1
|
Wu C, Zhang N, Li H, Wang H, Han L, Wang Y, Li F, Tian F. Preparation of immobilized xanthine oxidase with magnetic metal-organic framework and its application in screening of active ingredients in traditional Chinese medicine. Mikrochim Acta 2025; 192:319. [PMID: 40272571 DOI: 10.1007/s00604-025-07167-0] [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: 11/20/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025]
Abstract
Enzymes play a crucial role in the development and progression of various diseases, making them important targets for drug development. However, the stability issues associated with natural enzymes limit their broader application. Traditional methods for screening enzyme inhibitors from natural products are often time-consuming and labor-intensive. In this study, we designed and employed magnetic metal-organic frameworks (MOFs) to immobilize xanthine oxidase for the first time. By leveraging the porous structure and high specific surface area of MOFs, combined with the magnetic responsiveness of nanoparticles, we successfully developed a novel method for the efficient screening of potential enzyme inhibitors derived from natural products. By using 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide/N-hydroxysuccinimide (EDC/NHS) as a cross-linking agent, we achieved efficient immobilization of xanthine oxidase and identified baicalin as a potential inhibitor from the extract of Scutellaria baicalensis. In addition, we confirmed the adsorption capacity of this method for hordenine, demonstrated the specific adsorption of allopurinol, and also performed in vitro activity validation for baicalein. We not only successfully prepared the immobilized enzyme but also showcased that this method can efficiently screen and isolate potential enzyme inhibitors from traditional Chinese medicine, which provides a rapid and efficient new strategy for identifying enzyme inhibitors in natural products. This innovative approach offers a fresh perspective on the application of botanical medicine and the pharmacological treatment of hyperuricemia, which has important theoretical and practical significance. Graphical abstract Schematic diagram of synthesis of XO@MMOF (A) and ligand fishing process (B).
Collapse
Affiliation(s)
- Chenxi Wu
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China
| | - Na Zhang
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China
| | - Hailin Li
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China
| | - Hao Wang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Lifeng Han
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China
| | - Yuefei Wang
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China
| | - Fuyi Li
- South Australian Immunogenomics Cancer Institute, The University of Adelaide, 4 North Terrace, Adelaide, SA, 5000, Australia
| | - Fei Tian
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China.
| |
Collapse
|
2
|
Kang HG, Wei ML, Wang JL, Ma CP, Zhang X, Huang BW, Xin LS, Bai CM. Berberine, a Natural Compound That Demonstrates Antiviral Effects Against Ostreid Herpesvirus 1 Infection in Anadara broughtonii. Viruses 2025; 17:282. [PMID: 40007037 PMCID: PMC11860497 DOI: 10.3390/v17020282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/26/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Ostreid herpesvirus 1 (OsHV-1) infection is the primary viral disease responsible for large-scale mortality in bivalve mollusks worldwide, and effective strategies to control the outbreaks of this disease are still lacking. Berberine (BBR), a plant-derived alkaloid, has demonstrated antiviral activity against various vertebrate viruses, while its potential antiviral effects on molluscan herpesviruses remain to be fully elucidated. Therefore, the present study sought to investigate the potential of berberine hydrochloride (BBH) against OsHV-1 infection in blood clams (Anadara broughtonii). The most optimal BBH concentration was figured out according to virus replication and mortality rates during in vivo experimental infection. Quantitative PCR and reverse transcription quantitative PCR were utilized to monitor the OsHV-1 genomic copy numbers and viral gene transcription levels during the development of OsHV-1 infection in the BBH-treated and control groups. The results demonstrated that a 3 mg/L BBH bath immersion significantly suppressed OsHV-1 replication in blood clams. During the early stage of infection (24 h), BBH treatment significantly reduced the expression of OsHV-1 open reading frames (ORFs) related to early enzymes, putative membrane proteins, and nucleocapsid proteins. At 96 h post-infection, all untreated blood clams died, whereas the survival rate of BBH-treated individuals increased to 46.67%. This study provides preliminary evidence for the inhibitory effects of BBH on OsHV-1, paving the way for the development of pharmacological control technologies for OsHV-1 infections.
Collapse
Affiliation(s)
- Hui-Gang Kang
- Qingdao Nucleic Acid Rapid Detection Engineering Research Center, Qingdao Key Laboratory of Nucleic Acid Rapid Detection, College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; (H.-G.K.); (M.-L.W.); (J.-L.W.); (C.-P.M.)
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (X.Z.); (B.-W.H.)
| | - Mao-Le Wei
- Qingdao Nucleic Acid Rapid Detection Engineering Research Center, Qingdao Key Laboratory of Nucleic Acid Rapid Detection, College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; (H.-G.K.); (M.-L.W.); (J.-L.W.); (C.-P.M.)
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (X.Z.); (B.-W.H.)
| | - Jing-Li Wang
- Qingdao Nucleic Acid Rapid Detection Engineering Research Center, Qingdao Key Laboratory of Nucleic Acid Rapid Detection, College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; (H.-G.K.); (M.-L.W.); (J.-L.W.); (C.-P.M.)
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (X.Z.); (B.-W.H.)
| | - Cui-Ping Ma
- Qingdao Nucleic Acid Rapid Detection Engineering Research Center, Qingdao Key Laboratory of Nucleic Acid Rapid Detection, College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; (H.-G.K.); (M.-L.W.); (J.-L.W.); (C.-P.M.)
| | - Xiang Zhang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (X.Z.); (B.-W.H.)
| | - Bo-Wen Huang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (X.Z.); (B.-W.H.)
| | - Lu-Sheng Xin
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (X.Z.); (B.-W.H.)
| | - Chang-Ming Bai
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (X.Z.); (B.-W.H.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Shandong Center of Technology Innovation for Oyster Seed Industry, Qingdao 266105, China
| |
Collapse
|
3
|
Liu Y, Wang X, Chen Y, Zhou L, Wang Y, Li L, Wang Z, Yang L. Pharmacological mechanisms of traditional Chinese medicine against acute lung injury: From active ingredients to herbal formulae. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:155562. [PMID: 39536423 DOI: 10.1016/j.phymed.2024.155562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are the leading causes of acute respiratory failure in many critical diseases and are among the main respiratory diseases with high clinical mortality. The global outbreak of coronavirus disease 2019 (COVID-19) can cause severe ARDS, resulting in a steep rise in the number of patient deaths. Therefore, it is important to explore the pathogenesis of ALI and find effective therapeutic agents. In recent years, thanks to modern biomedical tools, some progress has been made in the application of traditional Chinese medicine (TCM) treatment principles based on syndromic differentiation and holistic concepts in clinical and experimental studies of ALI. More and more TCM effective components and formulae have been verified to have significant curative effects, which have a certain guiding significance for clinical practice. PURPOSE It is hoped to provide reference for the clinical research of ALI/ARDS and provide theoretical basis and technical support for the scientific application of TCM in respiratory related diseases. METHODS We performed a literature survey using traditional books of Chinese medicine and online scientific databases including PubMed, Web of Science, Google Scholar, ScienceDirect, China National Knowledge Infrastructure (CNKI), and others up to January 2023. RESULTS In recent years, thanks to modern biomedical tools, some progress has been made in the application of TCM treatment principles based on syndromic differentiation and holistic concepts in clinical and experimental studies of ALI. This paper mainly reviews the research progress of ALI/ARDS mechanism, the understanding of its etiology and pathogenesis by TCM, and the therapeutic effects of TCM formulae and active ingredients of Chinese medicine. A large number of studies have shown that the effective components and formulae of TCM can prevent or treat ALI/ARDS in vivo and in vitro experiments. CONCLUSION TCM effective components and formulae play an important role in the prevention and treatment of ALI/ARDS through multiple approaches and multiple targets, and provide necessary theoretical support for the further development and utilization of TCM resources.
Collapse
Affiliation(s)
- Yamin Liu
- The MOE Key Laboratory of Standardization of Chinese Medicines, the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xunjiang Wang
- The MOE Key Laboratory of Standardization of Chinese Medicines, the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yilin Chen
- The MOE Key Laboratory of Standardization of Chinese Medicines, the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Limei Zhou
- The MOE Key Laboratory of Standardization of Chinese Medicines, the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yining Wang
- The MOE Key Laboratory of Standardization of Chinese Medicines, the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Linnan Li
- The MOE Key Laboratory of Standardization of Chinese Medicines, the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Zhengtao Wang
- The MOE Key Laboratory of Standardization of Chinese Medicines, the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Li Yang
- The MOE Key Laboratory of Standardization of Chinese Medicines, the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
4
|
Gao T, Liu J, Huang N, Zhou Y, Li C, Chen Y, Hong Z, Deng X, Liang X. Sangju Cold Granule exerts anti-viral and anti-inflammatory activities against influenza A virus in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118521. [PMID: 38969152 DOI: 10.1016/j.jep.2024.118521] [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: 04/18/2024] [Revised: 06/22/2024] [Accepted: 07/03/2024] [Indexed: 07/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sangju Cold Granule (SJCG) is a classical traditional Chinese medicine (TCM) prescription described in "Item Differentiation of Warm Febrile Diseases". Historically, SJCG was employed to treat respiratory illnesses. Despite its popular usage, the alleviating effect of SJCG on influenza A virus infection and its mechanisms have not been fully elucidated. AIM OF THE STUDY Influenza is a severe respiratory disease that threatens human health. This study aims to assess the therapeutic potential of SJCG and the possible molecular mechanism underlying its activity against influenza A virus in vitro and in vivo. MATERIALS AND METHODS Ultrahigh-performance liquid chromatography (UPLC)-Q-Exactive was used to identify the components of SJCG. The 50% cytotoxic concentration of SJCG in MDCK and A549 cells were determined using the CCK-8 assay. The activity of SJCG against influenza A virus H1N1 was evaluated in vitro using plaque reduction and progeny virus titer reduction assays. RT-qPCR was performed to obtain the expression levels of inflammatory mediators and the transcriptional regulation of RIG-I and MDA5 in H1N1-infected A549 cells. Then, the mechanism of SJCG effect on viral replication and inflammation was further explored by measuring the expressions of proteins of the RIG-I/NF-kB/IFN(I/III) signaling pathway by Western blot. The impact of SJCG was explored in vivo in an intranasally H1N1-infected BALB/c mouse pneumonia model treated with varying doses of SJCG. The protective role of SJCG in this model was evaluated by survival, body weight monitoring, lung viral titers, lung index, lung histological changes, lung inflammatory mediators, and peripheral blood leukocyte count. RESULTS The main SJCG chemical constituents were flavonoids, carbohydrates and glycosides, amino acids, peptides, and derivatives, organic acids and derivatives, alkaloids, fatty acyls, and terpenes. The CC50 of SJCG were 24.43 mg/mL on MDCK cells and 20.54 mg/mL on A549 cells, respectively. In vitro, SJCG significantly inhibited H1N1 replication and reduced the production of TNF-α, IFN-β, IL-6, IL-8, IL-13, IP-10, RANTES, TRAIL, and SOCS1 in infected A549 cells. Intracellularly, SJCG reduced the expression of RIG-I, MDA5, P-NF-κB P65 (P-P65), P-IκBα, P-STAT1, P-STAT2, and IRF9. In vivo, SJCG enhanced the survival rate and decreased body weight loss in H1N1-infected mice. Mice with H1N1-induced pneumonia treated with SJCG showed a lower lung viral load and lung index than untreated mice. SJCG effectively alleviated lung damage and reduced the levels of TNF-α, IFN-β, IL-6, IP-10, RANTES, and SOCS1 in lung tissue. Moreover, SJCG significantly ameliorated H1N1-induced leukocyte changes in peripheral blood. CONCLUSIONS SJCG significantly reduced influenza A virus and virus-mediated inflammation through inhibiting the RIG-I/NF-kB/IFN(I/III) signaling pathway. Thus, SJCG could provide an effective TCM for influenza treatment.
Collapse
Affiliation(s)
- Taotao Gao
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jinbing Liu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, China; Department of Ultrasound Medicine, Liwan Central Hospital of Guangzhou, 35 Liwan Road, Guangzhou, 510000, Guangdong, China
| | - Nan Huang
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yingxuan Zhou
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China
| | - Conglin Li
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yintong Chen
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zifan Hong
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiaoyan Deng
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xiaoli Liang
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
5
|
Ma Q, Zhao G, Liu J, Chen IT, Wei Y, Liang M, Dai P, Nuez-Ortin WG, Xu H. Effects of a phytobiotic-based additive on the growth, hepatopancreas health, intestinal microbiota, and Vibrio parahaemolyticus resistance of Pacific white shrimp, Litopenaeus vannamei. Front Immunol 2024; 15:1368444. [PMID: 39185423 PMCID: PMC11341979 DOI: 10.3389/fimmu.2024.1368444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 07/16/2024] [Indexed: 08/27/2024] Open
Abstract
Vibrio genus is a common pathogen in aquaculture and causes acute hepatopancreatic necrosis disease (AHPND) and massive mortality of shrimp. Many studies have suggested that a single functional ingredient such as plant extract or organic acid can reduce the dependence on antibiotics and promote the growth and immunity of aquatic animals. In this study, we evaluated the effects of a phytobiotic-based compound additive (Sanacore® GM, SNGM), which had a successful trajectory of commercial application in fish farming. However, its effects on the hepatopancreas health and intestinal microbiota of shrimp after Vibrio challenge have not been well evaluated. In the present study, Pacific white shrimp were fed diets with or without supplementation of SNGM, and the SNGM grades were 0-g/kg (CON), 3-g/kg (SNGM3), and 5-g/kg (SNGM5) diets. The feed trial lasted 60 days, after which a Vibrio parahaemolyticus challenge was performed. The results showed that compared to the CON group, both the SNGM3 and SNGM5 groups had a significantly higher weight gain and a lower feed conversion ratio as well as higher survival after Vibrio parahaemolyticus challenge. In the growth trial, the SNGM3 group had a significantly increased total protein, albumin concentration, and acid phosphatase activity in hemolymph compared to the CON group. In the challenge experiment, the SNGM3 and SNGM5 groups had increased albumin and glucose contents as well as the activities of phenoloxidase, lysozyme, alkaline phosphatase, and superoxide dismutase in hemolymph. Both the SNGM3 and SNGM5 groups had improved morphology of the hepatopancreas and intestine. The SNGM5 group had alleviated gut microbiota dysbiosis induced by Vibrio infection by increasing the potential probiotic bacterium abundance (Shewanella) and decreasing the potential pathogenic bacteria abundance (Vibrio, Photobacteriuma, Pseudoalteromonas, and Candidatus_Bacilloplasma). In conclusion, the dietary phytobiotic-based additive at 3-g/kg level increased the growth and Vibrio parahaemolyticus resistance of Pacific white shrimp by promoting immune-related enzyme activities and improving the morphological structure of the hepatopancreas and intestine and the intestinal microbiota composition.
Collapse
Affiliation(s)
- Qiang Ma
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Guiping Zhao
- Adisseo Life Science (Shanghai) Co., Ltd, Shanghai, China
| | - Jiahao Liu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - I-Tung Chen
- Adisseo Life Science (Shanghai) Co., Ltd, Shanghai, China
| | - Yuliang Wei
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Mengqing Liang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Ping Dai
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | | | - Houguo Xu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| |
Collapse
|
6
|
Zhang Z, Wu W, Li Q, Du F, Wang X, Yang M, Zhang H. The effect of matrine and glycyrrhizic acid on porcine reproductive and respiratory syndrome virus in Vitro and in vivo. Virol J 2024; 21:150. [PMID: 38965549 PMCID: PMC11225320 DOI: 10.1186/s12985-024-02415-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 06/13/2024] [Indexed: 07/06/2024] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is endemic worldwide, seriously affecting the development of the pig industry, but vaccines have limited protective effects against PRRSV transmission. The aim of this study was to identify potential anti-PRRSV drugs. We examined the cytotoxicity of seven compounds formulated based on the mass ratio of glycyrrhizic acid to matrine and calculated their inhibition rates against PRRSV in vitro. The results showed that the seven compounds all had direct killing and therapeutic effects on PRRSV, and the compounds inhibited PRRSV replication in a time- and dose-dependent manner. The compound with the strongest anti-PRRSV effect was selected for subsequent in vivo experiments. Pigs were divided into a control group and a medication group for the in vivo evaluation. The results showed that pigs treated with the 4:1 compound had 100% morbidity after PRRSV challenge, and the mortality rate reached 75% on the 8th day of the virus challenge. These results suggest that this compound has no practical anti-PRRSV effect in vivo and can actually accelerate the death of infected pigs. Next, we further analyzed the pigs that exhibited semiprotective effects following vaccination with the compound to determine whether the compound can synergize with the vaccine in vivo. The results indicated that pigs treated with the compound had higher mortality rates and more severe clinical reactions after PRRSV infection (p < 0.05). The levels of proinflammatory cytokines (IL-6, IL-8, IL-1β, IFN-γ, and TNF-α) were significantly greater in the compound-treated pigs than in the positive control-treated pigs (p < 0.05), and there was no synergistic enhancement with the live attenuated PRRSV vaccine (p < 0.05). The compound enhanced the inflammatory response, prompted the body to produce excessive levels of inflammatory cytokines and caused body damage, preventing a therapeutic effect. In conclusion, the present study revealed that the in vitro effectiveness of these agents does not indicate that they are effective in vivo or useful for developing anti-PRRSV drugs. Our findings also showed that, to identify effective anti-PRRSV drugs, comprehensive drug screening is needed, for compounds with solid anti-inflammatory effects both in vitro and in vivo. Our study may aid in the development of new anti-PRRSV drugs.
Collapse
Affiliation(s)
- Zhilong Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou city, Henan, P.R. China
| | - Wenyi Wu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou city, Henan, P.R. China
| | - Qiannan Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou city, Henan, P.R. China
| | - Fangfang Du
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou city, Henan, P.R. China
- Key Laboratory for Animal-Derived Food Safety of Henan Province, Zhengzhou city, Henan, P.R. China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou city, Henan, P.R. China
| | - Xuebing Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou city, Henan, P.R. China
- Key Laboratory for Animal-Derived Food Safety of Henan Province, Zhengzhou city, Henan, P.R. China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou city, Henan, P.R. China
| | - Mingfan Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou city, Henan, P.R. China
- Key Laboratory for Animal-Derived Food Safety of Henan Province, Zhengzhou city, Henan, P.R. China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou city, Henan, P.R. China
| | - Hongying Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou city, Henan, P.R. China.
- Key Laboratory for Animal-Derived Food Safety of Henan Province, Zhengzhou city, Henan, P.R. China.
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou city, Henan, P.R. China.
| |
Collapse
|
7
|
Bai W, Zhu Q, Wang J, Jiang L, Guo D, Li C, Xing X, Sun D. Licorice extract inhibits porcine epidemic diarrhea virus in vitro and in vivo. J Gen Virol 2024; 105:001964. [PMID: 38471043 PMCID: PMC10999743 DOI: 10.1099/jgv.0.001964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) causes severe diarrhea and even death in piglets, resulting in significant economic losses to the pig industry. Because of the ongoing mutation of PEDV, there might be variations between the vaccine strain and the prevailing strain, causing the vaccine to not offer full protection against different PEDV variant strains. Therefore, it is necessary to develop anti-PEDV drugs to compensate for vaccines. This study confirmed the anti-PEDV effect of licorice extract (Le) in vitro and in vivo. Le inhibited PEDV replication in a dose-dependent manner in vitro. By exploring the effect of Le on the life cycle of PEDV, we found that Le inhibited the attachment, internalization, and replication stages of the virus. In vivo, all five piglets in the PEDV-infected group died within 72 h. In comparison, the Le-treated group had a survival rate of 80 % at the same time, with significant relief of clinical symptoms, pathological damage, and viral loads in the jejunum and ileum. Our results suggested that Le can exert anti-PEDV effects in vitro and in vivo. Le is effective and inexpensive; therefore it has the potential to be developed as a new anti-PEDV drug.
Collapse
Affiliation(s)
- Wenfei Bai
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing 163319, PR China
| | - Qinghe Zhu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing 163319, PR China
| | - Jun Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing 163319, PR China
| | - Limin Jiang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing 163319, PR China
| | - Donghua Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing 163319, PR China
| | - Chunqiu Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing 163319, PR China
| | - Xiaoxu Xing
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing 163319, PR China
| | - Dongbo Sun
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing 163319, PR China
| |
Collapse
|
8
|
Xue M, Yang C, Huang W, He Y, Yang C, Xue Y, Zheng Y, Diao X, Wang X. Pharmacokinetics and metabolite identification of 23-hydroxybetulinic acid in rats by using liquid chromatography-mass spectrometry method. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1234:124016. [PMID: 38266610 DOI: 10.1016/j.jchromb.2024.124016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/03/2024] [Accepted: 01/15/2024] [Indexed: 01/26/2024]
Abstract
23-hydroxybetulinic acid (23-HA), a main bioactive component isolated from Pulsatilla chinensis (Bunge) Regel, exhibits various pharmacological activities, such as antimelanoma, antileukemia, anti-colon cancer, and antihepatotoxicity. Although the main active ingredient anemoside B4 (AB4) from this plant has been well studied, research on its active metabolite 23-HA is limited. In the present study, a validated HPLC-QQQ-MS/MS method was established for the quantification of 23-HA in rat plasma. Pharmacokinetics analysis showed that the absorption and elimination of 23-HA in rats were rapid, with an oral bioavailability as 12.9 %. After oral administration with 50 mg/kg 23-HA for SD rats, the plasma, urine, feces, and bile samples were collected and analyzed by UPLC-Q Exactive Plus MS and HPLC-QQQ-MS/MS. Seventeen metabolites of 23-HA were identified, and its major metabolic pathways included oxidation, hydration, sulfation, and glucuronidation. This study highlights the first detailed investigation of 23-HA's pharmacokinetics in rats along with its metabolism in vivo, and will provide robust evidence for further research and clinical application of 23-HA.
Collapse
Affiliation(s)
- Mingzhen Xue
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou 362000, China
| | - Cheng Yang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wensi Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yifei He
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Chen Yang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yaru Xue
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yuandong Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Xingxing Diao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xiachang Wang
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou 362000, China.
| |
Collapse
|
9
|
Azizah NS, Irawan B, Kusmoro J, Safriansyah W, Farabi K, Oktavia D, Doni F, Miranti M. Sweet Basil ( Ocimum basilicum L.)-A Review of Its Botany, Phytochemistry, Pharmacological Activities, and Biotechnological Development. PLANTS (BASEL, SWITZERLAND) 2023; 12:4148. [PMID: 38140476 PMCID: PMC10748370 DOI: 10.3390/plants12244148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/09/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
An urgent demand for natural compound alternatives to conventional medications has arisen due to global health challenges, such as drug resistance and the adverse effects associated with synthetic drugs. Plant extracts are considered an alternative due to their favorable safety profiles and potential for reducing side effects. Sweet basil (Ocimum basilicum L.) is a valuable plant resource and a potential candidate for the development of pharmaceutical medications. A single pure compound or a combination of compounds exhibits exceptional medicinal properties, including antiviral activity against both DNA and RNA viruses, antibacterial effects against both Gram-positive and Gram-negative bacteria, antifungal properties, antioxidant activity, antidiabetic potential, neuroprotective qualities, and anticancer properties. The plant contains various phytochemical constituents, which mostly consist of linalool, eucalyptol, estragole, and eugenol. For centuries, community and traditional healers across the globe have employed O. basilicum L. to treat a wide range of ailments, including flu, fever, colds, as well as issues pertaining to digestion, reproduction, and respiration. In addition, the current research presented underscores the significant potential of O. basilicum-related nanotechnology applications in addressing diverse challenges and advancing numerous fields. This promising avenue of exploration holds great potential for future scientific and technological advancements, promising improved utilization of medicinal products derived from O. basilicum L.
Collapse
Affiliation(s)
- Nabilah Sekar Azizah
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor 45363, Indonesia; (N.S.A.); (B.I.); (J.K.); (F.D.)
| | - Budi Irawan
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor 45363, Indonesia; (N.S.A.); (B.I.); (J.K.); (F.D.)
| | - Joko Kusmoro
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor 45363, Indonesia; (N.S.A.); (B.I.); (J.K.); (F.D.)
| | - Wahyu Safriansyah
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor 45363, Indonesia; (W.S.); (K.F.)
| | - Kindi Farabi
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor 45363, Indonesia; (W.S.); (K.F.)
| | - Dina Oktavia
- Department of Transdisciplinary, Graduate School, Universitas Padjadjaran, Bandung 40132, Indonesia;
| | - Febri Doni
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor 45363, Indonesia; (N.S.A.); (B.I.); (J.K.); (F.D.)
| | - Mia Miranti
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor 45363, Indonesia; (N.S.A.); (B.I.); (J.K.); (F.D.)
| |
Collapse
|
10
|
Gong M, Xia X, Chen D, Ren Y, Liu Y, Xiang H, Li X, Zhi Y, Mo Y. Antiviral activity of chrysin and naringenin against porcine epidemic diarrhea virus infection. Front Vet Sci 2023; 10:1278997. [PMID: 38130439 PMCID: PMC10733469 DOI: 10.3389/fvets.2023.1278997] [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: 08/17/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is one of the critical pathogens causing diarrhea in piglets and has caused huge economic losses to the swine industry in worldwide. However, there is currently no effective therapeutic medication available for the treatment of PEDV. Natural compounds are a hot topic for researching and screening antiviral lead compounds due to their abundant sources, varied activities, and low toxicity. In this study, a total of 6 compounds from different plant sources were selected for in vitro anti-PEDV screening, including chrysin, naringenin, soy isoflavone, glycyrrhetinic acid, oleanolic acid, and geniposide. Then two active compounds, chrysin and naringenin, were further evaluated on PEDV infected cells at different stage. And the anti-PEDV mechanism was analyzed by molecule docking and molecular dynamics. The results showed that both chrysin and naringenin showed the most significant anti-PEDV activity by increasing the cell viability and decreasing the virus copy number. Both natural compounds could inhibit viral titer, mRNA and protein levels in the prophylactic and post-viral entry stages of PEDV infection. Furthermore, chrysin and naringenin mainly interacted with viral replicase proteins such as 3CLpro and PLP-2 through hydrogen bonds and hydrophobic forces. The complexes formed by chrysin and naringenin with the two PEDV replication proteases had high stability. These results suggested that chrysin and naringenin may exert antiviral effects by interacting with the virus 3CLpro protein or PLP2 protein, thereby affecting their role in the formation of PEDV non-structural proteins or interfering with virus replication. This study lays the foundation for developing chrysin and naringenin as novel anti-PEDV therapeutic drugs.
Collapse
Affiliation(s)
- Mengfei Gong
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Xuemei Xia
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Dishi Chen
- Center for Animal Disease Prevention and Control, Chengdu, China
| | - Yupeng Ren
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Yutong Liu
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Hua Xiang
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Xiaohuan Li
- Agricultural and Rural Bureau of Shizhong District, Leshan, China
| | - Yupeng Zhi
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Yu Mo
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| |
Collapse
|
11
|
Zou Q, Chen Y, Qin H, Tang R, Han T, Guo Z, Zhao J, Xu D. The role and mechanism of TCM in the prevention and treatment of infectious diseases. Front Microbiol 2023; 14:1286364. [PMID: 38033575 PMCID: PMC10682724 DOI: 10.3389/fmicb.2023.1286364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023] Open
Abstract
The constant presence of infectious diseases poses an everlasting threat to the entire world. In recent years, there has been an increased attention toward the application of traditional Chinese medicine (TCM) in the treatment of emerging infectious diseases, as it has played a significant role. The aim of this article is to provide a concise overview of the roles and mechanisms of TCM in treating infectious diseases. TCM possesses the ability to modulate relevant factors, impede signaling pathways, and inhibit microbial growth, thereby exhibiting potent antiviral, antibacterial, and anti-inflammatory effects that demonstrate remarkable efficacy against viral and bacterial infections. This article concludes that the comprehensive regulatory features of Chinese herbal medicines, with their various components, targets, and pathways, result in synergistic effects. The significance of Chinese herbal medicines in the context of infectious diseases should not be underestimated; however, it is crucial to also acknowledge their underutilization. This paper presents constructive suggestions regarding the challenges and opportunities faced by Chinese medicines. Particularly, it emphasizes the effectiveness and characteristics of Chinese medicines in the treatment of infectious diseases, specifying how these medicines' active substances can be utilized to target infectious diseases. This perspective is advantageous in facilitating researchers' pharmacological studies on Chinese medicines, focusing on the specific points of action. The mechanism of action of Chinese herbal medicines in the treatment of infectious diseases is comprehensively elucidated in this paper, providing compelling evidence for the superior treatment of infectious diseases through Chinese medicine. This information is favorable for advancing the development of TCM and its potential applications in the field of infectious diseases.
Collapse
Affiliation(s)
- Qifei Zou
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yitong Chen
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Huanxin Qin
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Rui Tang
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Taojian Han
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ziyi Guo
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Juanjuan Zhao
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Delin Xu
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
12
|
Liu T, Li Y, Wang L, Zhang X, Zhang Y, Gai X, Chen L, Liu L, Yang L, Wang B. Network pharmacology-based exploration identified the antiviral efficacy of Quercetin isolated from mulberry leaves against enterovirus 71 via the NF-κB signaling pathway. Front Pharmacol 2023; 14:1260288. [PMID: 37795035 PMCID: PMC10546324 DOI: 10.3389/fphar.2023.1260288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/06/2023] [Indexed: 10/06/2023] Open
Abstract
Introduction: Mulberry leaf (ML) is known for its antibacterial and anti-inflammatory properties, historically documented in "Shen Nong's Materia Medica". This study aimed to investigate the effects of ML on enterovirus 71 (EV71) using network pharmacology, molecular docking, and in vitro experiments. Methods: We successfully pinpointed shared targets between mulberry leaves (ML) and the EV71 virus by leveraging online databases. Our investigation delved into the interaction among these identified targets, leading to the identification of pivotal components within ML that possess potent anti-EV71 properties. The ability of these components to bind to the targets was verified by molecular docking. Moreover, bioinformatics predictions were used to identify the signaling pathways involved. Finally, the mechanism behind its anti-EV71 action was confirmed through in vitro experiments. Results: Our investigation uncovered 25 active components in ML that targeted 231 specific genes. Of these genes, 29 correlated with the targets of EV71. Quercetin, a major ingredient in ML, was associated with 25 of these genes. According to the molecular docking results, Quercetin has a high binding affinity to the targets of ML and EV71. According to the KEGG pathway analysis, the antiviral effect of Quercetin against EV71 was found to be closely related to the NF-κB signaling pathway. The results of immunofluorescence and Western blotting showed that Quercetin significantly reduced the expression levels of VP1, TNF-α, and IL-1β in EV71-infected human rhabdomyosarcoma cells. The phosphorylation level of NF-κB p65 was reduced, and the activation of NF-κB signaling pathway was suppressed by Quercetin. Furthermore, our results showed that Quercetin downregulated the expression of JNK, ERK, and p38 and their phosphorylation levels due to EV71 infection. Conclusion: With these findings in mind, we can conclude that inhibiting the NF-κB signaling pathway is a critical mechanism through which Quercetin exerts its anti-EV71 effectiveness.
Collapse
Affiliation(s)
- Tianrun Liu
- School of Medicine, Jiamusi University, Jiamusi, China
| | - Yingyu Li
- School of Medicine, Jiamusi University, Jiamusi, China
| | - Lumeng Wang
- School of Medicine, Jiamusi University, Jiamusi, China
| | | | - Yuxuan Zhang
- School of Medicine, Jiamusi University, Jiamusi, China
| | - Xuejie Gai
- The Affiliated First Hospital, Jiamusi University, Jiamusi, China
| | - Li Chen
- School of Medicine, Jiamusi University, Jiamusi, China
| | - Lei Liu
- School of Medicine, Jiamusi University, Jiamusi, China
| | - Limin Yang
- School of Medicine, Dalian University, Dalian, China
| | - Baixin Wang
- School of Medicine, Jiamusi University, Jiamusi, China
| |
Collapse
|
13
|
Wang Z, Cai X, Ren Z, Shao Y, Xu Y, Fu L, Zhu Y. Piceatannol as an Antiviral Inhibitor of PRV Infection In Vitro and In Vivo. Animals (Basel) 2023; 13:2376. [PMID: 37508153 PMCID: PMC10375968 DOI: 10.3390/ani13142376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Pseudorabies virus (PRV) belongs to the family Herpesviridae. PRV has a wide host range and can cause cytopathic effects (CPEs) in PK-15 cells. Therefore, PRV was used as a model to study the antiviral activity of piceatannol. The results showed that piceatannol could restrain PRV multiplication in PK-15 cells in a dose-dependent manner. The 50% inhibitory concentration (IC50) was 0.0307 mg/mL, and the selectivity index (SI, CC50/IC50) was 3.68. Piceatannol could exert an anti-PRV effect by reducing the transcription level of viral genes, inhibiting PRV-induced apoptosis and elevating the levels of IL-4, TNF-α and IFN-γ in the serum of mice. Animal experiments showed that piceatannol could delay the onset of disease, reduce the viral load in the brain and kidney and reduce the pathological changes in the tissues and organs of the mice to improve the survival rate of the mice (14.3%). Therefore, the anti-PRV activity of piceatannol in vivo and in vitro was systematically evaluated in this study to provide scientific data for developing a new alternative measure for controlling PRV infection.
Collapse
Affiliation(s)
- Zhiying Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150038, China
| | - Xiaojing Cai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150038, China
| | - Zhiyuan Ren
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150038, China
| | - Yi Shao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150038, China
| | - Yongkang Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150038, China
| | - Lian Fu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150038, China
| | - Yan Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150038, China
| |
Collapse
|
14
|
Xi YF, Bai M, Zhang X, Hou ZL, Lin B, Yao GD, Lou LL, Wang XB, Song SJ, Huang XX. Insight into tetrahydrofuran lignans from Isatis indigotica fortune with neuroprotective and acetylcholinesterase inhibitor activity. PHYTOCHEMISTRY 2023; 208:113609. [PMID: 36758886 DOI: 10.1016/j.phytochem.2023.113609] [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: 11/26/2022] [Revised: 02/04/2023] [Accepted: 02/05/2023] [Indexed: 06/18/2023]
Abstract
Nine tetrahydrofuran lignans, including three undescribed spiro-lignans, were isolated from Isatis indigotica Fortune (Brassicaceae). Extensive spectroscopic analyses achieved the structure elucidation of these tetrahydrofuran lignans, and quantum chemical calculation combined with the MAEΔΔδ parameter. Notably, isatispironeols A-B have a unique spiro[dienone-tetrahydrofuran] molecular core. These spiro[dienone-tetrahydrofuran] lignans showed comparable neuroprotective effects as the positive control in the H2O2-induced SH-SY5Y cells model. In addition, (-)-(7R,8S,1'R,7'R,8'R)-isatispironeol A possessed more significant AChE inhibitory activity, further interact sites were also predicted by the in silico assay.
Collapse
Affiliation(s)
- Yu-Fei Xi
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Ming Bai
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xin Zhang
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zi-Lin Hou
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Bin Lin
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Guo-Dong Yao
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Li-Li Lou
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Xiao-Bo Wang
- Chinese People's Liberation Army Logistics Support Force No. 967 Hospital, Dalian, 116021, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xiao-Xiao Huang
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
15
|
Feng F, Pan L, Wu J, Liu M, He L, Yang L, Zhou W. Schisantherin A inhibits cell proliferation by regulating glucose metabolism pathway in hepatocellular carcinoma. Front Pharmacol 2022; 13:1019486. [PMID: 36425581 PMCID: PMC9679220 DOI: 10.3389/fphar.2022.1019486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/26/2022] [Indexed: 08/06/2023] Open
Abstract
Schisantherin A (STA) is a traditional Chinese medicine extracted from the plant Schisandra chinensis, which has a wide range of anti-inflammatory, antioxidant, and other pharmacological effects. This study investigates the anti-hepatocellular carcinoma effects of STA and the underlying mechanisms. STA significantly inhibits the proliferation and migration of Hep3B and HCCLM3 cells in vitro in a concentration-dependent manner. RNA-sequencing showed that 77 genes are upregulated and 136 genes are downregulated in STA-treated cells compared with untreated cells. KEGG pathway analysis showed significant enrichment in galactose metabolism as well as in fructose and mannose metabolism. Further gas chromatography-mass spectrometric analysis (GC-MS) confirmed this, indicating that STA significantly inhibits the glucose metabolism pathway of Hep3B cells. Tumor xenograft in nude mice showed that STA has a significant inhibitory effect on tumor growth in vivo. In conclusion, our results indicate that STA can inhibit cell proliferation by regulating glucose metabolism, with subsequent anti-tumor effects, and has the potential to be a candidate drug for the treatment of liver cancer.
Collapse
Affiliation(s)
- Fan Feng
- National Innovation and Attracting Talents “111” Base, Key Laboratory of Biorheological Science and Technology, College of Bioengineering, Ministry of Education, Chongqing University, Chongqing, China
| | - Lianhong Pan
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing, China
| | - Jiaqin Wu
- National Innovation and Attracting Talents “111” Base, Key Laboratory of Biorheological Science and Technology, College of Bioengineering, Ministry of Education, Chongqing University, Chongqing, China
| | - Mingying Liu
- School of Comprehensive Health Management, XiHua University, Chengdu, Sichuan, China
| | - Long He
- School of Artificial Intelligence, Chongqing University of Education, Chongqing, China
| | - Li Yang
- National Innovation and Attracting Talents “111” Base, Key Laboratory of Biorheological Science and Technology, College of Bioengineering, Ministry of Education, Chongqing University, Chongqing, China
| | - Wei Zhou
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
16
|
Mechanism of Zhinao Capsule in Treating Alzheimer’s Disease Based on Network Pharmacology Analysis and Molecular Docking Validation. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:5708769. [PMID: 36032542 PMCID: PMC9410932 DOI: 10.1155/2022/5708769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/02/2022]
Abstract
Objective This study aimed to determine the active components of Zhinao capsule (ZNC) and the targets in treating Alzheimer's disease (AD) so as to investigate and explore the mechanism of ZNC for AD. Methods The active components and targets of ZNC were determined from the traditional Chinese medicine systems pharmacology database (TCMSP). The target genes of AD were searched for in GeneCards. Cytoscape was used to construct an herb-component-target-disease network. A protein-protein interaction (PPI) network was constructed by STRING. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed using the OmicShare. UCSF Chimera and SwissDock were used for molecular docking verification. Finally, four key target genes were validated by Western blotting. Results In total, 55 active components, 287 targets of active components, 1197 disease genes, and 134 common genes were screened, which were significantly enriched in 3975 terms of biological processes (BP), 284 terms of cellular components (CC), 433 terms of molecular functions (MF), and 245 signaling pathways. Caspase-3 (CASP3) and beta-sitosterol, tumor necrosis factor-alpha (TNF-α) and quercetin, vascular endothelial growth factor A (VEGFA) and baicalein, and mitogen-activated protein kinase 1 (MAPK1) and quercetin showed good-to-better docking. Moreover, ZNC not only downregulated CASP3 and TNF-α protein expression but also upregulated the protein expression of VEGFA and MAPK1. Conclusions The active components of ZNC, such as beta-sitosterol, quercetin, and baicalein may act on multiple targets like CASP3, VEGFA, MAPK1, and TNF-α to affect T cell receptor (TCR), TNF, and MAPK signaling pathway, thereby achieving the treatment of AD. This study provides a scientific basis for further exploring the potential mechanism of ZNC in the treatment of AD and a reference for its clinical application.
Collapse
|