1
|
Jin Q, Shen JS, Wu XR, Peng HZ, Fu ZH, Chen LQ, Zhao YL, Ye M, Luo XD. Antithrombotic macrocyclic sesquiterpene pyridine alkaloids from Tripterygium hypoglaucum. PHYTOCHEMISTRY 2025; 236:114516. [PMID: 40268176 DOI: 10.1016/j.phytochem.2025.114516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/20/2025] [Accepted: 04/20/2025] [Indexed: 04/25/2025]
Abstract
Six previously undescribed macrocyclic sesquiterpene pyridine alkaloids (SPAs) derivatives, named triptocumines A-F (1-6), as well as eighteen known analogs, were isolated from Tripterygium hypoglaucum. The structures were assigned based on analysis of spectroscopic data and electron circular dichroism calculations. Furthermore, compounds 1-6, 8, and 24 could effectively inhibit adenosine diphosphate-induced platelet aggregation, alleviate thrombosis and oxidative stress in zebrafish, reduce endothelin-1 level, protect endothelial cells from oxidative damage, and promote the formation of lumen structure.
Collapse
Affiliation(s)
- Qiong Jin
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Jia-Shan Shen
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Southwest United Graduate School, Kunming, 650500, PR China; Southwest United Graduate School Kunming 650092, China
| | - Xian-Run Wu
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Southwest United Graduate School, Kunming, 650500, PR China
| | - Hui-Zhen Peng
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Southwest United Graduate School, Kunming, 650500, PR China
| | - Zi-Hao Fu
- Institute of International Rivers and Eco-security, Yunnan University, Kunming 650091, China
| | - Li-Qiang Chen
- Institute of International Rivers and Eco-security, Yunnan University, Kunming 650091, China
| | - Yun-Li Zhao
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Southwest United Graduate School, Kunming, 650500, PR China.
| | - Min Ye
- Southwest United Graduate School Kunming 650092, China; State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China.
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Southwest United Graduate School, Kunming, 650500, PR China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
| |
Collapse
|
2
|
Li J, Liang H, Liu L, Gao X, Liu Y, Zhang M, Yuan X, Ren S, Zhang W. Structural diversity and biological activities of terpenoids derived from Tripterygium wilfordii Hook. f. RSC Adv 2025; 15:12594-12608. [PMID: 40264893 PMCID: PMC12012611 DOI: 10.1039/d4ra09048a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 04/05/2025] [Indexed: 04/24/2025] Open
Abstract
Terpenoids, a heterogeneous group of natural products, have garnered considerable attention in the field of drug discovery. This is attributed to their vast diversity, intricate structural features, and extensive biological activities. Tripterygium wilfordii Hook. f., a traditional medicinal plant with widespread application in East Asia, is particularly enriched in terpenoids, which can be classified into sesquiterpenoids, diterpenoids, and triterpenoids. The present review provides a comprehensive elaboration of the chemical structures and biological activities of 217 terpenoids isolated from T. wilfordii. The purpose is to shed light on their potential in pharmacological research and to stimulate innovative drug discovery as well as clinical applications. These terpenoids display a broad spectrum of biological activities, such as antitumor, anti - inflammatory, immunosuppressive, and other therapeutic effects. Nevertheless, their clinical application is impeded by issues related to toxicity and poor bioavailability. Future research efforts should be concentrated on exploring effective strategies to alleviate toxicity and enhance drug delivery systems. In addition, in - depth investigation into the structure-activity relationships and the identification of new active constituents are crucial for the development of more potent and safer drugs. This review serves as an exhaustive reference for the discovery and development of novel drugs based on the natural active products of T. wilfordii, providing valuable insights and guidance for researchers in the relevant field.
Collapse
Affiliation(s)
- Jiping Li
- School of Public Health, Qiqihar Medical University Qiqihar 161006 China
| | - Hong Liang
- School of Public Health, Qiqihar Medical University Qiqihar 161006 China
| | - Likun Liu
- Research Institute of Medicine of Pharmacy, Qiqihar Medical University Qiqihar 161006 China
| | - Xiuli Gao
- Research Institute of Medicine of Pharmacy, Qiqihar Medical University Qiqihar 161006 China
| | - Yang Liu
- Office of Academic Research, Qiqihar Medical University Qiqihar 161006 China
| | - Meng Zhang
- Office of Academic Research, Qiqihar Medical University Qiqihar 161006 China
| | - Xiaoan Yuan
- Research Institute of Medicine of Pharmacy, Qiqihar Medical University Qiqihar 161006 China
| | - Shan Ren
- Research Institute of Medicine of Pharmacy, Qiqihar Medical University Qiqihar 161006 China
| | - Wei Zhang
- Office of Academic Research, Qiqihar Medical University Qiqihar 161006 China
| |
Collapse
|
3
|
Wang H, Zou J, Li Y, Liu J, Guo F. Tripterygium glycoside tablets and triptolide alleviate experimental autoimmune encephalomyelitis mice involving the PACAP/cAMP signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119748. [PMID: 40188893 DOI: 10.1016/j.jep.2025.119748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygiumwilfordii, a traditional Chinese herbal medicine, has been used for treating autoimmune diseases, including rheumatoid arthritis and systemic lupus erythematosus. Tripterygium glycoside tablets (TGT), derived from this herb, is widely used in clinical practice in China. However, the therapeutic effects of TGT on Multiple sclerosis (MS), particularly through its active component triptolide (TP), remain insufficiently understood. AIM OF THE STUDY This study aimed to investigate the therapeutic effects of TGT and TP on experimental autoimmune encephalomyelitis (EAE) and elucidate the underlying molecular mechanisms. MATERIALS AND METHODS TGT and TWPT were chemically characterized using liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. The therapeutic effect of TGT, TWPT, and TP was evaluated in the EAE model. Proteomics analysis and Western blot analysis were validated the signaling pathways. RESULTS TGT and TP significantly alleviated EAE symptoms in mice, including reduced weight loss and neurological deficits, whereas TWPT (TGT without triptolide) shows no significant therapeutic effect. Histological analysis revealed that TGT and TP reduced demyelination and inflammatory cell infiltration in the spinal cord. TGT and TP decreased systemic inflammatory cytokines (IL-17A, IFN-γ, TNF-α, and IL-6) and the mRNA expression of the transcription factors T-bet and ROR-γt in the spinal cord. Proteomic analysis indicated that TP significantly upregulated the expression of PACAP and activated the cAMP signaling pathway. Furthermore, TGT and TP modulate PKA, PI3K-AKT, NF-κB, and apoptosis-related signaling pathways, contributing to the reducing inflammation, apoptosis and demyelination in EAE mice. CONCLUSION TGT and TP exert anti-inflammatory and demyelination-improving effects to alleviate both clinical and pathological manifestations of EAE in mice via the PACAP/cAMP signaling axis, suggesting TGT as promising therapeutic strategies for MS.
Collapse
Affiliation(s)
- Hong Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China
| | - Juan Zou
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China
| | - Yiming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China
| | - Jingwen Liu
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China.
| | - Fujiang Guo
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China; Shanghai Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, PR China.
| |
Collapse
|
4
|
Zhong X, Deng X, Yang Y, Xie X, Li B, Peng X. Immuno-engineered macrophage membrane-coated nanodrug to restore immune balance for rheumatoid arthritis treatment. Acta Biomater 2025:S1742-7061(25)00197-7. [PMID: 40097125 DOI: 10.1016/j.actbio.2025.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/02/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
Current immunosuppressive therapies for rheumatoid arthritis (RA) lack disease specificity, primarily targeting inflammation while causing debilitating side effects. To address this limitation, we developed a biomimetic nanodrug MP@NEs/CT to induce antigen-specific immune tolerance for precise, effective and safe RA immunotherapy. MP@NEs/CT features a core of multiepitope citrullinated peptide (CitP) and triptolide (TPL) co-loaded nanoemulsion and coated with a macrophage membrane harvested from IFN-γ treated RAW264.7 cells. CitP, an RA autoantigen, specifically targets the immune response, while TPL acts as an immunosuppressant by inhibiting dendritic cells (DCs) maturation. IFN-γ treatment upregulates programmed death-ligand 1 (PD-L1) expression, facilitating MP@NEs/CT accumulation within inflamed tissues via programmed death-1 (PD-1) binding following intravenous administration. Additionally, the immune-engineered macrophage membrane sequesters proinflammatory cytokines, further dampening local inflammation. A significant reduction of CII-specific IgG levels in collagen-induced arthritis (CIA) mice model provides the evidence of CitP in restoring antigen-specific immune tolerance. Importantly, a low dose of TPL within MP@NEs/CT promotes tolerogenic DCs and generation of anti-inflammatory cytokines, ultimately leading to upregulation of antigen-specific regulatory T cells (Tregs) and B cells (Bregs) and a reduction in pro-inflammatory cytokine levels. Consequently, the nanodrug demonstrates synergistic and effective anti-inflammatory and immunosuppressive effects, alleviating autoimmune damage in a CIA mice model. STATEMENT OF SIGNIFICANCE: : Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by dysregulated immune responses, leading to synovial hyperplasia, tissue destruction, and irreversible disability. Early work in RA therapy mainly applying anti-inflammatory drugs which focuses on delaying joint deformity, but have no effects on the aberrant immune response. However, these drugs often require high doses and long-term administration, leading to potential adverse effects. In this work, we reported a therapeutic system that co-delivery of autoantigens with immune modulators promotes antigen-specific tolerance for effective and safe RA immunotherapy.
Collapse
Affiliation(s)
- Xiaofang Zhong
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China
| | - Xiaoyu Deng
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China
| | - Yongqing Yang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China
| | - Xin Xie
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China
| | - Bowen Li
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 117585, Singapore.
| | - Xinsheng Peng
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China.
| |
Collapse
|
5
|
Zhang X, Zhang J, Xun G, Gao Y, Zhao J, Fu Y, Su S, Kong D, Wang Q, Wang X. Alleviation effect of macrophage depletion on hepatotoxicity of triptolide: A new insight based on metabolomics and proteomics. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119485. [PMID: 39947369 DOI: 10.1016/j.jep.2025.119485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/14/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Triptolide (TP) is an abietane-type diterpenoid isolated from the traditional Chinese herb Tripterygium wilfordii Hook. F, which is used to relieve rheumatism, alleviate joint pain and swelling, and promote blood circulation for more than 600 years in China. The most common preparations containing TP from Tripterygium wilfordii Hook F, which are Tripterygium tablets and Tripterygium glycoside tablets, are widely used in clinical for treating rheumatoid arthritis and other autoimmune diseases at present. However, the clinical application is hindered by severe systemic toxicity induced by TP, especially hepatotoxicity. It is crucial to discover potent and specific detoxification strategy for TP. AIM OF STUDY According to our previous study, TP-induced hepatotoxicity is primarily related to macrophages. This study aimed to investigate the alleviation effects of macrophage depletion on the TP-induced liver injury in mice and to explore the related mechanisms by integration of metabolomics and proteomics. MATERIALS AND METHODS Mice were treated with clodronate liposomes to deplete macrophage before administration of triptolide. The alleviation effects were evaluated by biochemical analysis of serum and histopathology observation of the hepatic tissues. Metabolomics and proteomics were carried out to explore the mechanism of macrophage depletion on triptolide-induced liver injury. The levels of mRNA and protein of TLR4- MyD88-NF-κB axis were further detected. RESULTS The altered levels of biochemistry indicators, including aminotransferase (ALT) and aspartate aminotransferase (AST), albumin (ALB), and γ-glutamyltranspeptidase (GGT) were significantly recovered, and histopathological liver injury also showed restoring tendency in mice with macrophage depletion compared to mice with TP-treatment. The inflammation indicator interleukin-6 (IL-6) and interleukin-1β (IL-1β) were recovered significantly after depletion of macrophage. Results of metabolomics and proteomics demonstrated that macrophage depletion exerted protective effects on triptolide-induced liver injury by regulating 85 metabolites and 202 proteins. Joint analysis of multi-omics data suggested macrophage depletion could regulate lipid metabolism and maintain inflammatory homeostasis. The increased expression of NF-κB, TLR4, and MyD88 were decreased after depletion of macrophage. CONCLUSION TP-induced hepatotoxicity is mainly associated with dysfunction of macrophages and imbalance of inflammatory homeostasis. The findings of this study may help facilitate the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoguang Zhang
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Jia Zhang
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Ge Xun
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Yanhua Gao
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, PR China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Jie Zhao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, PR China
| | - Yan Fu
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, Hebei, PR China; School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Suwen Su
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Dezhi Kong
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Qiao Wang
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, PR China.
| | - Xu Wang
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, PR China.
| |
Collapse
|
6
|
Niu Z, Zhang H, Cai C, Yang T, Ma T, Xu D, Cui D, Tang Y. The mechanisms of tripterygium glycosides-induced reproductive toxicity and detoxification strategies. Reprod Toxicol 2025; 132:108830. [PMID: 39778665 DOI: 10.1016/j.reprotox.2025.108830] [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: 10/26/2024] [Revised: 12/15/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
Tripterygium glycosides (TG) is a widely used preparation in the treatment of rheumatoid arthritis (RA), nephrotic syndrome and diabetic nephropathy. Although the clinical efficacy is definite, the side-effects on reproductive system limit its wide application. It is of great significance to take measures to alleviate its reproductive toxicity and expand its clinical use. The mechanism of TG-induced reproductive toxicity involves oxidative stress, inflammation, apoptosis, and metabolism imbalance, which lead to adverse effects on male and female reproductive organs. To mitigate these effects, detoxification strategies including combining TG with other agents have been proved to counteract its toxicity. This review will provide information for the studies of TG-induced reproductive toxicity, and also provide insights for developing novel strategies to alleviate the reproductive side effects of TG.
Collapse
Affiliation(s)
- Zechen Niu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province 712046, China
| | - Huanhuan Zhang
- Shaanxi University of International Trade & Commerce, Xianyang, Shaanxi 712046, China
| | - Chunzhou Cai
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province 712046, China
| | - Ting Yang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province 712046, China
| | - Tian Ma
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province 712046, China
| | - Dingqiao Xu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province 712046, China
| | - Dongxiao Cui
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province 712046, China.
| | - Yuping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province 712046, China.
| |
Collapse
|
7
|
Zhu BY, Liu ZC, Zhao ZX, Huang HP, Zhang N, Xia J, Chen WW. Pharmacological Mechanism of Chinese Medicine in Systemic Lupus Erythematosus: A Narrative Review. Chin J Integr Med 2025; 31:157-169. [PMID: 39240290 DOI: 10.1007/s11655-024-3762-0] [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] [Accepted: 06/24/2024] [Indexed: 09/07/2024]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder affecting multiple systems, characterized by the development of harmful autoantibodies and immune complexes that lead to damage in organs and tissues. Chinese medicine (CM) plays a role in mitigating complications, enhancing treatment effectiveness, and reducing toxicity of concurrent medications, and ensuring a safe pregnancy. However, CM mainly solves the disease comprehensively through multi-target and multi-channel regulation process, therefore, its treatment mechanism is often complicated, involving many molecular links. This review introduces the research progress of pathogenesis of SLE from the aspects of genetics, epigenetics, innate immunity and acquired immunity, and then discusses the molecular mechanism and target of single Chinese herbal medicine and prescription that are commonly used and effective in clinic to treat SLE.
Collapse
Affiliation(s)
- Bo-Yu Zhu
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Zhi-Chao Liu
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Zhen-Xi Zhao
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Hui-Ping Huang
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Na Zhang
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Jia Xia
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Wei-Wei Chen
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| |
Collapse
|
8
|
Zhou GL, Su SL, Yu L, Shang EX, Hua YQ, Yu H, Duan JA. Exploring the liver toxicity mechanism of Tripterygium wilfordii extract based on metabolomics, network pharmacological analysis and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118888. [PMID: 39368758 DOI: 10.1016/j.jep.2024.118888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/06/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygii wilfordii Radix, (TW) as a toxic herbal medicine, is the root of Tripterygium wilfordii Hook. F. , which commonly used in China for the treatment of rheumatoid arthritis and autoimmune diseases, but its severe toxicity, particularly hepatotoxicity, significantly impacts its clinical application. AIM OF THE STUDY The hepatotoxicity and its molecular mechanism of 70% TW ethanol extract (TWE) on male mice were demonstrated based on metabolomics, network pharmacological analysis and experimental validation. MATERIALS AND METHODS The toxic and bioactive ingredients in TWE were quantitative analyzed by Triple quadrupole (TQ) mass spectrometry method. The liver organ index, as well as the liver function indexes AST and ALT were evaluated after administering different doses of TWE for 24 h, and a pathological change was analyzed in liver tissue. Non-targeted metabolomics using UPLC-QTOF/MS was performed on both the plasma and liver tissue samples in combination with network toxicology to screen for key targets related to TWE toxicity in the liver. These key targets including caspase 3, NF-κB, TLR4, TNF-α, NQO1, and Bcl2 were subsequently verified through Western blotting experiments. RESULTS The six toxic and active ingredients of raphenolactone, ranolactone, triptolide tripterine, wilforlide A, demethylzeylasterain in TWE for the contents of 0.709, 1.408, 0.353, 0.354, 0.882, 0.227 mg g-1, respectively. Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels increased and liver index decreased after administration of TWE for 24 h. Pathological analysis showed that TWE could produce toxicity to mouse liver, and its toxicity was dose-dependent. In the high-dose group, TW-D (11.23 g/kg) and TW-E (22.46 g/kg) caused a large amount of rupture in mouse liver nucleus and a large amount of inflammatory infiltration at the same time. Furthermore, 64 metabolites in plasma and 59 metabolites in the liver tissue were identified. The main metabolic pathways involved glycerol phospholipid metabolism, glycosylphosphatidylinositol-ether lipid metabolism, fatty acid metabolism, sphingomyelin metabolism, and ether lipid metabolism in plasma and liver tissue. Through analysis of the top 10 correlated targets, 6 out of the top 10 selected target proteins exhibited consistent expression patterns with liver injury. The levels of Bcl2 and NQO1 decreased with increasing exposure dose. The expression of Caspase 3, NF-κB, TLR4, and TNF-α increased with increasing dose. These findings suggest that protein expression has a regulatory effect at different doses groups compared to the control group.These findings suggest a regulatory effect of protein expression in different dose groups compared to the control group. CONCLUSION The hepatotoxic effects of TWE can increase ALT and AST levels in plasma, leading to hepatic oxidative damage and inflammatory response. The toxic mechanisms that produce are closely related to the regulating of the abnormal metabolites in plasma and liver tissue. Furthermore, the regulating the expression levels of targeted proteins of TNF-α, NF-κB, Caspase 3, NQO1, and Bcl2 were confirmed by examining the liver tissue. These data clearly elucidate the toxicity mechanism of TW, laying the foundation for ensuring the quality and safety of drugs.
Collapse
Affiliation(s)
- Guo-Liang Zhou
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Pharmacy, School of Life and Health Sciences, Anhui Science and Technology University, Bengbu, 233100, China
| | - Shu-Lan Su
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Li Yu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Er-Xin Shang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yong-Qing Hua
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hao Yu
- Department of Pharmacy, School of Life and Health Sciences, Anhui Science and Technology University, Bengbu, 233100, China
| | - Jin-Ao Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
9
|
Qiang L, Lee SH, Xiao P, Chunhui L, Lei G, Shaoli C, Tingjie Y, Guangli D, Wei X, Guofu Z. Novel detoxifier of spironolactone against triptolide-induced hepatotoxicity through inhibition of RPB1 degradation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118722. [PMID: 39182704 DOI: 10.1016/j.jep.2024.118722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Triptolide is a major bioactive and toxic ingredient isolated from the traditional Chinese herb Tripterygium wilfordii (T. wilfordii) Hook F. It exhibits potent antitumor, immunosuppressive, and anti-inflammatory biological activities; however, its clinical application is hindered by severe systemic toxicity. Two preparations of T. wilfordii, including T. wilfordii glycoside tablets and T. wilfordii tablets, containing triptolide, are commonly used in clinical practice. However, their adverse side effects, particularly hepatotoxicity, limit their safe use. Therefore, it is crucial to discover potent and specific detoxification medicines for triptolide. AIM OF THE STUDY This study aimed to investigate the detoxification effects and potential mechanism of action of spironolactone on triptolide-induced hepatotoxicity to provide a potential detoxifying strategy for triptolide, thereby promoting the safe applications of T. wilfordii preparations in clinical settings. MATERIALS AND METHODS Cell viability was assessed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and crystal violet staining. Nuclear fragmentation was visualized using 4',6-diamidino-2-phenylindole (DAPI) staining, and protein expression was analyzed by Western blotting. The inhibitory effect of spironolactone on triptolide-induced hepatotoxicity was evaluated by examining the effects of spironolactone on serum alanine aminotransferase and aspartate aminotransferase levels, as well as liver pathology in a mouse model of triptolide-induced acute hepatotoxicity. Furthermore, a survival assay was performed to investigate the effects of spironolactone on the survival rate of mice exposed to a lethal dose of triptolide. The effect of spironolactone on triptolide-induced global transcriptional repression was assessed through 5-ethynyl uridine staining. RESULTS Triptolide treatment decreased the cell viability, increased the nuclear fragmentation and the cleaved caspase-3 levels in both hepatoma cells and hepatocytes. It also increased the alanine aminotransferase and aspartate aminotransferase levels, induced the hepatocyte swelling and necrosis, and led to seven deaths out of 11 mice. The above effects could be mitigated by pretreatment with spironolactone. Additionally, molecular mechanism exploration unveiled that spironolactone inhibited triptolide-induced DNA-directed RNA polymerase II subunit RPB1 degradation, consequently increased the fluorescence intensity of 5-ethynyl uridine staining for nascent RNA. CONCLUSIONS This study shows that spironolactone exhibits a potent detoxification role against triptolide hepatotoxicity, through inhibition of RPB1 degradation induced by triptolide and, in turn, retardation of global transcriptional inhibition in affected cells. These findings suggest a potential detoxification strategy for triptolide that may contribute to the safe use of T. wilfordii preparations.
Collapse
Affiliation(s)
- Li Qiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Sau Har Lee
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia.
| | - Peng Xiao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Li Chunhui
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Guo Lei
- Taizhou Hospital of Traditional Chinese Medicine, Taizhou, China.
| | - Chen Shaoli
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Ye Tingjie
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Du Guangli
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xu Wei
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zhu Guofu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
10
|
Cheng L, Rong X. Emodin promotes the recovery of rheumatoid arthritis by regulating the crosstalk between macrophage subsets and synovial fibroblast subsets. Animal Model Exp Med 2025; 8:44-56. [PMID: 38369605 PMCID: PMC11798747 DOI: 10.1002/ame2.12387] [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: 09/25/2023] [Accepted: 01/05/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND To study the relationships among emodin, synovial fibroblasts (FLSs), and macrophages (STMs) to provide guidance for the use of emodin in rheumatoid arthritis (RA) treatment. METHODS RA clinical samples from patients with different pathological processes were collected, and the correlations between the subsets of FLSs and STMs and pathological processes were analyzed via flow cytometry. In vitro experimental methods such as enzyme linked immunosorbent assay (ELISA), Western blotting, Transwell assays, CCK-8 assays and cell coculture were used to assess cell proliferation, migration and secretion of inflammatory factors. A collagen-induced arthritis mouse model was constructed to investigate the therapeutic potential of emodin in RA by flow cytometry, micro-CT and staining. RESULTS Unique subsets of FLSs and STMs, namely, FAPα+THY1- FLSs, FAPα+THY1+ FLSs, and MerTKposTREM2high STMs, were identified in synovial tissues from RA patients. The number of MerTKposTREM2high STMs was negatively correlated with the degree of damage in RA, while the number of FAPα+THY1- FLSs was positively correlated with damage. On the one hand, emodin promoted the aggregation of MerTKposTREM2high STMs. Moreover, MerTKposTREM2high STM-mediated secretion of exosomes was promoted, which can inhibit the secretion of pro-inflammatory factors by FAPα+THY1+ FLSs and promote the secretion of anti-inflammatory factors by FAPα+THY1+ FLSs, thereby inhibiting FAPα+THY1-FLS proliferation and migration, improving the local immune microenvironment, and inhibiting RA damage. CONCLUSION Emodin was shown to regulate the aggregation of STM subsets and exosome secretion, affecting the secretion, proliferation and migration of inflammatory factors in FLS subsets, and ultimately achieving good therapeutic efficacy in RA patients, suggesting that it has important clinical value.
Collapse
Affiliation(s)
- Lianying Cheng
- Department of Integrated Traditional Chinese and Western MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaofeng Rong
- Department of Integrated Traditional Chinese and Western MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
11
|
Pu X, Ye Q. Triptophenolide Improves Rheumatoid Arthritis and Progression by Inducing Macrophage Toxicity. J Biochem Mol Toxicol 2025; 39:e70096. [PMID: 39722460 DOI: 10.1002/jbt.70096] [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: 08/01/2024] [Revised: 10/15/2024] [Accepted: 12/08/2024] [Indexed: 12/28/2024]
Abstract
To investigate the role and mechanism of triptophenolide (TRI) in resisting rheumatoid arthritis (RA). Network pharmacology analysis results suggested that TRI was related to multiple inflammation-related signaling proteins, and possessed the stable structural configuration. In animal experiments, TRI suppressed RA in mice, inhibited tissue inflammation, and improved synovial injury. Moreover, TRI can suppress RA via multiple signaling pathways, and inhibiting pyroptosis is one of the feasible treatments for improving RA.
Collapse
Affiliation(s)
- Xiuxiu Pu
- Rheumatology and Immunology Department, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
- Jiaxing University Graduate Joint Training Center, Zhejiang Chinese Medical University, Jiaxing, China
| | - Qiao Ye
- Rheumatology and Immunology Department, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
12
|
Shen P, Zhang L, Jiang X, Yu B, Zhang J. Targeting HMGB1 and Its Interaction with Receptors: Challenges and Future Directions. J Med Chem 2024; 67:21671-21694. [PMID: 39648929 DOI: 10.1021/acs.jmedchem.4c01912] [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: 12/10/2024]
Abstract
High mobility group box 1 (HMGB1) is a nonhistone chromatin protein predominantly located in the nucleus. However, under pathological conditions, HMGB1 can translocate from the nucleus to the cytoplasm and subsequently be released into the extracellular space through both active secretion and passive release mechanisms. The distinct cellular locations of HMGB1 facilitate its interaction with various endogenous and exogenous factors, allowing it to perform diverse functions across a range of diseases. This Perspective provides a comprehensive overview of the structure, release mechanisms, and multifaceted roles of HMGB1 in disease contexts. Furthermore, it introduces the development of both small molecule and macromolecule inhibitors targeting HMGB1 and its interaction with receptors. A detailed analysis of the predicted pockets is also presented, aiming to establish a foundation for the future design and development of HMGB1 inhibitors.
Collapse
Affiliation(s)
- Pingping Shen
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Libang Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xuewa Jiang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Jian Zhang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, P. R. China
| |
Collapse
|
13
|
Guo L, Yang Y, Ma J, Xiao M, Cao R, Xi Y, Li T, Huang T, Yan M. Triptolide induces hepatotoxicity by promoting ferroptosis through Nrf2 degradation. Cell Biol Toxicol 2024; 40:94. [PMID: 39503881 PMCID: PMC11541276 DOI: 10.1007/s10565-024-09930-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/02/2024] [Indexed: 11/09/2024]
Abstract
BACKGROUND Triptolide (TP), a principal active substance from Tripterygium wilfordii, exhibits various pharmacological effects. However, its potential hepatotoxicity has always been a significant concern in clinical applications. PURPOSE This research aimed to explore the involvement of ferroptosis in TP-mediated hepatic injury and the underlying mechanisms. METHODS In this study, in vitro and in vivo experiments were involved. Hepatocyte damage caused by TP was evaluated using MTT assays, liver enzyme measurement and H&E staining technique. Ferroptosis was assessed by measuring iron level, lipid peroxide, glutathione (GSH), mitochondrial morphology and the key protein/mRNA expression implicated in ferroptosis. To verify the contribution of ferroptosis to TP-induced liver damage, the ferroptosis inhibitor Ferrostatin-1 (Fer-1) and a plasmid for overexpressing glutathione peroxidase 4 (GPX4) were employed. Subsequently, nuclear factor erythroid 2-related factor 2 (Nrf2) knockout mice and Nrf2 overexpression plasmid were utilized to investigate the underlying mechanisms. Nontargeted lipidomics was used to analyze lipid metabolism in mouse liver. Moreover, the cellular thermal shift assay (CETSA), cycloheximide (CHX) and MG132 treatments, and immunoprecipitation (IP) assays were applied to validate the binding of TP to Nrf2 and their interactions. RESULTS TP triggered ferroptosis in hepatocytes, as indicated by iron accumulation and lipid peroxidation. Ferroptosis was responsible for TP-induced hepatic injury. During the process of TP-induced liver damage, the Nrf2 signaling pathway was significantly suppressed. Notably, the deletion of Nrf2 in mice aggravated the extent of liver injury and ferroptosis associated with TP, whereas enhancing Nrf2 expression in cells significantly reduced TP-induced ferroptosis. Additionally, dysregulation of lipid metabolism was associated with TP-induced liver injury. TP may directly bind to Nrf2 and enhance its degradation through the ubiquitin-proteasome pathway, thereby inhibiting or reducing Nrf2 expression. CONCLUSION In summary, the suppression of Nrf2 by TP facilitated the occurrence of ferroptosis, resulting in liver damage.
Collapse
Affiliation(s)
- Lin Guo
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yan Yang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Department of Pharmacy, Wuzhou Gongren Hospital, Wuzhou, 543000, China
| | - Jiating Ma
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Mingxuan Xiao
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Rong Cao
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410011, China
| | - Yang Xi
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Tao Li
- Department of Pharmacy, Wuzhou Gongren Hospital, Wuzhou, 543000, China
| | - Tianlong Huang
- Department of Orthopaedic Surgery, the Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Miao Yan
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
14
|
Liu Y, Qu Y, Liu C, Zhang D, Xu B, Wan Y, Jiang P. Neutrophil extracellular traps: Potential targets for the treatment of rheumatoid arthritis with traditional Chinese medicine and natural products. Phytother Res 2024; 38:5067-5087. [PMID: 39105461 DOI: 10.1002/ptr.8311] [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: 02/27/2024] [Revised: 07/06/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease. Abnormal formation of neutrophil extracellular traps (NETs) at the synovial membrane leads to the release of many inflammatory cytokines, including IL-1β, IL-6, and TNF-α. Elastase, histone H3, and myeloperoxidase, which are carried by NETs, damage the soft tissues of the joints and aggravate the progression of RA. The balance of NET formation coordinates the pro-inflammatory and anti-inflammatory effects and plays a key role in the development of RA. Therefore, when NETs are used as effector targets, highly targeted drugs with fewer side effects can be developed to treat RA without damaging the host immune system. Currently, an increasing number of studies have shown that traditional Chinese medicines and natural products can regulate the formation of NETs through multiple pathways to counteract RA, which shows great potential for the treatment of RA and has a promising future for clinical application. In this article, we review the latest biological progress in understanding NET formation, the mechanism of NETs in RA, and the potential targets or pathways related to the modulation of NET formation by Chinese medicines and natural products. This review provides a relevant basis for the use of Chinese medicines and natural products as natural adjuvants in the treatment of RA.
Collapse
Affiliation(s)
- Yuan Liu
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Yuan Qu
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Chuanguo Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Zhang
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bing Xu
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yakun Wan
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ping Jiang
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
15
|
Shen P, Deng X, Chen Z, Chen M, Han L, Chen X, Tu S. Demethylzeylasteral ameliorates podocyte damage in murine lupus by inhibiting inflammation and enhancing autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155966. [PMID: 39241387 DOI: 10.1016/j.phymed.2024.155966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 06/07/2024] [Accepted: 08/15/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with multiorgan and tissue involvement. Lupus nephritis (LN), an inflammatory condition of the kidneys associated with SLE, represents a significant cause of morbidity and mortality in SLE patients. Current immunosuppressive therapies for LN have limited efficacy and can lead to significant side effects. Demethylzeylasteral (DML) has shown promise in the treatment of LN, but its precise mechanism of action remains unclear. PURPOSE To assess the therapeutic effects and potential molecular mechanisms of DML in LN METHODS: The study evaluated the renal protective effects of DML in MRL/lpr mice through assessments of immune complex levels, renal function, and pathological changes. Network pharmacology and transcriptomics approaches were used to elucidate the underlying mechanisms. Molecular docking, biacore assay, monoclonal antibody blocking experiments, and in vitro studies were conducted to verify the mechanisms of action. RESULTS DML treatment reduced levels of anti-Sm and anti-dsDNA IgG antibodies, as well as serum creatinine and blood urea nitrogen levels. DML also mitigated glomerular damage and fibrosis. Mechanistically, DML alleviated podocyte damage by suppressing inflammation and enhancing autophagy through inhibition of the IL-17A/JAK2-STAT3 pathways. Additionally, DML exhibited high binding affinity with IL17A, JAK2, and STAT3. CONCLUSION These findings provide strong evidence for the beneficial effects of DML in LN, suggesting its potential as a novel therapeutic strategy for improving renal function in autoimmune kidney diseases.
Collapse
Affiliation(s)
- Pan Shen
- Department of Dermatology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology; Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University
| | - Xuan Deng
- Department of Nephrology, Zhongnan Hospital, Wuhan University
| | - Zhe Chen
- Department of Integrated Chinese Traditional and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology
| | - Min Chen
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University
| | - Liang Han
- Department of Integrated Chinese Traditional and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology.
| | - Xiaoqi Chen
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University.
| | - Shenghao Tu
- Department of Integrated Chinese Traditional and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology.
| |
Collapse
|
16
|
Zhang S, Hou B, Xu A, Wen Y, Zhu X, Cai W, Han Z, Chen J, Nhamdriel T, Mi M, Qiu L, Sun H. Ganlu formula ethyl acetate extract (GLEE) blocked the development of experimental arthritis by inhibiting NLRP3 activation and reducing M1 type macrophage polarization. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118377. [PMID: 38782307 DOI: 10.1016/j.jep.2024.118377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Tibetan medicine Ganlu Formula, as a classic prescription, is widely used across the Qinghai-Tibet Plateau area of China, which has a significant effect on relieving the course of rheumatoid arthritis (RA). However, the active compounds and underlying mechanisms of Ganlu Formula in RA treatment remain largely unexplored. AIM OF THE STUDY This study aimed to elucidate the active substances and potential mechanisms of the ethyl acetate extract of Ganlu Formula ethyl acetate extract (GLEE) in the treatment of RA. MATERIALS AND METHODS Ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS) was utilized to analyze and identify the chemical constituents within GLEE. Discovery Studio molecular virtual docking technology was utilized to dock the interaction of GLEE with inflammation-related pathway proteins. The GLEE gene library was obtained by transcriptome sequencing. Collagen-induced arthritic(CIA) rats were utilized to assess the antiarthritic efficacy of GLEE. Micro-CT imaging was employed to visualize the rat paw, and ultrasound imaging revealed knee joint effusion. Evaluation of synovial tissue pathological changes was conducted through hematoxylin-eosin staining and saffranine solid green staining, while immunohistochemical staining was employed to assess NLRP3 expression along with inflammatory markers. Immunofluorescence staining was utilized to identify M1 macrophages. RESULTS Metabolomic analysis via UPLC-Q-TOF-MS identified 28 potentially bioactive compounds in GLEE, which interacted with the active sites of key proteins such as NLRP3, NF-κB, and STAT3 through hydrogen bonds, C-H bonds, and electrostatic attractions. In vitro analyses demonstrated that GLEE significantly attenuated NLRP3 inflammasome activation and inhibited the polarization of bone marrow-derived macrophages (BMDMs) towards the M1 phenotype. In vivo, GLEE not only prevented bone mineral density (BMD) loss but also reduced ankle swelling in CIA rats. Furthermore, it decreased the expression of the NLRP3 inflammasome and curtailed the release of inflammatory mediators within the knee joint. CONCLUSION GLEE effectively mitigated inflammatory responses in both blood and knee synovial membranes of CIA rats, potentially through the down-regulation of the NLRP3/Caspase-1/IL-1β signaling pathway and reduction in M1 macrophage polarization.
Collapse
Affiliation(s)
- Shijie Zhang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Bao Hou
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Anjing Xu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yuanyuan Wen
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Xuexue Zhu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Weiwei Cai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Zhijun Han
- Department of Clinical Research Center, Jiangnan University Medical Center, Wuxi, 214001, Jiangsu Province, China
| | - Jing Chen
- Department of Basic Medicine, Tibet University of Medicine, 850000, Lhasa, China
| | - Tsedien Nhamdriel
- Department of Basic Medicine, Tibet University of Medicine, 850000, Lhasa, China
| | - Ma Mi
- Department of Basic Medicine, Tibet University of Medicine, 850000, Lhasa, China.
| | - Liying Qiu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
| | - Haijian Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China.
| |
Collapse
|
17
|
Han P, Liu X, He J, Han L, Li J. Overview of mechanisms and novel therapies on rheumatoid arthritis from a cellular perspective. Front Immunol 2024; 15:1461756. [PMID: 39376556 PMCID: PMC11456432 DOI: 10.3389/fimmu.2024.1461756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/02/2024] [Indexed: 10/09/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation of joints in response to autoimmune disorders. Once triggered, many factors were involved in the development of RA, including both cellular factors like osteoclasts, synovial fibroblasts, T cells, B cells, and soluble factors like interleukin-1 (IL-1), IL-6, IL-17 and tumor necrosis factor-α (TNF-α), etc. The complex interplay of those factors results in such pathological abnormality as synovial hyperplasia, bone injury and multi-joint inflammation. To treat this chronic life-affecting disease, the primary drugs used in easing the patient's symptoms are disease-modifying antirheumatic drugs (DMARDs). However, these traditional drugs could cause serious side effects, such as high blood pressure and stomach ulcers. Interestingly, recent discoveries on the pathogenesis of RA have led to various new kinds of drugs or therapeutic strategies. Therefore, we present a timely review of the latest development in this field, focusing on the cellular aspects of RA pathogenesis and new therapeutic methods in clinical application. Hopefully it can provide translational guide to the pre-clinical research and treatment for the autoimmune joint disease.
Collapse
Affiliation(s)
- Peng Han
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Xiaoying Liu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Jiang He
- Key Laboratory of Uygur Medicine, Xinjiang Institute of Materia Medica, Urumqi, China
| | - Luyang Han
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| |
Collapse
|
18
|
Luo Z, Zhou W, Xie T, Xu W, Shi C, Xiao Z, Si Y, Ma Y, Ren Q, Di L, Shan J. The role of botanical triterpenoids and steroids in bile acid metabolism, transport, and signaling: Pharmacological and toxicological implications. Acta Pharm Sin B 2024; 14:3385-3415. [PMID: 39220868 PMCID: PMC11365449 DOI: 10.1016/j.apsb.2024.04.027] [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: 12/22/2023] [Revised: 03/28/2024] [Accepted: 04/22/2024] [Indexed: 09/04/2024] Open
Abstract
Bile acids (BAs) are synthesized by the host liver from cholesterol and are delivered to the intestine, where they undergo further metabolism by gut microbes and circulate between the liver and intestines through various transporters. They serve to emulsify dietary lipids and act as signaling molecules, regulating the host's metabolism and immune homeostasis through specific receptors. Therefore, disruptions in BA metabolism, transport, and signaling are closely associated with cholestasis, metabolic disorders, autoimmune diseases, and others. Botanical triterpenoids and steroids share structural similarities with BAs, and they have been found to modulate BA metabolism, transport, and signaling, potentially exerting pharmacological or toxicological effects. Here, we have updated the research progress on BA, with a particular emphasis on new-found microbial BAs. Additionally, the latest advancements in targeting BA metabolism and signaling for disease treatment are highlighted. Subsequently, the roles of botanical triterpenoids in BA metabolism, transport, and signaling are examined, analyzing their potential pharmacological, toxicological, or drug interaction effects through these mechanisms. Finally, a research paradigm is proposed that utilizes the gut microbiota as a link to interpret the role of these important natural products in BA signaling.
Collapse
Affiliation(s)
- Zichen Luo
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Children’s Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei Zhou
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tong Xie
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Children’s Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weichen Xu
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Children’s Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chen Shi
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Children’s Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zihan Xiao
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Children’s Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu Si
- Jiangsu CM Clinical Medicine Innovation Center for Obstetrics, Gynecology, and Reproduction, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210001, China
| | - Yan Ma
- National Institute of Biological Sciences, Beijing 102206, China
| | - Qingling Ren
- Jiangsu CM Clinical Medicine Innovation Center for Obstetrics, Gynecology, and Reproduction, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210001, China
| | - Liuqing Di
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinjun Shan
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Children’s Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
19
|
Sun Q, Gu Q, Jiang H, Li W, Lin Z, Li C, Ying Z. Is JAK effective in treating recurrent SAPHO syndrome? TwHF might be a good choice. Medicine (Baltimore) 2024; 103:e38848. [PMID: 39029083 PMCID: PMC11398812 DOI: 10.1097/md.0000000000038848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/17/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Recently, JAKi has also been widely proved to be an effective alternative to conventional treatment for Synovitis acne pustulosis-hyperostosis-osteitis (SAPHO) cases, after failure of multiple drugs including those described above. But what to do when all these treatments fail? We report a case of remission from Tripterygium wilfordii Hook (TwHF) treatment. METHODS The patient was treated with nonsteroidal anti-inflammatory drugs, oral prednisone, minocycline, bisphosphonate injection, etanercept, and tofacitinib, but the symptoms did not change significantly. Treatment with TwHF (1.0 mg/kg/day, patient weight 60 kg) was started for 24 weeks. RESULTS After 50 months of unsatisfactory treatment, this patient was finally treated with herbal TwHF, and after 6 months of treatment, the patient's magnetic resonance imaging and inflammatory indexes were significantly improved, indicating that the disease had been better controlled. CONCLUSION In this study, TwHF was successful in treating a patient with refractory SAPHO syndrome who was refractory to multiple Western medications without significant adverse effects or toxicities, but further follow-up is needed to determine long-term efficacy. More case reports as well as clinical trials are still needed to confirm whether TwHF can effectively treat refractory SAPHO syndrome.
Collapse
Affiliation(s)
- Qiong Sun
- The Second College of Clinical Medicine, Zhejiang Chinese Medical University, Zhejiang 310014, China
- Department of Rheumatology and Immunology, Center for General Practice Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Zhejiang 310014, China
| | - Qinchen Gu
- The Second College of Clinical Medicine, Zhejiang Chinese Medical University, Zhejiang 310014, China
- Department of Rheumatology and Immunology, Center for General Practice Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Zhejiang 310014, China
| | - Haixu Jiang
- School of Chinese Materia, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Weizhong Li
- Department of Rheumatology, Fangshan Hospital, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Zhimin Lin
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Chen Li
- Department of Rheumatology, Fangshan Hospital, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Zhenhua Ying
- Department of Rheumatology and Immunology, Center for General Practice Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Zhejiang 310014, China
- Institute of Rheumatology and Immunology, Hangzhou Medical College, Zhejiang 310014, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, Zhejiang 310014, China
| |
Collapse
|
20
|
Chen Y, Liu M, Lu M, Luo L, Han Z, Liu X. Exploring the impact of m 6A modification on immune diseases: mechanisms and therapeutic implication. Front Immunol 2024; 15:1387582. [PMID: 39072324 PMCID: PMC11272477 DOI: 10.3389/fimmu.2024.1387582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 06/28/2024] [Indexed: 07/30/2024] Open
Abstract
N6-methyladenosine (m6A) is a chemical modification of RNA and has become a widely discussed topic among scientific researchers in recent years. It is distributed in various organisms, including eukaryotes and bacteria. It has been found that m6A is composed of writers, erasers and readers and is involved in biological functions such as splicing, transport and translation of RNA. The balance of the human immune microenvironment is important for human health abnormalities. Increasing studies have found that m6A affects the development of immune diseases such as inflammatory enteritis and systemic lupus erythematosus (SLE) by participating in the homeostatic regulation of the immune microenvironment in vivo. In this manuscript, we introduce the composition, biological function, regulation of m6A in the immune microenvironment and its progression in various immune diseases, providing new targets and directions for the treatment of immune diseases in clinical practice.
Collapse
Affiliation(s)
- Yutong Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Min Liu
- Department of Traditional Chinese Medicine, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, China
| | - Miao Lu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Linling Luo
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xide Liu
- Department of Traditional Chinese Medicine, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
21
|
Yang L, Guo CW, Luo QM, Guo ZF, Chen L, Ishihama Y, Li P, Yang H, Gao W. Thermostability-assisted limited proteolysis-coupled mass spectrometry for capturing drug target proteins and sites. Anal Chim Acta 2024; 1312:342755. [PMID: 38834267 DOI: 10.1016/j.aca.2024.342755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/28/2024] [Accepted: 05/20/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Identifying drug-binding targets and their corresponding sites is crucial for drug discovery and mechanism studies. Limited proteolysis-coupled mass spectrometry (LiP-MS) is a sophisticated method used for the detection of compound and protein interactions. However, in some cases, LiP-MS cannot identify the target proteins due to the small structure changes or the lack of enrichment of low-abundant protein. To overcome this drawback, we developed a thermostability-assisted limited proteolysis-coupled mass spectrometry (TALiP-MS) approach for efficient drug target discovery. RESULTS We proved that the novel strategy, TALiP-MS, could efficiently identify target proteins of various ligands, including cyclosporin A (a calcineurin inhibitor), geldanamycin (an HSP90 inhibitor), and staurosporine (a kinase inhibitor), with accurately recognizing drug-binding domains. The TALiP protocol increased the number of target peptides detected in LiP-MS experiments by 2- to 8-fold. Meanwhile, the TALiP-MS approach can not only identify both ligand-binding stability and destabilization proteins but also shows high complementarity with the thermal proteome profiling (TPP) and machine learning-based limited proteolysis (LiP-Quant) methods. The developed TALiP-MS approach was applied to identify the target proteins of celastrol (CEL), a natural product known for its strong antioxidant and anti-cancer angiogenesis effect. Among them, four proteins, MTHFD1, UBA1, ACLY, and SND1 were further validated for their strong affinity to CEL by using cellular thermal shift assay. Additionally, the destabilized proteins induced by CEL such as TAGLN2 and CFL1 were also validated. SIGNIFICANCE Collectively, these findings underscore the efficacy of the TALiP-MS method for identifying drug targets, elucidating binding sites, and even detecting drug-induced conformational changes in target proteins in complex proteomes.
Collapse
Affiliation(s)
- Liu Yang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Chen-Wan Guo
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Qi-Ming Luo
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Zi-Fan Guo
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Ling Chen
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Yasushi Ishihama
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Ping Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Hua Yang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Wen Gao
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan.
| |
Collapse
|
22
|
Chen Z, Wang J, Zhou J, Yu H, Zheng L, Chen Y, Wan X, Zhang W. Tripterygium drug-loaded liposome alleviates renal function by promoting vascularization and inhibiting fibrosis. Front Chem 2024; 12:1427670. [PMID: 39010937 PMCID: PMC11246911 DOI: 10.3389/fchem.2024.1427670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/07/2024] [Indexed: 07/17/2024] Open
Abstract
Introduction: Tripterygium species have been traditionally used in Chinese medicine for treating various conditions. The aim of the study was to construct a drug-modified renal infarction targeting liposome (rTor-LIP) containing Tripterygium in order to improve the therapeutic effect on renal injury. Methods: rTor-LIP was prepared using the extruder method containing Tripterygium solution. The preparation was characterized by transmission electron microscopy, Marvin laser particle size analyzer, and Western blotting. In vitro experiments were conducted to verify the biocompatibility of rTor-LIP, and in vivo experiments were conducted to verify the therapeutic effect of rTor- LIP on renal injury. Results and discussion: The surface of rTor-LIP was regular and oval. In vitro results showed that after co-incubation with rTor-LIP, endothelial cells did not show significant apoptosis, and there were no significant abnormalities in the mitochondrial metabolism. The in vivo results showed that the morphology of endothelial cells in the rTor-LIP group was uniform and the cytoplasmic striations were clear, but the local striations had disappeared. Thus, rTor-LIP nano-targeted liposomes can effectively target hypoxic kidney tissue, providing a new idea for the treatment of renal infarction.
Collapse
Affiliation(s)
- Ziwei Chen
- Department of Nephrology, Taizhou Central Hospital, Affiliated to Taizhou University, Taizhou, China
| | - Jiajia Wang
- Department of Traditional Chinese Medicine, Taizhou Hospital of Zhejiang Province, Taizhou, China
| | - Jianyu Zhou
- Department of Ultrasound, Taizhou Central Hospital, Affiliated to Taizhou University, Taizhou, China
| | - Haifeng Yu
- Department of Nephrology, Taizhou Central Hospital, Affiliated to Taizhou University, Taizhou, China
| | - Lu Zheng
- Department of Nephrology, Taizhou Central Hospital, Affiliated to Taizhou University, Taizhou, China
| | - Yuan Chen
- Department of Nephrology, Taizhou Central Hospital, Affiliated to Taizhou University, Taizhou, China
| | - Xiaoqing Wan
- Department of Nephrology, Taizhou Central Hospital, Affiliated to Taizhou University, Taizhou, China
| | - Wei Zhang
- Department of Nephrology, Taizhou Central Hospital, Affiliated to Taizhou University, Taizhou, China
| |
Collapse
|
23
|
Chen Y, Wang Y, Xu Y, Ma S, Yang H, Liu Y, Wu X. Quality Evaluation of Tripterygium Glycoside Tablets Based on Quantitative Band-Selective 2D 1H- 13C HSQC and 1H NMR Fingerprinting. ACS OMEGA 2024; 9:27321-27328. [PMID: 38947815 PMCID: PMC11209881 DOI: 10.1021/acsomega.4c01878] [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: 02/26/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 07/02/2024]
Abstract
Tripterygium glycoside tablets (TGTs) are preparations extracted and purified from Tripterygium wilfordii Hook. F and are extensively utilized in the treatment of autoimmune diseases, such as rheumatoid arthritis (RA). However, variations in production processes among manufacturers can lead to challenges in quality control and clinical utilization of TGTs. A band-selective 2D 1H-13C HSQC quantification method was applied for the determination of 13 active ingredients in TGTs. This method was validated following the guidelines of USP-NF 2022. The results demonstrated that the quantitative method exhibited excellent signal resolution, as well as sufficient accuracy, sensitivity, and stability. In addition, the 1H NMR spectra of TGTs from three manufacturers underwent analysis using principal component analysis and orthogonal partial least-squares discriminant analysis. The results revealed significant differences among the TGTs from the three manufacturers, with manufacturer 2 and manufacturer 3 demonstrating superior product consistency compared to manufacturer 1. A quality evaluation system for TGTs was developed based on band-selective 2D 1H-13C HSQC and 1H NMR, encompassing both quality markers and fingerprinting. This system offers reliable approaches and insights for enhancing the quality control of natural products.
Collapse
Affiliation(s)
- Youwen Chen
- School
of Chinese Materia Medica, Beijing University
of Chinese Medicine, Beijing 100102, P.R. China
- National
Institutes for Food and Drug Control, Beijing 102629, P.R. China
| | - Yadan Wang
- National
Institutes for Food and Drug Control, Beijing 102629, P.R. China
| | - Yiwen Xu
- National
Institutes for Food and Drug Control, Beijing 102629, P.R. China
| | - Shuangcheng Ma
- National
Institutes for Food and Drug Control, Beijing 102629, P.R. China
| | - Huiying Yang
- National
Institutes for Food and Drug Control, Beijing 102629, P.R. China
| | - Yuanyan Liu
- School
of Chinese Materia Medica, Beijing University
of Chinese Medicine, Beijing 100102, P.R. China
| | - Xianfu Wu
- National
Institutes for Food and Drug Control, Beijing 102629, P.R. China
| |
Collapse
|
24
|
Zhang L, Lu L, Jiang S, Yin Z, Tan G, Ning F, Qin Z, Huang J, Huang M, Jin J. Salvianolic acid extract prevents Tripterygium wilfordii polyglycosides-induced acute liver injury by modulating bile acid metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:117939. [PMID: 38382651 DOI: 10.1016/j.jep.2024.117939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/16/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygium wilfordii polyglycosides (TWP) tablet is the most widely used traditional Chinese medicine preparation for the treatment of rheumatoid arthritis (RA), but the hepatotoxicity often limits its widespread application. In traditional use, Salvia miltiorrhiza has cardioprotective and hepatoprotective effects. Salvianolic acid extract (SA) is a hydrophilic component of Salvia miltiorrhiza and has significant antioxidant and hepatoprotective effects. AIM OF THE STUDY To investigate the protective effects of SA on the TWP-induced acute liver injury in rats and to explore the related mechanisms by integration of metabolomics and transcriptomics. MATERIALS AND METHODS SA and TWP extracts were identified by UPLC-Q/TOF-MS. SA (200 mg/kg) was administered for consecutive 7 days. On day 7, TWP (360 mg/kg) was administered by gavage to induce the acute liver injury in rats. Serum biochemical assay and H&E staining were used to evaluate liver damage. Liver metabolomics and transcriptomics were used to explore the potential mechanisms, and further molecular biological experiments such as qPCR and IHC were utilized to validate the relevant signaling pathways. RESULTS SA can prevent liver injury symptoms caused by TWP, such as elevated liver index, elevated ALT and AST, and pathological changes in liver tissue. Liver metabolomics studies showed that TWP can significantly alter the content of individual bile acid in the liver and SA had the most significant impact on the biosynthetic pathway of bile acids. The transcriptomics results of the liver indicated that the genes changed in the SA + TWP group were mainly involved in sterol metabolism, lipid regulation and bile acid homeostasis pathways. The gene expression of Nr1h4, which encodes farnesoid X receptor (FXR), an important regulator of bile acid homeostasis, was significantly changed. Further studies confirmed that SA can prevent the downregulation of FXR and its downstream signaling induced by TWP, thereby regulating bile acid metabolism, ultimately preventing acute liver injury caused by TWP. CONCLUSION Our results demonstrated that SA could protect the liver from TWP-induced hepatic injury by modulation of the bile acid metabolic pathway. SA may provide a new strategy for the protection against TWP-induced acute liver injury.
Collapse
Affiliation(s)
- Lei Zhang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Langqing Lu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shiqin Jiang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhaokun Yin
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Guoyao Tan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Fangqing Ning
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhiyan Qin
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Junyuan Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jing Jin
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
25
|
Zhu Y, Yao L, Guo Y, Zhang J, Xia Y, Wei Z, Dai Y. Bergenin attenuates triptolide-caused premature ovarian failure in mice based on the antioxidant activity. Reprod Toxicol 2024; 126:108608. [PMID: 38735593 DOI: 10.1016/j.reprotox.2024.108608] [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: 09/06/2023] [Revised: 04/16/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
Tripterygium wilfordii (TW) preparations have been utilized in China for treating rheumatoid arthritis and autoimmune diseases. However, their clinical use is limited due to reproductive toxicity, notably premature ovarian failure (POF). Our study aimed to investigate the effect and mechanism of bergenin in attenuating POF induced by triptolide in mice. POF was induced in female ICR mice via oral triptolide administration (50 μg/kg) for 60 days. Mice received bergenin (25, 50, 100 mg/kg, i.g.) or estradiol valerate (EV) (0.1 mg/kg, i.g.) daily, 1 h before triptolide treatment. In vitro, ovarian granulosa cells (OGCs) were exposed to triptolide (100 nM) and bergenin (1, 3, 10 μM). Antioxidant enzyme activity, protein expression, apoptosis rate, and reactive oxygen species (ROS) levels were assessed. The results showed that triptolide-treated mice exhibited evident atrophy, along with an increase in atretic follicles. Bergenin (50, 100 mg/kg) and EV (0.1 mg/kg), orally administered, exerted significant anti-POF effect. Bergenin and EV also decreased apoptosis in mouse ovaries. In vitro, bergenin (1, 3, 10 μM) attenuated triptolide-induced OGCs apoptosis by reducing levels of apoptosis-related proteins. Additionally, bergenin reduced oxidative stress through downregulation of antioxidant enzymes activity and overall ROS levels. Moreover, the combined use with Sh-Nrf2 resulted in a reduced protection of bergenin against triptolide-induced apoptosis of OGCs. Together, bergenin counteracts triptolide-caused POF in mice by inhibiting Nrf2-mediated oxidative stress and preventing OGC apoptosis. Combining bergenin with TW preparations may effectively reduce the risk of POF.
Collapse
Affiliation(s)
- Yanrong Zhu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Lichen Yao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Yilei Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Jing Zhang
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Yufeng Xia
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China.
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China.
| |
Collapse
|
26
|
Gao C, Song XD, Chen FH, Wei GL, Guo CY. The protective effect of natural medicines in rheumatoid arthritis via inhibit angiogenesis. Front Pharmacol 2024; 15:1380098. [PMID: 38881875 PMCID: PMC11176484 DOI: 10.3389/fphar.2024.1380098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/10/2024] [Indexed: 06/18/2024] Open
Abstract
Rheumatoid arthritis is a chronic immunological disease leading to the progressive bone and joint destruction. Angiogenesis, accompanied by synovial hyperplasia and inflammation underlies joint destruction. Delaying or even blocking synovial angiogenesis has emerged as an important target of RA treatment. Natural medicines has a long history of treating RA, and numerous reports have suggested that natural medicines have a strong inhibitory activity on synovial angiogenesis, thereby improving the progression of RA. Natural medicines could regulate the following signaling pathways: HIF/VEGF/ANG, PI3K/Akt pathway, MAPKs pathway, NF-κB pathway, PPARγ pathway, JAK2/STAT3 pathway, etc., thereby inhibiting angiogenesis. Tripterygium wilfordii Hook. f. (TwHF), sinomenine, and total glucoside of Paeonia lactiflora Pall. Are currently the most representative of all natural products worthy of development and utilization. In this paper, the main factors affecting angiogenesis were discussed and different types of natural medicines that inhibit angiogenesis were systematically summarized. Their specific anti-angiogenesis mechanisms are also reviewed which aiming to provide new perspective and options for the management of RA by targeting angiogenesis.
Collapse
Affiliation(s)
- Chang Gao
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| | - Xiao-Di Song
- Gannan Medical University, Jiangxi, Ganzhou, China
| | - Fang-Hui Chen
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| | - Gui-Lin Wei
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| | - Chun-Yu Guo
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| |
Collapse
|
27
|
Sun J, Du J, Liu X, An J, Hu Y, Wang J, Zhu F, Feng H, Cheng S, Tian H, Mei X, Wu C. Chondroitin sulfate-modified tragacanth gum-gelatin composite nanocapsules loaded with curcumin nanocrystals for the treatment of arthritis. J Nanobiotechnology 2024; 22:270. [PMID: 38769551 PMCID: PMC11104008 DOI: 10.1186/s12951-024-02540-2] [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: 02/19/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease of yet undetermined etiology that is accompanied by significant oxidative stress, inflammatory responses, and damage to joint tissues. In this study, we designed chondroitin sulfate (CS)-modified tragacanth gum-gelatin composite nanocapsules (CS-Cur-TGNCs) loaded with curcumin nanocrystals (Cur-NCs), which rely on the ability of CS to target CD44 to accumulate drugs in inflamed joints. Cur was encapsulated in the form of nanocrystals into tragacanth gum-gelatin composite nanocapsules (TGNCs) by using an inborn microcrystallization method, which produced CS-Cur-TGNCs with a particle size of approximately 80 ± 11.54 nm and a drug loading capacity of 54.18 ± 5.17%. In an in vitro drug release assay, CS-Cur-TGNCs showed MMP-2-responsive properties. During the treatment of RA, CS-Cur-TGNCs significantly inhibited oxidative stress, promoted the polarization of M2-type macrophages to M1-type macrophages, and decreased the expression of inflammatory factors (TNF-α, IL-1β, and IL-6). In addition, it also exerted excellent anti-inflammatory effects, and significantly alleviated the swelling of joints during the treatment of gouty arthritis (GA). Therefore, CS-Cur-TGNCs, as a novel drug delivery system, could lead to new ideas for clinical therapeutic regimens for RA and GA.
Collapse
Affiliation(s)
- Junpeng Sun
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Jiaqun Du
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Xiaobang Liu
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Jinyu An
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Yu Hu
- Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Jing Wang
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Fu Zhu
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Huicong Feng
- Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Shuai Cheng
- Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - He Tian
- Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
| | - Xifan Mei
- Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
- Liaoning Provincial Key Laboratory of Medical Tissue Engineering, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
| | - Chao Wu
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
- Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
- Liaoning Provincial Key Laboratory of Medical Tissue Engineering, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
| |
Collapse
|
28
|
Su QY, Li HC, Jiang XJ, Jiang ZQ, Zhang Y, Zhang HY, Zhang SX. Exploring the therapeutic potential of regulatory T cell in rheumatoid arthritis: Insights into subsets, markers, and signaling pathways. Biomed Pharmacother 2024; 174:116440. [PMID: 38518605 DOI: 10.1016/j.biopha.2024.116440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024] Open
Abstract
Rheumatoid arthritis (RA) is a complex autoimmune inflammatory rheumatic disease characterized by an imbalance between immunological reactivity and immune tolerance. Regulatory T cells (Tregs), which play a crucial role in controlling ongoing autoimmunity and maintaining peripheral tolerance, have shown great potential for the treatment of autoimmune inflammatory rheumatic diseases such as RA. This review aims to provide an updated summary of the latest insights into Treg-targeting techniques in RA. We focus on current therapeutic strategies for targeting Tregs based on discussing their subsets, surface markers, suppressive function, and signaling pathways in RA.
Collapse
Affiliation(s)
- Qin-Yi Su
- The Second Hospital of Shanxi Medical University, Department of Rheumatology, Taiyuan, China; Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Huan-Cheng Li
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Xiao-Jing Jiang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Zhong-Qing Jiang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Yan Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - He-Yi Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Sheng-Xiao Zhang
- The Second Hospital of Shanxi Medical University, Department of Rheumatology, Taiyuan, China; Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China.
| |
Collapse
|
29
|
Pan W, Yang B, He D, Chen L, Fu C. Functions and targets of miRNAs in pharmacological and toxicological effects of major components of Tripterygium wilfordii Hook F. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1997-2019. [PMID: 37831113 DOI: 10.1007/s00210-023-02764-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/29/2023] [Indexed: 10/14/2023]
Abstract
Tripterygium wilfordii Hook F (TwHF) has a long history of use as a traditional Chinese medicine and has been widely administered to treat various inflammatory and autoimmune diseases. MicroRNAs (miRNAs) are endogenous, short, non-coding RNAs that regulate gene expression post-transcriptionally. They participate in the efficacies and even toxicities of the components of TwHF, rendering miRNAs an appealing therapeutic strategy. This review summarizes the recent literature related to the roles and mechanisms of miRNAs in the pharmacological and toxicological effects of main components of TwHF, focusing on two active compounds, triptolide (TP) and celastrol (CEL). Additionally, the prospects for the "You Gu Wu Yun" theory regarding TwHF nephrotoxicity are presented.
Collapse
Affiliation(s)
- Wei Pan
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, People's Republic of China
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Bo Yang
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Dongxiu He
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, People's Republic of China
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, People's Republic of China
| | - Chengxiao Fu
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, People's Republic of China.
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
30
|
Xie H, Zhang A, Li J, Mou X, He T, Yeung TC, Lau CBS, Zuo Z, Li P, Kennelly EJ, Leung PC, Tang Y, Fan X, Wang CC, Li L. Cycasin derivative: a potential embryotoxic component of Atractylodes macrocephala rhizome for limb malformation. Toxicol Res (Camb) 2024; 13:tfae057. [PMID: 38623091 PMCID: PMC11015991 DOI: 10.1093/toxres/tfae057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/19/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
Objective The rhizome of Atractylodes macrocephala Koidz. (Asteraceae), called Atractylodes macrocephala rhizome (AMR) and known by its traditional name Bai Zhu, is a prominent Chinese herbal medicine employed for preventing miscarriage. However, our previous study revealed that high dosages of AMR administered during pregnancy could cause embryotoxicity but the specific embryotoxic components and their underlying mechanisms remain unclear. This study aimed to screen and identify the potential embryotoxic components of AMR. Methods The AMR extracts and sub-fractions were analyzed by thin layer chromatography and subsequently screened by in vitro mouse limb bud micromass and mouse whole embryo culture bioassays. The embryotoxic fractions from AMR were further evaluated in vivo using a pregnant mouse model. The structures of the potential embryotoxic components were analyzed using matrix-assisted laser desorption/ionization tandem time-of-flight mass spectrometry (MALDI-TOF/TOF-MS). Results In vitro and in vivo bioassays revealed that AMR glycoside-enriched sub-fractions (AMR-A-IIa and AMR-A-IIb) exhibited potential embryotoxicity. These sub-fractions, when administered to pregnant animals, increased the incidence of stillbirth and congenital limb malformations. MS spectrometry analysis identified cycasin derivatives in both sub-fractions, suggesting their possible role in the observed limb malformations. However, further experiments are necessary to validate this hypothesis and to elucidate the underlying mechanisms. Conclusions Our study provides significant scientific evidence on the pharmacotoxicity of AMR, which is important for the safe clinical application of commonly used Chinese herbal medicines during pregnancy.
Collapse
Affiliation(s)
- Hongliang Xie
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, No. 866 Yuhangtang Road, West Lake District, Hangzhou City, Zhejiang Province, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, No. 828 Zhongxing Road, Xitang Town, Jiaxing City, Zhejiang Province, 314100, China
| | - Aolin Zhang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, No. 866 Yuhangtang Road, West Lake District, Hangzhou City, Zhejiang Province, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, No. 828 Zhongxing Road, Xitang Town, Jiaxing City, Zhejiang Province, 314100, China
| | - Junwei Li
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, No. 866 Yuhangtang Road, West Lake District, Hangzhou City, Zhejiang Province, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, No. 828 Zhongxing Road, Xitang Town, Jiaxing City, Zhejiang Province, 314100, China
| | - Xuan Mou
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, No. 866 Yuhangtang Road, West Lake District, Hangzhou City, Zhejiang Province, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, No. 828 Zhongxing Road, Xitang Town, Jiaxing City, Zhejiang Province, 314100, China
| | - Tao He
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, No. 866 Yuhangtang Road, West Lake District, Hangzhou City, Zhejiang Province, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, No. 828 Zhongxing Road, Xitang Town, Jiaxing City, Zhejiang Province, 314100, China
| | - Tsz Ching Yeung
- Department of Obstetrics and Gynaecology; Li Ka Shing Institute of Health Sciences; School of Biomedical Sciences; Sichuan University-Chinese University of Hong Kong Joint Reproductive Medicine Laboratory, The Chinese University of Hong Kong, Shatin, New Territories, 999077, Hong Kong
| | - Clara Bik San Lau
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, 999077, Hong Kong SAR, China
| | - Zhong Zuo
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, 999077, Hong Kong SAR, China
| | - Ping Li
- Department of Biological Sciences, Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, New York, 10468, United States
| | - Edward J Kennelly
- Department of Biological Sciences, Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, New York, 10468, United States
| | - Ping Chung Leung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, 999077, Hong Kong SAR, China
| | - Yu Tang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, No. 866 Yuhangtang Road, West Lake District, Hangzhou City, Zhejiang Province, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, No. 828 Zhongxing Road, Xitang Town, Jiaxing City, Zhejiang Province, 314100, China
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, No. 866 Yuhangtang Road, West Lake District, Hangzhou City, Zhejiang Province, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, No. 828 Zhongxing Road, Xitang Town, Jiaxing City, Zhejiang Province, 314100, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, No. 866 Yuhangtang Road, West Lake District, Hangzhou City, Zhejiang Province, 310058, China
- Modern Chinese Medicine and Reproductive Health Joint Innovation Center, Innovation Center of Yangtze River Delta, No. 828 Zhongxing Road, Xitang Town, Jiaxing City, Zhejiang Province, 314100, China
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology; Li Ka Shing Institute of Health Sciences; School of Biomedical Sciences; Sichuan University-Chinese University of Hong Kong Joint Reproductive Medicine Laboratory, The Chinese University of Hong Kong, Shatin, New Territories, 999077, Hong Kong
- Modern Chinese Medicine and Reproductive Health Joint Innovation Center, Innovation Center of Yangtze River Delta, No. 828 Zhongxing Road, Xitang Town, Jiaxing City, Zhejiang Province, 314100, China
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou City, Zhejiang Province, 310053, China
| | - Lu Li
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, No. 866 Yuhangtang Road, West Lake District, Hangzhou City, Zhejiang Province, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, No. 828 Zhongxing Road, Xitang Town, Jiaxing City, Zhejiang Province, 314100, China
- Department of Obstetrics and Gynaecology; Li Ka Shing Institute of Health Sciences; School of Biomedical Sciences; Sichuan University-Chinese University of Hong Kong Joint Reproductive Medicine Laboratory, The Chinese University of Hong Kong, Shatin, New Territories, 999077, Hong Kong
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, 999077, Hong Kong SAR, China
- Modern Chinese Medicine and Reproductive Health Joint Innovation Center, Innovation Center of Yangtze River Delta, No. 828 Zhongxing Road, Xitang Town, Jiaxing City, Zhejiang Province, 314100, China
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine; Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, No. 3 Qingchun East Road, Shangcheng District, Hangzhou City, Zhejiang Province, 310016, China
| |
Collapse
|
31
|
Huang J, Cai H, Ye X, Zhang G, Ye L, Yang C, Wang J, Jin M. Demethylzeylasteral (T-96) Alleviates Allergic Asthma via Inhibiting MAPK/ERK and NF-κB Pathway. Int Arch Allergy Immunol 2024; 185:631-640. [PMID: 38527438 PMCID: PMC11216359 DOI: 10.1159/000537837] [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: 03/11/2023] [Accepted: 02/12/2024] [Indexed: 03/27/2024] Open
Abstract
INTRODUCTION Demethylzeylasteral (T-96), a new extract of Tripterygium wilfordii Hook F, exerted immunomodulatory properties in autoimmune diseases, but its effect on airway inflammatory diseases remains unclear. Our study aims to explore the protective effect and underlying mechanism of T-96 in allergic asthma. METHODS The OVA-induced asthmatic mice were administered by gavage with T-96 (0.1 mg/10 g, 0.3 mg/10 g, or 0.6 mg/10 g) 1 h before each challenge. The airway hyperresponsiveness was assessed, pathological changes were evaluated by HE and PAS staining, and expressions of Th2 cytokines were determined by PCR and ELISA. The activation of MAPK/ERK and NF-κB pathway was assessed by western blot. RESULTS T-96 significantly relieved airway hyperresponsiveness in asthmatic mice, evidenced by reduced airway resistance (Raw) and increased lung compliance dynamic compliance (Cdyn). Also, enhanced inflammatory infiltration and mucus hypersecretion were ameliorated in lungs of asthmatic mice following increasing doses of T-96 treatment, accompanied by decreased eosinophils in bronchoalveolar lavage fluid (BALF), IgE and OVA-specific IgE levels in serum, and downregulated IL-5 and IL-13 expressions in BALF and lung tissues as well. Notably, phosphorylation levels of p38 MAPK, ERK, and p65 NF-κB were obviously increased in asthmatic mice compared with the control group, which were then abrogated upon T-96 treatment. CONCLUSION This study first revealed that T-96 alleviated allergic airway inflammation and airway hyperresponsiveness via inhibiting MAPK/ERK and NF-κB pathway. Thus, T-96 could potentially act as a new anti-inflammatory agent in allergic asthma.
Collapse
Affiliation(s)
- Jianan Huang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hui Cai
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaofen Ye
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ge Zhang
- Department of Pulmonary Medicine, Xuhui Central Hospital, Shanghai Clinical Research Center, Shanghai, China
| | - Ling Ye
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunxin Yang
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Meiling Jin
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
32
|
Dao TNP, Onikanni SA, Fadaka AO, Klein A, Tran VD, Le MH, Wang CH, Chang HH. In silico identification of compounds from Piper sarmentosum Roxb leaf fractionated extract inhibit interleukin-6 to prevent rheumatoid arthritis. Front Pharmacol 2024; 15:1358037. [PMID: 38576490 PMCID: PMC10991700 DOI: 10.3389/fphar.2024.1358037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024] Open
Abstract
Objective: Medicinal herbs with a phytonutrient background has been applied globally as major alternatives to ameliorate the continuous increase in rheumatoid arthritis cases worldwide. We herein aimed to critically examine the bioactive components of the medicinal herb Piper sarmentosum Roxb leaf fractionated extract for its potential to inhibit the influx of interleukin-6 (IL-6) in rheumatoid arthritis. Methods: The Schrödinger platform was employed as the main computational acumen for the screening of bioactive compounds identified and reference compounds subjected to molecular simulation (MDS) for analyzing the stability of docked complexes to assess fluctuations and conformational changes during protein-ligand interactions. Results: The values of the simulatory properties and principal component analysis (PCA) revealed the good stability of these phytochemicals in the active pocket of interleukin-6 (IL-6). Discussion: Our findings reveal new strategies in which these phytochemicals are potential inhibitory agents that can be modified and further evaluated to develop more effective agents for the management of rheumatoid arthritis, thereby providing a better understanding and useful model for the reproduction and/or discovery of new drugs for the management of rheumatoid arthritis and its complications.
Collapse
Affiliation(s)
- Tran Nhat Phong Dao
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Faculty of Traditional Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Sunday Amos Onikanni
- College of Medicine, Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Chemical Sciences, Biochemistry Unit, Afe-Babalola University, Ado-Ekiti, Nigeria
| | | | - Ashwil Klein
- Department of Biotechnology, University of the Western Cape, Bellville, South Africa
| | - Van De Tran
- Department of Health Organization and Management, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Minh Hoang Le
- Faculty of Traditional Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Chih-Hao Wang
- College of Medicine, Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Cell Biology, China Medical University, Taichung, Taiwan
| | - Hen-Hong Chang
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
33
|
Lin J, Liu J, O’Fee A, Pandey C, Benna-Doyle S, Maunder A, Rao V, Alesi S, Ng B, Ee C. The effectiveness and safety of lifestyle medicine and integrative therapies in inflammatory arthritis: an umbrella review using a hierarchical evidence gathering approach. Front Med (Lausanne) 2024; 11:1357914. [PMID: 38545510 PMCID: PMC10965540 DOI: 10.3389/fmed.2024.1357914] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/13/2024] [Indexed: 11/11/2024] Open
Abstract
Objective An umbrella review was conducted to provide a comprehensive evaluation of the evidence on lifestyle medicine and integrative therapies for inflammatory arthritis. Methods Five electronic databases were searched for umbrella reviews, meta-analyses, and systematic reviews of randomised controlled trials on acupuncture, diet, exercise, herbal medicine, nutrient supplements, and mind-body therapies for rheumatoid arthritis, spondyloarthritis, and gout published from January 2012 to December 2022. The primary outcomes were functional status and quality of life. Quality assessment was performed using the A MeaSurement Tool to Assess systematic Reviews (AMSTAR-2) tool, and the certainty of evidence for our primary outcomes was assessed using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach where possible. Results We included 52 reviews. Exercise was beneficial for functional status in both rheumatoid arthritis and spondyloarthritis, with moderate certainty of evidence. Chinese herbal medicine in combination with disease-modifying anti-rheumatic drugs may improve functional status in rheumatoid arthritis (very low certainty evidence). Acupuncture may improve functional status in rheumatoid arthritis and pain in both rheumatoid arthritis and gout; however, the evidence is of very low certainty. Evidence for other therapies was not clinically significant; however, it suggests possible benefits from quercetin and polyunsaturated fatty acids. Yoga may result in a moderate improvement in functional status when used as an adjunct to medication; however, the certainty of evidence is very low. Diet interventions offered inconsistent improvements to functional status in rheumatoid arthritis, spondyloarthritis, and gout with low to very low certainty. Conclusion Exercise should be prescribed for people with rheumatoid arthritis and spondyloarthritis. More research is needed to confirm or refute evidence for Chinese herbal medicine, acupuncture, yoga, and anti-inflammatory diets.
Collapse
Affiliation(s)
- Joshua Lin
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Jing Liu
- NICM Health Research Institute, Western Sydney University, Sydney, NSW, Australia
| | - Allana O’Fee
- NICM Health Research Institute, Western Sydney University, Sydney, NSW, Australia
| | - Chhiti Pandey
- NICM Health Research Institute, Western Sydney University, Sydney, NSW, Australia
| | - Sarah Benna-Doyle
- NICM Health Research Institute, Western Sydney University, Sydney, NSW, Australia
| | - Alison Maunder
- NICM Health Research Institute, Western Sydney University, Sydney, NSW, Australia
| | - Vibhuti Rao
- NICM Health Research Institute, Western Sydney University, Sydney, NSW, Australia
| | - Simon Alesi
- NICM Health Research Institute, Western Sydney University, Sydney, NSW, Australia
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, VIC, Australia
| | - Beverly Ng
- Department of Rheumatology, Westmead Hospital, Sydney, NSW, Australia
| | - Carolyn Ee
- NICM Health Research Institute, Western Sydney University, Sydney, NSW, Australia
| |
Collapse
|
34
|
Long C, He P, Tu R, Song X, Li H, Huang W, Liu J, Zhang L, Guo Y. Subchronic toxicity evaluation of Huobahuagen extract and plasma metabolic profiling analysis combined with conventional pathology methods. J Appl Toxicol 2024; 44:201-215. [PMID: 37697829 DOI: 10.1002/jat.4532] [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: 07/02/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 09/13/2023]
Abstract
Huobahua, namely, Tripterygium hypoglaucum (Levl.) Hutch, known as a traditional Chinese herbal medicine, especially its underground parts, has been widely developed into several Tripterygium agents for the treatment of rheumatoid arthritis and other autoimmune diseases. It has sparked wide public concern about its safety, such as multi-organ toxicity. However, the toxic characteristics and damage mechanism of Huobahuagen extract (HBHGE) remain unclear. In the present study, subchronic oral toxicity study of HBHGE (10.0 g crude drug/kg/day for 12 weeks) was performed in male rats. Hematological, serum biochemical, and histopathological parameters, urinalysis, and plasma metabolic profiling were assessed. The single-dose subchronic toxicity results related to HBHGE exhibited obvious toxicity to the testis and epididymis of male rats. Furthermore, plasma metabolomics analysis suggested that a series of metabolic disorders were induced by oral administration of HBHGE, mainly focusing on amino acid (glutamate, phenylalanine, and tryptophan) metabolisms, pyrimidine metabolism, glutathione metabolism, and steroid hormone biosynthesis. Moreover, it appeared that serum testosterone in male rats treated with HBHGE for 12 weeks, decreased significantly, and was susceptible to the toxic effects of HBHGE. Taken together, conventional pathology and plasma metabolomics for preliminarily exploring subchronic toxicity and underlying mechanism can provide useful information about the reduction of toxic risks from HBHGE and new insights into the development of detoxification preparations.
Collapse
Affiliation(s)
- Chengyan Long
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Peilin He
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Ruxia Tu
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Xiaoxian Song
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Henghua Li
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Wentao Huang
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Jianyi Liu
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Li Zhang
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Yanlei Guo
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| |
Collapse
|
35
|
Shu Y, Yang R, Wen H, Dong Q, Chen Z, Xiang Y, Wu H. Myricetin reduces neutrophil extracellular trap release in a rat model of rheumatoid arthritis, which is associated with a decrease in disease severity. Innate Immun 2024; 30:66-78. [PMID: 38780369 PMCID: PMC11165658 DOI: 10.1177/17534259241255439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/01/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic disease characterized by joint inflammation and severe disability. However, there is a lack of safe and effective drugs for treating RA. In our previous study, we discovered that myricetin (MC) and celecoxib have a synergistic effect in the treatment of RA. We conducted in vitro and in vivo experiments to further investigate the effects and mechanisms of action of MC. Our findings demonstrated that MC treatment effectively reduced the release of neutrophil extracellular traps (NETs) and alleviated the inflammatory response in RA. Mechanistic studies showed that MC prevents the entry of PADI4 and MPO into the cell nucleus, thereby protecting DNA from decondensation. In a rat arthritis model, MC improved histological changes in ankle joints and suppressed NET-related signaling factors. In conclusion, MC protects the ankle joints against arthritis by inhibiting MPO and PADI4, thereby reducing NET release. The pharmacological mechanism of MC in RA involves the inhibition of NET release.
Collapse
Affiliation(s)
- Yiqin Shu
- Medical College of Hubei Enshi College, Enshi, China
| | - Rui Yang
- Medical College of Hubei Enshi College, Enshi, China
- Institute of anti-rheumatism Tujia Medicine, Enshi, China
| | - Huijie Wen
- Medical College of Hubei Enshi College, Enshi, China
| | - Qiannan Dong
- Medical College of Hubei Enshi College, Enshi, China
- Institute of anti-rheumatism Tujia Medicine, Enshi, China
| | - Zhiqi Chen
- Medical College of Hubei Enshi College, Enshi, China
| | - Yang Xiang
- Medical College of Hubei Enshi College, Enshi, China
- Institute of anti-rheumatism Tujia Medicine, Enshi, China
- Hubei Provincial Key Laboratory of Rheumatic Disease Development and Intervention, Enshi, China
| | - Hao Wu
- Medical College of Hubei Enshi College, Enshi, China
- Institute of anti-rheumatism Tujia Medicine, Enshi, China
- Hubei Provincial Key Laboratory of Rheumatic Disease Development and Intervention, Enshi, China
| |
Collapse
|
36
|
Zhou L, Yang Y, Fu X, Xia B, Li C, Lu C, Qi Y, Zhang H, Liu T. The protective effect and molecular mechanism of glycyrrhizic acid glycosides against Tripterygium glycosides induced nephrotoxicity based on the RhoA/ROCK1 signalling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117100. [PMID: 37648177 DOI: 10.1016/j.jep.2023.117100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/27/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygium glycosides (TG), which are extracted from the traditional Chinese medicine, Tripterygium wilfordii Hook F. (TwHF), has promising applications in the treatment of renal diseases; however, since its active components exerts bidirectional kidney toxicity, its clinical application is severely restricted. AIM OF THE STUDY Recent investigations have demonstrated definite toxicity-reducing effects from glycyrrhizic acid glycosides (GA) when combined with TG; however, the mechanism remains unclear. To our knowledge, this is the first study to investigate the specific molecular mechanism by which GA alleviates TG-induced renal toxicity from the perspective of tight junctions. MATERIALS AND METHODS Dynamic analyses, which investigated the changes in kidney toxicity biomarkers for different combinations and concentrations of TG and GA, were conducted for three weeks on SD rats and renal tissue structural changes were examined after three weeks of administration. Additionally, the transcription and translation levels of the relevant tight junctions and RhoA/ROCK1/MLC signalling proteins were analysed in HK-2 cells. RESULTS Our study showed that TG can cause transient tubulotoxicity at certain doses, and that the combined application of GA and TG can repair tight junction structures by regulating the key factors in the RhoA/ROCK1/MLC signalling pathway, thus reducing TG-induced nephrotoxicity. CONCLUSIONS Overall, this study provides a new strategy to reduce TG-induced toxicity by protecting renal tight junctions.
Collapse
Affiliation(s)
- Liu Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China.
| | - Yifei Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China.
| | - Xiaotong Fu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China.
| | - Bing Xia
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China.
| | - Chun Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China.
| | - Chenna Lu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China.
| | - Ying Qi
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China.
| | - Haijing Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China.
| | - Ting Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China.
| |
Collapse
|
37
|
Yu Q, Xu C, Song J, Jin Y, Gao X. Mechanisms of Traditional Chinese medicine/natural medicine in HR-positive Breast Cancer: A comprehensive Literature Review. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117322. [PMID: 37866466 DOI: 10.1016/j.jep.2023.117322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 10/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE With the emergence of endocrine resistance, the survival and good prognosis of HR-positive breast cancer (HR + BC) patients are threatened. As a common complementary and alternative therapy in cancer treatment, traditional Chinese medicine (TCM) has been widely used, and its internal mechanisms have been increasingly explored. AIM OF THE REVIEW In this review, the development status and achievements in understanding of the mechanisms related to the anti-invasion and anti-metastasis effects of TCM against HR + BC and the reversal of endocrine drug resistance by TCM in recent years have been summarized to provide ideas for antitumour research on the active components of TCM/natural medicine. METHODS We searched the electronic databases PubMed, Web of Science, and China National Knowledge Infrastructure database (CNKI) (from inception to July 2023) with the key words "HR-positive breast cancer" or "HR-positive breast carcinoma", "HR + BC" and "traditional Chinese medicine", "TCM", or "natural plant", "herb", etc., with the aim of elucidating the intrinsic mechanisms of traditional Chinese medicine and natural medicine in the treatment of HR + BC. RESULTS TCM/natural medicine monomers and formulas can regulate the expression of related genes and proteins through the PI3K/AKT, JAK2/STAT3, MAPK, Wnt and other signalling pathways, inhibit the proliferation and metastasis of HR + BC tumours, play a synergistic role in combination with endocrine drugs, and reverse endocrine drug resistance. CONCLUSION The wide variety of TCM/natural medicine components makes the research and development of new methods of TCM for BC treatments more selective and innovative. Although progress has been made on research on TCM/natural medicine, there are still many problems in clinical and basic experimental designs, and more in-depth scientific explorations and research are still needed.
Collapse
Affiliation(s)
- Qinghong Yu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| | - Chuchu Xu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| | - Jiaqing Song
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| | - Ying Jin
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| | - Xiufei Gao
- The First Affiliated Hospital of Zhejiang Chinese Medical University, NO. 54 Youdian Road, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
38
|
Liu X, Lv T, Li X, Xue J, Lin L, Lu L, Li X, Yang Y, Wu Y, Wei Q, Cao W, Li T. Comprehensive transcriptomic analyses identify the immunosuppressive effects of LLDT-8 in ART-treated SIV-infected rhesus macaques. Int Immunopharmacol 2024; 126:111173. [PMID: 37984249 DOI: 10.1016/j.intimp.2023.111173] [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: 07/10/2023] [Revised: 10/15/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Chronic immune activation plays a significant role in the pathogenesis and disease progression of human immunodeficiency virus (HIV), and the existing interventions to address this issue are limited. In a phase II clinical trial, (5R)-5-hydroxytriptolide (LLDT-8) demonstrated promising potential in enhancing CD4+ T cell recovery. However, the therapeutical effects of LLDT-8 remained to be systemic explored. METHODS To assess the treatment effects of LLDT-8, we conducted flow cytometry and RNA-seq analyses on eight Chinese rhesus monkeys infected with simian immunodeficiency virus (SIV). Additionally, we performed comprehensive transcriptomic analyses, including cross-sectional and longitudinal differentially expressed gene (DEG) analysis, gene set enrichment analysis (GSEA), weighted gene co-expression network analysis (WGCNA), and deconvolution analysis using peripheral blood mononuclear cell (PBMC) samples from 14-time points. These findings were further validated with RNA-seq analysis on patients who received LLDT-8 treatment, along with in vitro cellular experiments using human PBMCs. RESULTS Flow cytometry analysis revealed that LLDT-8 treatment significantly reduced the percentage of HLA-DR+CD38+CD8+ T cells in SIV-infected rhesus monkeys (P < 0.001). The cross-sectional and longitudinal analysis identified 2531 and 1809 DEGs, respectively. GSEA analysis indicated that LLDT-8 treatment led to significant downregulation of proliferation-related pathways, such as E2F targets, G2M checkpoint, and mitotic spindle pathways. WGCNA analysis identified two modules and 202 hub genes associated with CD8 activation levels. Deconvolution analysis showed a significant decrease in the proportion of CD8+ T cells and activated CD4+ T cells during LLDT-8 treatment. Gene ontology results demonstrated that the common DEGs between LLDT-8-treated patients and rhesus monkeys were primarily enriched in cell activation and cell cycle progression. Furthermore, in vitro cellular experiments validated the consistent impact of LLDT-8 in inhibiting proliferation, activation (HLA-DR and CD38 expression), exhaustion (PD-1 expression), and IFN-γ production in human CD4+ and CD8+ T cells. CONCLUSION LLDT-8 exhibited notable efficacy in alleviating immune activation in both an in vivo animal model and in vitro human cell experiments. These findings suggest that LLDT-8 may hold potential as a drug for managing systemic immune activation associated with SIV/HIV infection, warranting further prospective clinical exploration.
Collapse
Affiliation(s)
- Xiaosheng Liu
- Tsinghua-Peking Center for Life Sciences, Beijing, China; Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China; Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Tingxia Lv
- Department of Infectious Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiuxia Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| | - Jing Xue
- Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Ling Lin
- Department of Infectious Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lianfeng Lu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaodi Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Yang
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuanni Wu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Qiang Wei
- Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Wei Cao
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| | - Taisheng Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China; Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China; Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| |
Collapse
|
39
|
Dong X, Zhu LW, Zhang Z, Cao R, Liu P, Shu X, Cao X, Hu Y, Bao X, Xu L, Li C, Xu Y. LLDT-8 ameliorates experimental autoimmune encephalomyelitis by mediating macrophage functions in the priming stage. Eur J Pharmacol 2024; 962:176201. [PMID: 37984728 DOI: 10.1016/j.ejphar.2023.176201] [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: 07/05/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/22/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease in the central nervous system caused by T cell activation mediated by peripheral macrophages, resulting in severe neurological deficits and disability. Due to the currently limited and expensive treatments for MS, we here introduce an economic Chinese medicine extract, (5R)-5-Hydroxytriptolide (LLDT-8), which shows low toxicity and high immunosuppressive activity. We used the widely accepted mouse model of MS, experimental autoimmune encephalomyelitis (EAE), to examine the immunosuppressive effect of LLDT-8 in vivo. Through the RNA-sequence analysis of peripheral macrophages in EAE mice, we discovered that LLDT-8 alleviates the symptoms of EAE by inhibiting the proinflammatory effect of macrophages, thereby blocking the activation and proliferation of T cells. In all, we found that LLDT-8 could be a potential treatment for MS.
Collapse
Affiliation(s)
- Xiaohong Dong
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China; Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210008, China; The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Li-Wen Zhu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China; Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Zhi Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China; Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Runjing Cao
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210008, China; Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Pinyi Liu
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Xin Shu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China; Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Xiang Cao
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Yujie Hu
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Xinyu Bao
- Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Lushan Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China; Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Chenggang Li
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China; Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China; Department of Neurology, Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China.
| |
Collapse
|
40
|
Gao Y, Wang X, Gao Y, Bai J, Zhao Y, Wang R, Wang H, Zhu G, Wang X, Han X, Zhang Y, Wang H. The Lnc-ENST00000602558/IGF1 axis as a predictor of response to treatment with tripterygium glycosides in rheumatoid arthritis patients. Immun Inflamm Dis 2024; 12:e1098. [PMID: 38270302 PMCID: PMC10790680 DOI: 10.1002/iid3.1098] [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: 06/26/2023] [Revised: 11/04/2023] [Accepted: 11/12/2023] [Indexed: 01/26/2024] Open
Abstract
AIMS Growing clinical evidence suggests that not all patients with rheumatoid arthritis (RA) benefit to the same extent by treatment with tripterygium glycoside (TG), which highlights the need to identify RA-related genes that can be used to predict drug responses. In addition, single genes as markers of RA are not sufficiently accurate for use as predictors. Therefore, there is a need to identify paired expression genes that can serve as biomarkers for predicting the therapeutic effects of TG tablets in RA. METHODS A total of 17 pairs of co-expressed genes were identified as candidates for predicting an RA patient's response to TG therapy, and genes involved in the Lnc-ENST00000602558/GF1 axis were selected for that purpose. A partial-least-squares (PLS)-based model was constructed based on the expression levels of Lnc-ENST00000602558/IGF1 in peripheral blood. The model showed high efficiency for predicting an RA patient's response to TG tablets. RESULTS Our data confirmed that genes co-expressed in the Lnc-ENST00000602558/IGF1 axis mediate the efficacy of TG in RA treatment, reduce tumor necrosis factor-α induced IGF1 expression, and decrease the inflammatory response of MH7a cells. CONCLUSION We found that genes expressed in the Lnc-ENST00000602558/IGF1 axis may be useful for identifying RA patients who will not respond to TG treatment. Our findings provide a rationale for the individualized treatment of RA in clinical settings.
Collapse
Affiliation(s)
- Yang Gao
- Department of Chinese MedicineTsinghua University HospitalBeijingChina
| | - Xiaoyue Wang
- Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Yanfeng Gao
- Department of DermatologyThe Second Mongolian Medical Hospital of Traditional Chinese MedicineChi Feng CityInner MongoliaChina
| | - Jian Bai
- Guizhou University of Traditional Chinese Medicine Graduate SchoolGuiyang CityGuizhouChina
| | - Yanpeng Zhao
- Guizhou University of Traditional Chinese Medicine Graduate SchoolGuiyang CityGuizhouChina
| | - Renyi Wang
- Guizhou University of Traditional Chinese Medicine Graduate SchoolGuiyang CityGuizhouChina
| | - Hanzhou Wang
- Department of Rheumatology, Guang'anmen HospitalChina Medical SciencesBeijingChina
| | - Guangzhao Zhu
- Department of RheumatologyQinghai Hospital of TCMXining CityQinghaiChina
| | - Xixi Wang
- Guizhou University of Traditional Chinese Medicine Graduate SchoolGuiyang CityGuizhouChina
| | - Xiaochen Han
- Department of Internal MedicineBeijing Fengsheng Hospital of Traditional Medical Traumatology & OrthopedicsBeijingChina
| | - Yanqiong Zhang
- Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Hailong Wang
- Department of Rheumatology, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
| |
Collapse
|
41
|
Luo F, Gau SY, Wu YX, Liao HL, Tang F, Zhong Q, Huang Y, Hou L, Liu ZQ, Cai JL, Cao YP, Lu DM, An Y, Lan WY, Liu C, Chen CM, Jia ET, Yao XM, Wei JCC, Ma WK. Efficacy of adalimumab combined with Tripterygium wilfordii Hook F in the treatment of patient with rheumatoid arthritis: A multicenter, open-label, randomized-controlled trial. Int J Rheum Dis 2024; 27:e15031. [PMID: 38287544 DOI: 10.1111/1756-185x.15031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/08/2023] [Accepted: 12/23/2023] [Indexed: 01/31/2024]
Abstract
OBJECTIVES To evaluate the efficacy and safety of adalimumab (ADA) combined with Tripterygium wilfordii Hook F (TwHF) in the treatment of methotrexate (MTX)-inadequate response patients with rheumatoid arthritis (RA). METHODS In this multicenter, open-label, randomized controlled clinical trial, 64 RA patients with inadequate response to MTX were 1:1 randomly assigned into treatment or control groups. The treatment group was treated with ADA in combination with TwHF, and the control group was treated with ADA in combination with MTX for 24 weeks. The primary endpoint was the percentage of patients having low disease activity (2.6 ≤ DAS28-ESR < 3.2) and remission rates (DAS28-ESR < 2.6) at week 24. RESULTS In total, 53 of the 64 patients (82.8%) completed this 24-week clinical trial. By intent-to-treat (ITT) analysis, a comparable outcome was observed between the two groups. The percentage of patients achieving low disease activity in the treatment group and control group were 43.8% and 46.9% (95% CI, 21.28 to 27.48, p = .802). Percentage of patients achieving low disease activity rates were respectively 28.1% and 31.3% in the treatment group and control group (95% CI, 19.18 to 25.58, p = .784). In per-protocol (PP) analysis, the results were consistent with the ITT model. The incidence of adverse events was comparable between the two groups. CONCLUSIONS There were no significant differences in efficacy and safety between ADA combined with TwHF versus ADA combined with MTX in the treatment of RA. TwHF might be an alternative treatment for RA patients who are intolerant to MTX.
Collapse
Affiliation(s)
- Feng Luo
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Shuo-Yan Gau
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Xia Wu
- Department of Rheumatology and Immunology, QiandongnanZhou People's Hospital, Kaili, China
| | - Hou-Li Liao
- Department of Rheumatism and Immunology, Xingyi People's Hospital, Xingyi, China
| | - Fang Tang
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qin Zhong
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Ying Huang
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Lei Hou
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zheng-Qi Liu
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jin-Long Cai
- Department of Rheumatology and Immunology, QiandongnanZhou People's Hospital, Kaili, China
| | - Yue-Peng Cao
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Dao-Min Lu
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yang An
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Wei-Ya Lan
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Can Liu
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Chang-Ming Chen
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Er-Tao Jia
- The Department of Rheumatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Xue-Ming Yao
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Department of Liupanshui Hospital of Traditional Chinese Medicine, Liupanshui, China
| | - James Cheng-Chung Wei
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Rheumatology & Immunology, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Wu-Kai Ma
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
42
|
Huang Y, Peng Y, Li H, Li C, Wu Y, Wang X, Chang J, Miao C. Wilforine inhibits rheumatoid arthritis pathology through the Wnt11/β-catenin signaling pathway axis. Arthritis Res Ther 2023; 25:243. [PMID: 38098062 PMCID: PMC10720104 DOI: 10.1186/s13075-023-03224-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Wilforine (WFR) is a monomeric compound of the anti-RA plant Tripterygium wilfordii Hook. f. (TwHF). Whether WFR has anti-RA effect, its molecular mechanism has not been elucidated. AIM OF THE STUDY Our study aims to clarify how WFR inhibits fibroblast-like synovial cells (FLS) activation and improves RA through Wnt11 action on the Wnt11/β-catenin signaling pathway. METHODS The therapeutic effect of WFR on collagen-induced arthritis (CIA) rats was evaluated using methods such as rat arthritis score. The inhibitory effects and signaling pathways of WFR on the proliferation and inflammatory response of CIA FLS and RA FLS were studied using ELISA, CCK-8, RT-qPCR, Western blot, and immunofluorescence methods. RESULTS WFR could effectively alleviate the arthritis symptoms of CIA rats; reduce the levels of IL-6, IL-1β, and TNF-α in the peripheral blood of CIA rats; and inhibit the expression of MMP3 and fibronectin. The data showed that WFR has a significant inhibitory effect on FLS proliferation. Furthermore, WFR inhibited the activation of Wnt/β-catenin signaling pathway and decreased the expression of Wnt11, β-catenin, CCND1, GSK-3β, and c-Myc, while the effects of WFR were reversed after overexpression of Wnt11. CONCLUSIONS WFR improves RA by inhibiting the Wnt11/β-catenin signaling pathway, and Wnt11 is the direct target of WFR. This study provides a new molecular mechanism for WFR to improve RA and contributes to the clinical promotion of WFR.
Collapse
Affiliation(s)
- Yurong Huang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui Province, China
| | - Yanhui Peng
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui Province, China
| | - Hui Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui Province, China
| | - Chen Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui Province, China
| | - Yajie Wu
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui Province, China
| | - Xiaomei Wang
- Department of Humanistic Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China.
| | - Jun Chang
- Department of Orthopaedics, the First Affiliated Hospital, Anhui Medical University, Hefei, 230032, China.
- Anhui Public Health Clinical Center, Hefei, China.
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui Province, China.
| |
Collapse
|
43
|
Zhang L, Jiang S, Guan Z, Huang J, Yin Z, Tan G, Wang Y, Zhao Z, Huang M, Jin J. Effect of Salvia miltiorrhiza Bunge extracts on improving the efficacy and reducing the toxicity of Tripterygium wilfordii polyglycosides in the treatment of rheumatoid arthritis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116782. [PMID: 37321427 DOI: 10.1016/j.jep.2023.116782] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygium wilfordii polyglycosides (TWP), extracted from the traditional Chinese herb Tripterygium wilfordii, has been widely used in the treatment of rheumatoid arthritis (RA). However, the toxicity of TWP to a variety of organs such as liver, kidney and testis greatly limits its clinical application. Salvia miltiorrhiza Bunge is often used in the treatment of RA due to its blood circulation promoting, stasis resolving, and anti-inflammatory effects. Salvia miltiorrhiza Bunge has also been reported to possess multiple organ protective effects. AIM OF THE STUDY To investigate the influences of two main components of Salviorrhiza miltiorrhiza Bunge, hydrophilic salvianolic acids (SA) and lipophilic tanshinones (Tan), on the efficacy and toxicity of TWP in treating RA and to explore the underlying mechanisms. MATERIALS AND METHODS SA and Tan were extracted from Salvia miltiorrhiza Bunge and the extracts were quantitated by HPLC and identified by UPLC-Q/TOF-MS. Then, a collagen-induced arthritis (CIA) rat model was established using bovine type II collagen (CII) and incomplete Freund's adjuvant (IFA). CIA rats were treated with TWP and/or SA/Tan. After 21 days of continuous treatment, arthritis symptoms and organs toxicity were evaluated. Meanwhile, serum metabolomics were investigated by the UPLC-Q/TOF-MS to understand the underlying mechanism. RESULTS SA and Tan extracts could significantly alleviate arthritis symptoms in CIA rats and decrease the serum levels of inflammatory factors TNF-α, IL-1β and IL-6 when combined with TWP. Meanwhile, both extracts alleviated injury of liver, kidney and testis caused by TWP, and the hydrophilic extract SA was superior. Moreover, a total of 38 endogenous differential metabolites were identified between the CIA model group and the TWP group, among which 33 metabolites were significantly recovered after the combination of SA or Tan. Metabolic pathway analysis showed that SA and Tan can affect metabolic pathways including linoleic acid metabolism, glycerophospholipid metabolism, sphingolipid metabolism and steroid biosynthesis metabolism pathway. CONCLUSIONS Our findings indicated for the first time that two Salviorrhiza miltiorrhiza Bunge extracts could improve the efficacy and reduce the toxicity of TWP in the treatment of RA by adjusting metabolic pathways, and the hydrophilic extract SA was superior.
Collapse
Affiliation(s)
- Lei Zhang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shiqin Jiang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zehao Guan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Junyuan Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhaokun Yin
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Guoyao Tan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yuanyuan Wang
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhongxiang Zhao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jing Jin
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
44
|
Wen J, Liu J, Wan L, Wang F. Long noncoding RNA/circular RNA regulates competitive endogenous RNA networks in rheumatoid arthritis: molecular mechanisms and traditional Chinese medicine therapeutic significances. Ann Med 2023; 55:973-989. [PMID: 36905646 PMCID: PMC10795602 DOI: 10.1080/07853890.2023.2172605] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/20/2023] [Indexed: 03/13/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic and autoimmune disease that is mainly featured abnormal fibroblast-like synoviocyte (FLS) proliferation and inflammatory cell infiltration. Abnormal expression or function of long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) are closely related to human diseases, including RA. There has been increasing evidence showing that in the competitive endogenous RNA (ceRNA) networks, both lncRNA and circRNA are vital in the biological functions of cells. Nevertheless, the exact mechanism of ceRNA in RA remains to be investigated. Herein, we summarized the molecular potencies of lncRNA/circRNA-mediated ceRNA networks in RA, with emphasis on the phenotypic regulation of ceRNA in the progression of RA, including regulation of proliferation, invasion, inflammation and apoptosis, as well as the role of ceRNA in traditional Chinese medicine (TCM) in the treatment of RA. In addition, we also discussed the future direction and potential clinical value of ceRNA in the treatment of RA, which may provide potential reference value for clinical trials of TCM therapy for the treatment of RA.Key messagesLong noncoding RNA/circular RNA can work as the competitive endogenous RNA sponge and participate in the pathogenesis of rheumatoid arthritis.Traditional Chinese medicine and its agents have shown potential roles in the prevention and treatment of rheumatoid arthritis via competitive endogenous RNA.
Collapse
Affiliation(s)
- Jianting Wen
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, Anhui, China
- Key Laboratory of Xin’an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jian Liu
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, Anhui, China
- Department of Internal Medicine Application Foundation Research and Development, Anhui Province—Key Laboratory of Modern Chinese Medicine, Hefei, Anhui, China
| | - Lei Wan
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, Anhui, China
- Department of Internal Medicine Application Foundation Research and Development, Anhui Province—Key Laboratory of Modern Chinese Medicine, Hefei, Anhui, China
| | - Fanfan Wang
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, Anhui, China
- Department of Internal Medicine Application Foundation Research and Development, Anhui Province—Key Laboratory of Modern Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
45
|
Zhou J, Li M, Yu Z, Li C, Zhou L, Zhou X. Protective effect of Qingluotongbi formula against Tripterygium wilfordii induced liver injury in mice by improving fatty acid β-oxidation and mitochondrial biosynthesis. PHARMACEUTICAL BIOLOGY 2023; 61:80-88. [PMID: 36541729 PMCID: PMC9788700 DOI: 10.1080/13880209.2022.2157842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/21/2022] [Accepted: 12/07/2022] [Indexed: 06/17/2023]
Abstract
CONTEXT Qingluotongbi formula (QLT) is a Chinese medicine compound consisting of Tripterygium wilfordii Hook. f. (Celastraceae, TW), Panax notoginseng (Burkill) F.H.Chen (Araliaceae, PN), Rehmannia glutinosa (Gaertn.) DC. (Orobanchaceae, RG), Sinomenium acutum (Thunb.) Rehder & E.H. Wilson (Menispermaceae, SA), and Bombyx mori L. (Bombycidae, BM). OBJECTIVE This study investigated the protective effect and possible mechanism of QLT against TW-induced liver injury in mice. MATERIALS AND METHODS To establish the model of TW-induced liver injury in mice, C57BL/6J mice were randomly divided into 4 groups: control group, low-dose TW group, middle-dose TW group, and high-dose TW group. To observe the effects of QLT and its individual ingredients against TW-induced liver injury, C57BL/6J mice were randomly divided into 7 groups: control group, TW group, QLT group, PN group, RG group, SA group, BM group.After administration for 7 days, C57BL/6J mice were tested for biochemical indicators and liver pathological changes. Then, we evaluated the mitochondrial function and analysed the gene and protein expression related to the peroxisome proliferator-activated receptor alpha (PPARα)/peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) pathway by quantitative real-time PCR (qRT-PCR) and Western blotting. RESULTS Compared with the control group (0.30 ± 0.35), TW significantly increased mice liver histological score (L, 0.95 ± 1.14; M, 1.25 ± 1.16; H, 4.00 ± 1.13). QLT and its ingredients significantly improved the pathology scores (CON, 0.63 ± 0.74; TW, 4.19 ± 1.53; QLT, 1.56 ± 0.62; PN, 1.94 ± 0.68; RG, 2.75 ± 1.39; SA, 4.13 ± 0.99; BM, 4.13 ± 0.99). Western blot and qRT-PCR analysis revealed that QLT and its ingredients reversed TW-induced suppression of PPARα/PGC1-α pathway.Discussion and conclusions: These findings provide valuable information for compound compatibility studies and TW clinical applications.
Collapse
Affiliation(s)
- Jie Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming Li
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Zhichao Yu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Changqing Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lingling Zhou
- Jiangsu Provincial Key Laboratory of Pharmacology and Safety Evaluation of Material Medical, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xueping Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
46
|
Felipe JL, Bonfá IS, Lossavaro PKMB, Lencina JS, B Carvalho D, Candeloro L, Ferreira GIS, das Neves AR, Souza MIL, Silva-Filho SE, Baroni ACM, Toffoli-Kadri MC. 1,4-Diaryl-1,2,3-triazole neolignan-celecoxib hybrids inhibit experimental arthritis induced by zymosan. Inflammopharmacology 2023; 31:3227-3241. [PMID: 37806984 DOI: 10.1007/s10787-023-01345-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes cartilage damage. Anti-inflammatories are widely used in the management of RA, but they can have side effects such as gastrointestinal and/or cardiovascular disorders. Studies published by our group showed that the synthesis of hybrid triazole analogs neolignan-celecoxib containing the substituent groups sulfonamide (L15) or carboxylic acid (L18) exhibited anti-inflammatory activity in an acute model of inflammation, inhibited expression of P-selectin related to platelet activation and did not induce gastric ulcer, minimizing the related side effects. In continuation, the present study evaluated the anti-inflammatory effects of these analogs in an experimental model of arthritis and on the functions of one of the important cells in this process, macrophages. Mechanical hyperalgesia, joint edema, leukocyte recruitment to the joint and damage to cartilage in experimental arthritis and cytotoxicity, spread of disease, phagocytic activity and nitric oxide (NO) and hydrogen peroxide production by macrophages were evaluated. Pre-treatment with L15 and L18 reduced mechanical hyperalgesia, joint edema and the influx of leukocytes into the joint cavity after different periods of the stimulus. The histological evaluation of the joint showed that L15 and L18 reduced cartilage damage and there was no formation of rheumatoid pannus. Furthermore, L15 and L18 were non-cytotoxic. The analogs inhibited the spreading, the production of NO and hydrogen peroxide. L15 decreased the phagocytosis. Therefore, L15 and L18 may be potential therapeutic prototypes to treat chronic inflammatory diseases such as RA.
Collapse
Affiliation(s)
- Josyelen L Felipe
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, 79070-900, Brazil
| | - Iluska S Bonfá
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, 79070-900, Brazil
| | - Paloma K M B Lossavaro
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, 79070-900, Brazil
| | - Joyce S Lencina
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, 79070-900, Brazil
| | - Diego B Carvalho
- Laboratory of Synthesis and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, Brazil
| | - Luciane Candeloro
- Laboratory of Hystology, Institute of Biosciences, Federal University of Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Giovanni I S Ferreira
- Laboratory of Hystology, Institute of Biosciences, Federal University of Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Amarith R das Neves
- Laboratory of Synthesis and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, Brazil
| | - Maria Inês L Souza
- Department of Biophysiopharmacology, Institute of Biosciences, Federal University of Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Saulo E Silva-Filho
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, 79070-900, Brazil
| | - Adriano C M Baroni
- Laboratory of Synthesis and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, Brazil.
| | - Mônica C Toffoli-Kadri
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, 79070-900, Brazil.
| |
Collapse
|
47
|
Jiang X, Yuan C, Ding R, Lu D, Peng X, Dong Z, Zhu C, Lin Y, Wu C, Xie Q. Toxic metabolites and metabolic soft spots of celastrol based on glutathione metabolic capture and high-resolution mass spectrometry. Expert Opin Drug Metab Toxicol 2023; 19:1023-1032. [PMID: 38145500 DOI: 10.1080/17425255.2023.2294042] [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: 10/10/2023] [Accepted: 12/06/2023] [Indexed: 12/27/2023]
Abstract
BACKGROUND Celastrol is known as one of the most medicinally valuable compounds. However, the pharmaceutical application of celastrol is significantly limited due to high toxicity, while there are few reports on the mechanism of toxicity. METHODS This study searched for possible toxic metabolites through phase I in vitro metabolism and glutathione capture experiments. Then in vivo metabolism experiments in mice and rats were conducted to look for metabolites in vivo. Finally, mice in vivo toxicity experiment was conducted to verify the toxicity of different doses of celastrol to mice. RESULTS In the in vivo and in vitro metabolism experiments, we found 7 phase I metabolites in vitro, 9 glutathione conjugation metabolites in vitro, and 20 metabolites in vivo. The metabolic soft points of celastrol could be the quinone methyl structure at C3-OH and C6. In vivo toxicity experiments show that celastrol causes weight loss, diarrhea, gastrointestinal tract and liver inflammation in mice. CONCLUSIONS This study analyzed the metabolites and possible metabolic soft spots of celastrol, and its hepatotoxicity and gastrointestinal toxicity were demonstrated through in vivo studies for the first time. The results might provide an important basis for potential structural modification to increase the druggability of celastrol.
Collapse
Affiliation(s)
- Xiaojuan Jiang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Caixia Yuan
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Rong Ding
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Di Lu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Xiaoyu Peng
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Zhihao Dong
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Chunyan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yihua Lin
- Department of Respiratory and Critical Care Medicine, The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Qiang Xie
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| |
Collapse
|
48
|
Jin F, Ni X, Yu S, Jiang X, Zhou J, Mao D, Liu Y, Wu F. Network pharmacology‑based investigation of potential targets of triptonodiol acting on non-small-cell lung cancer. Eur J Med Res 2023; 28:547. [PMID: 38017514 PMCID: PMC10683219 DOI: 10.1186/s40001-023-01453-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 10/17/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Triptonodiol is a very promising antitumor drug candidate extracted from the Chinese herbal remedy Tripterygium wilfordii Hook. F., and related studies are underway. METHODS To explore the mechanism of triptonodiol for lung cancer treatment, we used network pharmacology, molecular docking, and ultimately protein validation. Gene ontology (GO) analysis and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway enrichment analysis were performed through the David database. Molecular docking was performed using PyMoL2.3.0 and AutoDock Vina software. After screening, the major targets of triptonodiol were identified for the treatment of lung cancer. Target networks were established, Protein-protein interaction (PPI) network topology was analyzed, then KEGG pathway enrichment analysis was performed. Useful proteins were screened by survival analysis, and Western blot analysis was performed. RESULTS Triptonodiol may regulate cell proliferation, drug resistance, metastasis, anti-apoptosis, etc., by acting on glycogen synthase kinase 3 beta (GSK3B), protein kinase C (PKC), p21-activated kinase (PAK), and other processes. KEGG pathway enrichment analysis showed that these targets were associated with tumor, erythroblastic oncogene B (ErbB) signaling, protein phosphorylation, kinase activity, etc. Molecular docking showed that the target protein GSK has good binding activity to the main active component of triptonodiol. The protein abundance of GSK3B was significantly downregulated in non-small-cell lung cancer cells H1299 and A549 treated with triptonodiol for 24 h. CONCLUSION The cellular-level studies combined with network pharmacology and molecular docking approaches provide new ideas for the development and therapeutic application of triptonodiol, and identify it as a potential GSK inhibitor.
Collapse
Affiliation(s)
- Feng Jin
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Xiaochen Ni
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Shilong Yu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou, 225001, People's Republic of China
| | - Xiaomin Jiang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, People's Republic of China
| | - Jun Zhou
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Defang Mao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Yanqing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, People's Republic of China
| | - Feng Wu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
49
|
Gao J, Ma L, Liu Y, Tu L, Wu X, Wang J, Li D, Zhang X, Gao W, Zhang Y, Liu C. CYP72D19 from Tripterygium wilfordii catalyzes C-2 hydroxylation of abietane-type diterpenoids. PLANT CELL REPORTS 2023; 42:1733-1744. [PMID: 37615706 DOI: 10.1007/s00299-023-03059-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023]
Abstract
KEY MESSAGE CYP72D19, the first functional gene of the CYP72D subfamily, catalyzes the C-2 hydroxylation of abietane-type diterpenoids. The abietane-type diterpenoids, e.g., triptolide, tripdiolide, and 2-epitripdiolide, are the main natural products for the anti-tumor, anti-inflammatory, and immunosuppressive activities of Tripterygium wilfordii, while their biosynthetic pathways are not resolved. Here, we cloned and characterized the CYP72D19-catalyzed C-2 hydroxylation of dehydroabietic acid, a compound that has been proven to be a biosynthetic intermediate in triptolide biosynthesis. Through molecular docking and site-directed mutagenesis, L386, L387, and I493 near the active pocket were found to have an important effect on the enzyme activity, which also indicates that steric hindrance of residues plays an important role in function. In addition, CYP72D19 also catalyzed a variety of abietane-type diterpenoids with benzene ring, presumably because the benzene ring of the substrate molecule stabilized the C-ring, allowing the protein and the substrate to form a relatively stable spatial structure. This is the first demonstration of CYP72D subfamily gene function. Our research provides important genetic elements for the structural modification of active ingredients and the heterologous production of other 2-hydroxyl abietane-type natural products.
Collapse
Affiliation(s)
- Jie Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- National Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Lin Ma
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Yuan Liu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Lichan Tu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Xiaoyi Wu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Jian Wang
- National Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Dan Li
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xianan Zhang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Wei Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Yifeng Zhang
- National Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Changli Liu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
50
|
Huang Y, Xue Q, Chang J, Wang Y, Cheng C, Xu S, Wang X, Miao C. M6A methylation modification in autoimmune diseases, a promising treatment strategy based on epigenetics. Arthritis Res Ther 2023; 25:189. [PMID: 37784134 PMCID: PMC10544321 DOI: 10.1186/s13075-023-03149-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 08/24/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) methylation modification is involved in the regulation of various biological processes, including inflammation, antitumor, and antiviral immunity. However, the role of m6A modification in the pathogenesis of autoimmune diseases has been rarely reported. METHODS Based on a description of m6A modification and the corresponding research methods, this review systematically summarizes current insights into the mechanism of m6A methylation modification in autoimmune diseases, especially its contribution to rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). RESULTS By regulating different biological processes, m6A methylation is involved in the pathogenesis of autoimmune diseases and provides a promising biomarker for the diagnosis and treatment of such diseases. Notably, m6A methylation modification is involved in regulating a variety of immune cells and mitochondrial energy metabolism. In addition, m6A methylation modification plays a role in the pathological processes of RA, and m6A methylation-related genes can be used as potential targets in RA therapy. CONCLUSIONS M6A methylation modification plays an important role in autoimmune pathological processes such as RA and SLE and represents a promising new target for clinical diagnosis and treatment, providing new ideas for the treatment of autoimmune diseases by targeting m6A modification-related pathways.
Collapse
Affiliation(s)
- Yurong Huang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1 Qianjiang Road, Xinzhan District, Hefei, 230012, Anhui Province, China
| | - Qiuyun Xue
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1 Qianjiang Road, Xinzhan District, Hefei, 230012, Anhui Province, China
| | - Jun Chang
- Department of Orthopaedics, the First Affiliated Hospital, Anhui Medical University, Hefei, 230032, China.
- Anhui Public Health Clinical Center, Hefei, China.
| | - Yuting Wang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1 Qianjiang Road, Xinzhan District, Hefei, 230012, Anhui Province, China
| | - Chenglong Cheng
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1 Qianjiang Road, Xinzhan District, Hefei, 230012, Anhui Province, China
| | - Suowen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230027, China
| | - Xiao Wang
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China.
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1 Qianjiang Road, Xinzhan District, Hefei, 230012, Anhui Province, China.
| |
Collapse
|