1
|
Zhang S, Tan H, Zhang Z, Wang Z, Zhang C, Zhang J, Jia E. Sinomenine alleviates gouty inflammation by inhibiting macrophage M1 polarization and neutrophil extracellular trap formation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04161-4. [PMID: 40381009 DOI: 10.1007/s00210-025-04161-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/08/2025] [Indexed: 05/19/2025]
Abstract
Gout is a common inflammatory arthropathy characterized by the deposition of monosodium urate (MSU) crystals, leading to severe pain and swelling. Sinomenine (SIN) is the major active component of Sinomenium acutum. SIN has been demonstrated to exert preventive and therapeutic effects on arthritis in cell-based, animal, and clinical studies. The present study focused on the efficacy and role of SIN in relieving symptoms of gouty inflammation in vivo and in vitro. The anti-inflammatory effects of SIN were evaluated in mice with MSU-induced air-pouch via hematoxylin-eosin (HE) staining, and enzyme-linked immunosorbent assay (ELISA). Transcriptomic analysis revealed that SIN modulates a range of inflammatory pathways associated with gout pathogenesis. Notably, the NOD-like receptor pathway and neutrophil extracellular trap (NET) formation were significantly enriched with the occurrence of gout and significantly improved after SIN treatment. THP-1 macrophages were stimulated with PBS or MSU, with or without SIN. Immunofluorescence (IF) and western blotting (WB) results indicated that SIN suppressed NOD-like receptor thermal protein domain associated protein 3 (NLRP3)/interleukin-1β (IL-1β) expression. Additionally, SIN inhibited macrophage M1 polarization and NET formation. In summary, SIN ameliorates gouty inflammation, likely by regulating the NLRP3/IL-1β pathway, M1 macrophage polarization, and NET formation. Thus, SIN is a promising drug for treating gout.
Collapse
Affiliation(s)
- Shan Zhang
- Shenzhen Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Shenzhen, 518033, People's Republic of China
| | - Haibo Tan
- Shenzhen Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Shenzhen, 518033, People's Republic of China
| | - Zhihao Zhang
- Department of Rheumatism, The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangdong Second Hospital of Traditional Chinese Medicine, Guangzhou, 510000, People's Republic of China
| | - Ziyu Wang
- Department of Rheumatism, The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangdong Second Hospital of Traditional Chinese Medicine, Guangzhou, 510000, People's Republic of China
| | - Chenyang Zhang
- Department of Rheumatism, The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangdong Second Hospital of Traditional Chinese Medicine, Guangzhou, 510000, People's Republic of China
| | - Jianyong Zhang
- Shenzhen Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Shenzhen, 518033, People's Republic of China
- Department of Rheumatism, The Fourth Clinical Medical College of Guangzhou, University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, People's Republic of China
| | - Ertao Jia
- Department of Rheumatism, The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangdong Second Hospital of Traditional Chinese Medicine, Guangzhou, 510000, People's Republic of China.
| |
Collapse
|
2
|
Tang J, Li S, Wang Y, Yuan M, Wan Y, Liang X, Guo L, Guo Y. Study of Folate-Modified Carboxymethyl Chitosan-Sinomenine-Curcumin Nanopolymer for Targeted Treatment of Rheumatoid Arthritis. Biomacromolecules 2025. [PMID: 40324022 DOI: 10.1021/acs.biomac.4c01701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Sinomenine hydrochloride (SH) has been clinically utilized for many years to treat rheumatoid arthritis (RA) in both oral and injectable forms. However, its low bioavailability, poor targeting, high dosage requirements, and side effects, present significant challenges. This study developed folic acid-carboxymethyl chitosan-modified sinomenine-curcumin nanopolymers (named SCNP) for the targeted treatment of RA, to reduce dosage and side effects. The design of SCNP employs folic acid (FA) as a targeting moiety, facilitating specific binding to the folate receptor (FR) on the surface of macrophages and enabling internalization into activated macrophages via endocytosis, thereby achieving targeted delivery to sites of inflammation. In a rat and cell model of RA, SCNP was found to decrease reactive oxygen species (ROS) and pro-inflammatory factors while increasing the anti-inflammatory factor IL-10 through the NF-κB/NLRP3 pathway. These findings indicate that SCNP has the potential to lower drug dosage, enhance therapeutic efficacy, and minimize side effects such as diarrhea and rash, thereby highlighting its promise as an inflammation-targeting nanopolymer.
Collapse
Affiliation(s)
- Jiamei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Sihui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yulu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Minghao Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yan Wan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xue Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yiping Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
3
|
Izzo AA, Stefanska B. Natural products and cancer: From drug discovery to prevention and therapy. Br J Pharmacol 2025; 182:2069-2074. [PMID: 40122586 DOI: 10.1111/bph.70014] [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: 03/25/2025] Open
Abstract
LINKED ARTICLES This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Angelo A Izzo
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Barbara Stefanska
- Faculty of Land and Food Systems, Food, Nutrition and Health Program, The University of British Columbia, Vancouver, Canada
| |
Collapse
|
4
|
He Y, Huang H, Li G, Zhang Y, He J, Lin Y, Wu F, Yan J, Cai X, Liu L. Safety Profile, Toxicokinetic, and Intestinal Absorption Differences of a Naturally-Derived Anti-Rheumatic Drug, Sinomenine Hydrochloride, in Normal and Arthritic Rats. Pharmaceutics 2025; 17:484. [PMID: 40284479 PMCID: PMC12030057 DOI: 10.3390/pharmaceutics17040484] [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: 02/27/2025] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objective: Sinomenine hydrochloride (SH), a natural anti-rheumatic drug derived from the Chinese medicinal plant Sinomenium acutum, demonstrates disease-modifying properties but lacks comprehensive safety and toxicokinetic (TK) comparisons between physiological and pathological states. This study evaluated SH's safety profile, TK parameters, and intestinal absorption differences in adjuvant-induced arthritis (AIA) and normal rats. Methods: Safety assessments determined median lethal doses (LD50) in female Sprague Dawley rats. TK parameters were analyzed via a validated ultrahigh performance liquid chromatography-tandem mass spectrometry approach after single oral administration of 600 mg/kg SH. Plasma protein binding (PPB) were measured using equilibrium dialysis. Intestinal absorption was evaluated through everted gut sac experiments, with P-glycoprotein (P-gp) inhibition tested via verapamil co-administration. Results: LD50 values revealed AIA rats tolerated SH better than normal rats (1179 vs. 805 mg/kg). TK analysis showed that Cmax, AUC(0-t), and AUC(0-∞) of SIN in normal rats were 2.01, 1.94, and 2.14 times higher than in AIA rats, respectively, while CL/F and V/F in AIA rats were 2.24 times greater. In addition, the PPB of SIN in normal rats was 2 times greater than that in AIA rats. AIA rats exhibited significantly lower SH absorption in the jejunum and ileum compared to normal rats. Notably, verapamil co-administration markedly increased SH absorption across most intestinal segments. Conclusions: Pathological states significantly alter SH's safety and TK profiles. Enhanced tolerance in AIA rats correlates with reduced intestinal absorption via altered P-gp activity and decreased PPB. These findings emphasize the necessity of disease-specific evaluations for optimizing SH's therapeutic safety in pathological contexts.
Collapse
Affiliation(s)
- Yini He
- Institute of Innovation and Applied Research in Chinese Medicine, School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.H.); (H.H.); (G.L.); (Y.Z.); (J.H.); (Y.L.); (J.Y.)
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Hong Huang
- Institute of Innovation and Applied Research in Chinese Medicine, School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.H.); (H.H.); (G.L.); (Y.Z.); (J.H.); (Y.L.); (J.Y.)
| | - Gejing Li
- Institute of Innovation and Applied Research in Chinese Medicine, School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.H.); (H.H.); (G.L.); (Y.Z.); (J.H.); (Y.L.); (J.Y.)
| | - Ye Zhang
- Institute of Innovation and Applied Research in Chinese Medicine, School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.H.); (H.H.); (G.L.); (Y.Z.); (J.H.); (Y.L.); (J.Y.)
| | - Junjie He
- Institute of Innovation and Applied Research in Chinese Medicine, School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.H.); (H.H.); (G.L.); (Y.Z.); (J.H.); (Y.L.); (J.Y.)
| | - Ye Lin
- Institute of Innovation and Applied Research in Chinese Medicine, School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.H.); (H.H.); (G.L.); (Y.Z.); (J.H.); (Y.L.); (J.Y.)
| | - Feichi Wu
- Hunan ZhenQin Pharmaceutical Group Co., Ltd., Huaihua 418000, China;
| | - Jianye Yan
- Institute of Innovation and Applied Research in Chinese Medicine, School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.H.); (H.H.); (G.L.); (Y.Z.); (J.H.); (Y.L.); (J.Y.)
| | - Xiong Cai
- Institute of Innovation and Applied Research in Chinese Medicine, School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.H.); (H.H.); (G.L.); (Y.Z.); (J.H.); (Y.L.); (J.Y.)
| | - Liang Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
5
|
Yang Q, Chen Q, Zhang KB, Liu Y, Zheng JC, Hu DX, Luo J. Sinomenine alleviates neuroinflammation in chronic cerebral hypoperfusion by promoting M2 microglial polarization and inhibiting neuronal pyroptosis via exosomal miRNA-223-3p. Acta Neuropathol Commun 2025; 13:48. [PMID: 40045356 PMCID: PMC11881310 DOI: 10.1186/s40478-025-01950-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Chronic cerebral hypoperfusion (CCH) is a major contributor to vascular dementia, with neuroinflammation playing a central role in its pathogenesis. Sinomenine (SINO), a natural alkaloid derived from traditional Chinese medicine, has shown significant anti-inflammatory and neuroprotective properties. However, its efficacy and mechanism of action in CCH remain unclear. In this study, we established a CCH rat model through bilateral common carotid artery occlusion and administered 10 mg/kg of SINO daily. Behavioral tests demonstrated that SINO significantly improved cognitive and memory functions in CCH rats. Histological analysis revealed that SINO effectively reduced neuroinflammation and damage in the hippocampal CA1, CA3, and DG regions. Mechanistically, SINO promoted microglial polarization from the M1 to M2 phenotype, markedly inhibiting the release of pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α. Further exploration of its neuroprotective mechanism showed that exosomes from SINO-treated microglia were enriched with miRNA-223-3p, which suppressed NLRP3-mediated pyroptosis in neurons. While our findings highlight the therapeutic potential of SINO, further studies are needed to validate its safety and efficacy in diverse populations and chronic settings. In summary, this study not only demonstrates SINO's regulatory effect on microglial polarization in CCH but also unveils a novel neuroprotective mechanism through exosomal miRNA-223-3p delivery, providing a solid theoretical foundation for SINO's potential as a treatment for CCH.
Collapse
Affiliation(s)
- Qu Yang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Precision Cell Therapy, Nanchang, Jiangxi, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- The National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Qi Chen
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Kai-Bing Zhang
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yu Liu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- The National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Jia-Cheng Zheng
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Dong-Xia Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- The National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Jun Luo
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- Jiangxi Province Key Laboratory of Precision Cell Therapy, Nanchang, Jiangxi, 330006, China.
- The National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
6
|
Li H, Li K, Cheng W, Liu M, Wen L, Zhang Z, Zhang W, Su J, Cai W. Rapid Characterization of the Potential Active of Sinomenine in Rats by Ultra-High-Performance Liquid Chromatography-Quadrupole-Exactive Orbitrap Mass Spectrometry and Molecular Docking. J Sep Sci 2024; 47:e202400486. [PMID: 39375918 DOI: 10.1002/jssc.202400486] [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: 07/01/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 10/09/2024]
Abstract
Sinomenium acutum (Thunb.) Rehd. et Wils is widely used in the treatment of rheumatoid arthritis, with its alkaloid compound sinomenine (SIN) being renowned for its significant anti-inflammatory properties. However, despite its widespread application, the in vivo anti-inflammatory mechanisms and metabolic pathways of SIN remain incompletely understood. This study established a rapid and reliable method based on an ultra-high-performance liquid chromatography method coupled with Quadrupole-Exactive Orbitrap mass spectrometry and molecular docking to identify and characterize SIN and 69 metabolites in rat plasma, urine, and feces, revealing primary metabolic pathways of hydroxylation, demethylation, sulfation, and glucuronidation. Molecular docking results revealed that phase I reactions, including dedimethylation, demethylation, dehydrogenation, and dihydroxylation, along with their composite reactions, were pivotal in influencing SIN's in vivo anti-inflammatory activity. M28, M36, and M59 are potentially the most anti-inflammatory active metabolites of SIN in vivo. This comprehensive analysis unveils SIN's metabolic pathways, offering insights into its biological processes and suggesting a novel approach for exploring active drug constituents. These findings pave the way for further understanding SIN's anti-inflammatory mechanisms, contributing significantly to the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Haixia Li
- College Pharmacy, Jiamusi University, Jiamusi, China
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - KaiLin Li
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
- School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Wenhui Cheng
- College Pharmacy, Jiamusi University, Jiamusi, China
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - Mingjuan Liu
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - Linwen Wen
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - Zexu Zhang
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - Wendan Zhang
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - Jin Su
- College Pharmacy, Jiamusi University, Jiamusi, China
| | - Wei Cai
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| |
Collapse
|
7
|
Liu Q, Wang J, Ding C, Chu Y, Jiang F, Hu Y, Li H, Wang Q. Sinomenine Alleviates Rheumatoid Arthritis by Suppressing the PI3K-Akt Signaling Pathway, as Demonstrated Through Network Pharmacology, Molecular Docking, and Experimental Validation. Drug Des Devel Ther 2024; 18:3523-3545. [PMID: 39135759 PMCID: PMC11317229 DOI: 10.2147/dddt.s475959] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/25/2024] [Indexed: 08/15/2024] Open
Abstract
Purpose Sinomenine (SIN) is commonly used in Traditional Chinese Medicine (TCM) as a respected remedy for rheumatoid arthritis (RA). Nevertheless, the therapeutic mechanism of SIN in RA remains incompletely understood. This study aimed to delve into the molecular mechanism of SIN in the treatment of RA. Methods The potential targets of SIN were predicted using the TCMSP server, STITCH database, and SwissTarget Prediction. Differentially expressed genes (DEGs) in RA were obtained from the GEO database. Enrichment analyses and molecular docking were conducted to explore the potential mechanism of SIN in the treatment of RA. In vitro and in vivo studies were conducted to validate the intervention effects of SIN on rheumatoid arthritis, as determined through network pharmacology analyses. Results A total of 39 potential targets associated with the therapeutic effects of SIN in RA were identified. Enrichment analysis revealed that these potential targets are primarily enriched in PI3K-Akt signaling pathway, and the molecular docking suggests that SIN may act on specific proteins in the pathway. Experimental results have shown that exposure to SIN inhibits cytokine secretion, promotes apoptosis, reduces metastasis and invasion, and blocks the activation of the PI3K-Akt signaling pathway in RA fibroblast-like synoviocytes (RA-FLS). Moreover, SIN treatment alleviated arthritis-related symptoms and regulated the differentiation of CD4+ T cells in the spleen of collagen-induced arthritis (CIA) mice. Conclusion By utilizing network pharmacology, molecular modeling, and in vitro/in vivo validation, this study demonstrates that SIN can alleviate RA by inhibiting the PI3K-Akt signaling pathway. These findings enhance the understanding of the therapeutic mechanisms of SIN in RA, offering a stronger theoretical foundation for its future clinical application.
Collapse
Affiliation(s)
- Qingyang Liu
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Jian Wang
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Chunhui Ding
- Department of Pharmacy, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Ying Chu
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Fengying Jiang
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Yunxia Hu
- Department of Rheumatology and Immunology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Haifeng Li
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Qiubo Wang
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| |
Collapse
|
8
|
Qin Y, Huang Y, Ji X, Gong L, Luo S, Gao J, Liu R, Zhang T. N-demethylsinomenine metabolite and its prototype sinomenine activate mast cells via MRGPRX2 and aggravate anaphylaxis. Front Pharmacol 2024; 15:1389761. [PMID: 39144634 PMCID: PMC11322065 DOI: 10.3389/fphar.2024.1389761] [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: 03/14/2024] [Accepted: 07/04/2024] [Indexed: 08/16/2024] Open
Abstract
Sinomenine hydrochloride (SH) is commonly used in the treatment of rheumatoid arthritis. It activates mast cells and induces anaphylaxis in the clinical setting. Adverse drug reactions can be caused by activation of MAS-associated G protein-coupled receptor X2 (MRGPRX2) on mast cells. Because the ligand binding site of MRGPRX2 is easily contacted in dilute solvents, it can be activated by many opioid drug structures. N-Demethylsinomenine (M-3) has a similar chemical structure to that of the opioid scaffold and is a major metabolite of SH. We sought to clarify whether M-3 induces anaphylaxis synergistically with its prototype in a mouse model. Molecular docking computer simulations suggested a similar binding effect between M-3 and SH. M-3 was chemically synthesized and analyzed by surface plasmon resonance to reveal its affinity for MRGPRX2. Temperature monitoring, in vivo hindlimb swelling and exudation test, and in vitro mast cell degranulation test were used to explore the mechanism of MRGPrx2 mediated allergic reaction triggered by M-3. Reduced M-3-induced inflammation was evident in MrgprB2 (the ortholog of MRGPRX2) conditional (Cpa3-Cre/MrgprB2flox) knockout (MrgprB2-CKO) mice. Additionally, LAD2 human mast cells with MRGPRX2 knockdown showed reduced degranulation. M-3 activated LAD2 cells synergistically with SH as regulated by GRK2 signaling and IP3R/PLC/PKC/P38 molecular signaling pathways. The results indicate that the M-3 metabolite can activate mast cells synergistically with its prototype SH via MRGPRX2 and aggravate anaphylaxis. These findings provide important insights into drug safety.
Collapse
Affiliation(s)
- Youfa Qin
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, China
- The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, China
| | - Yihan Huang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Xiaolan Ji
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Ling Gong
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Shiqiong Luo
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Jiapan Gao
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Rui Liu
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Tao Zhang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
9
|
Nie Z, Wu J, Xie J, Yin W. Sinomenine ameliorates bleomycin-induced pulmonary fibrosis by inhibiting the differentiation of fibroblast into myofibroblast. Heliyon 2024; 10:e33314. [PMID: 39050413 PMCID: PMC11268178 DOI: 10.1016/j.heliyon.2024.e33314] [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: 03/18/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/27/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) represents a severe interstitial lung disease characterized by limited therapeutic interventions. Recent study has suggested that sinomenine (SIN), an alkaloid derived from the roots of Sinomenium acutum, demonstrates efficacy in interrupting aerobic glycolysis, a predominant metabolic pathway in myofibroblasts. However, its pharmacological potential in the context of pulmonary fibrosis remains inadequately explored. In the present study, we established a bleomycin (BLM)-induced pulmonary fibrosis mouse model and subjected the mice to a one-week regimen of SIN treatment to assess its efficacy. Additionally, a TGF-β1-induced primary lung fibroblast model was employed to investigate the molecular mechanism underlying the effects of SIN. Our observations revealed robust anti-pulmonary fibrosis properties associated with SIN treatment, as evidenced by reduced extracellular matrix deposition, diminished hydroxyproline contents, improved Ashcroft scores, and enhanced lung function parameters. Furthermore, SIN administration significantly impeded TGF-β1-induced fibroblast-to-myofibroblast differentiation. Mechanistically, SIN exerted its beneficial effects by mitigating aerobic glycolysis, achieved through the inhibition of the expression of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (Pfkfb3). Notably, the protective effects of SIN on fibroblasts were reversed upon ectopic overexpression of Pfkfb3. In conclusion, our data underscore the potential of SIN to attenuate fibroblast-to-myofibroblast differentiation by modulating Pfkfb3-associated aerobic glycolysis and SIN emerges as a promising anti-fibrotic agent for pulmonary fibrosis in clinical practice.
Collapse
Affiliation(s)
| | | | - Jun Xie
- Department of Geriatrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Wanling Yin
- Department of Geriatrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| |
Collapse
|
10
|
Li S, Lin X, Duan L. Harnessing the power of natural alkaloids: the emergent role in epilepsy therapy. Front Pharmacol 2024; 15:1418555. [PMID: 38962319 PMCID: PMC11220463 DOI: 10.3389/fphar.2024.1418555] [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: 04/16/2024] [Accepted: 05/31/2024] [Indexed: 07/05/2024] Open
Abstract
The quest for effective epilepsy treatments has spotlighted natural alkaloids due to their broad neuropharmacological effects. This review provides a comprehensive analysis of the antiseizure properties of various natural compounds, with an emphasis on their mechanisms of action and potential therapeutic benefits. Our findings reveal that bioactive substances such as indole, quinoline, terpenoid, and pyridine alkaloids confer medicinal benefits by modulating synaptic interactions, restoring neuronal balance, and mitigating neuroinflammation-key factors in managing epileptic seizures. Notably, these compounds enhance GABAergic neurotransmission, diminish excitatory glutamatergic activities, particularly at NMDA receptors, and suppress proinflammatory pathways. A significant focus is placed on the strategic use of nanoparticle delivery systems to improve the solubility, stability, and bioavailability of these alkaloids, which helps overcome the challenges associated with crossing the blood-brain barrier (BBB). The review concludes with a prospective outlook on integrating these bioactive substances into epilepsy treatment regimes, advocating for extensive research to confirm their efficacy and safety. Advancing the bioavailability of alkaloids and rigorously assessing their toxicological profiles are essential to fully leverage the therapeutic potential of these compounds in clinical settings.
Collapse
Affiliation(s)
- Siyu Li
- Department of Neurosurgery, Clinical Trial Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyu Lin
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lijuan Duan
- Department of Neurosurgery, Clinical Trial Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Di Dalmazi G, Giuliani C, Bucci I, Mascitti M, Napolitano G. Promising Role of Alkaloids in the Prevention and Treatment of Thyroid Cancer and Autoimmune Thyroid Disease: A Comprehensive Review of the Current Evidence. Int J Mol Sci 2024; 25:5395. [PMID: 38791433 PMCID: PMC11121374 DOI: 10.3390/ijms25105395] [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: 03/21/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Thyroid cancer (TC) and thyroid autoimmune disorders (AITD) are among the most common diseases in the general population, with higher incidence in women. Chronic inflammation and autoimmunity play a pivotal role in carcinogenesis. Some studies, indeed, have pointed out the presence of AITD as a risk factor for TC, although this issue remains controversial. Prevention of autoimmune disease and cancer is the ultimate goal for clinicians and scientists, but it is not always feasible. Thus, new treatments, that overcome the current barriers to prevention and treatment of TC and AITD are needed. Alkaloids are secondary plant metabolites endowed with several biological activities including anticancer and immunomodulatory properties. In this perspective, alkaloids may represent a promising source of prophylactic and therapeutic agents for TC and AITD. This review encompasses the current published literature on alkaloids effects on TC and AITD, with a specific focus on the pathways involved in TC and AITD development and progression.
Collapse
Affiliation(s)
- Giulia Di Dalmazi
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Cesidio Giuliani
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Ines Bucci
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Marco Mascitti
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Giorgio Napolitano
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
12
|
Hou W, Huang L, Huang H, Liu S, Dai W, Tang J, Chen X, Lu X, Zheng Q, Zhou Z, Zhang Z, Lan J. Bioactivities and Mechanisms of Action of Sinomenine and Its Derivatives: A Comprehensive Review. Molecules 2024; 29:540. [PMID: 38276618 PMCID: PMC10818773 DOI: 10.3390/molecules29020540] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/13/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Sinomenine, an isoquinoline alkaloid extracted from the roots and stems of Sinomenium acutum, has been extensively studied for its derivatives as bioactive agents. This review concentrates on the research advancements in the biological activities and action mechanisms of sinomenine-related compounds until November 2023. The findings indicate a broad spectrum of pharmacological effects, including antitumor, anti-inflammation, neuroprotection, and immunosuppressive properties. These compounds are notably effective against breast, lung, liver, and prostate cancers, exhibiting IC50 values of approximately 121.4 nM against PC-3 and DU-145 cells, primarily through the PI3K/Akt/mTOR, NF-κB, MAPK, and JAK/STAT signaling pathways. Additionally, they manifest anti-inflammatory and analgesic effects predominantly via the NF-κB, MAPK, and Nrf2 signaling pathways. Utilized in treating rheumatic arthritis, these alkaloids also play a significant role in cardiovascular and cerebrovascular protection, as well as organ protection through the NF-κB, Nrf2, MAPK, and PI3K/Akt/mTOR signaling pathways. This review concludes with perspectives and insights on this topic, highlighting the potential of sinomenine-related compounds in clinical applications and the development of medications derived from natural products.
Collapse
Affiliation(s)
- Wen Hou
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Lejun Huang
- College of Rehabilitation, Gannan Medical University, Ganzhou 341000, China;
| | - Hao Huang
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Shenglan Liu
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Wei Dai
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Jianhong Tang
- Laboratory Animal Engineering Research Center of Ganzhou, Gannan Medical University, Ganzhou 341000, China;
| | - Xiangzhao Chen
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Xiaolu Lu
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Qisheng Zheng
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Zhinuo Zhou
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Ziyun Zhang
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Jinxia Lan
- College of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|