1
|
Jiang H, Lu Q, Huang X, Zhang H, Zeng J, Wang M, Xu J, Yuan Z, Wei Q, Xiao E, Wang P, Huang G, Xu A. Sinomenine-glycyrrhizic acid self-assembly enhanced the anti-inflammatory effect of sinomenine in the treatment of rheumatoid arthritis. J Control Release 2025; 382:113718. [PMID: 40220871 DOI: 10.1016/j.jconrel.2025.113718] [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: 01/07/2025] [Revised: 03/23/2025] [Accepted: 04/08/2025] [Indexed: 04/14/2025]
Abstract
Rheumatoid arthritis (RA) is a common chronic systemic autoimmune disease that causes cartilage and bone damage in multiple joints, ultimately leading to disability. There is an urgent need to develop multidimensional strategies to treat RA. Sinomenine (SIN) has the distinctive pharmacological activity in treating RA, but its broader clinical application is limited by its exceedingly short half-life and adverse digestive tract effects. To overcome this obstacle, a self-assembled nanohydrogel (S-G hydrogel) was designed and produced with sinomenine (SIN) and glycyrrhizic acid (GA) without carriers or catalysts through noncovalent bonding. The S-G hydrogel could promote the absorption of SIN probably by protecting SIN from releasing and degrading in the acid circumstances. Oral intake of the S-G hydrogel significantly suppressed the overactivation of neutrophil via the Nf-κb and Mapk pathways in mice with RA. Furthermore, the S-G hydrogel regulated neutrophil activity by reversing apoptosis delay and decreasing autophagy-dependent NET formation. In summary, this study presents a self-assembled hydrogel with promising potential for clinical application, and offers a novel strategy to develop new drugs from the existing patent medicine composed of compounds from traditional Chinese medicine, as well as a special insight to elucidate the herb-matching mechanism in decoction prescriptions.
Collapse
Affiliation(s)
- Haixu Jiang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China; School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Qingyi Lu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xuemei Huang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Honglin Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jia Zeng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Mengdan Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhihua Yuan
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Qiuzhu Wei
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Enfan Xiao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Penglong Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China.
| | - Guangrui Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.
| | - Anlong Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China; Hong Kong Institute of Advanced Studies, Sun Yat-sen University, Hong Kong, China.
| |
Collapse
|
2
|
Guo WY, Wu QM, Zeng HF, Chen YL, Xu J, Yu ZY, Shu YK, Yang XN, Zhang CH, He XZ, Mi JN, Chen S, Chen XM, Wu JQ, Yao HQ, Liu L, Pan HD. A sinomenine derivative alleviates bone destruction in collagen-induced arthritis mice by suppressing mitochondrial dysfunction and oxidative stress via the NRF2/HO-1/NQO1 signaling pathway. Pharmacol Res 2025; 215:107686. [PMID: 40088961 DOI: 10.1016/j.phrs.2025.107686] [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: 12/23/2024] [Revised: 02/09/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
Bone destruction in rheumatoid arthritis (RA) leads to significant disability, yet effective treatments are limited. Sinomenine (Sino) demonstrates anti-arthritic and bone-protective effects but requires high doses. In this study, we developed a Sino derivative, SINX, and evaluated its efficacy in RA. Safety assessments in mice confirmed its suitability for further study. In vitro, SINX inhibited osteoclast differentiation by reducing TRAP-positive cells, disrupting F-actin ring formation, and suppressing bone resorption pits, alongside downregulating osteoclast-specific genes. It also showed strong anti-inflammatory properties by reducing inflammatory cytokine levels. In vivo, using a collagen-induced arthritis (CIA) mouse model, SINX improved bone integrity by reducing joint inflammation, maintaining trabecular bone density, and preventing erosion. Histological and micro-CT analyses confirmed its effects, including suppressed osteoclast activity and reduced bone resorption-related gene expression. Mechanistically, SINX ameliorated mitochondrial dysfunction, decreased ROS levels, and activated the NRF2/HO-1/NQO1 pathway, enhancing antioxidant defenses. Compared to Sino, SINX achieved similar results at lower doses. These findings highlight the potential of SINX as a safe, effective treatment for RA-related bone destruction.
Collapse
Affiliation(s)
- Wan-Yi Guo
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China; State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Qi-Min Wu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, Jiangning District, Nanjing 211198, China
| | - Hao-Feng Zeng
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China
| | - Yu-Lian Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China
| | - Jie Xu
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Zhen-Yi Yu
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Yong-Kang Shu
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Xiao-Nan Yang
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Chuan-Hai Zhang
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Xi-Zi He
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China
| | - Jia-Ning Mi
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China
| | - Si Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China
| | - Xiao-Man Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China
| | - Jia-Qi Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China; State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - He-Quan Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, Jiangning District, Nanjing 211198, China
| | - Liang Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China; State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Hu-Dan Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China; State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao.
| |
Collapse
|
3
|
Li S, Wang S, Zhang L, Ka Y, Zhou M, Wang Y, Tang Z, Zhang J, Wang W, Liu W. Research progress on pharmacokinetics, anti-inflammatory and immunomodulatory effects of kaempferol. Int Immunopharmacol 2025; 152:114387. [PMID: 40054326 DOI: 10.1016/j.intimp.2025.114387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/24/2025]
Abstract
Chronic inflammation (an abnormal state) and autoimmune disease (AD) can both cause multiple organ damage. AD is a heterogeneous group of diseases due to immune dysfunction. Chronic inflammation is closely related to AD and is an important part of AD. With the increasing prevalence of AD, researchers are constantly exploring new drugs with small side effects, considerable curative effects, and lower costs. Kaempferol, a flavonoid, possesses a range of biological functions, including antioxidant, anti-inflammatory, anti-neoplastic, and immunomodulatory capabilities. This compound is prevalent in a variety of plant sources, such as vegetables, fruits, and medicinal herbs traditionally used in Chinese medicine. A plethora of empirical evidence from animal-based research supports the assertion that this particular substance exhibits both anti-inflammatory and immunomodulatory effects, with the curative effect being significant and application prospects. This article mainly summarizes and discusses the pharmacokinetics, drug delivery system, and the mechanism of kaempferol on immune cells, cytokines, signaling pathways, and other aspects. This paper summarizes the existing kaempferol drug delivery system, analyzes the possibility and limitations of kaempferol as a new anti-inflammatory and immunomodulatory drug, and discusses how to apply it in clinical practice. Therefore, kaempferol can more effectively exert its anti-inflammatory and immune-modulating effects, thereby demonstrating therapeutic potential in clinical settings, while reducing patient burden.
Collapse
Affiliation(s)
- Suiran Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Siwei Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Lei Zhang
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, China
| | - Yuxiu Ka
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Meijiao Zhou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yiwen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Zhuo Tang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Jiamin Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
4
|
Li B, Dong B, Xie L, Li Y. Exploring Advances in Natural Plant Molecules for Allergic Rhinitis Immunomodulation in Vivo and in Vitro. Int J Gen Med 2025; 18:529-565. [PMID: 39911299 PMCID: PMC11796455 DOI: 10.2147/ijgm.s493021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/20/2024] [Indexed: 02/07/2025] Open
Abstract
Allergic rhinitis (AR) is a prevalent allergic disease that imposes significant economic burdens and life pressures on individuals, families, and society, particularly in the context of accelerating globalization and increasing pathogenic factors. Current clinical therapies for AR include antihistamines, glucocorticoids administered via various routes, leukotriene receptor antagonists, immunotherapy, and several decongestants. These treatments have demonstrated efficacy in alleviating clinical symptoms and pathological states. However, with the growing awareness of AR and rising expectations for improvements in quality of life, these treatments have become associated with a higher incidence of side effects and an elevated risk of drug resistance. Furthermore, the development of AR is intricately associated with dysregulation of the immune system, yet the underlying pathogenetic mechanisms remain incompletely understood. In contrast, widely available natural plant molecules offer multiple targeting pathways that uniquely modify the typical pathophysiology of AR through immunomodulatory processes. This review presents a comprehensive analysis of both in vivo and in vitro studies on natural plant molecules that modulate immunity for treating AR. Additionally, we examine their specific mechanisms of action in animal models to provide new insights for developing safe and effective targeted therapies while guiding experimental and clinical applications against AR.
Collapse
Affiliation(s)
- Bingquan Li
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Boyang Dong
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Liangzhen Xie
- Ear-Nose-Throat Department, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Yan Li
- Ear-Nose-Throat Department, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| |
Collapse
|
5
|
Yu F, Peng Z, Gao N, Tang Z, Liao Z, Zhao S, Zhong S, Umwiza G, Huang H, Long W, He Z. Sinomenine attenuates uremia vascular calcification by miR-143-5p. Sci Rep 2025; 15:1798. [PMID: 39806038 PMCID: PMC11730593 DOI: 10.1038/s41598-025-86055-2] [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: 10/23/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
Vascular calcification is considered to be a killer of the cardiovascular system, involved inflammation and immunity. There is no approved therapeutic strategy for the prevention of vascular calcification. Sinomenine exhibited anti-inflammatory and immunosuppressive effects. Objective of this study was to investigate the effect of sinomenine in vascular calcification and its potential molecular mechanism. Adenine-induced uremic rats were constructed and administrated with sinomenine. Optical clearing of aortas, alizarin red staining, von Kossa staining, calcification quantification, micro-CT analyses of vascular calcification were performed to analyze calcification in aortas. Administration of 40 mg/kg/d sinomenine effectively alleviated vascular calcification in uremic rats. The miRNA sequencing revealed differentially expressed miRNAs in aortas and bioinformatic analysis assisted with miRNA screening. We screened 9 differential expressed miRNAs and their predicted target genes. By qRT-PCR, we validated that the expression of rno-miR-143-5p was corresponding to our prediction. Sinomenine inhibited vascular smooth muscle cells (VSMCs) calcification, accompanied with miR-143-5p upregulation. MiR-143-5p mimic decreased VSMCs calcification in high phosphate condition. On the contrary, miR-143-5p inhibitor increased VSMCs calcification in high phosphate condition, which was inhibited by sinomenine. In chronic kidney disease patients with vascular calcification, the expression level of circulating miR-143-5p was lower than those without vascular calcification. Sinomenine significantly inhibited vascular calcification in VSMCs and uremic rat. MiR-143-5p was one of the collection of miRNAs modified by sinomenine in vascular calcification. Reduction of miR-143-5p in VSMCs was not only a concomitant phenomenon in pro-calcification condition but also contribute to VSMCs calcification. Circulating miR-143-5p was supposed to be a potential biomarker for vascular calcification in chronic kidney disease patients. In conclusion, sinomenine effectively alleviated vascular calcification, which was attributed to miR-143-5p regulation partly.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Morphinans/pharmacology
- Animals
- Vascular Calcification/drug therapy
- Vascular Calcification/genetics
- Vascular Calcification/etiology
- Vascular Calcification/pathology
- Vascular Calcification/metabolism
- Uremia/complications
- Uremia/drug therapy
- Uremia/genetics
- Rats
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Humans
- Rats, Sprague-Dawley
- Aorta/pathology
- Aorta/metabolism
- Aorta/drug effects
- Disease Models, Animal
Collapse
Affiliation(s)
- Fengyi Yu
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
- Department of Gastroenterology, Yiyang Central Hospital, Yiyang, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhong Peng
- Department of Gastroenterology, Yiyang Central Hospital, Yiyang, Hunan, China
| | - Ning Gao
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
| | - Zixu Tang
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zihao Liao
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Song Zhao
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shuzhu Zhong
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Gloria Umwiza
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hong Huang
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wei Long
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
| | - Zhangxiu He
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China.
- Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
6
|
Wu S, Zhou G, Wang X, Zhang X, Guo S, Ma Y, Liu H, Li W. Development of Sinomenine Hydrochloride Sustained-release Pellet With Multiple Release Characteristics. AAPS PharmSciTech 2024; 25:224. [PMID: 39322795 DOI: 10.1208/s12249-024-02949-w] [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: 06/19/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024] Open
Abstract
Due to the gastrointestinal side effects, the clinical application of sinomenine hydrochloride (SH) in rheumatoid arthritis is limited. The elderly population constitutes the primary group affected by this disease, and within this demographic, there are significant variations in gastric emptying time. To reduce the influence of individual differences on drug efficacy and concurrently alleviate gastrointestinal side effects, the SH sustained-release pellets with multiple release characteristics were developed, which comprised both regular sustained-release pellets and enteric-coated sustained-release pellets. The drug-loaded layer formulation was optimized by full factorial design. With the optimal formulation, the drug-loaded pellets achieved a yield of 96.05%, an encapsulation efficiency of 83.36% for SH, a relative standard deviation of 3.26% in SH content distribution, an average roundness of 0.971 for the pellets, and the particle size span of 0.808. The pellets with a 4 h SH release profile in an acidic environment and pellets displaying 4 h acid resistance followed by an 8 h SH release behavior in the intestinal environment were individually prepared through in vitro dissolution tests. The results demonstrated stable and compliant dissolution behavior of the formulation, along with excellent stability and physical appearance. This research offers novel insights and references for the innovative formulation of SH.
Collapse
Affiliation(s)
- Sijun Wu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Guoming Zhou
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xi Wang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiaoyang Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shubo Guo
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yongqiang Ma
- Xinyite Science and Technology Co., Ltd, Guangdong, 518083, China
| | - Hai Liu
- Xinyite Science and Technology Co., Ltd, Guangdong, 518083, China
| | - Wenlong Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China.
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
7
|
Feng Y, Pan M, Li R, He W, Chen Y, Xu S, Chen H, Xu H, Lin Y. Recent developments and new directions in the use of natural products for the treatment of inflammatory bowel disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155812. [PMID: 38905845 DOI: 10.1016/j.phymed.2024.155812] [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: 04/10/2024] [Revised: 05/13/2024] [Accepted: 06/06/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) represents a significant global health challenge, and there is an urgent need to explore novel therapeutic interventions. Natural products have demonstrated highly promising effectiveness in the treatment of IBD. PURPOSE This study systematically reviews the latest research advancements in leveraging natural products for IBD treatment. METHODS This manuscript strictly adheres to the PRISMA guidelines. Relevant literature on the effects of natural products on IBD was retrieved from the PubMed, Web of Science and Cochrane Library databases using the search terms "natural product," "inflammatory bowel disease," "colitis," "metagenomics", "target identification", "drug delivery systems", "polyphenols," "alkaloids," "terpenoids," and so on. The retrieved data were then systematically summarized and reviewed. RESULTS This review assessed the different effects of various natural products, such as polyphenols, alkaloids, terpenoids, quinones, and others, in the treatment of IBD. While these natural products offer promising avenues for IBD management, they also face challenges in terms of clinical translation and drug discovery. The advent of metagenomics, single-cell sequencing, target identification techniques, drug delivery systems, and other cutting-edge technologies heralds a new era in overcoming these challenges. CONCLUSION This paper provides an overview of current research progress in utilizing natural products for the treatment of IBD, exploring how contemporary technological innovations can aid in discovering and harnessing bioactive natural products for the treatment of IBD.
Collapse
Affiliation(s)
- Yaqian Feng
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Mengting Pan
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Ruiqiong Li
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Weishen He
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yangyang Chen
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Shaohua Xu
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| | - Hui Chen
- Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, China.
| | - Huilong Xu
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| | - Yao Lin
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| |
Collapse
|
8
|
Lin Y, Tang Y, Yi O, Zhu J, Su Z, Li G, Zhou H, Liu L, Liu B, Cai X. Graphene oxide quantum dots-loaded sinomenine hydrochloride nanocomplexes for effective treatment of rheumatoid arthritis via inducing macrophage repolarization and arresting abnormal proliferation of fibroblast-like synoviocytes. J Nanobiotechnology 2024; 22:383. [PMID: 38951875 PMCID: PMC11218134 DOI: 10.1186/s12951-024-02645-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/16/2024] [Indexed: 07/03/2024] Open
Abstract
The characteristic features of the rheumatoid arthritis (RA) microenvironment are synovial inflammation and hyperplasia. Therefore, there is a growing interest in developing a suitable therapeutic strategy for RA that targets the synovial macrophages and fibroblast-like synoviocytes (FLSs). In this study, we used graphene oxide quantum dots (GOQDs) for loading anti-arthritic sinomenine hydrochloride (SIN). By combining with hyaluronic acid (HA)-inserted hybrid membrane (RFM), we successfully constructed a new nanodrug system named HA@RFM@GP@SIN NPs for target therapy of inflammatory articular lesions. Mechanistic studies showed that this nanomedicine system was effective against RA by facilitating the transition of M1 to M2 macrophages and inhibiting the abnormal proliferation of FLSs in vitro. In vivo therapeutic potential investigation demonstrated its effects on macrophage polarization and synovial hyperplasia, ultimately preventing cartilage destruction and bone erosion in the preclinical models of adjuvant-induced arthritis and collagen-induced arthritis in rats. Metabolomics indicated that the anti-arthritic effects of HA@RFM@GP@SIN NPs were mainly associated with the regulation of steroid hormone biosynthesis, ovarian steroidogenesis, tryptophan metabolism, and tyrosine metabolism. More notably, transcriptomic analyses revealed that HA@RFM@GP@SIN NPs suppressed the cell cycle pathway while inducing the cell apoptosis pathway. Furthermore, protein validation revealed that HA@RFM@GP@SIN NPs disrupted the excessive growth of RAFLS by interfering with the PI3K/Akt/SGK/FoxO signaling cascade, resulting in a decline in cyclin B1 expression and the arrest of the G2 phase. Additionally, considering the favorable biocompatibility and biosafety, these multifunctional nanoparticles offer a promising therapeutic approach for patients with RA.
Collapse
Affiliation(s)
- Ye Lin
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yuanyuan Tang
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
- College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Ouyang Yi
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Junping Zhu
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
- College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Zhaoli Su
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
- College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Gejing Li
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Hua Zhou
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, University of Chinese Medicine, Guangzhou, 510006, China
| | - Liang Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, University of Chinese Medicine, Guangzhou, 510006, China.
| | - Bin Liu
- College of Biology, Hunan University, Changsha, 410082, Hunan, China.
| | - Xiong Cai
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
9
|
Jiang S, Li S, Pang S, Liu M, Sun H, Zhang N, Liu J. A systematic review: Sinomenine. Heliyon 2024; 10:e29976. [PMID: 38765107 PMCID: PMC11098800 DOI: 10.1016/j.heliyon.2024.e29976] [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: 01/23/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/21/2024] Open
Abstract
Sinomenine (SIN), an alkaloid derived from the traditional Chinese medicine, Caulis Sinomenii, has been used as an anti-inflammatory drug in China for over 30 years. With the continuous increase in research on the pharmacological mechanism of SIN, it has been found that, in addition to the typical rheumatoid arthritis (RA) treatment, SIN can be used as a potentially effective therapeutic drug for anti-tumour, anti-renal, and anti-nervous system diseases. By reviewing a large amount of literature and conducting a summary analysis of the literature pertaining to the pharmacological mechanism of SIN, we completed a review that focused on SIN, found that the current research is insufficient, and offered an outlook for future SIN development. We hope that this review will increase the public understanding of the pharmacological mechanisms of SIN, discover SIN research trial shortcomings, and promote the effective treatment of immune diseases, inflammation, and other related diseases.
Collapse
Affiliation(s)
- Shan Jiang
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin City, Heilongjiang Province, 150040, PR China
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua City, Hunan Province, 418000, PR China
| | - Shuang Li
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua City, Hunan Province, 418000, PR China
- College Pharmacy, Jiamusi University, Jiamusi City, Heilongjiang Province, 154000, PR China
| | - Siyuan Pang
- Hunan Zhengqing Pharmaceutical Company Group Ltd, Huaihua City, Hunan Province, 418000, PR China
| | - Mei Liu
- School of Pharmaceutical Sciences, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - Huifeng Sun
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin City, Heilongjiang Province, 150040, PR China
| | - Ning Zhang
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin City, Heilongjiang Province, 150040, PR China
| | - Jianxin Liu
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua City, Hunan Province, 418000, PR China
- School of Pharmaceutical Sciences, University of South China, Hengyang City, Hunan Province, 421001, PR China
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha City, Hunan Province, 410208, PR China
| |
Collapse
|
10
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [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
|
11
|
Li P, Wang C, Huo H, Xu C, Sun H, Wang X, Wang L, Li L. Prodrug-based nanomedicines for rheumatoid arthritis. DISCOVER NANO 2024; 19:9. [PMID: 38180534 PMCID: PMC10769998 DOI: 10.1186/s11671-023-03950-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/25/2023] [Indexed: 01/06/2024]
Abstract
Most antirheumatic drugs with high toxicity exhibit a narrow therapeutic window due to their nonspecific distribution in the body, leading to undesirable side effects and reduced patient compliance. To in response to these challenges, prodrug-based nanoparticulate drug delivery systems (PNDDS), which combines prodrug strategy and nanotechnology into a single system, resulting their many advantages, including stability for prodrug structure, the higher drug loading capacity of the system, improving the target activity and bioavailability, and reducing their untoward effects. PNDDS have gained attention as a method for relieving arthralgia syndrome of rheumatoid arthritis in recent years. This article systematically reviews prodrug-based nanocarriers for rheumatism treatment, including Nano systems based on prodrug-encapsulated nanomedicines and conjugate-based nanomedicines. It provides a new direction for the clinical treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Pei Li
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Cong Wang
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Hongjie Huo
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Chunyun Xu
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Huijun Sun
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Xinyu Wang
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Li Wang
- College of Pharmacy, Dalian Medical University, Dalian, China.
| | - Lei Li
- College of Pharmacy, Dalian Medical University, Dalian, China.
| |
Collapse
|
12
|
Liu Y, Wang W, Zhang J, Gao S, Xu T, Yin Y. JAK/STAT signaling in diabetic kidney disease. Front Cell Dev Biol 2023; 11:1233259. [PMID: 37635867 PMCID: PMC10450957 DOI: 10.3389/fcell.2023.1233259] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
Diabetic kidney disease (DKD) is the most important microvascular complication of diabetes and the leading cause of end-stage renal disease (ESRD) worldwide. The Janus kinase/signal transducer and activator of the transcription (JAK/STAT) signaling pathway, which is out of balance in the context of DKD, acts through a range of metabolism-related cytokines and hormones. JAK/STAT is the primary signaling node in the progression of DKD. The latest research on JAK/STAT signaling helps determine the role of this pathway in the factors associated with DKD progression. These factors include the renin-angiotensin system (RAS), fibrosis, immunity, inflammation, aging, autophagy, and EMT. This review epitomizes the progress in understanding the complicated explanation of the etiologies of DKD and the role of the JAK/STAT pathway in the progression of DKD and discusses whether it can be a potential target for treating DKD. It further summarizes the JAK/STAT inhibitors, natural products, and other drugs that are promising for treating DKD and discusses how these inhibitors can alleviate DKD to explore possible potential drugs that will contribute to formulating effective treatment strategies for DKD in the near future.
Collapse
Affiliation(s)
- Yingjun Liu
- Clinical Medicine Department, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenkuan Wang
- Clinical Medicine Department, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jintao Zhang
- Clinical Medicine Department, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuo Gao
- Clinical Medicine Department, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tingting Xu
- Clinical Medicine Department, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yonghui Yin
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
13
|
Li W, Xu X. Advances in mitophagy and mitochondrial apoptosis pathway-related drugs in glioblastoma treatment. Front Pharmacol 2023; 14:1211719. [PMID: 37456742 PMCID: PMC10347406 DOI: 10.3389/fphar.2023.1211719] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant tumor of the central nervous system (CNS). It is a leading cause of death among patients with intracranial malignant tumors. GBM exhibits intra- and inter-tumor heterogeneity, leading to drug resistance and eventual tumor recurrence. Conventional treatments for GBM include maximum surgical resection of glioma tissue, temozolomide administration, and radiotherapy, but these methods do not effectively halt cancer progression. Therefore, development of novel methods for the treatment of GBM and identification of new therapeutic targets are urgently required. In recent years, studies have shown that drugs related to mitophagy and mitochondrial apoptosis pathways can promote the death of glioblastoma cells by inducing mitochondrial damage, impairing adenosine triphosphate (ATP) synthesis, and depleting large amounts of ATP. Some studies have also shown that modern nano-drug delivery technology targeting mitochondria can achieve better drug release and deeper tissue penetration, suggesting that mitochondria could be a new target for intervention and therapy. The combination of drugs targeting mitochondrial apoptosis and autophagy pathways with nanotechnology is a promising novel approach for treating GBM.This article reviews the current status of drug therapy for GBM, drugs targeting mitophagy and mitochondrial apoptosis pathways, the potential of mitochondria as a new target for GBM treatment, the latest developments pertaining to GBM treatment, and promising directions for future research.
Collapse
|
14
|
Wang S, Zhang L, Zhou Y, Huang J, Zhou Z, Liu Z. A review on pharmacokinetics of sinomenine and its anti-inflammatory and immunomodulatory effects. Int Immunopharmacol 2023; 119:110227. [PMID: 37119677 DOI: 10.1016/j.intimp.2023.110227] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/02/2023] [Accepted: 04/19/2023] [Indexed: 05/01/2023]
Abstract
Autoimmune diseases (ADs), with significant effects on morbidity and mortality, are a broad spectrum of disorders featured by body's immune responses being directed against its own tissues, resulting in chronic inflammation and tissue damage. Sinomenine (SIN) is an alkaloid isolated from the root and stem of Sinomenium acutum which is mainly used to treat pain, inflammation and immune disorders for centuries in China. Its potential anti-inflammatory role for treating immune-related disorders in experimental animal models and in some clinical applications have been reported widely, suggesting an inspiring application prospect of SIN. In this review, the pharmacokinetics, drug delivery systems, pharmacological mechanisms of action underlying the anti-inflammatory and immunomodulatory effects of SIN, and the possibility of SIN as adjuvant to disease-modifying anti-rheumatic drugs (DMARDs) therapy were summarized and evaluated. This paper aims to reveal the potential prospects and limitations of SIN in the treatment of inflammatory and immune diseases, and to provide ideas for compensating its limitations and reducing the side effects, and thus to make SIN better translate to the clinic.
Collapse
Affiliation(s)
- Siwei Wang
- Medical Department, Yangtze University, Jingzhou 434023, Hubei Province, China; Honghu Hospital of Traditional Chinese Medicine, Honghu 433299, Hubei Province, China
| | - Lvzhuo Zhang
- Medical Department, Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Yanhua Zhou
- Honghu Hospital of Traditional Chinese Medicine, Honghu 433299, Hubei Province, China
| | - Jiangrong Huang
- Medical Department, Yangtze University, Jingzhou 434023, Hubei Province, China; Jingzhou Central Hospital Affiliated to Yangtze University, Jingzhou 434020, Hubei Province, China.
| | - Zushan Zhou
- Medical Department, Yangtze University, Jingzhou 434023, Hubei Province, China; Honghu Hospital of Traditional Chinese Medicine, Honghu 433299, Hubei Province, China.
| | - Zhenzhen Liu
- Medical Department, Yangtze University, Jingzhou 434023, Hubei Province, China.
| |
Collapse
|
15
|
Liu X, Chen H, Chen X, Wu P, Zhang J. Identification of Potential Targets and Mechanisms of Sinomenine in Allergic Rhinitis Treatment Based on Network Pharmacology and Molecular Docking. Crit Rev Immunol 2023; 43:1-10. [PMID: 37830189 DOI: 10.1615/critrevimmunol.2023049479] [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: 10/14/2023]
Abstract
This study aimed to investigate the potential targets and molecular mechanism of sinomenine in treating allergic rhinitis (AR) using network pharmacology and molecular docking. Relevant targets of sinomenine and AR were obtained from public databases, and differentially expressed genes (DEGs) for AR were identified in the Gene Expression Omnibus database. Using VennDiagram, we identified 22 potential targets of sinomenine against AR by crossing disease targets, drug targets, and DEGs. Functional analysis revealed that sinomenine may act via its anti-inflammatory and immunosuppressive effects, and its action pathways may include the MAPK, HIF-1, and JAK-STAT pathways. Furthermore, hub targets were identified using EPC, MCC, and MNC algorithms, and six hub targets (STAT3, EGFR, NFKB1, HIF1A, PTGS2, and JAK1) were selected by integrating the top 10 hub genes and 22 potential targets. Molecular docking analysis indicated that STAT3, EGFR, PTGS2, and JAK1 may be key targets of sinomenine against AR. Overall, our results suggest that sinomenine has potential therapeutic effects against AR, and its mechanism of action may involve the regulation of key targets and pathways related to inflammation and immunity.
Collapse
Affiliation(s)
- Xuemei Liu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Hong Chen
- Department of Rehabilitation, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Xiaobo Chen
- Department of Rehabilitation, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Peng Wu
- Department of Internal Medicine, Ganzhou Hospital of TCM, Ganzhou, 341000, China
| | - Jianhua Zhang
- Academic Affairs Office, The First Affiliated Hospital of Gannan Medical University, No. 23 Qingnian Road, Ganzhou, 341000, China
| |
Collapse
|