pylori infection by potential binding to Cag A protein and regulating adhesion. Front Microbiol 2024; 15:1416794. [PMID: 39421559 PMCID: PMC11483367 DOI: 10.3389/fmicb.2024.1416794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Background Terminalia chebula Retz, known as the King of Tibet, is considered a functional food in China, celebrated for its antioxidant, immune-modulating, antibacterial, and anti-inflammatory properties. Chebulinic acid, derived from aqueous extracts of Terminalia chebula Retz, is known for its anti-inflammatory properties. However, its potential as an anti-Helicobacter pylori (HP) agent has not been fully explored. Methods Herein, we extracted the main compound from Terminalia chebula Retz using a semi-preparative liquid chromatography (LC) system and identified compound 5 as chebulinic acid through Ultra-high performance liquid chromatography-MS/MS (UPLC-MS/MS) and Nuclear Magnetic Resonance (NMR). To evaluate its role, we conducted minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) assays, scanning electron microscope (SEM) imaging, inhibiting kinetics curves, urea fast test, cell counting kit-8 (CCK-8) assay, western blot analysis, griess reagent system, and molecular docking. Results Our results showed that chebulinic acid effectively inhibited the growth of the HP strain ATCC 700392, damaged the HP structure, and exhibited selective antimicrobial activity without affecting normal epithelial cells GES-1. Importantly, it suppressed the expression of Cytotoxin-associated gene A (Cag A) protein, a crucial factor in HP infection. Molecular docking analysis predicted a strong affinity (-9.7 kcal/mol) between chebulinic acid and Cag A protein. Conclusion Overall, our findings suggest that chebulinic acid acts as an anti-adhesive agent, disrupting the adhesion of HP to host cells, which is a critical step in HP infection. It also suppresses the Cag A protein. These results highlight the potential of chebulinic acid against HP infections.
Collapse
Affiliation(s)
- Ling Ou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
| | - Yajie Hao
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, China
| | - Hengrui Liu
- Cancer Institute, Jinan University, Guangzhou, China
- Yinuo Biomedical Company, Tianjin, China
| | - Zhixiang Zhu
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, China
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Qingwei Li
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, China
| | - Qingchang Chen
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Ruixia Wei
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, China
| | - Zhong Feng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, China
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, China
| | - Guimin Zhang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, China
| | - Meicun Yao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
3
|
Chen S, Zhai D, Li Y, Tan Y, Tang X, Pu X, Chai Y, Li L. Study on the mechanism of inhibition of Escherichia coli by Polygonum capitatum based on network pharmacology and molecular docking technology: A review. Medicine (Baltimore) 2024; 103:e38536. [PMID: 38875382 PMCID: PMC11175921 DOI: 10.1097/md.0000000000038536] [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: 01/15/2024] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 06/16/2024] Open
Abstract
This study aims to analyze the effective components of Polygonum capitatum (PC) inhibiting Escherichia coli based on network pharmacology methods and predict its molecular mechanism of action. PC compounds and targets were collected from the TCMSP database, Swiss Target Prediction, and the literature. E coli targets were searched using the GeneCards database. The targets of E coli and the targets of the active ingredients of PC were taken as intersections to obtain the intersecting targets. The resulting overlapping targets were uploaded to the STRING database to construct the protein interaction network diagram of E coli target inhibition. The key targets for the inhibitory effect of PC on E coli were obtained. Gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed by uploading key targets into the DAVID database. The results showed that there were 50 targets for PC to inhibit E coli. Among them, there are 5 core targets, mainly including AKT1, TNF, EGFR, JUN, and ESR1. A total of 196 gene ontology functional analysis results and 126 Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis results were obtained. These include cellular response to cadmium-ion, cellular response to reactive oxygen species, pathways in cancer, prostate cancer, and PI3K-Akt signaling pathway. Molecular docking results indicate that Lutedin, Hirsutin, Flazin, and Ellagic acid in PC have high affinity for the target genes AKT1, TNF, MAPK3 and EGFR. PC exerts its inhibitory effect on E coli through multi-targets and multi-pathways, which provides a new basis for the new use of PC as an old medicine.
Collapse
Affiliation(s)
- Shunhuan Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Dongyan Zhai
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yuan Li
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yong Tan
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xiaoke Tang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xiang Pu
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yihui Chai
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Lailai Li
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
4
|
Shi G, Lu X, Zheng Y, Yang T, Zhu E, Song Y, Huang P. Insights into the potential dual-antibacterial mechanism of Kelisha capsule on Escherichia coli. BMC Complement Med Ther 2024; 24:207. [PMID: 38807130 PMCID: PMC11134901 DOI: 10.1186/s12906-024-04500-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/13/2024] [Indexed: 05/30/2024] Open
Abstract
Traditional Chinese medicine (TCM), AYURVEDA and Indian medicine are essential in disease prevention and treatment. Kelisha capsule (KLSC), a TCM formula listed in the Chinese Pharmacopoeia, has been clinically proven to possess potent antibacterial properties. However, the precise antimicrobial mechanism of KLSC remained unknown. This study aimed to elucidate the dual antibacterial mechanism of KLSC using network pharmacology, molecular docking, and experimental validation. By analyzing the growth curve of Escherichia coli (E. coli), it was observed that KLSC significantly inhibited its growth, showcasing a remarkable antibacterial effect. Furthermore, SEM and TEM analysis revealed that KLSC damaged the cell wall and membrane of E. coli, resulting in cytoplasmic leakage, bacterial death, and the exertion of antibacterial effects. The network pharmacology analysis revealed that KLSC exhibited an effect on E. coli ATP synthase, thereby influencing the energy metabolism process. The molecular docking outcomes provided evidence that the active compounds of KLSC could effectively bind to the ATP synthase subunit. Subsequently, experimental findings substantiated that KLSC effectively suppressed the activity of ATP synthase in E. coli and consequently decreased the ATP content. This study highlighted the dual antibacterial mechanism of KLSC, emphasizing its effects on cell structure and energy metabolism, suggesting its potential as a natural antibacterial agent for E. coli-related infections. These findings offered new insights into exploring the antibacterial mechanisms of TCM by focusing on the energy metabolism process.
Collapse
Affiliation(s)
- Guolin Shi
- Department of Ultrasound in Medicine, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Zhejiang University, 88 Jiefang Road, Shangcheng District, Hangzhou, 310009, China
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, China
| | - Xiao Lu
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, China
| | - Yuanhang Zheng
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, China
| | - Tao Yang
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, China
| | - Enyuan Zhu
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, China
| | - Yanhong Song
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Zhejiang University, 88 Jiefang Road, Shangcheng District, Hangzhou, 310009, China.
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310053, China.
| |
Collapse
|
5
|
Xun M, Feng Z, Li H, Yao M, Wang H, Wei R, Jia J, Fan Z, Shi X, Lv Z, Zhang G. In vitro anti-Helicobacter pylori activity and antivirulence activity of cetylpyridinium chloride. PLoS One 2024; 19:e0300696. [PMID: 38603679 PMCID: PMC11008818 DOI: 10.1371/journal.pone.0300696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
The primary treatment method for eradicating Helicobacter pylori (H. pylori) infection involves the use of antibiotic-based therapies. Due to the growing antibiotic resistance of H. pylori, there has been a surge of interest in exploring alternative therapies. Cetylpyridinium chloride (CPC) is a water-soluble and nonvolatile quaternary ammonium compound with exceptional broad-spectrum antibacterial properties. To date, there is no documented or described specific antibacterial action of CPC against H. pylori. Therefore, this study aimed to explore the in vitro activity of CPC against H. pylori and its potential antibacterial mechanism. CPC exhibited significant in vitro activity against H. pylori, with MICs ranging from 0.16 to 0.62 μg/mL and MBCs ranging from 0.31 to 1.24 μg/mL. CPC could result in morphological and physiological modifications in H. pylori, leading to the suppression of virulence and adherence genes expression, including flaA, flaB, babB, alpA, alpB, ureE, and ureF, and inhibition of urease activity. CPC has demonstrated in vitro activity against H. pylori by inhibiting its growth, inducing damage to the bacterial structure, reducing virulence and adherence factors expression, and inhibiting urease activity.
Collapse
Affiliation(s)
- Mingjin Xun
- National Engineering and Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, Shandong, China
| | - Zhong Feng
- National Engineering and Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, Shandong, China
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Hui Li
- National Engineering and Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, Shandong, China
| | - Meicun Yao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Haibo Wang
- National Engineering and Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, Shandong, China
| | - Ruixia Wei
- National Engineering and Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
| | - Junwei Jia
- National Engineering and Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, Shandong, China
| | - Zimao Fan
- National Engineering and Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, Shandong, China
| | - Xiaoyan Shi
- National Engineering and Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, Shandong, China
| | - Zhanzhu Lv
- National Engineering and Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, Shandong, China
| | - Guimin Zhang
- National Engineering and Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, Shandong, China
| |
Collapse
|
6
|
Ou L, Zhu Z, Hao Y, Li Q, Liu H, Chen Q, Peng C, Zhang C, Zou Y, Jia J, Li H, Wang Y, Su B, Lai Y, Chen M, Chen H, Feng Z, Zhang G, Yao M. 1,3,6-Trigalloylglucose: A Novel Potent Anti- Helicobacter pylori Adhesion Agent Derived from Aqueous Extracts of Terminalia chebula Retz. Molecules 2024; 29:1161. [PMID: 38474673 DOI: 10.3390/molecules29051161] [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: 01/24/2024] [Revised: 02/28/2024] [Accepted: 03/02/2024] [Indexed: 03/14/2024] Open
Abstract
1,3,6-Trigalloylglucose is a natural compound that can be extracted from the aqueous extracts of ripe fruit of Terminalia chebula Retz, commonly known as "Haritaki". The potential anti-Helicobacter pylori (HP) activity of this compound has not been extensively studied or confirmed in scientific research. This compound was isolated using a semi-preparative liquid chromatography (LC) system and identified through Ultra-high-performance liquid chromatography-MS/MS (UPLC-MS/MS) and Nuclear Magnetic Resonance (NMR). Its role was evaluated using Minimum inhibitory concentration (MIC) assay and minimum bactericidal concentration (MBC) assay, scanning electron microscope (SEM), inhibiting kinetics curves, urea fast test, Cell Counting Kit-8 (CCK-8) assay, Western blot, and Griess Reagent System. Results showed that this compound effectively inhibits the growth of HP strain ATCC 700392, damages the HP structure, and suppresses the Cytotoxin-associated gene A (Cag A) protein, a crucial factor in HP infection. Importantly, it exhibits selective antimicrobial activity without impacting normal epithelial cells GES-1. In vitro studies have revealed that 1,3,6-Trigalloylglucose acts as an anti-adhesive agent, disrupting the adhesion of HP to host cells, a critical step in HP infection. These findings underscore the potential of 1,3,6-Trigalloylglucose as a targeted therapeutic agent against HP infections.
Collapse
Affiliation(s)
- Ling Ou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Zhixiang Zhu
- School of Medicine and Pharmacy (Qingdao), Ocean University of China, Qingdao 266003, China
- International Pharmaceutical Engineering Lab of Shandong Province, Linyi 273400, China
| | - Yajie Hao
- International Pharmaceutical Engineering Lab of Shandong Province, Linyi 273400, China
| | - Qingwei Li
- International Pharmaceutical Engineering Lab of Shandong Province, Linyi 273400, China
| | - Hengrui Liu
- Regenerative Medicine Research Center, Future Homo Sapiens Institute of Regenerative Medicine Co., Ltd., Guangzhou 510535, China
| | - Qingchang Chen
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore
| | - Chang Peng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Chuqiu Zhang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yuanjing Zou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Junwei Jia
- International Pharmaceutical Engineering Lab of Shandong Province, Linyi 273400, China
| | - Hui Li
- International Pharmaceutical Engineering Lab of Shandong Province, Linyi 273400, China
| | - Yanhua Wang
- International Pharmaceutical Engineering Lab of Shandong Province, Linyi 273400, China
| | - Bingmei Su
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yuqian Lai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Meiyun Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Haobo Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Zhong Feng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
- School of Medicine and Pharmacy (Qingdao), Ocean University of China, Qingdao 266003, China
- International Pharmaceutical Engineering Lab of Shandong Province, Linyi 273400, China
| | - Guimin Zhang
- Lunan Pharmaceutical Group Co., Ltd., Linyi 276006, China
| | - Meicun Yao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
7
|
Feng Z, Li H, Hao Y, Peng C, Ou L, Jia J, Xun M, Zou Y, Chen M, Zhang G, Yao M. In vitro anti- Helicobacter pylori activity and the underlining mechanism of an empirical herbal formula - Hezi Qingyou. Front Microbiol 2024; 15:1355460. [PMID: 38440143 PMCID: PMC10910045 DOI: 10.3389/fmicb.2024.1355460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Background Helicobacter pylori (H. pylori) is thought to primarily colonize the human stomach and lead to various gastrointestinal disorders, such as gastritis and gastric cancer. Currently, main eradication treatment is triple or quadruple therapy centered on antibiotics. Due to antibiotic resistance, the eradication rate of H. pylori is decreasing gradually. Therefore, searching for anti-H. pylori drugs from herbal sources has become a strategy for the treatment. Our team proposed a Hezi Qingyou Formula (HZQYF), composed of Chebulae Fructus, Ficus hirta Vahl and Cloves, and studied its anti-H. pylori activity and mechanism. Methods Chemical components of HZQYF were studied using UHPLC-MS/MS and HPLC. Broth microdilution method and agar dilution method were used to evaluate HZQYF's antibacterial activity. The effects of HZQYF on expression of adhesion genes (alpA, alpB, babA), urease genes (ureE, ureF), and flagellar genes (flaA, flaB) were explored using Reverse Transcription-quantitative Polymerase Chain Reaction (RT-qPCR) technology. Effects on morphology and permeability of the extracellular membrane were studied using scanning electron microscopy (SEM) and N-phenylnaphthalen-1-amine (NPN) uptake. Effect on urease activity was studied using a urease kinetics analysis in vitro. Immunofluorescence staining method was used to examine the effect on adhesion. Western blot was used to examine the effect on cagA protein. Results Minimum inhibitory concentration (MIC) values of the formula against H. pylori clinical strains and standard strains were 80-160 μg/mL, and minimum bactericidal concentration (MBC) values were 160-320 μg/mL. The formula could down-regulate the expression of adhesion genes (alpA, alpB, babA), urease genes (ureE, ureF) and flagellar genes (flaA, flaB), change the morphology of H. pylori, increase its extracellular membrane permeability, and decrease its urease activity. Conclusion Present studies confirmed that HZQYF had promising in vitro anti-H. pylori activities and demonstrated its possible mechanism of action by down-regulating the bacterial adhesion, urease, and flagellar gene expression, which provided scientific bases for further clinical investigations.
Collapse
Affiliation(s)
- Zhong Feng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, China
| | - Hui Li
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, China
| | - Yajie Hao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, China
| | - Chang Peng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
| | - Ling Ou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
| | - Junwei Jia
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, China
| | - Mingjin Xun
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, China
| | - Yuanjing Zou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
| | - Meiyun Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
| | - Guimin Zhang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
- International Pharmaceutical Engineering Laboratory in Shandong Province, Shandong New Time Pharmaceutical Co., Ltd., Linyi, China
| | - Meicun Yao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
- Nanchang Research Institute, Sun Yat-sen University, Jiangxi, China
| |
Collapse
|
8
|
Ou L, Liu HR, Shi XY, Peng C, Zou YJ, Jia JW, Li H, Zhu ZX, Wang YH, Su BM, Lai YQ, Chen MY, Zhu WX, Feng Z, Zhang GM, Yao MC. Terminalia chebula Retz. aqueous extract inhibits the Helicobacter pylori-induced inflammatory response by regulating the inflammasome signaling and ER-stress pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117428. [PMID: 37981121 DOI: 10.1016/j.jep.2023.117428] [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: 09/05/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 11/21/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Terminalia chebula Retz., known as the King of Traditional Tibetan Medicine, is widely used for treating various ailments, particularly stomach disorders. It exhibited inhibitory activity against helicobacter pylori. AIM OF THE STUDY The exact mechanism by which T. chebula combats H. pylori infection remains unclear. Therefore, this study aimed to investigate its mechanism of action and the key pathways and targets involved. MATERIAL AND METHODS Minimum inhibitory concentration (MIC) assay, scanning electron microscope, and inhibiting kinetics curves were conducted. The mRNA expressions were measured by RNA-seq analysis and RT-QPCR. ELISA and Western blot were used to detect the changes in proteins. The main compounds were analyzed by High-performance Liquid Chromatography. The interaction between the compound and target was predicted by Molecular Docking. RESULTS The study revealed that T. chebula disrupted the structure of H. pylori bacteria and inhibited Cag A protein expression. Additionally, T. chebula can reduce the expression of flaA, flaB, babA, alpA, alpB, ureE, and ureF genes. Furthermore, T. chebula demonstrated its effectiveness in inhibiting the H. pylori-induced inflammatory response by regulating the inflammasome signaling and ER-stress pathway. Moreover, the study discovered that chebulagic acid has anti-HP activity and inhibits the expression of Cag A protein. CONCLUSIONS T. chebula acts as a natural remedy for combating H. pylori infection. Its ability to disrupt the bacterial structure, inhibit key proteins, regulate inflammatory pathways, and the presence of chebulagic acid contribute to its anti-H. pylori activity.
Collapse
Affiliation(s)
- Ling Ou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| | - Heng-Rui Liu
- Regenerative Medicine Research Center, Future Homo Sapiens Institute of Regenerative Medicine Co., Ltd, Guangzhou, China.
| | - Xiao-Yan Shi
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, 273400, Shandong, China.
| | - Chang Peng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| | - Yuan-Jing Zou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| | - Jun-Wei Jia
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, 273400, Shandong, China.
| | - Hui Li
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, 273400, Shandong, China.
| | - Zhi-Xiang Zhu
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, 273400, Shandong, China.
| | - Yan-Hua Wang
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, 273400, Shandong, China.
| | - Bing-Mei Su
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| | - Yu-Qian Lai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| | - Mei-Yun Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| | - Wei-Xing Zhu
- Qingyuan Hospital' of Traditional Chinese Medicine, Qingyuan, 511500, Guangdong, China.
| | - Zhong Feng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, Guangdong, China; Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Gui-Min Zhang
- Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Mei-Cun Yao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
9
|
Wang C, Jiang S, Zheng H, An Y, Zheng W, Zhang J, Liu J, Lin H, Wang G, Wang F. Integration of gut microbiome and serum metabolome revealed the effect of Qing-Wei-Zhi-Tong Micro-pills on gastric ulcer in rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117294. [PMID: 37839771 DOI: 10.1016/j.jep.2023.117294] [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: 09/03/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qing-Wei-Zhi-Tong Micro-pills (QWZT) is herbal compound used in the treatment of GU, whose functions include clearing the stomach and fire, softening the liver and relieving pain. However, its mechanistic profile on host intestinal microbiota and metabolism has not been determined. AIM OF THE STUDY The present study aimed to observe the healing effect of QWZT on acetic acid-induced gastric ulcer in a rat model and to preliminarily elucidate its possible therapeutic mechanism from the perspective of host intestinal microbiota and metabolism. MATERIALS AND METHODS The Wistar male rats (7 weeks old; weight 180-200 g) were randomly divided into normal control group (NC), acetic acid-induced gastric ulcer group (GU), and QWZT treatment group (High dose: 1250 mg/kg/day, Middle dose: 625 mg/kg/day, Low dose: 312.5 mg/kg/day) of 6 rats each. An acetic acid-induced gastric ulcer rat model was constructed based on anatomical surgery. QWZT (High dose, Middle dose, and Low dose) was used to treat gastric ulcer rats for 7 days by gavage. At the end of treatment, the body weight, macroscopic condition of gastric tissue ulcers, pathological changes (HE staining), inflammatory factors, oxidative stress factors, and endocrine factors were assessed in each group of rats. Fresh feces and serum from each group of rats were collected for microbiome and metabolome analysis on the machine, respectively. Drug-disease common targets and functional pathways were captured based on network pharmacology. The complex network of Herbs-Targets-Pathways-Metabolites-Microbiota interactions was constructed. Ultimately, Fecal Microbiota Transplantation (FMT) evaluated the contribution of gut microbiota in disease. RESULTS QWZT increased the abundance of beneficial bacteria (Bacteroides, Alloprevotella, Rikenellaceae_RC9_gut_group, Lactobacillus, Lachnospiraceae_NK4A136_group, Parabacteroides, etc.), reduced the abundance of harmful bacteria (Micromonospora, Geobacter, Nocardioides, and Arenimonas, etc.), reduced the levels of inflammatory mediators (12,13-EpOME, 9,10-Epoxyoctadecenoic acid, SM(d18:1/16:0) and Leukotriene A4, etc.), restored host metabolic disorders (Linoleic acid metabolism, Glycerophospholipid metabolism, and Arachidonic acid metabolism), and regulated the level of cytokines (IL-6, TNF-a, SOD, MDA, PEG-2 and NO), ultimately exerting an anti-ulcer effect. Apart from that, FMT improved acetic acid-induced gastric ulcers in rats. CONCLUSION QWZT improved acetic acid-induced gastric ulcers in rats by remodeling intestinal microbiota and regulating host metabolism. This work may promote the process of developing and utilizing clinical applications of QWZT.
Collapse
Affiliation(s)
- Chao Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
| | - Shengyu Jiang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China; Department of Laboratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| | - Haoyu Zheng
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
| | - Yiming An
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
| | - Wenxue Zheng
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
| | - Jiaqi Zhang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
| | - Jianming Liu
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China; Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| | - Hongqiang Lin
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China; Jilin Provincial Engineering Laboratory of Precision Prevention and Control for Common Diseases, Changchun, 130021, China.
| | - Guoqiang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China; Jilin Provincial Engineering Laboratory of Precision Prevention and Control for Common Diseases, Changchun, 130021, China.
| | - Fang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China; Jilin Provincial Science and Technology Innovation Centre for Secondary Development of Proprietary Chinese Medicines, Changchun, 130021, China; Jilin Provincial Engineering Laboratory of Precision Prevention and Control for Common Diseases, Changchun, 130021, China.
| |
Collapse
|
10
|
Chen P, Chen M, Peng C, Yan J, Shen X, Zhang W, Yuan Y, Gan G, Luo X, Zhu W, Yao M. In vitro anti-bactrical activity and its preliminary mechanism of action of the non-medicinal parts of Sanguisorba officinalis L. against Helicobacter pylori infection. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116981. [PMID: 37574016 DOI: 10.1016/j.jep.2023.116981] [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: 06/08/2023] [Revised: 07/20/2023] [Accepted: 07/29/2023] [Indexed: 08/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sanguisorba officinalis L. (S. officinalis L.), known as Di Yu (DY) in Traditional Chinese Medicine (TCM), are used to treat burns, vomiting of blood, asthma, intestinal infections, and dermatitis. It has been reported that the root of DY has a significant inhibitory effect on Helicobacter pylori (H. pylori). However, there is currently little research on the composition analysis and anti-H. pylori infection properties of the non-medicinal parts of DY, such as its stems, leaves, and flowers. AIM OF STUDY The commonly used eradication therapies for H. pylori infection are antibiotic-based therapies. With the increasing antibiotic resistance of H. pylori, it is urgent to find effective alternative therapies. To find alternative therapies and increase the utilization of DY, this study aims to investigate the phytochemistry profile, in vitro anti-H. pylori activity, and preliminary antibacterial mechanism of the non-medicinal parts of DY. MATERIALS AND METHODS The non-medicinal parts of DY extracts were obtained by using hot water reflux method. The chemical composition of these extracts was analyzed using colorimetric method, high-performance liquid chromatography (HPLC), and ultra-high-performance liquid chromatography-electrospray ionization-mass spectrometry (UPLC-ESI-MS). The in vitro anti-H. pylori activity was investigated using broth microdilution method, checkerboard dilution method, time-kill curve, time-inhibition curve, scanning electron microscopy, and transmission electron microscopy. Transcriptional sequencing technology was used to study the effect of DY stems and flowers on the gene expression of H. pylori and explore possible antibacterial mechanisms. RESULTS The non-medicinal parts of DY contain abundant phytochemicals, such as total phenols and total flavonoids, and possess strong inhibitory and bactericidal activity against both standard and clinical strains of H. pylori in vitro. The MIC was 80-1280 μg/mL and the MBC was 80-2560 μg/mL, and the strength of the antibacterial effects was dependent on the concentration of phytochemicals (total polyphenols, gallic acid and ellagic acid). In addition, the combination of non-medicinal parts of DY with antibiotics, such as amoxicillin, metronidazole, levofloxacin, and clarithromycin, did not result in any antagonistic effects. All of them could disrupt the morphology, internal microscopic and cell wall structures of H. pylori thereby acting as an inhibitor. The mechanism of action was found to be the disruption of H. pylori morphology, internal microstructure, and cell wall. Transcriptomic analysis showed that the non-medicinal parts of DY significantly regulated the gene expression of H. pylori, especially the metabolic pathway. CONCLUSIONS This study analyzed the chemical composition of the non-medicinal parts of DY and confirmed its inhibitory and bactericidal activities against H. pylori, both standard and clinical strains. Additional, the mechanism of inhibition involves disrupting the structure of H. pylori cells, altering gene expression, and interfering with bacterial metabolic pathways. This study provides a reference for further resource utilization and the development of H. pylori drugs using the non-medicinal parts of DY.
Collapse
Affiliation(s)
- Pengting Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Meiyun Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Chang Peng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Jiahui Yan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Xue Shen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Weijia Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Yuemei Yuan
- School of Earth Sciences and Engineering, Sun Yat-sen University, Zhuhai 519080, China.
| | - Guoxing Gan
- Qingyuan Hospital of Traditional Chinese Medicine, Qingyuan 511500, China.
| | - Xiaojun Luo
- Lianzhou Hospital of Traditional Chinese Medicine, Qingyuan 513400, China.
| | - Weixing Zhu
- Qingyuan Hospital of Traditional Chinese Medicine, Qingyuan 511500, China.
| | - Meicun Yao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
11
|
Peng C, Feng Z, Ou L, Zou Y, Sang S, Liu H, Zhu W, Gan G, Zhang G, Yao M. Syzygium aromaticum enhances innate immunity by triggering macrophage M1 polarization and alleviates Helicobacter pylori-induced inflammation. J Funct Foods 2023; 107:105626. [DOI: 10.1016/j.jff.2023.105626] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024] Open
|
12
|
Zahra N, Iqbal J, Arif M, Abbasi BA, Sher H, Nawaz AF, Yaseen T, Ydyrys A, Sharifi-Rad J, Calina D. A comprehensive review on traditional uses, phytochemistry and pharmacological properties of Paeonia emodi Wall. ex Royle: current landscape and future perspectives. Chin Med 2023; 18:23. [PMID: 36859262 PMCID: PMC9979516 DOI: 10.1186/s13020-023-00727-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 02/10/2023] [Indexed: 03/03/2023] Open
Abstract
Paeonia emodi Wall. ex Royle is commonly known as Himalayan paeony has great importance as a food and medicine. The practice of Paeonia emodi Wall. ex Royle is very ancient and it is conventionally used for a wide range of illnesses in the folk system of medicine because of its wide beneficial phytochemical profile. The main purpose of the current review was the synthesis of recent data on botany, ethnopharmacology, phytochemistry and potential pharmacological mechanisms of action of Paeonia emodi Wall. ex Royle, thus offering new prospects for the development of new adjuvant natural therapies. Using scientific databases such as PubMed/MedLine, Scopus, Web of Science, ScienceDirect, Google Scholar, Springer, and Wiley, a comprehensive literature search was performed for Paeonia emodi Wall. ex Royle. For searching, we used the next MeSH terms: "Biological Product/isolation and purification", "Biological Products/pharmacology", "Drug Discovery/methods", "Ethnopharmacology, Medicine", "Traditional/methods", "Paeonia/chemistry", "Plant Extracts/pharmacology", "Phytochemicals/chemistry", "Phytochemicals/pharmacology", "Plants, Medicinal". The results of the most recent studies were analyzed and the most important data were summarized in tables and figures. Phytochemical research of Paeonia emodi Wall. ex Royle has led to the isolation of triterpenes, monoterpenes, phenolic acids, fatty acids, organic compounds, steroids, free radicals and some other classes of primary metabolites. In addition, diverse pharmacological activities like antibacterial, antifungal, anticoagulant, airway relaxant lipoxygenase and beta-glucuronidase inhibiting activity, radical scavenging activity, phytotoxic and insecticidal activities have been reported for Paeonia emodi Wall. ex Royle. Different bioactive compounds of Paeonia emodi Wall. ex Royle has proven their therapeutic potential in modern pharmacological and biomedical research to cure numerous gastrointestinal and nervous disorders. In future, further in vitro and in vivo therapeutic studies are required to identify new mechanisms of action, pharmacokinetics studies, and new pharmaceutical formulations for target transport and possible interaction with allopathic drugs. Also, new research regarding quality evaluation, toxicity and safety data in humans is needed.
Collapse
Affiliation(s)
- Nida Zahra
- Department of Biotechnology, University of Mianwali, Mianwali, 42200 Pakistan
| | - Javed Iqbal
- Department of Botany, Bacha Khan University, Charsadda, Khyber Pakhtunkhwa Pakistan
| | - Muhammad Arif
- Department of Biotechnology, University of Mianwali, Mianwali, 42200 Pakistan
| | - Banzeer Ahsan Abbasi
- Department of Botany, Rawalpindi Women University, 6th Road, Satellite Town, Rawalpindi, 46300 Pakistan
| | - Hassan Sher
- Center for Plant Sciences and Biodiversity, University of Swat, Kanju, 19201 Pakistan
| | - Ayesha Fazal Nawaz
- National Institute of Genomics and Advanced Biotechnology (NIGAB), National Agricultural Research Center (NARC), Park Road, Islamabad, Pakistan
| | - Tabassum Yaseen
- Department of Botany, Bacha Khan University, Charsadda, Khyber Pakhtunkhwa Pakistan
| | - Alibek Ydyrys
- Biomedical Research Centre, Al-Farabi Kazakh National University, Al-Farabi Ave. 71, 050040 Almaty, Kazakhstan
- The Elliott School of International Affairs, George Washington University, 1957 E St NW, Washington, DC 20052 USA
| | | | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
13
|
Bao Z, Wu G, Du J, Ye Y, Zheng Y, Wang Y, Ji R. The comparative efficacy and safety of 9 traditional Chinese medicines combined with standard quadruple therapy for Helicobacter pylori-associated gastritis: a systematic review and network meta-analysis. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1349. [PMID: 36660649 PMCID: PMC9843361 DOI: 10.21037/atm-22-5421] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/08/2022] [Indexed: 01/01/2023]
Abstract
Background There are 9 traditional Chinese medicines (TCMs) combined with standard quadruple (SQ) available for the treatment of Helicobacter pylori (Hp)-associated gastritis, but their relative efficacy and best options in clinical decision making are unknown due to a lack of high-quality head-to-head randomized controlled trials (RCTs). This study aimed to explore which formulas are the most effective and/or safest for Hp-associated gastritis. Methods We performed a search of electronic databases including PubMed, Web of Science, Cochrane Library, Embase, Chinese databases and South Korean database from inception to March 2022 to identify all relevant RCTs on the comparison between TCM combined with SQ and SQ for Hp-associated gastritis. Efficacy outcomes were the eradication rate of Hp and therapeutic response rate, and safety outcome was incidence of adverse reactions. Publication bias was assessed quantitatively using Egger's regression analysis and qualitatively using trim-and-fill method. Quality assessment was performed using Cochrane Risk of Bias, version 2 (ROB 2) tool. The Bayesian methods were applied to compare each treatment. Results A total of 55 trials with 6,187 patients were involved. The experimental group included 9 TCMs combined with SQ. The control group was SQ. The pair-wise meta-analysis demonstrated that compared with control group, 8 TCMs combined with SQ could statistically improve the eradication rate of Hp in patients with gastritis, 9 TCMs combined with SQ could significantly improve the therapeutic response rate. Additionally, Banxia Xiexin decoction combined with SQ (BXS) could statistically decrease the incidence of adverse reactions. The network meta-analysis results showed that BXS, Xiangsha Liujunzi combined with SQ (XSS), and Huangqi Jianzhong decoction combined with SQ (HQS) was the best measures to effectively eradicate Hp, enhance therapeutic effect, and decrease adverse reactions, respectively. The results of trim-and-fill method indicated that the results were stable and less affected by publication bias. Conclusions Compared with SQ, TCM combined with SQ generally has a better clinical effect and higher safety in patients with Hp-associated gastritis. BXS, XSS, and HQS are recommended based on the patient's condition and needs in clinical practice. Further high-quality double-blinded RCTs are warranted to validate the conclusions.
Collapse
Affiliation(s)
- Zhixian Bao
- The First Clinical Medical College, Lanzhou University, Lanzhou, China;,Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China;,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Guobing Wu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China;,Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China;,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Jie Du
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Yuwei Ye
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China;,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ya Zheng
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China;,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuping Wang
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China;,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Rui Ji
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China;,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
14
|
Jia X, Huang Q, Lin M, Chu Y, Shi Z, Zhang X, Ye H. Revealing the novel effect of Jinghua Weikang capsule against the antibiotic resistance of Helicobacter pylori. Front Microbiol 2022; 13:962354. [PMID: 36147839 PMCID: PMC9485998 DOI: 10.3389/fmicb.2022.962354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/15/2022] [Indexed: 11/21/2022] Open
Abstract
Background Helicobacter pylori (H. pylori) infects half of the human population globally. Eradication rates with triple or quadruple therapy have decreased owing to the increasing rate of antibiotic resistance. Jinghua Weikang capsule (JWC) is the first and most popular Chinese patent medicine approved by the state for the treatment of gastritis and peptic ulcers caused by H. pylori infection in China. Previous studies have found that JWC has a certain bactericidal effect on drug-resistant H. pylori and its major component, Chenopodium ambrosioides L. inhibits biofilm formation, but the mechanism remains unclear. This study focused on drug-resistant H. pylori and explored whether JWC could reverse drug resistance and its related mechanisms. Method The agar plate dilution method, E-test method, and killing kinetics assay were used to evaluate the bactericidal effect of JWC on antibiotic-resistant H. pylori and its effect on antibiotic resistance. Sanger sequencing was used to detect mutations in drug resistance genes. The crystal violet method, scanning electron microscopy, and confocal laser scanning microscopy were used to evaluate the effects of JWC on biofilms. qPCR was performed to evaluate the effect of JWC on the expression of efflux pump-related genes. qPCR and immunofluorescence were used to evaluate the effects of JWC on H. pylori adhesion. Results JWC showed considerable antibacterial activity against drug-resistant H. pylori strains, with minimum inhibitory concentration (MIC) values ranging from 64 to 1,024 μg/ml. The MIC of metronidazole (MTZ) against H. pylori 26,695–16R decreased from 64 to 6 μg/ml after treatment with 1/2 MIC of JWC. The resistance of H. pylori 26,695–16R to MTZ was reversed by JWC, and its effect was better than that of PaβN and CCCP. H. pylori 26,695–16R is a moderate biofilm-forming strain, and JWC (16–64 μg/ml) can inhibit the formation of biofilms in H. pylori 26,695–16R. JWC reduced the expression of HP0605-HP0607 (hefABC), HP0971-HP0969 (hefDEF), HP1327-HP1329 (hefGHI), and HP1489-HP1487. JWC reduced the adhesion of H. pylori to GES-1 cells and the expression of adhesives NapA, SabA, and BabA. Conclusion The reversal of MTZ resistance by JWC may be achieved through the adhesin/efflux pump-biofilm pathway.
Collapse
|
15
|
Salinas Ibáñez ÁG, Origone AL, Liggieri CS, Barberis SE, Vega AE. Asclepain cI, a proteolytic enzyme from Asclepias curassavica L., a south American plant, against Helicobacter pylori. Front Microbiol 2022; 13:961958. [PMID: 36060760 PMCID: PMC9433900 DOI: 10.3389/fmicb.2022.961958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Helicobacter pylori is a Gram negative bacterium most frequently associated with human gastrointestinal infections worldwide. The increasing occurrence of antibiotic-resistant isolates of H. pylori constitutes a challenge. The eradication of the microorganism is currently being considered a “high priority” by the World Health Organization (WHO). In this context, bioactive compounds found in natural products seem to be an effective therapeutic option to develop new antibiotics against the pathogen. In this study, we investigated the effect of asclepain cI, the main purified proteolytic enzyme of the latex of petioles and stems from Asclepia curassavica L. (Asclepiadaceae), a South American native plant, against H. pylori; in order to obtain a natural therapeutic adjuvant and a safe nutraceutical product. Asclepain cI showed antibacterial activity against reference strains and drug-resistant clinical isolates of H. pylori in vitro. A range of minimal inhibitory concentration (MIC) from 1 to 2 μg/ml and minimal bactericidal concentration (MBC) from 2 to 4 μg/ml was obtained, respectively. The action of asclepain cI on the transcription of omp18, ureA, flaA genes showed a significantly decreased expression of the selected pathogenic factors. Furthermore, asclepain cI did not induce toxic effects at the concentrations assayed. Asclepain cI could be considered a highly feasible option to be used as a natural therapeutic adjuvant and a safe nutraceutical product against H. pylori.
Collapse
Affiliation(s)
- Ángel Gabriel Salinas Ibáñez
- Laboratorio de Microbiología e Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
- Instituto de Física Aplicada (INFAP) - Centro Científico Tecnológico (CCT) San Luis - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis, Argentina
| | - Anabella L. Origone
- Instituto de Física Aplicada (INFAP) - Centro Científico Tecnológico (CCT) San Luis - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis, Argentina
- Laboratorio de Control de Calidad y Desarrollo de Bromatología, Universidad Nacional de San Luis, San Luis, Argentina
| | - Constanza S. Liggieri
- Centro de Investigación de Proteínas Vegetales (CIProVe), Universidad Nacional de La Plata, La Plata, Argentina
| | - Sonia E. Barberis
- Instituto de Física Aplicada (INFAP) - Centro Científico Tecnológico (CCT) San Luis - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis, Argentina
- Laboratorio de Control de Calidad y Desarrollo de Bromatología, Universidad Nacional de San Luis, San Luis, Argentina
- *Correspondence: Sonia E. Barberis
| | - Alba E. Vega
- Laboratorio de Microbiología e Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| |
Collapse
|
16
|
Yan Z, Zhang K, Zhang K, Wang G, Wang L, Zhang J, Qiu Z, Guo Z, Kang Y, Song X, Li J. Huang Bai Jian Pi decoction alleviates diarrhea and represses inflammatory injury via PI3K/Akt/NF-κB pathway: In vivo and in vitro studies. JOURNAL OF ETHNOPHARMACOLOGY 2022; 292:115212. [PMID: 35331876 DOI: 10.1016/j.jep.2022.115212] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 05/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huang Bai Jian Pi (HBJP) decoction, a Chinese herbal formula based on the Pulsatilla decoction (PD) and Si Junzi decoction, is efficacy to treat clinical diarrhea in calves. AIM OF THE STUDY The mechanism of HBJP decoction to treat calf diarrhea remains unclear. This study was to investigate the therapeutic effect and anti-inflammatory mechanism of HBJP decoction on diarrhea in rats. MATERIALS AND METHODS Thirty-six Sprague Dawley rats were randomly divided into control group, model group, PD group and three treated groups with HBJP decoction. The diarrheal model in rats was established by multiple factors including high-sugar and fat diet, high temperature and dampness environment, biological pathogenic factors. The diarrheal animals were treated with HBJP decoction or PD for 5 days. The inflammatory model of the intestinal epithelioid cell line 6 (IEC-6) was induced by TNF-α. The clinical symptoms, blood routine and biochemistry parameters, histopathology of main organs were detected. The proteins associated with PI3K/Akt/NF-κB pathway and the expression levels of cytokines associated with inflammation were detected in vivo and in vitro by Western blot and ELISA. RESULTS The model rats showed obvious diarrheal symptoms, and the obvious systemic inflammatory response accompanied with abnormal change in blood routine, biochemistry parameters and histopathology. HBJP decoction alleviated obviously the clinical symptoms, and pathological changes of the liver, colon and lung, and abnormal blood routine and biochemistry indexes in rats. The expression of P-PI3K, P-Akt, P-NF-κB, IL-1β, IL-6 was significantly increased, and the expression of IL-10 was markedly decreased in diarrheal rats and IEC-6 with inflammation. HBJP decoction significantly inhibited the PI3K/AKT/NF-κB signal pathway and adjusted the expression of these inflammatory cytokines. CONCLUSIONS The finding suggested that HBJP decoction alleviate the inflammation in diarrhea through inhibiting the PI3K/Akt/NF-κB signal pathway, which provides scientific evidences for the clinical application of HBJP decoction in diarrhea.
Collapse
Affiliation(s)
- Zunxiang Yan
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, 730050, Lanzhou, China; College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, China
| | - Kai Zhang
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, 730050, Lanzhou, China
| | - Kang Zhang
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, 730050, Lanzhou, China
| | - Guibo Wang
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, 730050, Lanzhou, China
| | - Lei Wang
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, 730050, Lanzhou, China
| | - Jingyan Zhang
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, 730050, Lanzhou, China
| | - Zhengying Qiu
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, 730050, Lanzhou, China
| | - Zhiting Guo
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, 730050, Lanzhou, China
| | - Yandong Kang
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, 730050, Lanzhou, China
| | - Xiaoping Song
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, China.
| | - Jianxi Li
- Engineering and Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, 730050, Lanzhou, China.
| |
Collapse
|
17
|
Peng C, Sang S, Shen X, Zhang W, Yan J, Chen P, Jiang C, Yuan Y, Zhu W, Yao M. In vitro anti-Helicobacter pylori activity of Syzygium aromaticum and the preliminary mechanism of action. JOURNAL OF ETHNOPHARMACOLOGY 2022; 288:114995. [PMID: 35032584 DOI: 10.1016/j.jep.2022.114995] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/28/2021] [Accepted: 01/11/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The dried flower bud of Syzygium aromaticum (L.) Merr. & L.M Perry (S. aromaticum) (Myrtaceae), also known as clove, was used in Traditional Chinese Medicine (TCM) to aid gastrointestinal function and treat stomach disorders including vomiting, flatulence and nausea. And it is a food homology medicine which is a promising candidate for H. pylori treatment. H. pylori is a Gram-negative bacterium that infects approximately 50% of the human population worldwide, which is closely related to multiple gastric diseases, including gastric cancer. However, there are still no sufficient studies on the anti-H. pylori activity of S. aromaticum, especially for the mechanism of action. AIM OF STUDY This study aimed to study the antibacterial activities of S. aromaticum extracts on both antibiotic-sensitive and -resistant H. pylori strains, and to explore the underlying mechanisms of action. MATERIALS AND METHODS The S. aromaticum extracts were obtained by heat reflux extraction and lyophilized to powder form. The phytochemical analyses were performed by High-performance liquid chromatography (HPLC) and UPLC-electrospray ionization mass spectrometry (ESI-MS). In vitro anti-H. pylori activity was evaluated by broth microdilution method. Mechanism of action studies included morphological observation using electron microscopy, determination of expression of virulence genes by reverse transcription quantitative polymerase chain reaction (RT-qPCR), genes expression profile identification by transcriptomic analysis, and exploration of anti-H. pylori infection mechanisms by network pharmacology analysis and western blotting validation. RESULTS The S. aromaticum extracts, aqueous extract (AE) and 75% hydroalcoholic extract (HE), exerted significant antibacterial activities against both antibiotic-sensitive and -resistant H. pylori strains with MICs of 160∼320 μg/ml, without developing drug resistance. Among them, AE was bactericide to all the tested strains with MBCs of less than 4MIC, while HE was merely bacteriostatic to most of the tested strains with MBCs of 2MIC∼16MIC. Besides, they showed no antagonistic effects in combination with clarithromycin, metronidazole, levofloxacin, and amoxicillin. Additionally, these extracts altered the morphology and ultrastructure and down-regulated the virulence genes expression of H. pylori. And transcriptomic analysis showed that they regulated genes expression of multiple H. pylori biological processes, including tricarboxylic acid cycle (TAC) and pyruvate metabolic pathways. Furthermore, these extracts combated the abnormal activation of PI3K-Akt and MAPK signaling pathways caused by H. pylori infection. CONCLUSIONS Overall, the present study firstly analyzed the chemical compositions of S. aromaticum extracts, and then confirmed their activities on both antibiotic-sensitive and -resistant H. pylori strains. In addition, the mechanisms of action of S. aromaticum extracts against H. pylori were found to be destroying the bacterial structure, down-regulating the expression of virulence genes, and interfering TAC and pyruvate metabolic pathways. Finally, S. aromaticum extracts were found to combated the abnormal activation of PI3K-Akt and MAPK signaling pathways to treat H. pylori infection. This study should accelerate further research and application of S. aromaticum against H. pylori infection.
Collapse
Affiliation(s)
- Chang Peng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Shuyi Sang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Xue Shen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Weijia Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Jiahui Yan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Pengting Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Cheng Jiang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Yuemei Yuan
- School of Ecology, Sun Yat-sen University, Shenzhen, 518107, China.
| | - Weixing Zhu
- Qingyuan Hospital of Traditional Chinese Medicine, Qingyuan, 511500, China.
| | - Meicun Yao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
18
|
Yan J, Peng C, Chen P, Zhang W, Jiang C, Sang S, Zhu W, Yuan Y, Hong Y, Yao M. In-vitro anti-Helicobacter pylori activity and preliminary mechanism of action of Canarium album Raeusch. fruit extracts. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114578. [PMID: 34464702 DOI: 10.1016/j.jep.2021.114578] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/17/2021] [Accepted: 08/27/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Canarium album Raeusch. belongs to the Burseraceae family. Its ripe fruits, known as Qing Guo (QG) in Traditional Chinese Medicine (TCM), are used to treat sore throat, cough, and fish or crab poisoning. QG was reported to have antibacterial activity, and it exerted excellent anti-Helicobacter pylori (H. pylori) activity in our screening of abundant TCM. However, few studies have reported its anti-H. pylori activity and mechanism. AIM OF STUDY The commonly used eradication therapies for H. pylori infection are antibiotic-based therapies. With the increasing antibiotic resistance of H. pylori, interest in finding alternative therapies has been aroused. This study investigated the phytochemistry profile, in vitro anti-H. pylori activity and possible anti-bacterial mechanism of QG extracts. MATERIALS AND METHODS QG extracts were obtained by heat reflux extraction, ultrasonic extraction or liquid-liquid extraction with different solvents. The quantitative and qualitative phytochemical analyses were performed by colorimetric determination, high-performance liquid chromatography (HPLC), and UPLC-electrospray ionization mass spectrometry (ESI-MS). In vitro anti- H. pylori activity was assessed by broth micro-dilution method. Mechanism of action studies included morphological observation using electron microscopy, urease inhibition assay and determination of expression of virulence genes by RT-qPCR. RESULTS All QG extracts especially ethyl acetate extract (QGEAE) were rich in phenolic components, with the minimum inhibitory concentrations (MICs) on H.pylori of 39-625 μg/ml and minimum bactericidal concentrations (MBCs) of 78-1250 μg/ml. Both aqueous extract (QGAE) and QGEAE could induce the morphological and structural changes of H. pylori, inhibit urease activity with IC50 of 1093 μg/ml and 332.90 μg/ml, respectively, and down-regulate the virulence genes, such as vacA and cagA. CONCLUSIONS QG may exhibit in vitro anti-H. pylori activity by inhibiting growth, destroying the bacterial structure and down-regulating the expression of virulence factors. Moreover, QG is the homology of food and TCM, which can be considered as a safe and convenient agent against H. pylori infection.
Collapse
Affiliation(s)
- Jiahui Yan
- School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Chang Peng
- School of Pharmaceutical Science (Shenzhen), Sun Yat-Sen University, Shenzhen, 518107, China
| | - Pengting Chen
- School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Weijia Zhang
- School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Cheng Jiang
- School of Pharmaceutical Science (Shenzhen), Sun Yat-Sen University, Shenzhen, 518107, China
| | - Shuyi Sang
- School of Pharmaceutical Science (Shenzhen), Sun Yat-Sen University, Shenzhen, 518107, China
| | - Weixing Zhu
- Qingyuan Hospital of Traditional Chinese Medicine, Qingyuan, 511500, China.
| | - Yuemei Yuan
- School of Ecology, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yanjun Hong
- School of Pharmaceutical Science (Shenzhen), Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Meicun Yao
- School of Pharmaceutical Science (Shenzhen), Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|