1
|
Lin SK, Chen ST, Zhan Y, Guo XY, Wu WT, Lin YT, Yu CX, Yang J. The alleviatory effects of koumine on MSU-induced gouty arthritis via the TLR4/NF-κB/NLRP3 pathway. Basic Clin Pharmacol Toxicol 2024; 135:133-147. [PMID: 38828789 DOI: 10.1111/bcpt.14037] [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: 12/11/2023] [Revised: 03/13/2024] [Accepted: 05/15/2024] [Indexed: 06/05/2024]
Abstract
The aim of this study was to validate the preventive effects of koumine (KM), a monoterpene indole alkaloid, on gouty arthritis (GA) and to explore its possible mechanisms. C57BL/6 mice were intraperitoneally administered KM (0.8, 2.4 or 7.2 mg/kg), colchicine (3.0 mg/kg) or sterile saline. One hour later, a monosodium urate (MSU) suspension was injected into the right hind paws of the mice to establish an acute gout model. Inflammation symptoms were evaluated at 0, 3, 6, 12 and 24 h, and the mechanical withdrawal threshold was evaluated at 0, 6 and 24 h. After 24 h, the mice were euthanized, and the joint tissue, kidney and blood were collected for subsequent experiments. Histological examination and antioxidant enzyme, kidney index and serum uric acid (UA) measurements were taken. The expression levels of the signalling pathway components were determined. KM effectively alleviated the symptoms of redness, swelling and pain; counteracted inflammatory cell infiltration; and increased antioxidant enzyme levels, reduced kidney index and serum UA levels through regulating UA excretion in MSU-induced mice. The expression of toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB)/nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing 3 (NLRP3) signalling pathway proteins and mRNA were reduced in the KM group. These results suggest that KM may be effective in alleviating GA through the TLR4/NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Shi-Kang Lin
- School of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shi-Ting Chen
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Ying Zhan
- School of Stomatology, Fujian Medical University, Fuzhou, China
| | - Xin-Yue Guo
- School of Stomatology, Fujian Medical University, Fuzhou, China
| | - Wen-Tao Wu
- School of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yi-Ting Lin
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Chang-Xi Yu
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Jian Yang
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
2
|
Xu J, Zhang S, Luo SH, Xiong CR, Zhu M, Chang J, Zou B, Ren B, Tian ZQ, Liu GK. Rapid Sample Pretreatment Facilitating SERS Detection of Trace Weak Organic Acids/Bases in Complex Matrices. Anal Chem 2024; 96:9399-9407. [PMID: 38804597 DOI: 10.1021/acs.analchem.4c00234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Fast and efficient sample pretreatment is the prerequisite for realizing surface-enhanced Raman spectroscopy (SERS) detection of trace targets in complex matrices, which is still a big issue for the practical application of SERS. Recently, we have proposed a highly performed liquid-liquid extraction (LLE)-back extraction (BE) for weak acids/bases extraction in drinking water and beverage samples. However, the performance efficiency decreased drastically on facing matrices like food and biological blood. Based on the total interaction energies among target, interferent, and extractant molecules, solid-phase extraction (SPE) with a higher selectivity was introduced in advance of LLE-BE, which enabled the sensitive (μg L-1 level) and rapid (within 10 min) SERS detection of both koumine (a weak base) and celastrol (a weak acid) in different food and biological samples. Further, the high SERS sensitivity was determined unmanned by Vis-CAD (a machine learning algorithm), instead of the highly demanded expert recognition. The generality of SPE-LLE-BE for various weak acids/bases (2 < pKa < 12), accompanied by the high efficiency, easy operation, and low cost, offers SERS as a powerful on-site and efficient inspection tool in food safety and forensics.
Collapse
Affiliation(s)
- Jing Xu
- State Key Laboratory of Marine Environmental Science, Fujian Provincial Key Laboratory for Coastal Ecology and Environmental Studies, Center for Marine Environmental Chemistry & Toxicology, College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Shu Zhang
- State Key Laboratory for Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Si-Heng Luo
- State Key Laboratory for Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Chen-Ru Xiong
- State Key Laboratory of Marine Environmental Science, Fujian Provincial Key Laboratory for Coastal Ecology and Environmental Studies, Center for Marine Environmental Chemistry & Toxicology, College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Minghuai Zhu
- Institute of Forensic Science, Xiamen Public Security Bureau, Xiamen 361000, China
| | - Jing Chang
- Institute of Forensic Science, Ministry of Public Security, Beijing 100038, China
| | - Bo Zou
- Institute of Forensic Science, Ministry of Public Security, Beijing 100038, China
| | - Bin Ren
- State Key Laboratory for Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Zhong-Qun Tian
- State Key Laboratory for Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Guo-Kun Liu
- State Key Laboratory of Marine Environmental Science, Fujian Provincial Key Laboratory for Coastal Ecology and Environmental Studies, Center for Marine Environmental Chemistry & Toxicology, College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| |
Collapse
|
3
|
Cai N, Gao X, Li W, Yang L, Zhao J, Qu J, Zhou Y. Novel trifluoromethyl ketone derivatives as oral cPLA 2/COX-2 dual inhibitors for resolution of inflammation in rheumatoid arthritis. Bioorg Chem 2024; 148:107453. [PMID: 38761708 DOI: 10.1016/j.bioorg.2024.107453] [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: 03/20/2024] [Revised: 04/25/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
Thirty-five trifluoromethyl hydrazones and seventeen trifluoromethyl oxime esters were designed and synthesized via molecular hybridization. All the target compounds were initially screened for in vitro anti-inflammatory activity by assessing their inhibitory effect on NO release in LPS-stimulated RAW264.7 cells, and the optimal compound was finally identified as 2-(3-Methoxyphenyl)-N'-((6Z,9Z,12Z,15Z)-1,1,1-trifluorohenicosa-6,9,12,15-tetraen-2-ylidene)acetohydrazide (F26, IC50 = 4.55 ± 0.92 μM) with no cytotoxicity. Moreover, F26 potently reduced the production of PGE2 in LPS-stimulated RAW264.7 cells compared to indomethacin. The interaction of F26 with COX-2 and cPLA2 was directly verified by the CETSA technique. F26 was found to modulate the phosphorylation levels of p38 MAPK and NF-κB p65, as well as the protein expression of IκB, cPLA2, COX-2, and iNOS in LPS-stimulated rat peritoneal macrophages. Additionally, F26 was observed to prevent the nuclear translocation of NF-κB p65 in LPS-stimulated rat peritoneal macrophages by immunofluorescence localization. Therefore, the aforementioned in vitro experiments demonstrated that F26 blocked the p38 MAPK and NF-κB pathways by binding to COX-2 and cPLA2. In the adjuvant-induced arthritis model, F26 demonstrated a significant effect in preventing arthritis symptoms and inflammatory status in rats, exerting an immunomodulatory role by regulating the homeostasis between Th17 and Treg through inhibition of the p38 MAPK/cPLA2/COX-2/PGE2 and NF-κB pathways. Encouragingly, F26 caused less acute ulcerogenicity in rats at a dose of 50 mg/kg compared to indomethacin. Overall, F26 is a promising candidate worthy of further investigation for treating inflammation and associated pain with lesser gastrointestinal irritation, as well as other symptoms in which cPLA2 and COX-2 are implicated in the pathophysiology.
Collapse
Affiliation(s)
- Nan Cai
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China
| | - Xiang Gao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China
| | - Wenjing Li
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China
| | - Li Yang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China.
| | - Jinfeng Zhao
- Instrumental Analysis Center, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China.
| | - Jingping Qu
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China.
| | - Yuhan Zhou
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China.
| |
Collapse
|
4
|
Han T, Sun Y, Zhao C, Wang HY, Yu H, Liu Y. Mitochondrial-Targeted Ratiometric Near-Infrared Fluorescence Probe for Monitoring Nitric Oxide in Rheumatoid Arthritis. J Med Chem 2024; 67:4026-4035. [PMID: 38359302 DOI: 10.1021/acs.jmedchem.3c02344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Rheumatoid arthritis (RA) is a destructive autoimmune disease, where nitric oxide (NO) is closely implicated in the inflammatory processes of RA. Therefore, direct visualization of NO is essential to assess the pathological changes in RA. Herein, a mitochondrial-targeted near-infrared ratiometric fluorescent probe (NFL-NH2), based on the intramolecular charge transfer effect, was synthesized and applied to monitor the changes of NO content in early RA. Specially, probe NFL-NH2 showed a 44-fold fluorescent intensity ratio (I705/I780) response toward NO with a detection limit of 0.536 nM, enabling qualitative and quantitative analysis of NO. Additionally, NFL-NH2 can accurately target mitochondria and sensitively detect exogenous and endogenous NO in RAW 264.7 cells. Notably, in vivo RA monitoring assays demonstrated that NFL-NH2 can rapidly detect NO levels associated with the inflammatory damage degree in RA mice models by ratiometric fluorescence imaging. These results validate that NFL-NH2 holds significant potential for diagnosing NO-mediated RA diseases.
Collapse
Affiliation(s)
- Tingting Han
- School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Ye Sun
- School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Chao Zhao
- School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Hai-Yan Wang
- School of Mechanical Engineering, Southeast University, Nanjing 211189, China
| | - Hui Yu
- School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Liu
- School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
5
|
Cai N, Gao X, Yang L, Li W, Sun W, Zhang S, Zhao J, Qu J, Zhou Y. Discovery of novel NSAID hybrids as cPLA 2/COX-2 dual inhibitors alleviating rheumatoid arthritis via inhibiting p38 MAPK pathway. Eur J Med Chem 2024; 267:116176. [PMID: 38286094 DOI: 10.1016/j.ejmech.2024.116176] [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: 11/22/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024]
Abstract
A series of NSAIDs hybrid molecules were synthesized and characterized, and their ability to inhibit NO release in LPS-induced RAW264.7 macrophages was evaluated. Most of the compounds showed significant anti-inflammatory activity in vitro, of which (2E,6Z,9Z,12Z,15Z)-1,1,1-trifluorohenicosa-2,6,9,12,15-pentaen-2-yl 2-(4-benzoylphenyl) propanoate (VI-60) was the most optimal (IC50 = 3.85 ± 0.25 μΜ) and had no cytotoxicity. In addition, VI-60 notably reduced the production of PGE2 in LPS-stimulated RAW264.7 cells compared to ketoprofen. Futhur more, VI-60 significantly inhibited the expression of iNOS, cPLA2, and COX-2 and the phosphorylation of p38 MAPK in LPS-stimulated RAW264.7 cells. The binding of VI-60 to cPLA2 and COX-2 was directly verified by the CETSA technique. In vivo studies illustrated that VI-60 exerted an excellent therapeutic effect on adjuvant-induced arthritis in rats by regulating the balance between Th17 and Treg through inhibiting the p38 MAPK/cPLA2/COX-2/PGE2 pathway. Encouragingly, VI-60 showed a lower ulcerative potential in rats at a dose of 50 mg/kg compared to ketoprofen. In conclusion, the hybrid molecules of NSAIDs and trifluoromethyl enols are promising candidates worthy of further investigation for the treatment of inflammation, pain, and other symptoms in which cPLA2 and COX-2 play a role in their etiology.
Collapse
Affiliation(s)
- Nan Cai
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Xiang Gao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Li Yang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Wenjing Li
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Wuding Sun
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Shuaibo Zhang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Jinfeng Zhao
- Instrumental Analysis Center, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Jingping Qu
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Yuhan Zhou
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| |
Collapse
|
6
|
Rana N, Gupta P, Singh H, Nagarajan K. Role of Bioactive Compounds, Novel Drug Delivery Systems, and Polyherbal Formulations in the Management of Rheumatoid Arthritis. Comb Chem High Throughput Screen 2024; 27:353-385. [PMID: 37711009 DOI: 10.2174/1386207326666230914103714] [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: 05/08/2023] [Revised: 07/04/2023] [Accepted: 07/24/2023] [Indexed: 09/16/2023]
Abstract
Rheumatoid Arthritis (RA) is an autoimmune disorder that generally causes joint synovial inflammation as well as gradual cartilage and degenerative changes, resulting in progressive immobility. Cartilage destruction induces synovial inflammation, including synovial cell hyperplasia, increased synovial fluid, and synovial pane development. This phenomenon causes articular cartilage damage and joint alkalosis. Traditional medicinal system exerts their effect through several cellular mechanisms, including inhibition of inflammatory mediators, oxidative stress suppression, cartilage degradation inhibition, increasing antioxidants and decreasing rheumatic biomarkers. The medicinal plants have yielded a variety of active constituents from various chemical categories, including alkaloids, triterpenoids, steroids, glycosides, volatile oils, flavonoids, lignans, coumarins, terpenes, sesquiterpene lactones, anthocyanins, and anthraquinones. This review sheds light on the utilization of medicinal plants in the treatment of RA. It explains various phytoconstituents present in medicinal plants and their mechanism of action against RA. It also briefs about the uses of polyherbal formulations (PHF), which are currently in the market and the toxicity associated with the use of medicinal plants and PHF, along with the limitations and research gaps in the field of PHF. This review paper is an attempt to understand various mechanistic approaches employed by several medicinal plants, their possible drug delivery systems and synergistic effects for curing RA with minimum side effects.
Collapse
Affiliation(s)
- Neha Rana
- KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, 201206, Uttar Pradesh, India
| | - Piyush Gupta
- Department of Chemistry, SRM Institute of Science and Technology, Faculty of Engineering and Technology, NCR Campus, Delhi-NCR Campus, Delhi-Meerut Road, Modinagar, 201204, Ghaziabad, Uttar Pradesh, India
| | - Hridayanand Singh
- Dr. K. N. Modi Institute of Pharmaceutical Education and Research, Modinagar, 201204, Uttar Pradesh, India
| | - Kandasamy Nagarajan
- KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, 201206, Uttar Pradesh, India
| |
Collapse
|
7
|
Wang L, Chen S, Gao X, Liang X, Lv W, Zhang D, Jin X. Recent progress in chemistry and bioactivity of monoterpenoid indole alkaloids from the genus gelsemium: a comprehensive review. J Enzyme Inhib Med Chem 2023; 38:2155639. [PMID: 36629436 PMCID: PMC9848241 DOI: 10.1080/14756366.2022.2155639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Monoterpenoid indole alkaloids (MIAs) represent a major class of active ingredients from the plants of the genus Gelsemium. Gelsemium MIAs with diverse chemical structures can be divided into six categories: gelsedine-, gelsemine-, humantenine-, koumine-, sarpagine- and yohimbane-type. Additionally, gelsemium MIAs exert a wide range of bioactivities, including anti-tumour, immunosuppression, anti-anxiety, analgesia, and so on. Owing to their fascinating structures and potent pharmaceutical properties, these gelsemium MIAs arouse significant organic chemists' interest to design state-of-the-art synthetic strategies for their total synthesis. In this review, we comprehensively summarised recently reported novel gelsemium MIAs, potential pharmacological activities of some active molecules, and total synthetic strategies covering the period from 2013 to 2022. It is expected that this study may open the window to timely illuminate and guide further study and development of gelsemium MIAs and their derivatives in clinical practice.
Collapse
Affiliation(s)
- Lin Wang
- School of Pharmacy, China Medical University, Shenyang, China
| | - Siyu Chen
- China Medical University-Queen’s University of Belfast Joint College, China Medical University, Shenyang, China
| | - Xun Gao
- Jiangsu Institute Marine Resources Development, Jiangsu Ocean University, Lianyungang, China
| | - Xiao Liang
- School of Pharmacy, Liaoning University, Shenyang, China
| | - Weichen Lv
- Department of Clinical Medicine, Dalian University, Dalian, China
| | - Dongfang Zhang
- School of Pharmacy, China Medical University, Shenyang, China,CONTACT Dongfang Zhang
| | - Xin Jin
- School of Pharmacy, China Medical University, Shenyang, China,Xin Jin School of Pharmacy, China Medical University, Shenyang, 110122, China
| |
Collapse
|
8
|
Lin YR, Zheng FT, Xiong BJ, Chen ZH, Chen ST, Fang CN, Yu CX, Yang J. Koumine alleviates rheumatoid arthritis by regulating macrophage polarization. JOURNAL OF ETHNOPHARMACOLOGY 2023; 311:116474. [PMID: 37031823 DOI: 10.1016/j.jep.2023.116474] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/30/2023] [Accepted: 04/07/2023] [Indexed: 06/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The imbalance between M1-and M2-polarized macrophages is one of the major pathophysiological changes in RA. Therefore, targeted macrophage polarization may be an effective therapy for RA. Koumine, an alkaloid monomer with the highest content and low toxicity in Gelsemium elegans Benth., has the effect of treating RA by playing an immunomodulatory role by influencing various immune cells. However, whether koumine affects macrophage polarization in RA and the associated molecular mechanisms remain unknown. AIM OF THE STUDY To investigate the mechanism of the anti-RA effect of koumine on macrophage polarization. MATERIALS AND METHODS The effect of koumine on macrophage polarization was investigated in vivo and in vitro. We first explored the effects of koumine on AIA rats and detected the levels of M1/M2 macrophage polarization markers in the spleen by western blotting. Then, we explored the regulatory effect of koumine on M1/M2 macrophage polarization and the effect on the PI3K/AKT signaling pathway in vitro. Finally, we verified the effects of koumine on macrophage polarization in CIA mice. RESULTS We found that koumine alleviated symptoms, including relieving pain, reducing joint redness and swelling in AIA rats and restoring the M1/M2 macrophage balance in vivo. Interestingly, koumine had an inhibitory effect on both M1 and M2 macrophage polarization in vitro, but it had a stronger inhibitory effect on M1 macrophage. In a mixed polarization experiment, koumine mainly inhibited M1 macrophage polarization and had an inhibitory effect on the PI3K/AKT signaling pathway. Finally, we found that koumine had therapeutic effects on CIA mice, regulated macrophage polarization and inhibited the PI3K/AKT signaling pathway. CONCLUSIONS Our results reveal that koumine regulates macrophage polarization through the PI3K/AKT signaling pathway. This may be one of the important mechanisms of its anti-RA effect, which provides a theoretical and scientific basis for the possible clinical application of koumine.
Collapse
Affiliation(s)
- Ya-Rong Lin
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| | - Feng-Ting Zheng
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| | - Bo-Jun Xiong
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| | - Ze-Hong Chen
- Laboratory of Medical Function, Basic Medical Experimental Teaching Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.
| | - Shi-Ting Chen
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| | - Chao-Nan Fang
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| | - Chang-Xi Yu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| | - Jian Yang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| |
Collapse
|
9
|
Hu Q, Fu XL, Dong YY, Ma J, Hua J, Li JT, Liu KX, Yang J, Yu CX. D-Optimal Design and Development of a Koumine-Loaded Microemulsion for Rheumatoid Arthritis Treatment: In vivo and in vitro Evaluation. Int J Nanomedicine 2023; 18:2973-2988. [PMID: 37304972 PMCID: PMC10255651 DOI: 10.2147/ijn.s406641] [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: 02/01/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Koumine (KME) is the most abundant active ingredient separated from Gelsemium elegans Benth and exhibits a significant therapeutic effect on rheumatoid arthritis (RA). It is a lipophilic compound with poor aqueous solubility, and there is an urgent need to develop novel dosage forms of KME and promote its clinical application for the treatment of RA. The aim of this study was to design and develop KME-loaded microemulsions (KME-MEs) for the effective management of RA. Methods The composition of the microemulsion was selected by carrying out a solubility study and generating pseudoternary phase diagrams, and further optimized by D-Optimal design. The optimized KME-MEs was evaluated for particle size, viscosity, drug release, storage stability, cytotoxicity, cellular uptake, Caco-2 cell transport and everted gut sac investigations. In vivo fluorescence imaging and the therapeutic effects of KME and KME-MEs on collagen-induced arthritis (CIA) rats were also evaluated. Results The optimized microemulsion contained 8% oil, 32% Smix (surfactant/cosurfactant) and 60% water and was used for in vivo and in vitro studies. The optimal KME-MEs exhibited a small globule size of 18.5 ± 0.14 nm and good stability over 3 months, and the release kinetics followed a first-order model. These KME-MEs had no toxic effect on Caco-2 cells but were efficiently internalized into the cytoplasm. Compared to KME, the KME-MEs displayed significantly increased permeability and absorption in Caco-2 cell monolayer assay and ex vivo everted gut sac experiment. As expected, the KME-MEs attenuated the progression of RA in CIA rats and were more effective than free KME with a reduced frequency of administration. Conclusion The KME-MEs improved the solubility and therapeutic efficacy of KME by employing formulation technology. These results provide a promising vehicle for the oral delivery of KME to treat RA and have attractive potential for clinical translation.
Collapse
Affiliation(s)
- Qing Hu
- School of Pharmacy, Fujian Medical University, Fuzhou, People’s Republic of China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Xiao-Ling Fu
- School of Pharmacy, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Yi-Yan Dong
- School of Pharmacy, Fujian Medical University, Fuzhou, People’s Republic of China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Ju Ma
- School of Pharmacy, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Jian Hua
- School of Pharmacy, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Jia-Ting Li
- School of Pharmacy, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Kai-Xin Liu
- School of Pharmacy, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Jian Yang
- School of Pharmacy, Fujian Medical University, Fuzhou, People’s Republic of China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Chang-Xi Yu
- School of Pharmacy, Fujian Medical University, Fuzhou, People’s Republic of China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, People’s Republic of China
| |
Collapse
|
10
|
Yuan Z, Yang M, Liang Z, Yang C, Kong X, Wu Y, Wang S, Fan H, Ning C, Xiao W, Sun Z, Wu J. PI3K/AKT/mTOR, NF-κB and ERS pathway participated in the attenuation of H 2O 2-induced IPEC-J2 cell injury by koumine. JOURNAL OF ETHNOPHARMACOLOGY 2023; 304:116028. [PMID: 36529250 DOI: 10.1016/j.jep.2022.116028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/13/2022] [Accepted: 12/03/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Koumine, an indole alkaloid extracted from Gelsemium elegans Benth, exerts anti-inflammation and antioxidant activities. However, the effects of koumine on intestinal injury induced by H2O2 and its potential molecular mechanisms need larger studies. AIM OF THE STUDY We established an IPEC-J2 cell damage model induced by H2O2 to explore the protective mechanism of koumine on intestinal injury. MATERIALS AND METHODS In the experiment, cell damage models were made with hydrogen peroxide. To assess the protective effect of koumine on H2O2-induced IPEC-J2 cell injury, CCK-8, the release of LDH and ROS, transmission electron microscopy and Annexin V-FITC/PI were employed. Western Blot and Quantitative Real-time PCR were used to determine the potential alleviated mechanism of koumine on H2O2-trigged IPEC-J2 cell damage. RESULTS The results of CCK-8 and LDH implied that koumine has a mitigative effect on H2O2-induced cell damage via upregulating cell viability and suppressing cell membrane fragmentation. Simultaneously, koumine notably inhibited the level of pro-inflammatory factors (IL-1β, IL-6, IL-8, TNF-α and TGF-β), the over-production of ROS along with decreasing the injury of mitochondrion, endoplasmic reticulum and lysosome induced by H2O2. Moreover, koumine dramatically attenuated H2O2-triggered IPEC-J2 cell apoptosis and autophagy. Subsequently, Western blot analysis identified NF-ΚB, PI3K and ERS as possible pathway responsible for the protective effect of koumine on H2O2-stimulated IPEC-J2 cell inflammation. CONCLUSIONS This in vitro experimental study suggests that koumine suppresses the H2O2-induced activation of inflammatory pathways, oxidative injury, ER stress, apoptosis and autophagy, which provide a rationale for therapeutically use in major intestinal diseases.
Collapse
Affiliation(s)
- Zhihang Yuan
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, PR China; Hunan Co-innovation Center of Animal Production Safety, Changsha, 410128, PR China
| | - Mengran Yang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, PR China; Hunan Co-innovation Center of Animal Production Safety, Changsha, 410128, PR China
| | - Zengenni Liang
- Department of Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, 410128, PR China
| | - Chenglin Yang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, PR China; Hunan Co-innovation Center of Animal Production Safety, Changsha, 410128, PR China
| | - Xiangyi Kong
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, PR China; Hunan Co-innovation Center of Animal Production Safety, Changsha, 410128, PR China
| | - You Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, PR China; Hunan Co-innovation Center of Animal Production Safety, Changsha, 410128, PR China
| | - Siqi Wang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, PR China; Hunan Co-innovation Center of Animal Production Safety, Changsha, 410128, PR China
| | - Hui Fan
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, PR China; Hunan Co-innovation Center of Animal Production Safety, Changsha, 410128, PR China
| | - Can Ning
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, PR China; Hunan Co-innovation Center of Animal Production Safety, Changsha, 410128, PR China
| | - Wenguang Xiao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, PR China; Hunan Co-innovation Center of Animal Production Safety, Changsha, 410128, PR China
| | - Zhiliang Sun
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, PR China; Hunan Co-innovation Center of Animal Production Safety, Changsha, 410128, PR China; Hunan Engineering Research Center of Veterinary Drug, Hunan Agricultural University, Changsha, 410128, PR China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, PR China.
| | - Jing Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, PR China; Hunan Co-innovation Center of Animal Production Safety, Changsha, 410128, PR China.
| |
Collapse
|
11
|
(+)/(-)-Yanhusamides A-C, three pairs of unprecedented benzylisoquinoline-pyrrole hetero-dimeric alkaloid enantiomers from Corydalis yanhusuo. Acta Pharm Sin B 2023; 13:754-764. [PMID: 36873186 PMCID: PMC9979263 DOI: 10.1016/j.apsb.2022.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/30/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
A chemical investigation on the aqueous extract of Corydalis yanhusuo tubers led to the isolation and structural elucidation of three pairs of trace enantiomeric hetero-dimeric alkaloids, (+)/(-)-yanhusamides A-C (1-3), featuring an unprecedented 3,8-diazatricylco[5.2.2.02,6]undecane-8,10-diene bridged system. Their structures were exhaustively characterized by X-ray diffraction, comprehensive spectroscopic data analysis, and computational methods. Guided by the hypothetical biosynthetic pathway for 1-3, a gram-scale biomimetic synthesis of (±)-1 was achieved in 3 steps using photoenolization/Diels-Alder (PEDA) [4+2] cycloaddition. Compounds 1‒3 exhibited potent inhibition of NO production induced by LPS in RAW264.7 macrophages. The in vivo assay showed that oral administration of 30 mg/kg of (±)-1 attenuated the severity of rat adjuvant-induced arthritis (AIA). Additionally, (±)-1 induced a dose-dependent antinociceptive effect in the acetic acid-induced mice writhing assay.
Collapse
|
12
|
Koumine ameliorates concanavalin A-induced autoimmune hepatitis in mice: involvement of the Nrf2, NF-κB pathways, and gut microbiota. Int Immunopharmacol 2023; 114:109573. [PMID: 36527886 DOI: 10.1016/j.intimp.2022.109573] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Gelsemiumelegans(Gardner. & Chapm.) Benth. has long been considered a traditional Chinese medicine effective against rheumatoid pain, cancer, cirrhosis, and skin diseases. Koumine (KM), the most abundant alkaloid in G.elegans Benth., demonstrates a variety of biological effects, including antitumor, analgesic, anxiolytic, anti-inflammatory, antidepressant, antioxidant, immunoregulatory, and hepatoprotective effects. Furthermore, the relatively low toxicity of KM makes it a promising drug candidate. This study aimed to investigate the protective effects of KM and its possible mechanisms using a concanavalin A (Con A)-induced autoimmune hepatitis (AIH) model in mice. Mice were orally administered different doses of KM for 14 d before Con A tail vein injections. The effects of KM on serum biochemical markers and liver histopathology were then evaluated 12 h after Con A exposure. The Nrf2 and NF-κB signaling pathways and alterations in gut microbiota were determined using western blotting, immunohistochemistry, and 16S rRNA sequencing to explore the underlying mechanisms of KM exposure. KM pretreatment dose-dependently decreased serum liver injury markers (Alanine aminotransferase, and aspartate aminotransferase) and cytokine levels (Tumor necrosis factor-α and interleukin-6), as well as the liver pathological damage triggered by Con A. Furthermore, the results of the multi-technique analysis indicated that KM activated the Nrf2 pathway, upregulated the expression of anti-oxidation factors HO-1 and Nrf2, and downregulated the expression of Keap1. Moreover, the NF-κB signaling pathway was inhibited. Interestingly, pre-treatment with KM also significantly improved the composition of the gut microbiota probably because it increases the richness of probiotics. Our findings suggest that KM pretreatment could attenuate Con A-induced AIH, the Nrf2 and NF-κB signaling pathways, and that gut microbiota are involved in the process of the hepatoprotective effect. This study provides a theoretical basis for the development of KM as an effective agent against AIH.
Collapse
|
13
|
Yang J, Lin YR, Xiong BJ, Chen ZH, Luo YF, Xu Y, Su YP, Huang HH, Yu CX. Regulation effect of koumine on T-helper cell polarization in rheumatoid arthritis. Eur J Pharmacol 2022; 937:175387. [DOI: 10.1016/j.ejphar.2022.175387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
|
14
|
Su Y, Shi D, Xiong B, Xu Y, Hu Q, Huang H, Yang J, Yu C. Solid-State Forms of Koumine Hydrochloride: Phase Transformations and the Crystal Structure and Properties of the Stable Form. ACS OMEGA 2022; 7:29692-29701. [PMID: 36061709 PMCID: PMC9434794 DOI: 10.1021/acsomega.2c02175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/02/2022] [Indexed: 06/15/2023]
Abstract
To investigate the solid-state forms of koumine hydrochloride (KMY), solid form screening was performed, and one amorphous form and five crystalline forms (forms A, B, C, D, and E) were identified by powder X-ray diffraction. Form A was the dominant crystal product, and its crystal structure and packing pattern were determined by single-crystal X-ray diffraction. The crystals displayed an orthorhombic crystal system and symmetry of space group P212121 with Z' = 1. The amorphous form transformed to form A at 105-120 °C or 75% RH, while forms B, C, D, and E could only be intermediate phases and readily transformed to form A at room temperature. Therefore, the phase transformations of KMY solid-state forms were established. The properties of the amorphous form and form A were further elucidated by applying vibrational spectroscopy, moisture sorption analysis, and thermal analysis. Accordingly, form A, the KMY anhydrate, was found to be the thermodynamically stable form with low hygroscopicity under ambient conditions. These characteristics are crucial in the manufacture and storage of active pharmaceutical ingredients.
Collapse
Affiliation(s)
- Yanping Su
- School
of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, People’s Republic of China
- Fujian
Key Laboratory of Drug Target Discovery and Structural and Functional
Research, School of Pharmacy, Fujian Medical
University, Fuzhou 350122, Fujian, People’s Republic of
China
| | - Dongmei Shi
- School
of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, People’s Republic of China
| | - Bojun Xiong
- School
of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, People’s Republic of China
| | - Ying Xu
- School
of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, People’s Republic of China
| | - Qing Hu
- School
of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, People’s Republic of China
- Fujian
Key Laboratory of Drug Target Discovery and Structural and Functional
Research, School of Pharmacy, Fujian Medical
University, Fuzhou 350122, Fujian, People’s Republic of
China
| | - Huihui Huang
- School
of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, People’s Republic of China
- Fujian
Key Laboratory of Drug Target Discovery and Structural and Functional
Research, School of Pharmacy, Fujian Medical
University, Fuzhou 350122, Fujian, People’s Republic of
China
| | - Jian Yang
- School
of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, People’s Republic of China
| | - Changxi Yu
- School
of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, People’s Republic of China
- Fujian
Key Laboratory of Drug Target Discovery and Structural and Functional
Research, School of Pharmacy, Fujian Medical
University, Fuzhou 350122, Fujian, People’s Republic of
China
| |
Collapse
|
15
|
Xiong B, Zhong Z, Chen C, Huang H, Lin J, Xu Y, Yang J, Yu C. The anxiolytic effect of koumine on a predatory sound stress-induced anxiety model and its associated molecular mechanisms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 103:154225. [PMID: 35689899 DOI: 10.1016/j.phymed.2022.154225] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 05/24/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Koumine is the most abundant alkaloid extracted from Gelsemium elegans Benth.. Preliminary studies by our research group have shown that koumine has significant anxiolytic effect, but this needs to be further confirmed. HYPOTHESIS/PURPOSE To investigate the potential anxiolytic effect of koumine on predatory sound (PS) stress-induced anxiety models and preliminarily explore its therapeutic targets and molecular mechanisms. STUDY DESIGN AND METHODS The anxiolytic effect of koumine in an animal model of acute PS stress-induced anxiety were determined. Then, neurosteroids levels in the main brain regions involved in anxiety disorders, as well as plasma adrenocorticotropic hormone (ACTH) and corticosterone (CORT) levels, were determinated. Finally, to clarify the effect of koumine on translocator protein 18 kDa (TSPO), the affinity between koumine and TSPO was evaluated by surface plasmon resonance (SPR) technology. RESULTS Koumine treatment mitigated anxiety-like behavior following acute PS stress in the open field test and elevated plus maze test. PS exposure significantly decreased progesterone and allopregnanolone levels in the PFC, Hip, and Amy and increased ACTH and CORT levels in plasma, and koumine administration significantly reversed these effects. Finally, the reliable SPR results showed that the KD of koumine with TSPO was 155.33 ± 11.0 μM, indicating that koumine is a human TSPO high-affinity ligand that has an affinity comparable to typical TSPO ligands. CONCLUSION Our results show that koumine has obvious anxiolytic effect in the PS-induced anxiety model. Targeting TSPO-neurosteroids-HPA axis may be an important mechanism by which koumine exerts its anxiolytic effect.
Collapse
Affiliation(s)
- Bojun Xiong
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Zhifeng Zhong
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China; Department of High Altitude Operational Medicine, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Chaojie Chen
- School of Chemical Engineering and Resource Recycling, Wuzhou University, Wuzhou 543002, China; Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou 350122, China
| | - Huihui Huang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Jinxiang Lin
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Ying Xu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Jian Yang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Changxi Yu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
16
|
Fe(III)-catalyzed regioselective and faster synthesis of isocoumarins with 3-oxoalkyl moiety at C-4: Identification of new inhibitors of PDE4. Bioorg Chem 2022; 121:105667. [DOI: 10.1016/j.bioorg.2022.105667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/24/2021] [Accepted: 02/06/2022] [Indexed: 11/29/2022]
|
17
|
Lin Y, Liu Q, Chen Z, Zheng F, Huang H, Yu C, Yang J. The immunomodulatory effect of koumine on B cells under dependent and independent responses by T cells. Eur J Pharmacol 2022; 914:174690. [PMID: 34890543 DOI: 10.1016/j.ejphar.2021.174690] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 11/03/2022]
Abstract
Dysregulated activation of polyclonal B cells and production of pathogenic antibodies are involved in the development of rheumatoid arthritis (RA). Therefore, targeted B cell therapy is effective against RA. Gelsemium elegans (Gardn. & Champ.) Benth., a toxic plant widely distributed in Southeast Asia, has been used for treating rheumatoid pain, neuropathic pain, spasticity, skin ulcers, and cancers for many years in traditional Chinese medicine. Koumine, an alkaloid monomer from Gelsemium elegans Benth., exerts therapeutic effects against RA. However, whether koumine affects B cells remains unknown. In this study, the effect of koumine on B cells under T cell-independent (TI) and T cell-dependent (TD) immune responses is investigated in vitro and in vivo. Mouse primary B cells were obtained by immunomagnetic bead sorting, and immunomodulatory effects of koumine on the activation, proliferation, and differentiation of B cells were determined in TI and TD models induced by lipopolysaccharide (LPS) and anti-CD40 antibodies in vitro, respectively. The humoral immune responses of TI and TD were established using NP-AECM-FICOLL and NP-CGG in C57BL/6J mice, respectively. We found that koumine inhibited B cell differentiation in the TI model and inhibited B cell activation and proliferation in the TD model in vitro. Koumine also inhibited antibody secretion in TI immune response, TD initial immune response, and in TD secondary immune response. Our results reveal that koumine has a direct and indirect immune regulatory effect on B cells, showing that it can directly inhibit the differentiation and secretion of autoantibodies after abnormal activation of B cells, and indirectly inhibit the activation and proliferation of TD B cells to reduce the secretion of antibodies. It may be an important mechanism for its anti-RA effect in mice, providing a rationale and laboratory data support for the application of koumine in anti-human RA therapy.
Collapse
Affiliation(s)
- Yarong Lin
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Qian Liu
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Zehong Chen
- Laboratory of Medical Function, Basic Medical Experimental Teaching Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Fengting Zheng
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Huihui Huang
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Changxi Yu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| | - Jian Yang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| |
Collapse
|
18
|
Si M, Ma Z, Zhang J, Li X, Li R, Wang C, Jia H, Luo S. Qingluoyin granules protect against adjuvant-induced arthritis in rats via downregulating the CXCL12/CXCR4-NF-κB signalling pathway. PHARMACEUTICAL BIOLOGY 2021; 59:1441-1451. [PMID: 34693865 PMCID: PMC8547818 DOI: 10.1080/13880209.2021.1991386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/02/2021] [Accepted: 10/05/2021] [Indexed: 06/13/2023]
Abstract
CONTEXT Qingluoyin (QLY) is a traditional Chinese medicine (TCM) formula which has been used in treating human rheumatoid arthritis (RA) for years in China. OBJECTIVE This study investigates the effect of QLY granules on adjuvant arthritis (AA) and the possible mechanism. MATERIALS AND METHODS Sprague-Dawley (SD) rats were injected with Complete Freund's adjuvant (CFA) to induce the AA model. After the onset of arthritis, rats received intragastric administrations of the QLY granules (1.35, 2.70, and 5.40 g/kg) or Tripterygium glycosides (TG) tablets (positive drug, 10 mg/kg) for 14 d. After 28 d immunization, the symptoms, inflammatory parameters and molecular mechanisms were investigated. RESULTS In the QLY granule (1.35, 2.70, and 5.40 g/kg) therapy groups, the arthritis index decreased to 6.30 ± 2.06, 5.80 ± 1.55, 5.30 ± 1.16 compared with the model (9.00 ± 3.01), paw swelling decreased to 1.56 ± 0.40, 1.28 ± 0.38, 1.12 ± 0.41 mL compared with the model (2.22 ± 0.73 mL). QLY granules (1.35, 2.70 and 5.40 g/kg) significantly reduced the thymus and the spleen indexes, inhibited the production of pro-inflammatory cytokines, and alleviated the pathological changes of joints compared with the model group. Furthermore, the treatment of QLY granules (2.70 and 5.40 g/kg) markedly inhibited CXCL12, CXCR4 (in spleen and synovium) and p-NF-κB p65 (in synovium) protein expression of AA rats. CONCLUSIONS QLY granules have obvious therapeutic effects on AA rats, which may be associated with downregulating the CXCL12/CXCR4-NF-κB signalling pathway. QLY granules can be used as a candidate for the treatment of RA, which deserves further study.
Collapse
Affiliation(s)
- Min Si
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Zheng Ma
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Jie Zhang
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Xinwei Li
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Rui Li
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Chao Wang
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Huiyu Jia
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Shengyong Luo
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| |
Collapse
|
19
|
Lin H, Qiu H, Cheng Y, Liu M, Chen M, Que Y, Que W. Gelsemium elegans Benth: Chemical Components, Pharmacological Effects, and Toxicity Mechanisms. Molecules 2021; 26:molecules26237145. [PMID: 34885727 PMCID: PMC8659130 DOI: 10.3390/molecules26237145] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/20/2021] [Accepted: 11/20/2021] [Indexed: 11/16/2022] Open
Abstract
Gelsemium elegans Benth (GEB), also known as heartbreak grass, is a highly poisonous plant belonging to the family Loganiaceae and genus Gelsemium that has broad application prospects in medicine. This article reviews its chemical components, pharmacological effects, toxicity mechanisms, and research progress in clinical applications in recent years. Indole alkaloids are the main active components of GEB and have a variety of pharmacological and biological functions. They have anti-tumor, anti-inflammatory, analgesic, and immunomodulation properties, with the therapeutic dose being close to the toxic dose. Application of small-dose indole alkaloids fails to work effectively, while high-dose usage is prone to poisoning, aggravating the patient’s conditions. Special caution is needed, especially to observe the changes in the disease condition of the patients in clinical practice. In-depth research on the chemical components and mechanisms of GEB is essential to the development of promising lead compounds and lays the foundation for extensive clinical application and safe usage of GEB in the future.
Collapse
Affiliation(s)
- Hailing Lin
- Department of Pharmacy, Fujian Medical University Union Hospital, 29 Xin Quan Rd, Gulou, Fuzhou 350001, China; (H.L.); (H.Q.); (Y.C.); (M.L.); (M.C.)
| | - Hongqiang Qiu
- Department of Pharmacy, Fujian Medical University Union Hospital, 29 Xin Quan Rd, Gulou, Fuzhou 350001, China; (H.L.); (H.Q.); (Y.C.); (M.L.); (M.C.)
| | - Yu Cheng
- Department of Pharmacy, Fujian Medical University Union Hospital, 29 Xin Quan Rd, Gulou, Fuzhou 350001, China; (H.L.); (H.Q.); (Y.C.); (M.L.); (M.C.)
| | - Maobai Liu
- Department of Pharmacy, Fujian Medical University Union Hospital, 29 Xin Quan Rd, Gulou, Fuzhou 350001, China; (H.L.); (H.Q.); (Y.C.); (M.L.); (M.C.)
| | - Maohua Chen
- Department of Pharmacy, Fujian Medical University Union Hospital, 29 Xin Quan Rd, Gulou, Fuzhou 350001, China; (H.L.); (H.Q.); (Y.C.); (M.L.); (M.C.)
| | - Youxiong Que
- College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Correspondence: (Y.Q.); (W.Q.)
| | - Wancai Que
- Department of Pharmacy, Fujian Medical University Union Hospital, 29 Xin Quan Rd, Gulou, Fuzhou 350001, China; (H.L.); (H.Q.); (Y.C.); (M.L.); (M.C.)
- Correspondence: (Y.Q.); (W.Q.)
| |
Collapse
|
20
|
Hu Q, Fu X, Su Y, Wang Y, Gao S, Wang X, Xu Y, Yu C. Enhanced oral bioavailability of koumine by complexation with hydroxypropyl-β-cyclodextrin: preparation, optimization, ex vivo and in vivo characterization. Drug Deliv 2021; 28:2415-2426. [PMID: 34763595 PMCID: PMC8592623 DOI: 10.1080/10717544.2021.1998248] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Koumine (KME) is an active alkaloid extracted from Gelsemium elegans, and its diverse bioactivities have been studied for decades. However, KME exhibits poor solubility and low oral bioavailability, which hampers its potential therapeutic exploitation. This work aimed to develop optimized inclusion complexes to improve the bioavailability of KME. The KME/hydroxypropyl-β-cyclodextrin (KME/HP-β-CD) inclusion complexes were prepared by the solvent evaporation method and later optimized using the Box-Behnken design. The optimal KME/HP-β-CD was characterized by scanning electron microscopy, Fourier transforms infrared spectroscopy, differential scanning calorimetry, and nuclear magnetic resonance spectroscopy. The physicochemical characterization results revealed that the crystalline state of KME was transformed into an amorphous form, forming KME/HP-β-CD inclusion complexes. Compared with KME, the solubility and in vitro release rate of KME/HP-β-CD was significantly enhanced by 52.34- and 1.3-fold, respectively. Further research was performed to investigate the intestinal absorption characteristics and in vivo bioavailability in rats. The optimal KME/HP-β-CD showed enhanced absorptive permeability and relative bioavailability increased more than two-fold compared to that of raw KME. These results indicate that the optimal KME/HP-β-CD can be used as an effective drug carrier to improve the solubility, intestinal absorption, and bioavailability of KME.
Collapse
Affiliation(s)
- Qing Hu
- School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xiaoling Fu
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Yanping Su
- School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Yanfang Wang
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Sihuan Gao
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xiaoqin Wang
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Ying Xu
- School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Changxi Yu
- School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
21
|
Majeed M, Nagabhushanam K, Lawrence L, Nallathambi R, Thiyagarajan V, Mundkur L. Boswellia serrata Extract Containing 30% 3-Acetyl-11-Keto-Boswellic Acid Attenuates Inflammatory Mediators and Preserves Extracellular Matrix in Collagen-Induced Arthritis. Front Physiol 2021; 12:735247. [PMID: 34650445 PMCID: PMC8506213 DOI: 10.3389/fphys.2021.735247] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/27/2021] [Indexed: 11/13/2022] Open
Abstract
Boswellia serrata extracts have been traditionally employed for the treatment of inflammatory diseases. In the present study, we have evaluated the mechanism of activity of Boswellin Super® FJ (BSE), a standardized extract of B. serrata containing not less than 30% 3-acetyl-11-keto-β-boswellic acid along with other β-boswellic acids. The in vitro anti-inflammatory activities were carried out in RAW 264.7 macrophages or human peripheral blood mononuclear cells stimulated with bacterial lipopolysaccharides (LPS) and treated with 1.25-5μg/ml BSE. The anti-arthritic activity of the extract was evaluated in a rat model of collagen-induced arthritis. BSE at 40 and 80mg/kg and celecoxib 10mg/kg were orally dosed for 21days. BSE showed significant (p<0.05) inhibition of inflammation (TNF-α, IL-6, nitric oxide, and COX-2 secretion) and downregulates the mRNA levels of TNF-α, IL-6, IL1-β, and inducible nitric oxide synthase in macrophages. BSE treatment reduced the levels of phosphorylated-NF-κB (P65), suggesting an anti-inflammatory activity mediated by blocking this key signal transduction pathway. In addition, BSE showed inhibition (p<0.05) of collagenase, elastase, hyaluronidase enzymes, and a reduction in reactive oxygen species and matrix-degrading proteins in RAW 264.7 macrophages stimulated with LPS. BSE treatment significantly (p<0.05) reduced the arthritic index, paw volume, and joint inflammation comparable to celecoxib in collagen-induced arthritis (CIA) in rats. The circulating anti-collagen antibodies were reduced in BSE and celecoxib-treated animals as compared to the CIA. In confirmation with in vitro data, BSE showed a significant (p<0.05) dose-dependent effect on C-reactive protein, prostaglandin E2, and erythrocyte sedimentation rate, which is widely used as a blood marker of inflammation. Further, BSE treatment suppressed the cartilage oligomeric matrix protein and significantly enhanced the hyaluronan levels in synovial fluid. As observed by collagen staining in joints, the loss of matrix proteins was lower in BSE-treated animals, suggesting that BSE could preserve the extracellular matrix in RA. The extract showed inhibition of collagenase enzyme activity in vitro, further strengthening this hypothesis. BSE treatment was found to be safe, and rats displayed no abnormal behavior or activities. The results suggest that Boswellin Super® mediates its activity by preserving matrix proteins, reducing pro-inflammatory mediators, and oxidative stress.
Collapse
Affiliation(s)
- Muhammed Majeed
- Sami-Sabinsa Group Limited, Bangalore, India
- Sabinsa Corporation, East Windsor, NJ, United States
| | | | | | | | | | | |
Collapse
|
22
|
Xiong B, Jin G, Xu Y, You W, Luo Y, Fang M, Chen B, Huang H, Yang J, Lin X, Yu C. Identification of Koumine as a Translocator Protein 18 kDa Positive Allosteric Modulator for the Treatment of Inflammatory and Neuropathic Pain. Front Pharmacol 2021; 12:692917. [PMID: 34248642 PMCID: PMC8264504 DOI: 10.3389/fphar.2021.692917] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/07/2021] [Indexed: 01/08/2023] Open
Abstract
Koumine is an alkaloid that displays notable activity against inflammatory and neuropathic pain, but its therapeutic target and molecular mechanism still need further study. Translocator protein 18 kDa (TSPO) is a vital therapeutic target for pain treatment, and recent research implies that there may be allostery in TSPO. Our previous competitive binding assay hint that koumine may function as a TSPO positive allosteric modulator (PAM). Here, for the first time, we report the pharmacological characterization of koumine as a TSPO PAM. The results imply that koumine might be a high-affinity ligand of TSPO and that it likely acts as a PAM since it could delay the dissociation of 3H-PK11195 from TSPO. Importantly, the allostery was retained in vivo, as koumine augmented Ro5-4864-mediated analgesic and anti-inflammatory effects in several acute and chronic inflammatory and neuropathic pain models. Moreover, the positive allosteric modulatory effect of koumine on TSPO was further demonstrated in cell proliferation assays in T98G human glioblastoma cells. In summary, we have identified and characterized koumine as a TSPO PAM for the treatment of inflammatory and neuropathic pain. Our data lay a solid foundation for the use of the clinical candidate koumine to treat inflammatory and neuropathic pain, further demonstrate the allostery in TSPO, and provide the first proof of principle that TSPO PAM may be a novel avenue for the discovery of analgesics.
Collapse
Affiliation(s)
- Bojun Xiong
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Guilin Jin
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Ying Xu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Wenbing You
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Yufei Luo
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Menghan Fang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Bing Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Huihui Huang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Jian Yang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xu Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Changxi Yu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
23
|
Santiago LÂM, Neto RNM, Santos Ataíde AC, Fonseca DCSC, Soares EFA, de Sá Sousa JC, Mondego-Oliveira R, Ribeiro RM, de Sousa Cartágenes MDS, Lima-Neto LG, Carvalho RC, de Sousa EM. Flavonoids, alkaloids and saponins: are these plant-derived compounds an alternative to the treatment of rheumatoid arthritis? A literature review. CLINICAL PHYTOSCIENCE 2021. [DOI: 10.1186/s40816-021-00291-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AbstractRheumatoid arthritis (RA) is a systemic inflammatory disease characterized by synovial inflammation leading to progressive joint erosion and, eventually, joint deformities. RA treatment includes anti-inflammatories, corticosteroids, synthetic disease-modifying antirheumatic drugs (DMARDs), and immunosuppressants. Drug administration is associated with adverse reactions, as gastrointestinal ulcers, cardiovascular complications, and opportunistic infections. Wherefore, different plant-derived phytochemical compounds are studied like new therapeutic approach to treatment of RA. Among the phytochemical compounds of plants for treatment of RA, flavonoids, alkaloids and saponins are related for present anti-inflammatory activity and act as physiological and metabolic regulators. They have low toxicity compared to other active plant compounds, so their therapeutic properties are widely studied. The intention of the review is to present an overview of the therapeutics of flavonoids, alkaloids, and saponins for RA. An extensive literature survey was undertaken through different online platforms:PubMed, SciELO, and Virtual Health Library databases, to identify phytochemical compounds used in RA treatment and the descriptors used were medicinal plants, herbal medicines, and rheumatoid arthritis. Seventy-five research and review articles were found to be apt for inclusion into the review. The present study summarizes the phytochemicals isolated from plants that have therapeutic effects on RA models, in vitro and in vivo. The studied substances exerted anti-inflammatory, chondroprotective, immunoregulatory, anti-angiogenic, and antioxidant activities and the most compounds possess good therapeutic properties, valuable for further research for treatment of RA.
Collapse
|
24
|
Shahen SM, Mohamed MR, Ali MRK, Samaka RM, Hamdy GM, Talaat RM. Therapeutic potential of targeted-gold nanospheres on collagen-induced arthritis in rats. Clin Exp Pharmacol Physiol 2021; 48:1346-1357. [PMID: 34060659 DOI: 10.1111/1440-1681.13531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 11/29/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that causes functional disability due to bone destruction and severe joint pain. Current anti-rheumatic treatments develop severe complications and do not provide complete remission. Gold nanoparticles (AuNPs) have garnered attention because of their unique physical and chemical properties. In this study, we have evaluated the therapeutic effects of gold nanospheres (AuNSs) with two different ligands (targeted-nanoparticles) against collagen-induced arthritis (CIA) and compared the outcomes with conventional methotrexate (MTX) and biological (infliximab) treatments. Clinical evaluation was performed by radiographic and histological examinations. The bioaccumulation of AuNSs in vital organs was assessed. The mechanistic studies targeting pro-inflammatory/anti-inflammatory and angiogenic mediators' expressions were performed. Radiographic examination showed that the targeted AuNSs reduced joint space narrowing and bone erosion. Moreover, histopathological examination of rat ankle joints demonstrated that targeted AuNSs reduce bone and cartilage degeneration/inflammation. Gold nanospheres-conjugated with nucleus localized peptide (nuclear membrane-targeted) (AuNSs@NLS) has resolved bone destruction and inflammation compared to gold nanospheres-conjugated at polyethylene glycol (AuNSs@PEG). Although the AuNSs accumulated in different organs in both cases, they did not induce any toxicity or tissue damage. The two different targeted AuNSs significantly suppress inflammatory and angiogenic mediators' expression and induced anti-inflammatory cytokine production, but the AuNSs@NLS had superior therapeutic efficacy. In conclusion, these results suggested that nuclear membrane-targeted AuNSs effectively attenuated arthritis progression without systemic side effects.
Collapse
Affiliation(s)
- Samar M Shahen
- Genetic Engineering and Biotechnology Research Institute (GEBRI), Molecular Biology Department, University of Sadat City (USC, Sadat City, Egypt.,Faculty of Science, Biochemistry Department, Ain shams University, Cairo, Egypt
| | - Mohamed R Mohamed
- Faculty of Science, Biochemistry Department, Ain shams University, Cairo, Egypt
| | - Moustafa R K Ali
- Massachusetts Institute of Technology, Biological Engineering Department, Cambridge, MA, USA
| | - Rehab M Samaka
- Faculty of Medicine, Pathology Department, Menoufia University, Shebin El Kom, Egypt
| | - Germine M Hamdy
- Faculty of Science, Biochemistry Department, Ain shams University, Cairo, Egypt
| | - Roba M Talaat
- Genetic Engineering and Biotechnology Research Institute (GEBRI), Molecular Biology Department, University of Sadat City (USC, Sadat City, Egypt
| |
Collapse
|
25
|
Luo Y, Xiong B, Liu H, Chen Z, Huang H, Yu C, Yang J. Koumine Suppresses IL-1β Secretion and Attenuates Inflammation Associated With Blocking ROS/NF-κB/NLRP3 Axis in Macrophages. Front Pharmacol 2021; 11:622074. [PMID: 33542692 PMCID: PMC7851739 DOI: 10.3389/fphar.2020.622074] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/15/2020] [Indexed: 12/21/2022] Open
Abstract
Koumine (KM), one of the primary constituents of Gelsemium elegans, has been used for the treatment of inflammatory diseases such as rheumatoid arthritis, but whether KM impacts the activation of the NOD-like receptor protein 3 (NLRP3) inflammasome remains unknown. This study aimed to explore the inhibitory effect of KM on NLRP3 inflammasome activation and the underlying mechanisms both in vitro using macrophages stimulated with LPS plus ATP, nigericin or monosodium urate (MSU) crystals and in vivo using an MSU-induced peritonitis model. We found that KM dose-dependently inhibited IL-1β secretion in macrophages after NLRP3 inflammasome activators stimulation. Furthermore, KM treatment efficiently attenuated the infiltration of neutrophils and suppressed IL-1β production in mice with MSU-induced peritonitis. These results indicated that KM inhibited NLRP3 inflammasome activation, and consistent with this finding, KM effectively inhibited caspase-1 activation, mature IL-1β secretion, NLRP3 formation and pro-IL-1β expression in LPS-primed macrophages treated with ATP, nigericin or MSU. The mechanistic study showed that, KM exerted a potent inhibitory effect on the NLRP3 priming step, which decreased the phosphorylation of IκBα and p65, the nuclear localization of p65, and the secretion of TNF-α and IL-6. Moreover, the assembly of NLRP3 was also interrupted by KM. KM blocked apoptosis-associated speck-like protein containing a CARD (ASC) speck formation and its oligomerization and hampered the NLRP3-ASC interaction. This suppression was attributed to the ability of KM to inhibit the production of reactive oxygen species (ROS). In support of this finding, the inhibitory effect of KM on ROS production was completely counteracted by H2O2, an ROS promoter. Our results provide the first indication that KM exerts an inhibitory effect on NLRP3 inflammasome activation associated with blocking the ROS/NF-κB/NLRP3 signal axis. KM might have potential clinical application in the treatment of NLRP3 inflammasome-related diseases.
Collapse
Affiliation(s)
- Yufei Luo
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Bojun Xiong
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Haiping Liu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Zehong Chen
- Experimental Teaching Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Huihui Huang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Changxi Yu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Jian Yang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
26
|
Su Y, Lu W, Fu X, Xu Y, Ye L, Yang J, Huang H, Yu C. Formulation and Pharmacokinetic Evaluation of a Drug-in-Adhesive Patch for Transdermal Delivery of Koumine. AAPS PharmSciTech 2020; 21:297. [PMID: 33099696 DOI: 10.1208/s12249-020-01793-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 08/18/2020] [Indexed: 11/30/2022] Open
Abstract
The aim of this study was to develop a suitable drug-in-adhesive patch for transdermal delivery of koumine. Acrylic polymer Duro-Tak® 87-4287, which contains hydroxyl groups, may significantly enhance the skin permeation of koumine from transdermal patches containing 0.93-3.72% koumine. Among permeation enhancers, 10% azone showed the greatest potential and increased the flux of koumine to 1.48-fold that of the control. Therefore, an optimized patch formulation containing 3.72% koumine and 10% azone in Duro-Tak® 87-4287 that offers good physical properties was selected for an in vivo pharmacokinetic study using rats. The maximal plasma drug concentration (Cmax) of koumine after transdermal administration (4 mg/patch) was 25.80 ± 1.51 ng/mL, which was in the range of those after oral administration (3 mg/kg and 15 mg/kg). The time to the maximal concentration (Tmax) and the half-life (t1/2) of the drug with transdermal administration were 3.96 ± 0.46 h and 21.10 ± 1.36 h, respectively, which were longer than those with oral administration. Furthermore, the area under the concentration-time curve (AUC0-72 h) of 898.20 ± 45.57 ng·h/mL for the transdermal patch was much higher than that for oral administration (15 mg/kg). In conclusion, the drug-in-adhesive patch containing koumine provides a steady plasma koumine level and sustained release in vivo and can be an effective means of transdermal delivery for koumine.
Collapse
|
27
|
Ye LX, Xu Y, Zhang SH, Cao DX, Chen LF, Su YP, Huang HH, Yu CX. Orally Administered Koumine Persists Longer in the Plasma of Aged Rats Than That of Adult Rats as Assessed by Ultra-Performance Liquid Chromatography-Tandem Mass Spectrometry. Front Pharmacol 2020; 11:1113. [PMID: 32792950 PMCID: PMC7385321 DOI: 10.3389/fphar.2020.01113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/08/2020] [Indexed: 12/28/2022] Open
Abstract
Aging leads to changes in nearly all pharmacokinetic phases. Koumine (KM), an alkaloid derived from Gelsemium elegans Benth., is effective against age-associated chronic diseases, but its dose proportionality following oral administration in aged individuals remains unknown. Herein, we established and validated a simple method that requires low sample volumes to determine KM concentration in rats using ultra-performance liquid chromatography-tandem mass spectrometry. The maximum plasma concentration (Cmax) of 7 mg·kg−1 KM was ~12-fold and ~24-fold higher than that of 0.28 mg·kg−1 KM in adult and aged rats, respectively (P < 0.01). Time to reach Cmax (Tmax) for 7 mg·kg−1 KM was 4-fold longer in aged rats (P < 0.05). The area under the curve (AUC) of 7 mg·kg−1 KM was >17-fold and >43-fold higher than those of 0.28 mg·kg−1 KM in adult and aged rats, respectively (P < 0.01). The half-life (t1/2) of 7 mg·kg−1 KM was over 4-fold longer than that of 0.28 mg·kg−1 KM in adult rats (P < 0.01). The t1/2 of 1.4 and 7 mg·kg−1 KM were 1.5~2-fold longer, than that of 0.28 mg·kg−1 KM in aged rats (P < 0.05). The clearance rate of 7 mg·kg−1 KM was significantly lower in aged than in adult rats (P < 0.05). For 7.0 mg·kg−1 KM, the Cmax in aged rats was higher than in adult rats during the Tmax period (P < 0.05). In aged rats, the AUC for KM was >2.5-fold higher (P < 0.05) and the t1/2 was >60% longer than in adult rats (P < 0.05). These results help interpret the pharmacokinetics of KM in aging-associated diseases.
Collapse
Affiliation(s)
- Li-Xiang Ye
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Ying Xu
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Shui-Hua Zhang
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Da-Xuan Cao
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Ling-Fan Chen
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Yan-Ping Su
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Hui-Hui Huang
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Chang-Xi Yu
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, College of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
28
|
Wu J, Yang CL, Sha YK, Wu Y, Liu ZY, Yuan ZH, Sun ZL. Koumine Alleviates Lipopolysaccharide-Induced Intestinal Barrier Dysfunction in IPEC-J2 Cells by Regulating Nrf2/NF- κB Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:127-142. [PMID: 31931594 DOI: 10.1142/s0192415x2050007x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gelsemium elegans Benth. (G. elegans), a traditional Chinese medicine, has great potential as an effective growth promoter in animals, however, the mechanism of its actin remains unclear. Here, we evaluated the protective effects of koumine extract from G. elegans against lipopolysaccharide (LPS)-induced intestinal barrier dysfunction in IPEC-J2 cells through alleviation of inflammation and oxidative stress. MTT and LDH assays revealed that koumine significantly reduced LPS cytotoxicity. Transepithelial electrical resistance (TEER) and cell monolayer permeability assays showed that koumine treatment attenuated the LPS-induced intestinal barrier dysfunction with no particularly different effects in tight junction proteins such as ZO-1, claudin-1, and occludin. LPS-triggered inflammatory response was also suppressed by koumine, as evidenced by the downregulated inflammatory factors, including TNF-α, IL-6, IL-1β, NO, iNOS, and COX-2, which was closely connected with the inhibition of NF-κB pathway for the decrease of phosphorylation of IκBα and NF-κB and nuclear translocation of p-p65. Amount of reactive oxygen species (ROS) and MDA induced by LPS was also reduced by koumine through activation of Nrf2 pathway, and increased in the levels of Nrf2 and HO-1 degradation of keap-1 to promote anti-oxidants, including superoxide dismutase (SOD) and catalase (CAT). To summarize, koumine-reduced the oxidative stress and inflammatory reaction triggered by LPS through regulation of the Nrf2/NF-κB signaling pathway and preventing intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Jing Wu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China.,Hunan Collaborative Innovation for Utilization of Botanical Function Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China
| | - Cheng-Lin Yang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China.,Hunan Collaborative Innovation for Utilization of Botanical Function Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China
| | - Yuan-Kun Sha
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China.,Hunan Collaborative Innovation for Utilization of Botanical Function Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China
| | - Yong Wu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China.,Hunan Collaborative Innovation for Utilization of Botanical Function Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China.,Hunan Engineering Research Center of Veterinary Drug, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China
| | - Zhao-Ying Liu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China.,Hunan Collaborative Innovation for Utilization of Botanical Function Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China.,Hunan Engineering Research Center of Veterinary Drug, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China
| | - Zhi-Hang Yuan
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China.,Hunan Collaborative Innovation for Utilization of Botanical Function Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China.,Hunan Engineering Research Center of Veterinary Drug, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China
| | - Zhi-Liang Sun
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China.,Hunan Collaborative Innovation for Utilization of Botanical Function Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China.,Hunan Engineering Research Center of Veterinary Drug, Hunan Agricultural University, Changsha, Hunan 410128, P. R. China
| |
Collapse
|
29
|
Gene expression profile analysis of ileum transcriptomes in pigs fed Gelsemium elegans plants. Sci Rep 2019; 9:15756. [PMID: 31673142 PMCID: PMC6823445 DOI: 10.1038/s41598-019-52374-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 10/16/2019] [Indexed: 01/18/2023] Open
Abstract
Gelsemium elegans is a flowering plant in the Loganiaceae. Because it can promote the growth of pigs and sheep, it is widely used, including in veterinary clinics, but little information is available about its biological effects. Here, we used high-throughput sequencing to characterize the differentially expressed genes (DEGs) in the ileums of pigs between a control group and a group fed Gelsemium elegans for 45 days. We found that Gelsemium elegans affected many inflammatory and immune pathways, including biological processes such as defense responses, inflammation and immune responses. Moreover, this study identified several important genes related to the anti-inflammatory activity of Gelsemium elegans (e.g., CXCL-8, IL1A, and CSF2), which will be beneficial for further study of the pharmacological mechanisms and clinical applications of Gelsemium elegans.
Collapse
|
30
|
Jin J, Ji M, Fu R, Wang M, Xue N, Xiao Q, Hu J, Wang X, Lai F, Yin D, Chen X. Sphingosine-1-Phosphate Receptor Subtype 1 (S1P1) Modulator IMMH001 Regulates Adjuvant- and Collagen-Induced Arthritis. Front Pharmacol 2019; 10:1085. [PMID: 31607926 PMCID: PMC6761374 DOI: 10.3389/fphar.2019.01085] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/26/2019] [Indexed: 01/06/2023] Open
Abstract
Sphingosine-1-phosphate receptor subtype 1 (S1P1) is essential for lymphocyte egress from lymphoid organs into systemic circulation and provides a well-defined drug target for autoimmune disorders. IMMH001, also called SYL930, is a specific S1P1/S1P4/S1P5 modulator. Here, we investigated the potential therapeutic effect of IMMH001 on rheumatoid arthritis (RA). IMMH001 rendered periphery blood lymphocytes insensitive to the egress signal from secondary lymphoid organs. Importantly, in both rat adjuvant-induced arthritis and collagen-induced arthritis models, IMMH001 treatment significantly inhibited the progression of RA and RA-associated histological changes in the joints of Sprague-Dawley rats, including hind paw swelling and arthritic index, and thus reduced the pathological score. Furthermore, IMMH001 markedly decreased proinflammatory cytokine and chemokine release from the damaged joints. These data demonstrated that IMMH001 is a promising drug candidate for RA treatment.
Collapse
Affiliation(s)
- Jing Jin
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming Ji
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rong Fu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingjin Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nina Xue
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiong Xiao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingpin Hu
- Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaojian Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fangfang Lai
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dali Yin
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoguang Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
31
|
Zhao M, Yuan X, Pei YH, Ye HY, Peng AH, Tang MH, Guo DL, Deng Y, Chen LJ. Anti-inflammatory Ellagitannins from Cleidion brevipetiolatum for the Treatment of Rheumatoid Arthritis. JOURNAL OF NATURAL PRODUCTS 2019; 82:2409-2418. [PMID: 31419126 DOI: 10.1021/acs.jnatprod.8b00984] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Six new ellagitannins, brevipetins B-G (5 and 7-11), and a new phenolic glucoside, brevipetin A (4), along with six known compounds were isolated from the traditional Chinese medicinal plant Cleidion brevipetiolatum. Their structures and absolute configurations were determined by spectroscopic analyses, chemical methods, and TD-DFT-ECD calculations. Compounds 5-11 exhibited NO inhibitory effects with IC50 values of 1.9-8.2 μM, and 9 showed the most potent inhibitory effect (IC50: 1.9 μM). An in vivo anti-inflammatory assessment of 9 showed that it exerts therapeutic effects in both the carrageenan-induced rat paw edema and collagen-induced arthritis (CIA) models at 50 mg/kg oral administration. The enhanced protein and mRNA expression levels of iNOS (inducible nitric oxide synthase) and COX-2 (cyclooxygenase-2) in LPS-stimulated RAW 264.7 cells were dose-dependently suppressed by 9. An anti-inflammatory mechanistic study revealed that 9 suppressed NF-κB activity by inhibiting IκBα phosphorylation and blocking translocation of p65 from the cytosol to the nucleus. Therefore, 9 might have the potential to be developed as a lead compound for relieving rheumatoid arthritis.
Collapse
Affiliation(s)
- Min Zhao
- Laboratory of Natural Product Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital , Sichuan University , Chengdu 610041 , People's Republic of China
| | - Xue Yuan
- Laboratory of Natural Product Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital , Sichuan University , Chengdu 610041 , People's Republic of China
| | - Ying-He Pei
- Laboratory of Natural Product Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital , Sichuan University , Chengdu 610041 , People's Republic of China
| | - Hao-Yu Ye
- Laboratory of Natural Product Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital , Sichuan University , Chengdu 610041 , People's Republic of China
| | - Ai-Hua Peng
- Laboratory of Natural Product Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital , Sichuan University , Chengdu 610041 , People's Republic of China
| | - Ming-Hai Tang
- Laboratory of Natural Product Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital , Sichuan University , Chengdu 610041 , People's Republic of China
| | - Da-Le Guo
- The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, State Key Laboratory, Breeding Base of Systematic Research Development and Utilization of Chinese Medicine Resources, School of Pharmacy , Chengdu University of Traditional Chinese Medicine , Chengdu 611137 , People's Republic of China
| | - Yun Deng
- The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, State Key Laboratory, Breeding Base of Systematic Research Development and Utilization of Chinese Medicine Resources, School of Pharmacy , Chengdu University of Traditional Chinese Medicine , Chengdu 611137 , People's Republic of China
| | - Li-Juan Chen
- Laboratory of Natural Product Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital , Sichuan University , Chengdu 610041 , People's Republic of China.,The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, State Key Laboratory, Breeding Base of Systematic Research Development and Utilization of Chinese Medicine Resources, School of Pharmacy , Chengdu University of Traditional Chinese Medicine , Chengdu 611137 , People's Republic of China
| |
Collapse
|
32
|
Wang J, Zhang J, Zhang C, Sun X, Liao X, Zheng W, Yin Q, Yang J, Mao D, Wang B, Li Q, Chen X, Ding Q, Li J, Ma B. The qualitative and quantitative analyses of Gelsemium elegans. J Pharm Biomed Anal 2019; 172:329-338. [DOI: 10.1016/j.jpba.2019.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 05/03/2019] [Accepted: 05/05/2019] [Indexed: 01/25/2023]
|
33
|
Qiu H, Yu C, Cheng Y, Que W, Zeng X, Wang H, Liu M. Simultaneous Determination of Koumine and Gelsemine in Human Plasma Using HPLC-UV Assay and Its Clinical Application. CURR PHARM ANAL 2019. [DOI: 10.2174/1573412915666190222161942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Of two main alkaloids extracted from Gelsemium, koumine was shown to be a
promising analgesic, while gelsemine proved to be deleterious. Many patients suspected to be poisoned
by Gelsemium cannot be timely diagnosed due to the lack of UPLC-MS/MS. Additionally, the
concentration of alkaloids in humans has never been reported. The aim of this study was to establish a
more economical and accessible method using HPLC-UV for diagnosis and quantitative analysis of
Gelsemium poisoning.
Methods:
Plasma spiked with an internal standard, oxcarbazepine, was prepared with solid-phase extraction.
Koumine and gelsemine were separated on a C18 column using a mobile phase consisting of
methanol, water, and di-n-butylamine (58:42:0.01) pumped at a flow rate of 1.00 mL/min. The detection
wavelength was set at 263 nm. Plasma concentrations of two different times were determined for the
patients.
Results:
The calibration curves for both monomers possessed good linearity from 0.05-50 mg/L
(r=0.9997 and 0.9999, respectively). The extraction recoveries were greater than 88.5 %. Variation for
intraday and interday assays of koumine and gelsemine were less than 8.3% and 7.7%, respectively. The
concentrations of the two alkaloids were identified in 5 patients with Gelsemium poisoning by using the
established method.
Conclusion:
The established method by using HPLC-UV is applicable for diagnosis and quantitative
analysis of Gelsemium poisoning in such cases. TDM of koumine and gelsemine in patients with Gelsemium
poisoning may provide additional information for the clinic to improve rescue strategy.
Collapse
Affiliation(s)
- Hongqiang Qiu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Changxi Yu
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Yu Cheng
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Wancai Que
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaofang Zeng
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hui Wang
- Department of Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Maobai Liu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
34
|
Yue R, Jin G, Wei S, Huang H, Su L, Zhang C, Xu Y, Yang J, Liu M, Chu Z, Yu C. Immunoregulatory Effect of Koumine on Nonalcoholic Fatty Liver Disease Rats. J Immunol Res 2019; 2019:8325102. [PMID: 30915371 PMCID: PMC6402221 DOI: 10.1155/2019/8325102] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 11/13/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common and important chronic liver disease all over the world. In the present study, we found that koumine, the main and active ingredient isolated from Gelsemium elegans, has the potential therapeutic effect on NAFLD rats by immunomodulatory activity. Koumine could significantly reduce the level of TG, TC, LDL-C, ALT, and AST in the serum of NAFLD rats and increase the level of HDL-C, reduce the liver index, and improve the adipose-like lesions of liver cells in NAFLD rats. Furthermore, treatment with koumine inhibited the severity of NAFLD. In addition, koumine-treated rats significantly increased the proportion of CD4+/CD8+ T cells and also decreased the percentages of Th1 and Th17 cells and increased Th2 and Treg cells in the liver. Moreover, koumine reduced the production and mRNA expression of proinflammatory cytokines in vivo. This result showed that koumine could effectively modulate different subtypes of helper T cells and prevent NAFLD. The present study revealed the novel immunomodulatory activity of koumine and highlighted the importance to further investigate the effects of koumine on hepatic manifestation of the metabolic syndrome.
Collapse
Affiliation(s)
- Rongcai Yue
- School of Pharmacy, Fujian Medical University, Fuzhou, 350122 Fujian, China
| | - Guilin Jin
- School of Pharmacy, Fujian Medical University, Fuzhou, 350122 Fujian, China
| | - Shanshan Wei
- Naval Medical Research Institute, Second Military Medical University, Shanghai 200433, China
| | - Huihui Huang
- School of Pharmacy, Fujian Medical University, Fuzhou, 350122 Fujian, China
| | - Li Su
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Chenxi Zhang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Ying Xu
- School of Pharmacy, Fujian Medical University, Fuzhou, 350122 Fujian, China
| | - Jian Yang
- School of Pharmacy, Fujian Medical University, Fuzhou, 350122 Fujian, China
| | - Ming Liu
- School of Pharmacy, Fujian Medical University, Fuzhou, 350122 Fujian, China
| | - Zhiyong Chu
- Naval Medical Research Institute, Second Military Medical University, Shanghai 200433, China
| | - Changxi Yu
- School of Pharmacy, Fujian Medical University, Fuzhou, 350122 Fujian, China
| |
Collapse
|
35
|
De Paolis M, Reyes Loya D, Maddaluno J. Study toward an Asymmetric and Catalytic Synthesis of Koumine. HETEROCYCLES 2019. [DOI: 10.3987/com-18-s(f)83] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
36
|
Jin GL, Yue RC, He SD, Hong LM, Xu Y, Yu CX. Koumine Decreases Astrocyte-Mediated Neuroinflammation and Enhances Autophagy, Contributing to Neuropathic Pain From Chronic Constriction Injury in Rats. Front Pharmacol 2018; 9:989. [PMID: 30214411 PMCID: PMC6125371 DOI: 10.3389/fphar.2018.00989] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/10/2018] [Indexed: 01/23/2023] Open
Abstract
Koumine, an indole alkaloid, is a major bioactive component of Gelsemium elegans. Previous studies have demonstrated that koumine has noticeable anti-inflammatory and analgesic effects in inflammatory and neuropathic pain (NP) models, but the mechanisms involved are not well understood. This study was designed to explore the analgesic effect of koumine on chronic constriction injury (CCI)-induced NP in rats and the underlying mechanisms, including astrocyte autophagy and apoptosis in the spinal cord. Rats with CCI-induced NP were used to evaluate the analgesic and anti-inflammatory effects of koumine. Lipopolysaccharide (LPS)-induced inflammation in rat primary astrocytes was also used to evaluate the anti-inflammatory effect of koumine. We found that repeated treatment with koumine significantly reduced and inhibited CCI-evoked astrocyte activation as well as the levels of pro-inflammatory cytokines. Meanwhile, we found that koumine promoted autophagy in the spinal cord of CCI rats, as reflected by decreases in the LC3-II/I ratio and P62 expression. Double immunofluorescence staining showed a high level of colocalization between LC3 and GFAP-positive glia cells, which could be decreased by koumine. Intrathecal injection of an autophagy inhibitor (chloroquine) reversed the analgesic effect of koumine, as well as the inhibitory effect of koumine on astrocyte activation in the spinal cord. In addition, TUNEL staining suggested that CCI-induced apoptosis was inhibited by koumine, and this inhibition could be abolished by chloroquine. Western blot analysis revealed that koumine significantly increased the level of Bcl-xl while inhibiting Bax expression and decreasing cleaved caspase-3. In addition, we found that koumine could decrease astrocyte-mediated neuroinflammation and enhance autophagy in primary cultured astrocytes. These results suggest that the analgesic effects of koumine on CCI-induced NP may involve inhibition of astrocyte activation and pro-inflammatory cytokine release, which may relate to the promotion of astrocyte autophagy and the inhibition for apoptosis in the spinal cord.
Collapse
Affiliation(s)
- Gui-Lin Jin
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Rong-Cai Yue
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Sai-di He
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Li-Mian Hong
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Ying Xu
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Chang-Xi Yu
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
37
|
The analgesic effect and possible mechanisms by which koumine alters type II collagen-induced arthritis in rats. J Nat Med 2018; 73:217-225. [DOI: 10.1007/s11418-018-1229-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/20/2018] [Indexed: 01/28/2023]
|
38
|
Koumine Attenuates Neuroglia Activation and Inflammatory Response to Neuropathic Pain. Neural Plast 2018; 2018:9347696. [PMID: 29770147 PMCID: PMC5889871 DOI: 10.1155/2018/9347696] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/24/2017] [Accepted: 02/13/2018] [Indexed: 01/12/2023] Open
Abstract
Despite decades of studies, the currently available drugs largely fail to control neuropathic pain. Koumine—an alkaloidal constituent derived from the medicinal plant Gelsemium elegans Benth.—has been shown to possess analgesic and anti-inflammatory properties; however, the underlying mechanisms remain unclear. In this study, we aimed to investigate the analgesic and anti-inflammatory effects and the possible underlying mechanisms of koumine. The analgesic and anti-inflammatory effects of koumine were explored by using chronic constriction injury of the sciatic nerve (CCI) neuropathic pain model in vivo and LPS-induced injury in microglia BV2 cells in vitro. Immunofluorescence staining and Western blot analysis were used to assess the modulator effect of koumine on microglia and astrocyte activation after CCI surgery. Enzyme-linked immunosorbent assay (ELISA) was used to evaluate the levels of proinflammatory cytokines. Western blot analysis and quantitative real-time polymerase chain reaction (qPCR) were used to examine the modulator effect of koumine on microglial M1 polarization. We found that single or repeated treatment of koumine can significantly reduce neuropathic pain after nerve injury. Moreover, koumine showed inhibitory effects on CCI-evoked microglia and astrocyte activation and reduced proinflammatory cytokine production in the spinal cord in rat CCI models. In BV2 cells, koumine significantly inhibited microglia M1 polarization. Furthermore, the analgesic effect of koumine was inhibited by a TSPO antagonist PK11195. These findings suggest that the analgesic effects of koumine on CCI-induced neuropathic pain may result from the inhibition of microglia activation and M1 polarization as well as the activation of astrocytes while sparing the anti-inflammatory responses to neuropathic pain.
Collapse
|
39
|
Xiong BJ, Xu Y, Jin GL, Liu M, Yang J, Yu CX. Analgesic effects and pharmacologic mechanisms of the Gelsemium alkaloid koumine on a rat model of postoperative pain. Sci Rep 2017; 7:14269. [PMID: 29079733 PMCID: PMC5660238 DOI: 10.1038/s41598-017-14714-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/13/2017] [Indexed: 02/06/2023] Open
Abstract
Postoperative pain (POP) of various durations is a common complication of surgical procedures. POP is caused by nerve damage and inflammatory responses that are difficult to treat. The neuroinflammation-glia-steroid network is known to be important in POP. It has been reported that the Gelsemium alkaloid koumine possesses analgesic, anti-inflammatory and neurosteroid modulating activities. This study was undertaken to test the analgesic effects of koumine against POP and explore the underlying pharmacologic mechanisms. Our results showed that microglia and astroglia were activated in the spinal dorsal horn post-incision, along with an increase of proinflammatory cytokines (interleukin 1β, interleukin 6, and tumor necrosis factor α). Both subcutaneous and intrathecal (i.t.) koumine treatment after incision significantly prevented mechanical allodynia and thermal hyperalgesia, inhibited microglial and astroglial activation, and suppressed expression of proinflammatory cytokines. Moreover, the analgesic effects of koumine were antagonized by i.t. administration of translocator protein (18 kDa) (TSPO) antagonist PK11195 and GABAA receptor antagonist bicuculline. Together, koumine prevented mechanical allodynia and thermal hyperalgesia caused by POP. The pharmacologic mechanism of koumine-mediated analgesia might involve inhibition of spinal neuroinflammation and activation of TSPO. These data suggested that koumine might be a potential pharmacotherapy for the management of POP.
Collapse
Affiliation(s)
- Bo-Jun Xiong
- Department of Pharmacology and College of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Ying Xu
- Department of Pharmacology and College of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
- Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Gui-Lin Jin
- Department of Pharmacology and College of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
- Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Ming Liu
- Department of Pharmacology and College of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Jian Yang
- Department of Pharmacology and College of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
- Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Chang-Xi Yu
- Department of Pharmacology and College of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China.
- Fujian Key Laboratory of Natural Medicine Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China.
| |
Collapse
|