1
|
Liang X, Su W, Zhang W, Wang S, Wu X, Li X, Gao W. An overview of the research progress on Aconitum carmichaelii Debx.:active compounds, pharmacology, toxicity, detoxification, and applications. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118832. [PMID: 39306209 DOI: 10.1016/j.jep.2024.118832] [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: 04/06/2024] [Revised: 08/18/2024] [Accepted: 09/13/2024] [Indexed: 09/29/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aconitum carmichaelii Debx. is the most widely distributed species of Aconitum plants in China and has a long history of medicinal use. Because of its toxicity, A. carmichaelii is classified as lower class in the Shennong Bencao Jing (Shennong's Classic of Materia Medica). According to the theory of Chinese medicine, the roots can be used to revive yang for resuscitation, dispel wind, remove dampness, and relieve pain. AIMS OF THE REVIEW This review focuses on summarizing the latest reports on the components, pharmacology, toxicity, detoxification mechanism and application of A. carmichaelii. It aims to provide ideas for in-depth research on activity mechanism of A. carmichaelii and expanding the value of exploitation and utilization. MATERIALS AND METHODS Information was collected from the following online scientific databases: PubMed, Web of Science, Wiley Online Library, SciFinder, Scopus, PubChem, China National Knowledge Internet (CNKI), etc. Additional data were obtained from other Chinese medicine books. RESULTS In this review, 224 compounds were categorized and new compounds discovered in the last five years were highlighted. The main components of A. carmichaelii are C19-diterpene alkaloids(C19-DAs), among which diester-type aconitine is the most toxic and also the main active ingredient, while monoester diterpene alkaloids (MDAs) and aminol diterpene alkaloids (ADAs) are greatly toxicity reduced due to the loss of ester bond. Heating and compatibility are the means to increase the efficiency and reduce the toxicity of A. carmichaelii. In addition, it also contains abundant C20-diterpene alkaloids (C20-DAs). Like C19-DAs, these compounds also have cardiotonic, anticancer, anti-inflammatory and analgesic pharmacological effects, but their toxicity is weaker. The above-ground part contains not only a variety of MDAs and ADAs, but also contains abundant non-diterpenoid alkaloids and active polysaccharides. In addition to pharmacological effects, we further summarized the mechanisms of cardiotoxicity, neurotoxicity and other toxicity of A. carmichaelii. What's more, the application prospects are also discussed. Polysaccharides and diterpenoid alkaloids in A. carmichaelii and related traditional prescriptions have great promising prospects for the development of new drugs. CONCLUSION A. carmichaelii has rich alkaloids and polysaccharides, but the new compounds discovered in recent years are only in the activity screening stage. The toxic differences between C19- and C20- DAs and the dose that affect toxicity of A. carmichaelii are still not clear. The non-traditional medicinal parts, such as stems and leaves, show great potential for development and utilization. More extensive and in-depth exploration of low-toxic active compounds, as well as the mechanism of efficacy-enhancement and toxicity-attenuation, will help A. carmichaelii to be better and safer used for clinical.
Collapse
Affiliation(s)
- Xv Liang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Wenya Su
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Weimei Zhang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Shirui Wang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Xipei Wu
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Xia Li
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China.
| | - Wenyuan Gao
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China.
| |
Collapse
|
2
|
Zhao Y, Wu J, Li X, Zheng L, Chen Q, Zhang S, Chen J. Huangqi-Guizhi-Wuwutang protects against oligospermia in mice by promoting the proliferation of spermatogenic stem cells: A comprehensive study using HPLC-Q-TOF/MS and experimental pharmacology. Biomed Chromatogr 2024:e6023. [PMID: 39390901 DOI: 10.1002/bmc.6023] [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: 05/29/2024] [Revised: 08/24/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024]
Abstract
The classical traditional Chinese medicine formula Huangqi-Guizhi-Wuwutang (HGW) has been shown to enhance sperm production. However, the bioactive components and comprehensive mechanisms underlying the therapeutic effects remain unclear. The present study investigates the potential active ingredients and underlying mechanisms of HGW against spermatogenesis dysfunction. The chemical components of HGW were analyzed by mass spectrometry. And then the "components-targets-pathway-disease" network was constructed using network pharmacology research methods, which aimed to identify the key active components and potential targets of HGW in treating oligospermia. Experimental validation was finally conducted in animal model. The male-specific pathogen-free Kunming mice were divided into five groups: Sham group, Model group, and HGW groups (8, 16, and 32 g/kg of HGW by gavage for 35 days). Chemical profile and network pharmacology results revealed that potential bioactive compounds were dihydrocinnacasside, isomucronulatol, and 6-gingerol, and the mechanism of which was enriched in regulating spermatogenic stem cells (SSCs), endocrine function, and apoptosis. The administration of HGW significantly improved oligospermia in mice. HGW significantly upregulated the expression of marker proteins in SSCs and the potential targets within the testis simultaneously. Our data indicates that HGW enhances the proliferation of SSCs, and HGW can be a promising therapeutic candidate for oligospermia.
Collapse
Affiliation(s)
- Yuan Zhao
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Jinru Wu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Xiangbin Li
- Shenzhen Zhongshan Obstetrics and Gynecology Hospital, Shenzhen, 518031, China
| | - Lin Zheng
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Qiugu Chen
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Shangbin Zhang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Jianping Chen
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| |
Collapse
|
3
|
Rui Wang X, Ting Zhang J, Guang Jing W, Hua Li M, Han Guo X, Long Cheng X, Wei F. Digital identification and adulteration analysis of Pulsatilla Radix and Pulsatilla Cernua based on "digital identity" and UHPLC-QTOF-MS E. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1244:124257. [PMID: 39089066 DOI: 10.1016/j.jchromb.2024.124257] [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: 06/09/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/03/2024]
Abstract
Under the background of digitalization of traditional Chinese medicine (TCM), to realize the quick identification and adulteration analysis of Pulsatilla Radix (PR), adhering to digital conviction, this study conducted UHPLC-QTOF-MSE analysis on PR and its adulterant-Pulsatilla Cernua (PC) from different batches and based on digital conversion, the shared ions were extracted from different batches of PR and PC as their "ions representation", respectively. Further, the data set of unique ions of PR relative to PC and PC relative to PR were screened out as the "digital identities" of PR and PC respectively. Further, above the "digital identities" of PR and PC were used as the benchmarks for matching and identifying to feedback give a matching credibility (MC). The results showed that based on the "digital identities" of PR and PC, the digital identification of two herbal samples can be realized efficiently and accurately at the individual level with the MC≥70.00 %, even if 5 % of PC in the mixed samples can still be identified efficiently and accurately. The study is of great practical significance for improving the identification efficiency of PR and PC, cracking down on adulterated and counterfeit drugs, and strengthening the quality control of PR. In addition, it has important reference significance for developing non-targeted digital identification of herbal medicines at the individual level based on UHPLC-QTOF-MSE and the "digital identity", which was beneficial to the construction of digital Chinese medicine and digital quality control.
Collapse
Affiliation(s)
- Xian Rui Wang
- Institute for Control of Traditional Chinese Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing 102629, P. R. China
| | - Jia Ting Zhang
- Institute for Control of Traditional Chinese Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing 102629, P. R. China
| | - Wen Guang Jing
- Institute for Control of Traditional Chinese Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing 102629, P. R. China
| | - Ming Hua Li
- Institute for Control of Traditional Chinese Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing 102629, P. R. China
| | - Xiao Han Guo
- Institute for Control of Traditional Chinese Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing 102629, P. R. China
| | - Xian Long Cheng
- Institute for Control of Traditional Chinese Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing 102629, P. R. China.
| | - Feng Wei
- Institute for Control of Traditional Chinese Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing 102629, P. R. China.
| |
Collapse
|
4
|
Yan Q, Zhang F, Qiao Z, Jin Y, Zheng R, Wu J. Investigating the mechanism of PAD in the treatment of acne based on network pharmacology and molecular docking: A review. Medicine (Baltimore) 2024; 103:e38785. [PMID: 39029003 PMCID: PMC11398774 DOI: 10.1097/md.0000000000038785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2024] Open
Abstract
Acne is a common and chronic skin condition characterized by high incidence, recurrent symptoms and difficult cure. Summarizing the clinical treatment experience, it was found that the powder for ascending and descending was effective in the treatment of acne. Our aim was to use network pharmacology and molecular docking to reveal the hub genes, biological functions, and signaling pathways of powder for ascending and descending against acne. First, the chemical components and target genes of PAD were sifted using the TCMSP and HERB database. The targets of acne were obtained simultaneously from the CTD, OMIM and GeneCards database. The obtained drug targets and disease targets were imported into the R language software to draw Venn diagrams. Then, the potential targets were imported into the String website to construct a protein interaction network diagram. And Cytoscape software was used for topological analysis to screen the core targets, and the core targets were analyzed by GO functional enrichment and KEGG pathway enrichment. Finally, molecular docking was used to verify the predictions of key genes' reliability. The core targets of the treatment of acne were TNF, GADPH, IL-6 and so on. The results of enrichment analysis showed that the treatment of acne with PAD may be related to TNF signaling pathway and AGE-RAGE signaling pathway. The molecular docking verification showed that the components were well bound to the core targets of acne, and the docking ability of stigmasterol and TNF (-12.73 kcal/mol) was particularly outstanding.
Collapse
Affiliation(s)
- QianJun Yan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Fang Zhang
- Department of Integrated of Traditional Chinese and Western Medicine, Anqing 116 Hospital, Anqing, Anhui, China
| | - Zukang Qiao
- Department of Otorhinolaryngology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yangzi Jin
- Department of Otorhinolaryngology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ruyi Zheng
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Jiani Wu
- Department of Otorhinolaryngology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
ZHANG G, LIN Y, CHEN X, QIN J, HE Y, LIU T, ZHANG L, ZHANG L. Cinnamomi cortex extract mitigated monosodium urate-induced acute gouty arthritis in rats through nuclear factor-κB-NOD-like receptor thermal protein domain associated protein 3 signaling pathway. J Vet Med Sci 2024; 86:623-630. [PMID: 38030283 PMCID: PMC11187596 DOI: 10.1292/jvms.23-0085] [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: 05/12/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
Cinnamomi cortex was applied to mitigate joint injury since ancient China. However, the effect of Cinnamomi cortex on gouty arthritis (GA) was rarely reported. This study aimed to explore the effect of Cinnamomi cortex on monosodium urate (MSU)-induced acute GA (AGA) in rats, and clarify the underlying mechanism. The results showed that Cinnamomi cortex extract (CE) containing rich polyphenols and flavonoids alleviated joint swelling and inflammation by reducing programmed cell death in MSU-induced AGA rats. Network pharmacology analysis showed that CE's predictive inflammatory pathways included nuclear factor-κB (NF-κB) and necroptosis pathways. CE reduced expression of pyroptosis-related regulators including Gasdermin D and Caspase 1 via regulating NF-κB/NOD-like receptor thermal protein domain associated protein 3 signaling pathway in AGA rats. In conclusion, this study provided a theoretical basis for Cinnamomi cortex applied as a new veterinary medicine to protect against GA.
Collapse
Affiliation(s)
- Gengpeng ZHANG
- The Fourth Clinical Medical College of Guangzhou University
of Chinese Medicine, Shenzhen, China
- Department of Traditional Chinese Medicine, The Seventh
Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuejia LIN
- Department of Traditional Chinese Medicine, The Seventh
Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xianhua CHEN
- Department of Traditional Chinese Medicine, The Seventh
Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jian QIN
- Department of Traditional Chinese Medicine, The Seventh
Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuhai HE
- The Fourth Clinical Medical College of Guangzhou University
of Chinese Medicine, Shenzhen, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen,
China
| | - Taoli LIU
- Department of Traditional Chinese Medicine, The Seventh
Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Li ZHANG
- Department of Traditional Chinese Medicine, The Seventh
Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Lu ZHANG
- University of Chinese Academy of Sciences-Shenzhen Hospital,
Shenzhen, China
| |
Collapse
|
6
|
Yang Z, Lyu B, Ma B, Gao Y, Qin D. Screening of the effective sites of Cichorium glandulosum against hyperuricemia combined with hyperlipidemia and its network pharmacology analysis. Comput Biol Chem 2024; 110:108088. [PMID: 38685167 DOI: 10.1016/j.compbiolchem.2024.108088] [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: 02/02/2024] [Revised: 04/05/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024]
Abstract
Cichorium glandulosum, a common traditional Chinese medicine used by Uyghur and Mongolian ethnic groups, is recognized for its potential to ameliorate metabolic disorders. However, the specific efficacy and mechanisms of Cichorium glandulosum in treating the comorbidity of hyperuricaemia and hyperlipidaemia remain unexplored. This study aims to explore the pharmacological effects and mechanisms of Cichorium glandulosum on this comorbidity through a combination of animal experiments, network pharmacology, and molecular docking techniques. A rat model of hyperuricaemia combined with hyperlipidaemia was established through a high-fat and high-purine diet, and the effective parts of the aqueous extract of Cichorium glandulosum to reduce uric acid and lipid levels were screened and the components of the parts were analysed by LC-MS/MS. The active components, core targets, and key pathways were analysed using network pharmacology and validated by molecular docking. Animal experimental results indicated that the n-butanol extract of Cichorium glandulosum showed a significant therapeutic effect on this comorbidity. Analysis of the n-butanol extract yielded 35 active ingredients and 138 intersecting targets related to diseases. Key targets identified through compound-target-pathway (C-T-P) and Protein-Protein Interaction (PPI) analyses included RELA, CASP3, PTGS2, TNF, and ESR1. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses revealed 2515 functional items and 164 pathways, respectively. Molecular docking demonstrated that isochlorogenic acid A, baicalin, chicoric acid, and lactucopicrin showed the highest binding affinity to RELA and PTGS2. The n-butanol fraction from the aqueous extract of Cichorium glandulosum was found to reduce uric acid and lipid levels effectively. In summary, Cichorium glandulosum has a therapeutic effect on hyperuricaemia combined with hyperlipidaemia through its multi-component, multi-target, and multi-pathway characteristics.
Collapse
Affiliation(s)
- Zhiguo Yang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832002, China
| | - Bo Lyu
- The First Affiliated Hospital of School of Medicine, Shihezi University, Shihezi, Xinjiang 832000, China
| | - Bin Ma
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832002, China
| | - Yuefeng Gao
- College of Applied Engineering, Henan University of Science and Technology, Sanmenxia, Henan 472000, China
| | - Dongmei Qin
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832002, China.
| |
Collapse
|
7
|
Zhang T, Xie Y, Li T, Deng Y, Wan Q, Bai T, Zhang Q, Cai Z, Chen M, Zhang J. Phytochemical analysis and hepatotoxicity assessment of braised Polygoni Multiflori Radix (Wen-He-Shou-Wu). Biomed Chromatogr 2024; 38:e5768. [PMID: 38087457 DOI: 10.1002/bmc.5768] [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: 04/03/2023] [Revised: 09/26/2023] [Accepted: 10/09/2023] [Indexed: 01/26/2024]
Abstract
Polygoni Multiflori Radix (PMR) is a medicinal herb commonly used in China and Eastern Asia. Recently, the discovery of hepatotoxicity in PMR has received considerable attention from scientists. Processing is a traditional Chinese medicine technique used for the effective reduction of toxicity. One uncommon technique is the braising method-also known as 'Wen-Fa' in Chinese-which is used to prepare tonics or poisonous medications. Braised PMR (BPMR)-also known as 'Wen-He-Shou-Wu'-is one of the processed products of the braising method. However, the non-volatile components of BPMR have not been identified and examined in detail, and therefore, the hepatotoxic advantage of BPMR remains unknown. In this study, we compared the microscopic characteristics of different samples in powder form using scanning electron microscopy (SEM), investigated the non-volatile components, assessed the effects of different processed PMR products on the liver, and compared the differences between BPMR and PMR Praeparata recorded in the Chinese Pharmacopoeia (2020 edition). We found that the hepatotoxicity of BPMR was dramatically decreased, which may be related to an increase in polysaccharide content and a decrease in toxic substances. The present study provides an important foundation for future investigations of the processing mechanisms of BPMR.
Collapse
Affiliation(s)
- Tao Zhang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yating Xie
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Tao Li
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yaling Deng
- Department of Pharmacy, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Quan Wan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Tingting Bai
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Qing Zhang
- Jianchangbang Pharmaceutical Co., Ltd., Nanchang, China
- Key Laboratory of Traditional Chinese Medicine Processing (Braising Method), Nanchang, China
| | - Zhongxi Cai
- Jianchangbang Pharmaceutical Co., Ltd., Nanchang, China
- Key Laboratory of Traditional Chinese Medicine Processing (Braising Method), Nanchang, China
| | - Mingxia Chen
- Jianchangbang Pharmaceutical Co., Ltd., Nanchang, China
- Key Laboratory of Traditional Chinese Medicine Processing (Braising Method), Nanchang, China
- Beijing Scrianen Pharmaceutical Co., Ltd., Beijing, China
| | - Jinlian Zhang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
8
|
Sun Y, Liu J, Wang J, He M, Chen X, Chen L. Network pharmacology integrated with experimental validation revealed the mechanism of Fengshi Gutong Capsule in the treatment of osteoarthritis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117261. [PMID: 37788787 DOI: 10.1016/j.jep.2023.117261] [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: 06/21/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fengshi Gutong Capsule (FSGTC) is a commonly used Chinese medicine for the treatment of joint pain caused by osteoarthritis (OA). However, the mechanism of action of FSGTC for OA remains unclear. AIM OF THE STUDY This study aimed to explore the alleviating effects and potential mechanisms of action of FSGTC for OA through data mining, network pharmacology, and in vitro experiments. MATERIALS AND METHODS High-performance liquid chromatography (HPLC) was performed to establish the fingerprints of FSGTC and detect the components of FSGTC absorbed in the blood. The effects of FSGTC on inflammation, immunity, and liver and kidney functions in patients with OA were evaluated by mining clinical data. The potential targets and pathways of FSGTC were screened using network pharmacology. Subsequently, CCK-8 assay, flow cytometry, western blotting, RT-qPCR, ELISA, and immunofluorescence were performed in IL-1β-stimulated chondrocytes for further validation. RESULTS Eighty-seven common peaks and 10 components were identified using the HPLC fingerprints of 12 batches of samples, and the similarity was in the range of 0.973-0.998. Retrospective clinical analysis demonstrated a significant reduction in inflammatory response levels among patients with OA who received FSGTC treatment. Network pharmacology analysis revealed that FSGTC potentially targeted processes related to inflammation, oxidative stress, and apoptosis. The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT), The nuclear factor-κB (NF-κB), and janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathways were predicted to be the main pathways involved in the therapeutic effects of FSGTC in OA. In vitro, FSGTC-containing serum aided the proliferation of chondrocytes stimulated by IL-1β, while concurrently mitigating apoptosis, suppressing the expression of inflammatory cytokines and oxidative molecules, and inhibiting the degradation of the chondrocyte extracellular matrix (ECM). CONCLUSIONS FSGTC alleviates the inflammatory response in patients with OA. This therapeutic effect was attributed to its anti-inflammatory and antioxidant properties, and its ability to promote IL-1β-induced chondrocyte proliferation, inhibit apoptosis, and prevent the degradation of extracellular matrix. These favorable results were associated with the inhibition of the PI3K/AKT, NF-κB, and JAK2/STAT3 signaling pathways.
Collapse
Affiliation(s)
- Yanqiu Sun
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230038, Anhui Province, China; Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui Province, China.
| | - Jian Liu
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230038, Anhui Province, China; Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui Province, China.
| | - Jue Wang
- Sinopharm Group Jingfang (Anhui) Pharmaceutical Co., Ltd, Xuancheng City, 242000, Anhui Province, China.
| | - Mingyu He
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230038, Anhui Province, China; Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui Province, China.
| | - Xiaolu Chen
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230038, Anhui Province, China; Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui Province, China.
| | - Li Chen
- Pharmaceutical Department, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230038, Anhui Province, China.
| |
Collapse
|
9
|
Sun W, Xu JD, Zhang W, Guo MF, Kong M, Zhu H, Zhou SS, Wu CY, Li SL, Mao Q. Holistic quality evaluation of Callicarpae Formosanae Folium by multi-chromatography-based qualitative and quantitative analysis of polysaccharides and small molecules. J Pharm Biomed Anal 2023; 227:115282. [PMID: 36791651 DOI: 10.1016/j.jpba.2023.115282] [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: 12/17/2022] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023]
Abstract
Callicarpae Formosanae Folium (CFF), derived from the leaves of Callicarpa formosana Rolfe, is a common Chinese medicinal herb used for the treatment of hematemesis. Phytochemical studies found that phenylpropanoids, flavonoids, terpenoids and polysaccharides were the main ingredients of CFF. However, there is limited scientific information concerning holistic quality method and quality consistency evaluation of CFF. In this study, a strategy integrating HPGPC-ELSD, HPLC-PDA, UV-VIS and UPLC-QTOF-MS/MS was firstly developed to simultaneously qualify and quantify polysaccharides, as well as representative small molecules in CFF. HPGPC-ELSD was applied to characterize the molecular weight distribution of polysaccharides, HPLC-PDA was developed to qualitatively and quantitatively determine monosaccharides. UV-VIS was used to determine the total polysaccharides content, and UPLC-QTOF-MS/MS was established to characterize the small molecules. The quality consistency of commercial CFF (CM-CFF) was also evaluated. It was shown that the relative molecular weights, the compositional monosaccharides and small molecules composition in CM-CFF and self-collected CFF (SC-CFF) samples were similar. A total of 32 small molecules including 6 phenylpropanoids, 7 flavonoids and 19 terpenoids were characterized in CFF. However, the variation was observed in the content of polysaccharides, luteolin, ursolic acid, as well as total contents of terponoids in CM-CFF samples, which implied that the holistic quality of CM-CFF was inconsistent. The results suggested that the proposed evaluation strategy could be applied as a potential approach for the quality control of CFF. And the quality of CM-CFF should be improved by Good Agriculture Practice (GAP) base and standard processing method.
Collapse
Affiliation(s)
- Wen Sun
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Jin-Di Xu
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China
| | - Wei Zhang
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Meng-Fei Guo
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Ming Kong
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - He Zhu
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Shan-Shan Zhou
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Cheng-Yin Wu
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Song-Lin Li
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China.
| | - Qian Mao
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China.
| |
Collapse
|
10
|
Zhou YL, Zhang DN, Qiao JX, Wu J, Yan YM, Cao ZX, Peng C, Tan YZ. Diverse alkaloids from the aerial parts of Aconitum carmichaelii and antiproliferative activity of costemline via inhibiting SIRT1/ROCK1/P-STAT3 pathways. PHYTOCHEMISTRY 2023; 207:113558. [PMID: 36521584 DOI: 10.1016/j.phytochem.2022.113558] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 06/17/2023]
Abstract
Six undescribed alkaloids together with 15 known alkaloids were isolated from the aerial parts of Aconitum carmichaelii. Their structures were elucidated extensively by NMR and HRESIMS spectroscopy. The absolute configurations of N-formyllaurotetanine, and the known compounds glaucine-β-N-oxide and glaucine-α-N-oxide were established by electronic circular dichroism (ECD) spectra. Notably, it was the discovery of rare indole alkaloids from the genus Aconitum, and biosynthetic pathway of compounds 1 and 6 was deduced. Evaluation of the antiproliferative activity of these alkaloids demonstrated that costemline exhibited significant anti-proliferation effects against HCT116, SKOV3, and A549 cells with IC50 values of 5.6, 14.2, and 6.8 μM, respectively. Costemline could also inhibit the cell invasion activity of HCT116 cells. Mechanistic studies in HCT116 cells suggested that the antiproliferative activity of costemline was attributable to SIRT1/ROCK1/P-STAT3 pathways regulation. This study revealed the potential for developing and utilizing the aerial parts of Aconitum carmichaelii.
Collapse
Affiliation(s)
- Yin-Lin Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Duan-Na Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ji-Xu Qiao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jing Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yong-Ming Yan
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Zhi-Xing Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Yu-Zhu Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
11
|
Tang R, Wang L, Zhang J, Li X, Tan L, He W, Han H, Liu Y, Wang K, Wang M. Exploring the active ingredients and pharmacological mechanisms of the oral intake formula Huoxiang Suling Shuanghua Decoction on influenza virus type A based on network pharmacology and experimental exploration. Front Microbiol 2022; 13:1040056. [DOI: 10.3389/fmicb.2022.1040056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo investigate the active ingredients, underlying anti-influenza virus effects, and mechanisms of Huoxiang Suling Shuanghua Decoction (HSSD).Materials and methodsThe therapeutic effect of HSSD were confirmed through the survival rate experiment of H1N1-infected mice. Then, the HSSD solution and the ingredients absorbed into the blood after treatment with HSSD in rats were identified by UPLC/Q-TOF MS, while the main contents of ingredients were detected by high performance liquid chromatography (HPLC). Next, a systems pharmacology approach incorporating target prediction, gene ontology (GO) enrichment, kyoto encyclopedia of genes and genomes (KEGG) pathway analysis, and molecular docking were performed to screen out the active compounds and critical pathways of HSSD in treating influenza. According to prediction results, real-time quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry assay were used to detect the mRNA and protein expression levels of critical targets in H1N1-infected mice lungs.ResultsHuoxiang Suling Shuanghua Decoction improved the survival rate of H1N1-infected mice and prolonged the mice’s lifespan. Besides, HSSD exerts an antivirus effect by decreasing the levels of hemagglutinin (HA) and nucleoprotein (NP) to inhibit the replication and proliferation of H1N1, reducing the lung pathological state, inhibiting the cell apoptosis in the lung, and regulating the abnormal responses of peripheral blood, including GRA, LYM, white blood cell (WBC), PLT, and hemoglobin (HGB). Then, 87 compounds in the HSSD solution and 20 ingredients absorbed into the blood after treatment with HSSD were identified. Based on this, combined with the network analysis and previous research on antivirus, 16 compounds were screened out as the active components. Moreover, 16 potential targets were predicted by network pharmacology analysis. Next, molecular docking results showed stable binding modes between compounds and targets. Furthermore, experimental validation results indicated that HSSD regulates the contents of Immunoglobulin A (IgA), Immunoglobulin M (IgM), and Immunoglobulin G (IgG) in serum, modulating the levels of IFN-γ, IL-6, IL-10, MCP-1, MIP-1α, and IP-10 in the lung tissue, and significantly decreasing the mRNA and protein expressions of TLR4, CD14, MyD88, NF-κB p65, HIF1 α, VEGF, IL17A, and IL6 in the lung tissue.ConclusionHuoxiang Suling Shuanghua Decoction exerts an anti-influenza effect by affecting the expressions of mRNA and protein including TLR4, CD14, MyD88, NF-kB p65, HIF-1α, VEGF, IL17A, IL6, and inhibiting the accumulation of inflammation. Our study provided experimental pieces of evidence about the practical application of HSSD in treating influenza.
Collapse
|
12
|
Identification of Interleukin-1-Beta Inhibitors in Gouty Arthritis Using an Integrated Approach Based on Network Pharmacology, Molecular Docking, and Cell Experiments. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2322417. [PMID: 36193152 PMCID: PMC9526673 DOI: 10.1155/2022/2322417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022]
Abstract
Background This study aimed to investigate the molecular mechanism of Tongfengding capsule (TFDC) in treating immune-inflammatory diseases of gouty arthritis (GA) and interleukin-1-beta (IL-1β) inhibitors by using network pharmacology, molecular docking, and cell experiments. Methods In this study, the compounds of TFDC and the potential inflammatory targets of GA were obtained from Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP), Online Mendelian Inheritance in Man (OMIM), and GeneCards databases. The TFDC-GA-potential targets interaction network was accomplished by the STRING database. The TFDC-active compound-potential target-GA network was constructed using Cytoscape software. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were used to further explore the GA mechanism and therapeutic effects of TFDC. Quantitative real-time PCR (qPCR) was used to verify whether the TFDC inhibited IL-1β in GA. Molecular docking technology was used to analyze the optimal effective compounds from the TFDC for docking with IL-1β. Result 133 active compounds and 242 targets were screened from the TFDC, and 25 of the targets intersected with GA inflammatory targets, which were considered as potential therapeutic targets. Network pharmacological analysis showed that the TFDC active compounds such as quercetin, stigmasterol, betavulgarin, rutaecarpine, naringenin, dihydrochelerythrine, and dihydrosanguinarine had better correlation with GA inflammatory targets such as PTGS2, PTGS1, NOS2, SLC6A3, HTR3A, PPARG, MAPK14, RELA, MMP9, and MMP2. The immune-inflammatory signaling pathways of the active compounds for treating GA are IL-17 signaling pathway, TNF signaling pathway, NOD-like receptor signaling pathway, NF-kappa B signaling pathway, Toll-like receptor signaling pathway, HIF-1 signaling pathway, etc. The TFDC reduced IL-1β mRNA expression in GA by qPCR. Molecular docking results suggested that rutaecarpine was the most appropriate natural IL-1β inhibitor. Conclusion Our findings provide an essential role and bases for further immune-inflammatory studies on the molecular mechanisms of TFDC and IL-1β inhibitors development in GA.
Collapse
|
13
|
Zhao Z, Liu Y, Lu Y, Hou M, Shen X, Yang H, Shi Q, Zhang Y, He F, Zhu X. Gingko biloba-inspired lactone prevents osteoarthritis by activating the AMPK-SIRT1 signaling pathway. Arthritis Res Ther 2022; 24:197. [PMID: 35982488 PMCID: PMC9387049 DOI: 10.1186/s13075-022-02890-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Uncoupled extracellular matrix (ECM) causes cartilage degeneration and osteoarthritis (OA) by suppressing the synthesis and activating the degradation of ECM components. Gingko biloba is a natural Chinese herb with a variety of biological functions; however, the extent to which it can protect against OA and the mechanisms involved are unknown. METHODS In our study, using bioinformatics tools, we were able to identify an important lactone, bilobalide (BB), from Gingko biloba. In vitro experiments were performed to evaluate the potential therapeutic effects of BB on ECM homeostasis. In vivo experiments were conducted to assess the protection of systemic administration of BB on cartilage degeneration. Molecular mechanisms underlying BB-regulated anti-arthritic role were further explored. RESULTS In interleukin-1β-incubated human chondrocytes, in vitro treatment with BB increased the expression of cartilage anabolic proteins, while inhibiting the activities of ECM degrading enzymes. In a mice model, systemic administration of BB, in vivo, prevented post-traumatic cartilage erosion and attenuated the formation of abnormal osteophytes in the subchondral bone. Mechanistically, the activation of the adenosine 5'-monophosphate-activated protein kinase (AMPK)-sirtuin 1 (SIRT1) signaling pathway was involved in the anti-arthritic effects of BB. In vitro, blocking BB's chondroprotection with the AMPK-specific inhibitor Compound C abrogated it. CONCLUSIONS These results demonstrated that BB extracted from Gingko biloba regulates ECM balance to prevent OA by activating the AMPK-SIRT1 signaling pathway. This study proposed the monomer BB, a traditional Chinese medicine, as a de novo therapeutic insight for OA. Schematic representation of the experimental design. Based on the bioinformatic analysis, bilobalide (BB), a natural herb Gingko biloba-derived ingredient, was identified as a candidate for treating osteoarthritis. In vitro, BB treatment not only facilitates cartilage extracellular matrix synthesis but also inhibits proteolytic enzyme activities. In vivo intraperitoneal injection of BB improves cartilage degeneration and subchondral bone sclerosis. BB, in particular, had anti-arthritic effects by activating the AMPK-SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Zhijian Zhao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Yang Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Yingjie Lu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Mingzhuang Hou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Xu Shen
- Department of Orthopaedics, Suzhou Dushu Lake Hospital, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Qin Shi
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Yijian Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China. .,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China.
| | - Fan He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China. .,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China.
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China. .,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China.
| |
Collapse
|
14
|
Molecular Mechanism of Salvia miltiorrhiza Bunge in Treating Cerebral Infarction. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5992394. [PMID: 35392650 PMCID: PMC8983215 DOI: 10.1155/2022/5992394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 12/22/2021] [Indexed: 11/18/2022]
Abstract
Background Cerebral infarction (CI) is a common brain disease in clinical practice, which is mainly due to the pathological environment of ischemia and hypoxia caused by difficult cerebral circulation perfusion function, resulting in ischemic necrosis of local brain tissue and neurological impairment. In traditional Chinese medicine (TCM) theory, CI is mainly due to blood stasis in the brain. Therefore, blood-activating and stasis-dissipating drugs are often used to treat CI in clinical practice. Salvia miltiorrhiza Bunge (SMB) is a kind of traditional Chinese medicine with good efficacy in promoting blood circulation and removing blood stasis, and treatment of CI with it is a feasible strategy. Based on the above analysis, we chose network pharmacology to investigate the feasibility of SMB in the treatment of CI and to study the possible molecular mechanisms by providing some reference for the treatment of CI with TCM. Methods The active ingredients and related targets of SMB were obtained through the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database, and CI-related targets were obtained from the GeneCards and DisGeNET databases. The target of SMB for the treatment of CI was obtained using Cytoscape software and visualized. GO and KEGG enrichment analysis was performed based on “clusterProfiler” within R, and the prediction results were validated by molecular docking technique. Results By constructing a compound-target (C-T) network, it was found that the active components in SMB mainly treated CI by regulating key proteins such as AKT1, IL-6, and EGFR. These key proteins mainly involve in pathways such as immune regulation, cancer and lipid metabolism, such as lipid and atherosclerosis, chemical carcinogenesis-receptor activation pathways, and IL-17 signaling pathway. In the GO term, it mainly regulates the response to steroid hormones, membrane rafts, and G protein-amine receptor coupled activity. Eventually, we verified that the luteolin and tanshinone IIA components in SMB have a good possibility of action with AKT1 and IL-6 by in silico techniques, indicating that SMB has some scientificity in the treatment of CI. Conclusion SMB mainly treats CI by regulating 94 proteins involved in lipid and atherosclerosis, chemical carcinogenesis-receptor activation, and IL-17 signaling pathway. Our research strategy provided a template for the drug development of TCM for the treatment of CI.
Collapse
|
15
|
Li M, Zhao X, Xie J, Tong X, Shan J, Shi M, Wang G, Ye W, Liu Y, Unger BH, Cheng Y, Zhang W, Wu N, Xia XQ. Dietary Inclusion of Seabuckthorn ( Hippophae rhamnoides) Mitigates Foodborne Enteritis in Zebrafish Through the Gut-Liver Immune Axis. Front Physiol 2022; 13:831226. [PMID: 35464096 PMCID: PMC9019508 DOI: 10.3389/fphys.2022.831226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
To help prevent foodborne enteritis in aquaculture, several feed additives, such as herbal medicine, have been added to fish diets. Predictions of effective herb medicines for treating fish foodborne enteritis from key regulated DEGs (differentially expressed genes) in transcriptomic data can aid in the development of feed additives using the Traditional Chinese Medicine Integrated Database. Seabuckthorn has been assessed as a promising candidate for treating grass carp soybean-induced enteritis (SBMIE). In the present study, the SBMIE zebrafish model was used to assess seabuckthorn's therapeutic or preventative effects. The results showed that intestinal and hepatic inflammation was reduced when seabuckthorn was added, either pathologically (improved intestinal villi morphology, less oil-drops) or growth-related (body fat deposition). Moreover, seabuckthorn may block the intestinal p53 signaling pathway, while activating the PPAR signaling pathway and fatty acid metabolism in the liver. 16S rRNA gene sequencing results also indicated a significant increase in OTU numbers and skewed overlapping with the fish meal group following the addition of seabuckthorn. Additionally, there were signs of altered gut microbiota taxa composition, particularly for reduced TM7, Sphingomonas, and Shigella, following the addition of seabuckthorn. Hindgut imaging of fluorescent immune cells in SBMIE larvae revealed the immune regulatory mechanisms at the cellular level. Seabuckthorn may significantly inhibit the inflammatory gathering of neutrophils, macrophages, and mature T cells, as well as cellular protrusions' formation. On the other hand, in larvae, seabuckthorn inhibited the inflammatory aggregation of lck+ T cells but not immature lymphocytes, indicating that it affected intestinal adaptive immunity. Although seabuckthorn did not affect the distribution of intestinal CD4+ cells, the number of hepatic CD4+ cells were reduced in fish from the seabuckthorn supplementation group. Thus, the current data indicate that seabuckthorn may alleviate foodborne gut-liver symptoms by enhancing intestinal mucosal immunity and microbiota while simultaneously inhibiting hepatic adipose disposition, making it a potential additive for preventing fish foodborne gut-liver symptoms.
Collapse
Affiliation(s)
- Ming Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Xuyang Zhao
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Jiayuan Xie
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xinyu Tong
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Junwei Shan
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Mijuan Shi
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Guangxin Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Weidong Ye
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yuhang Liu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | | | - Yingyin Cheng
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Wanting Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Nan Wu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiao-Qin Xia
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
16
|
Li H, Zhang X, Gu L, Li Q, Ju Y, Zhou X, Hu M, Li Q. Anti-Gout Effects of the Medicinal Fungus Phellinus igniarius in Hyperuricaemia and Acute Gouty Arthritis Rat Models. Front Pharmacol 2022; 12:801910. [PMID: 35087407 PMCID: PMC8787200 DOI: 10.3389/fphar.2021.801910] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/16/2021] [Indexed: 12/23/2022] Open
Abstract
Background:Phellinus igniarius (P. igniarius) is an important medicinal and edible fungus in China and other Southeast Asian countries and has diverse biological activities. This study was performed to comparatively investigate the therapeutic effects of wild and cultivated P. igniarius on hyperuricaemia and gouty arthritis in rat models. Methods: UPLC-ESI-qTOF-MS was used to identify the chemical constituents of polyphenols from wild P. igniarius (WPP) and cultivated P. igniarius (CPP). Furthermore, WPP and CPP were evaluated in an improved hyperuricaemia rat model induced by yeast extract, adenine and potassium oxonate, which was used to examine xanthine oxidase (XO) activity inhibition and anti-hyperuricemia activity. WPP and CPP therapies for acute gouty arthritis were also investigated in a monosodium urate (MSU)-induced ankle swelling model. UHPLC-QE-MS was used to explore the underlying metabolic mechanisms of P. igniarius in the treatment of gout. Results: The main active components of WPP and CPP included protocatechuic aldehyde, hispidin, davallialactone, phelligridimer A, hypholomine B and inoscavin A as identified by UPLC-ESI-qTOF-MS. Wild P. igniarius and cultivated P. igniarius showed similar activities in reducing uric acid levels through inhibiting XO activity and down-regulating the levels of UA, Cr and UN, and they had anti-inflammatory activities through down-regulating the secretions of ICAM-1, IL-1β and IL-6 in the hyperuricaemia rat model. The pathological progression of kidney damage was also reversed. The polyphenols from wild and cultivated P. igniarius also showed significant anti-inflammatory activity by suppressing the expression of ICAM-1, IL-1β and IL-6 and by reducing the ankle joint swelling degree in an MSU-induced acute gouty arthritis rat model. The results of metabolic pathway enrichment indicated that the anti-hyperuricemia effect of WPP was mainly related to the metabolic pathways of valine, leucine and isoleucine biosynthesis and histidine metabolism. Additionally, the anti-hyperuricemia effect of CPP was mainly related to nicotinate and nicotinamide metabolism and beta-alanine metabolism. Conclusions: Wild P. igniarius and cultivated P. igniarius both significantly affected the treatment of hyperuricaemia and acute gouty arthritis models in vivo and therefore may be used as potential active agents for the treatment of hyperuricaemia and acute gouty arthritis.
Collapse
Affiliation(s)
- Hongxing Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Xinyue Zhang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Lili Gu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Qín Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Yue Ju
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Xuebin Zhou
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Min Hu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Qīn Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
17
|
Chen W, He L, Zhong L, Sun J, Zhang L, Wei D, Wu C. Identification of Active Compounds and Mechanism of Huangtu Decoction for the Treatment of Ulcerative Colitis by Network Pharmacology Combined with Experimental Verification. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:4125-4140. [PMID: 34616145 PMCID: PMC8487861 DOI: 10.2147/dddt.s328333] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/17/2021] [Indexed: 01/08/2023]
Abstract
Introduction Huangtu decoction (HTD) has been widely used in the treatment of gastrointestinal bleeding, ulcerative colitis (UC) and gastrointestinal tumors in China, but its active compounds and mechanism are still not clear yet. The present research aimed to identify the active compounds and mechanism of HTD for the treatment of UC. Methods Firstly, the chemical compounds of HTD were qualitatively identified based on Q Exactive Orbitrap LC-MS/MS, and their potential targets were predicted through SwissTargetPrediction. Secondly, the differential expressed genes (DEGs) in colon tissues of UC patients and normal controls were retrieved from the GEO database. Thirdly, the overlapping targets of DEGs and the predicted targets were obtained and subjected to GO and KEGG analysis. Finally, the key targets in the most significantly enriched pathway were verified by in vivo experiment, and the protein and mRNA expressions of matrix metalloproteinase-1 (MMP1), MMP3, MMP7, MMP9 and MMP12 were determined by immunohistochemistry (IHC), Western blotting (WB) and quantitative real-time-PCR (qRT-PCR). Results A total of 47 compounds were identified and 29 overlapping targets were obtained from HTD extract. The most significantly enriched pathway of overlapping targets involved was MMP. HTD improved the pathological damage in colon tissues of DSS-induced UC model and significantly decreased the serum levels of IL-1β and IL-6. The protein and mRNA expressions of MMP1, MMP3 and MMP9 in colon tissues were significantly decreased after HTD treatment. Conclusion HTD treatment can alleviate the colonic inflammation via inhibiting MMPs including MMP1, MMP3 and MMP9.
Collapse
Affiliation(s)
- Wenwen Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China.,Department of Pharmacy, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610091, People's Republic of China
| | - Lin He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Lian Zhong
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Jiayi Sun
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Lilin Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Daneng Wei
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Chunjie Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| |
Collapse
|