1
|
Yu J, You Z, Wong VKW, Chen M, Liu W. Novel Cinnamic Acid Derivatives Containing Naproxen as NLRP3 Inhibitors: Synthesis and Evaluation of Their Biological Activity. Chem Biodivers 2025; 22:e202402700. [PMID: 39737737 DOI: 10.1002/cbdv.202402700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/01/2025]
Abstract
Long-term use of naproxen can lead to serious side effects. Inspired by the biological activity of cinnamic acid, a series of cinnamic acid derivatives containing naproxen were designed and synthesized, and their anti-inflammatory activities and mechanisms were explored in vitro. Our results indicated that all of naproxen derivatives showed more significant inhibition against lipopolysaccharide (LPS)-induced nitric oxide (NO) production and had a lower degree of cytotoxicity than that of naproxen. The present studies revealed that compound 23 (IC50 = 5.66 ± 1.66 µM) markedly inhibited the LPS-induced NO production and the over-expression of pro-inflammatory cytokines, including interleukin (IL)-1β, inducible NO synthase (iNOS), and cyclooxygenase-2 (COX-2). Furthermore, it blocked the activation of NF-κB signaling pathway and pyrin domain-containing protein 3 (NLRP-3) inflammasome in a concentration-dependent manner. Additionally, docking studies confirmed that compound 23 exhibited a well-fitting into the NLRP3 active site. Considering these results, compound 23 might be a novel NLRP3 inhibitor to treat inflammatory diseases.
Collapse
Affiliation(s)
- Jialin Yu
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Zonglin You
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Vincent Kam Wai Wong
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Min Chen
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Wenfeng Liu
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| |
Collapse
|
2
|
Tang M, Li H, Chang S, Li Y, Nie H, Li F. Dysregulated circular RNAs in rheumatoid arthritis: Cellular roles and clinical prospects. Autoimmun Rev 2025; 24:103774. [PMID: 39956349 DOI: 10.1016/j.autrev.2025.103774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/27/2025] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
Rheumatoid arthritis (RA) is still a healthcare challenge, although current therapeutic strategies have substantially improved its clinical outcomes. The development of novel biomarkers and treatments can increase the likelihood of identification and disease remission in RA patients, especially for patients with seronegative RA and difficult-to-treat RA (D2T RA). Circular RNAs (circRNAs), a novel non-coding RNA species, have been reported to play crucial roles in various biological process of RA. The mechanistic functions of the dysregulated circRNAs in RA are primarily associated with miRNA sponging and regulating transcription. CircRNAs acting as miRNA sponges are further summarized by cell types, including fibroblast-like synoviocytes (FLSs), lymphocytes, macrophages, chondrocytes, and mesenchymal stem cells (MSCs)-/plasma-secreted exosomes. Besides, a description of dysregulated circRNAs in blood, synovial tissue and cartilage tissue suggests their diagnostic potential for RA. In addition, some directions for future research are provided to open the possibility that dysregulated cell- and tissue- specific circRNAs constituting a fresh reservoir of therapeutic targets, and biomarkers for diagnosis, predicting response to therapy, drug selection or patient stratification for RA.
Collapse
Affiliation(s)
- Mengshi Tang
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Clinical Medical Research Center for Systemic Autoimmune Disease in Hunan Province, Changsha, Hunan 410011, China
| | - Hongxing Li
- Department of Orthopaedics, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Department of Orthopaedics, the Central Hospital of Shaoyang, Shaoyang, Hunan 422099, China
| | - Siyuan Chang
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Clinical Medical Research Center for Systemic Autoimmune Disease in Hunan Province, Changsha, Hunan 410011, China
| | - Yuanyuan Li
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Clinical Medical Research Center for Systemic Autoimmune Disease in Hunan Province, Changsha, Hunan 410011, China
| | - Huiyu Nie
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Clinical Medical Research Center for Systemic Autoimmune Disease in Hunan Province, Changsha, Hunan 410011, China
| | - Fen Li
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Clinical Medical Research Center for Systemic Autoimmune Disease in Hunan Province, Changsha, Hunan 410011, China.
| |
Collapse
|
3
|
Wang T, Liu Y, Lu Y, Chi L. NTN-1 attenuates amyloid-β-mediated microglial neuroinflammation and memory impairment via the NF-κB pathway and NLRP3 inflammasome in a rat model of Alzheimer's disease. Front Aging Neurosci 2025; 17:1516399. [PMID: 40357234 PMCID: PMC12066592 DOI: 10.3389/fnagi.2025.1516399] [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: 10/24/2024] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Neuroinflammation driven by microglial activation represents a pivotal pathological mechanism underlying brain injury in Alzheimer's disease (AD), with NLRP3 inflammasome activation being a hallmark feature of this process. Netrin-1 (NTN-1) was recently shown to have potent anti-inflammatory and anti-apoptotic properties in a range of inflammatory diseases; however, its potential effect on neuroinflammation in AD treatment has not been well examined. Accordingly, this study aimed to investigate the effects of NTN-1 on cognitive impairment and to explore the anti-inflammatory properties related to the NLRP3 inflammasome and NF-κB signaling in Aβ1-42-induced rat models. Methods We assessed the effects of NTN-1 on neurobehavioral function, microglial activation and neuroinflammation mechanisms in Aβ1-42-treated rats using the Morris water maze test and Western blotting. Results Our results indicated that microinjections of NTN-1 attenuated Aβ1-42-induced memory and cognitive dysfunction and significantly inhibited microglial proliferation and NLRP3 inflammasome activation in the hippocampus and cortex of AD rats. Additionally, NTN-1 effectively prevented proinflammatory factor (IL1β and IL18) release and NF-κB signaling upstream activation. Discussion Overall, the results of the present study indicated that exogenous NTN-1 treatment prevented neuroinflammation and cognitive deficits by inhibiting microglial activation, which is possibly mediated by the NF-κB signaling pathway and NLRP3 inflammasome activation in Aβ1-42-simulated rat models. NTN-1 emerges as a promising therapeutic candidate for mitigating microglia-mediated neuropathology in AD through its anti-inflammatory properties.
Collapse
Affiliation(s)
| | | | | | - Lijun Chi
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
4
|
Huang YH, Qiu L, Peng Y, Cai R, Xiao QQ, Li MC, Xu LJ, Fu Q. Ecdysteroid-enriched fraction of Cyathula officinalis suppresses synovial proliferation and inflammation to ameliorate RA by inhibiting the AKT/PI3K/mTOR signaling pathway. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025:1-23. [PMID: 40257367 DOI: 10.1080/10286020.2025.2492357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/22/2025]
Abstract
This study investigated the chemical compositions isolated from Cyathula officinalis (CES) and elucidated its anti-RA effects and potential mechanisms. In vitro results revealed that CES suppressed interleukin (IL)-1β-stimulated proliferation, migration, and invasion of MH7A cells in a concentration-dependent manner. In vivo results indicated that CES could reduce the arthritis score, paw swelling, and improve histopathological deterioration. The levels of inflammatory factors, matrix metalloproteinases, and proteins expressed in rat synovial tissue were suppressed after treatment with CES. These findings illustrate that CES is involved in the PI3K/AKT/mTOR/SREBP1/SCD-1/GPX4 axis-mediated ferroptosis and anti-inflammation, exerting its protective effects against the progression of RA.
Collapse
Affiliation(s)
- Yue-Hui Huang
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
- School of Traditional Chinese Medicine and Nutritional Health Care, Meishan Pharmaceutical College, Meishan620000, China
| | - Lu Qiu
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| | - Yi Peng
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| | - Rui Cai
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| | - Qing-Qing Xiao
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| | - Mei-Chen Li
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| | - Li-Jia Xu
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| | - Qiang Fu
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| |
Collapse
|
5
|
Zhong S, Zhong S, Wu Y, Liu Z, Yang Y, Wang X, Li S, Wu Y, Huang X, Zhu Y, Zhou Z, Xu Y, Wen L, Yao X. Macrophage-targeting nano-formulated bicalutamide alleviates colitis by inducing MAP3K1-mediated degradation of NLRP3. J Control Release 2025; 380:417-432. [PMID: 39892647 DOI: 10.1016/j.jconrel.2025.01.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/13/2025] [Accepted: 01/25/2025] [Indexed: 02/04/2025]
Abstract
The excessive activation of the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) in macrophages has been recognized as a critical factor in the exacerbation of severe inflammatory bowel disease (IBD). Consequently, the modulation of macrophage NLRP3 activity may serve as an effective strategy for mitigating IBD. Our study has indicated that bicalutamide, a clinically administered agent, has the capacity to reduce inflammation by promoting the degradation of NLRP3 in a macrophage-specific manner. However, the therapeutic efficacy of bicalutamide in treating colitis has remained limited. In an effort to enhance the precision of NLRP3 regulation, a nano-bicalutamide system targeting macrophages has been developed, which has shown potential to significantly improve the therapeutic impact on colitis. Mechanistically, it has been found that this system degrades the NLRP3 protein through the autophagy pathway by recruiting the E3 ligase, mitogen-activated protein kinase kinase 1 (MAP3K1), and the autophagy receptor protein optineurin (OPTN). Furthermore, our findings have indicated that the degradation of macrophage NLRP3 inhibits its M1-type polarization, which in turn hinders the colitis process. The system that we have devised has demonstrated potential to address the urgent need for the treatment of colitis, as well as other diseases related to macrophage NLRP3 dysregulation.
Collapse
Affiliation(s)
- Suqin Zhong
- School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Department of General Surgery, Guangdong Provincial People's Hospital Ganzhou Hospital (Ganzhou Municipal Hospital), Ganzhou 341000, China
| | - Suzhen Zhong
- School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Yi Wu
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui 230051, China
| | - Zhiyuan Liu
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Yinyin Yang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Xinfeng Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Shanshan Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Yu Wu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Xiaowan Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yangyang Zhu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Zhengang Zhou
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Youcui Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Research and Industrialization of New Drug Release Technology Joint Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| | - Longping Wen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China.
| | - Xueqing Yao
- School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Department of General Surgery, Guangdong Provincial People's Hospital Ganzhou Hospital (Ganzhou Municipal Hospital), Ganzhou 341000, China.
| |
Collapse
|
6
|
Ou H, Wu Z, Ning J, Huang Q, Wang W, Yang G, Zhou Y, Hou A, Li P, Chen L, Jin WB. In vitro and in vivo characterization of oridonin analogs as anti-inflammatory agents that regulate the NF-κB and NLRP3 inflammasome axis. Front Pharmacol 2025; 16:1512740. [PMID: 40083382 PMCID: PMC11903421 DOI: 10.3389/fphar.2025.1512740] [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: 10/17/2024] [Accepted: 01/28/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction A series of oridonin hybrids were synthesized and evaluated for anti-inflammatory potential, focusing on their ability to inhibit NO production in RAW264.7 cells and their therapeutic prospects for NLRP3-driven disorders. Methods Anti-inflammatory activity was assessed by measuring NO inhibition in LPS-stimulated RAW264.7 cells. The most active compound, 4c, was further analyzed using ELISA and WB to evaluate its effects on inflammatory proteins (p-NF-κB, p-IκB, NLRP3, IL-6, IL-1β, COX-2, iNOS). In vivo efficacy was tested in a murine acute lung injury model, with RT‒qPCR and WB used to assess inflammatory markers in lung tissues. Molecular docking predicted 4c's binding mode with NLRP3, while RNA-seq and RT‒qPCR identified differentially expressed genes. Results Compound 4c significantly inhibited NO production and suppressed key inflammatory proteins in vitro. In vivo, it alleviated acute lung injury, reduced IL-6 and TNF-α mRNA levels, and inhibited NLRP3, p-NF-κB, and IL-6 protein expression. Docking suggested covalent binding to NLRP3. RNA-seq revealed 4c upregulated Trdc, Stfa2, and Gsta2 while downregulating Spib, Csf2, and Nr4a1. Discussion Compound 4c demonstrates potent anti-inflammatory effects via NLRP3 pathway inhibition and modulation of inflammatory genes. These findings highlight oridonin hybrids, particularly 4c, as promising candidates for NLRP3-driven inflammatory disorders, warranting further investigation.
Collapse
Affiliation(s)
- Huiping Ou
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, Guangdong, China
| | - Zhanpan Wu
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Jinhua Ning
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Qiufeng Huang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, Guangdong, China
| | - Wancun Wang
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Guochun Yang
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yingxun Zhou
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Anguo Hou
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Peng Li
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, Guangdong, China
| | - Lingyun Chen
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Wen Bin Jin
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
7
|
Zuo CY, Zhang CS, Zhang HX, Gou CY, Lei H, Tian FW, Wang ZX, Yin HY, Yu SG. Moxibustion Alleviates Inflammation via SIRT5-mediated Post-translational Modification and Macrophage Polarization. Inflammation 2025:10.1007/s10753-025-02239-y. [PMID: 39899130 DOI: 10.1007/s10753-025-02239-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/30/2024] [Accepted: 01/06/2025] [Indexed: 02/04/2025]
Abstract
Macrophage polarisation is influenced by Sirtuin5 (SIRT5), which is crucial for regulating anti-inflammatory processes. Moxibustion, a traditional Chinese medicine therapy, exerts anti-inflammatory effects by altering the succinate/α-ketoglutarate (α-KG) ratio, an indicator of the M1 to M2 macrophage shift. Glutamate dehydrogenase 1 (GLUD1), a key enzyme involved in α-KG production, is desuccinylated by SIRT5. Currently, the potential influence of moxibustion on SIRT5-GLUD1-α-KG-mediated macrophage polarization in inflammatory diseases remains unexplored. C57BL/6 J and Sirt5 knockout mice were used as complete Freund's adjuvant (CFA)-induced adjuvant arthritis models. Moxibustion and acupoint injections of MC3482 were administered. Paw capacity asssays and ELISA were performed to quantify inflammatory effects and the expression of succinate, and α-KG expressions. Flow cytometry (FCM) and immunofluorescence were used to assesss the expression of M1- and M2-like macrophages. LC-MS/MS-based proteomic analysis was performed, and GLUD1 was identified desuccinylated protein associated with SIRT5. Western blotting and immunoprecipitation (IP) were used to detect SIRT5, GLUD1, and succinylated GLUD1expressions. Moxibustion and the SIRT5-mediated desuccinylation inhibitor MC3482 decreased inflammation by increasing the number of M2 macrophages and reducing the number of M1 macrophage in the CFA model. The potential mechanism may be related to the effects of moxibustion and SIRT5 inhibition, which inverted succinate and α-KG levels in the CFA group, resulting in low succinate, high α-KG, and increased GLUD1 succinylation after treatment. These findings suggest that the anti-inflammatory effects moxibustion are related to the impact of macrophage conversion after SIRT5-mediated post-translational modification.
Collapse
Affiliation(s)
- Chuan-Yi Zuo
- Department of Acupuncture, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Cheng-Shun Zhang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Han-Xiao Zhang
- Faculty of Medicine, Université Paris-Saclay, 94800, Villejuif, France
| | - Chun-Yan Gou
- Department of Acupuncture, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Hong Lei
- Department of Acupuncture, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Feng-Wei Tian
- Department of Acupuncture, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Zhu-Xing Wang
- Department of Acupuncture, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Hai-Yan Yin
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
| | - Shu-Guang Yu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
| |
Collapse
|
8
|
Karmakar V, Chain M, Majie A, Ghosh A, Sengupta P, Dutta S, Mazumder PM, Gorain B. Targeting the NLRP3 inflammasome as a novel therapeutic target for osteoarthritis. Inflammopharmacology 2025; 33:461-484. [PMID: 39806051 DOI: 10.1007/s10787-024-01629-2] [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: 08/20/2024] [Accepted: 12/07/2024] [Indexed: 01/16/2025]
Abstract
Osteoarthritis, the most common arthritic condition, is an age-related progressive disease characterized by the loss of cartilage and synovial inflammation in the knees and hips. Development of pain, stiffness, and considerably restricted mobility of the joints are responsible for the production of matrix metalloproteinases and cytokines. Although several treatments are available for the management of this disease condition, they possess limitations at different levels. Recently, efforts have focused on regulating the production of the NLRP3 inflammasome, which plays a critical role in the disease's progression due to its dysregulation. Inhibition of NLRP3 inflammasome has shown the potential to modulate the production of MMP-13, caspase-1, IL-1β, etc., which has been reflected by positive responses in different preclinical and clinical studies. Aiming inhibition of this NLRP3 inflammasome, several compounds are in different stages of research owing to bring a novel agent for the treatment of osteoarthritis. This review summarizes the mechanistic pathways linking NLRP3 activation to osteoarthritis development and discusses the progress in new therapeutics aimed at effective treatment.
Collapse
Affiliation(s)
- Varnita Karmakar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Mayukh Chain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Ankit Majie
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Arya Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Pallav Sengupta
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Sulagna Dutta
- Basic Medical Sciences Department, College of Medicine, Ajman University, Ajman, United Arab Emirates
| | - Papiya Mitra Mazumder
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India.
| |
Collapse
|
9
|
Zhang W, Wu H, Liao Y, Zhu C, Zou Z. Caspase family in autoimmune diseases. Autoimmun Rev 2025; 24:103714. [PMID: 39638102 DOI: 10.1016/j.autrev.2024.103714] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Programmed cell death (PCD) plays a crucial role in maintaining tissue homeostasis, with its primary forms including apoptosis, pyroptosis, and necroptosis. The caspase family is central to these processes, and its complex functions across different cell death pathways and other non-cell death roles have been closely linked to the pathogenesis of autoimmune diseases. This article provides a comprehensive review of the role of the caspase family in autoimmune diseases such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), type 1 diabetes (T1D), and multiple sclerosis (MS). It particularly emphasizes the intricate functions of caspases within various cell death pathways and their potential as therapeutic targets, thereby offering innovative insights and a thorough discussion in this field. In terms of therapy, strategies targeting caspases hold significant promise. We emphasize the importance of a holistic understanding of caspases in the overall concept of cell death, exploring their unique functions and interrelationships across multiple cell death pathways, including apoptosis, pyroptosis, necroptosis, and PANoptosis. This approach transcends the limitations of previous studies that focused on singular cell death pathways. Additionally, caspases play a key role in non-cell death functions, such as immune cell activation, cytokine processing, inflammation regulation, and tissue repair, thereby opening new avenues for the treatment of autoimmune diseases. Regulating caspase activity holds the potential to restore immune balance in autoimmune diseases. Potential therapeutic approaches include small molecule inhibitors (both reversible and irreversible), biological agents (such as monoclonal antibodies), and gene therapies. However, achieving specific modulation of caspases to avoid interference with normal physiological functions remains a major challenge. Future research must delve deeper into the regulatory mechanisms of caspases and their associated complexes linked to PANoptosis to facilitate precision medicine. In summary, this article offers a comprehensive and in-depth analysis, providing a novel perspective on the complex roles of caspases in autoimmune diseases, with the potential to catalyze breakthroughs in understanding disease mechanisms and developing therapeutic strategies.
Collapse
Affiliation(s)
- Wangzheqi Zhang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Huang Wu
- Basic Medical University, Naval Medical University, Shanghai 200433, China
| | - Yan Liao
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Chenglong Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| | - Zui Zou
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| |
Collapse
|
10
|
Chen Y, Zhang Z, Yao Y, Zhou X, Ling Y, Mao L, Gu Z. Methyl Canthin-6-one-2-carboxylate Inhibits the Activation of the NLRP3 Inflammasome in Synovial Macrophages by Upregulating Nrf2 Expression. Curr Issues Mol Biol 2025; 47:38. [PMID: 39852153 PMCID: PMC11763762 DOI: 10.3390/cimb47010038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder that leads to severe cartilage deterioration and synovial impairment in the joints. Previous studies have indicated that the aberrant activation of the NLRP3 inflammasome in synovial macrophages plays a significant role in the pathogenesis of RA and has been regarded as a therapeutic target for the disease. In this study, we synthesized a novel canthin-6-one alkaloid, namely methyl canthin-6-one-2-carboxylate (Cant), and assessed its effects on NLRP3 inflammasome activation in macrophages. Our data reveal that exposure to Cant significantly suppressed the transcription and secretion of multiple pro-inflammatory mediators, including IL-1β, IL-6, IL-18, TNF-α, NO, and COX2, in a dose-dependent manner. These alterations were associated with changes in the activation of various signaling pathways, including NF-kB, MAPK, and PI3K-AKT pathways. Notably, pretreatment with Cant significantly reduced LPS/ATP-induced activation of the NLRP3 inflammasome, as evidenced by the decline in the cleaved forms of IL-1β and caspase-1 in cell culture supernatants of BMDMs. Regarding the mechanisms, our data show that Cant could enhance the expression of Nrf2 in macrophages, which play an inhibitory role in ROS production. Collectively, our data demonstrate that Cant might suppress the activation of the NLRP3 inflammasome by upregulating the production of Nrf2, suggesting that Cant could serve as a candidate for the further development of anti-RA drugs.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Graduate School, Dalian Medical University, Dalian 116044, China;
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong 226019, China
| | - Zongying Zhang
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226019, China; (Z.Z.); (Y.Y.); (X.Z.)
| | - Yuan Yao
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226019, China; (Z.Z.); (Y.Y.); (X.Z.)
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226019, China; (Z.Z.); (Y.Y.); (X.Z.)
| | - Yong Ling
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China;
| | - Liming Mao
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226019, China; (Z.Z.); (Y.Y.); (X.Z.)
- Basic Medical Research Center, School of Medicine, Nantong University, Nantong 226019, China
| | - Zhifeng Gu
- Graduate School, Dalian Medical University, Dalian 116044, China;
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong 226019, China
| |
Collapse
|
11
|
Liang J, Wang J, Zhang Y, Jin X, Dong H, Zhang L, Hao J, Pang X, Tang S, Hou H. Arthritis and Public Health Monitoring: Longitudinal Changes of Triglyceride-Glucose Index Associated with Arthritis in a Cohort of Older Chinese Adults. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025; 29:18-27. [PMID: 39692618 DOI: 10.1089/omi.2024.0177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
The current decade 2021-2030 was designated by the United Nations as the decade of healthy aging, which underlines the need for public health innovation for arthritis clinical care. The triglyceride-glucose (TyG) index is a novel and emerging parameter closely associated with diabetes and cardiovascular diseases and has been suggested to indicate the risk of arthritis. This study examined the longitudinal changes of TyG levels in relation to arthritis among a nationwide cohort of older Chinese adults. We recruited 1257 participants from a national cohort of older Chinese adults, the Chinese Longitudinal Healthy Longevity Survey. On the basis of the longitudinal changes in TyG between 2012 and 2014, we performed a k-means clustering analysis to classify the participants into four TyG groups: Class 1 with moderate and stable levels of TyG; Class 2 with low but rising level of TyG; Class 3 with consistently high TyG; and Class 4 with high and TyG-level rise compared with the baseline. After a 2-year follow-up, logistic regression was used to identify the association between TyG and the onset of arthritis. Compared with individuals in Class 1, those in Class 3 and Class 4 experienced a higher risk of arthritis, with an odds ratio (OR) of 2.823 (95% confidence interval [CI]: 1.113-7.160) and 2.848 (95% CI: 1.299-6.246), respectively. To the best of our knowledge, this is the first study exploring the association between dynamic longitudinal changes in TyG and arthritis. Further studies on world populations are called for.
Collapse
Affiliation(s)
- Jiuqin Liang
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Wang
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yue Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University, Jinan, China
| | - Xiangqian Jin
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hualei Dong
- Department of Sanatorium, Shandong Provincial Taishan Hospital, Taian, China
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, China
| | - Jiheng Hao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, China
| | - Xiaohong Pang
- Department of Journal Center, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Shaocan Tang
- Department of Rehabilitation Medicine, Shandong Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, China
| | - Haifeng Hou
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
12
|
He X, Wu T, He H, Chen L, Han K, Zheng J, Zhang Z, Yuan S, Wang Y, Zhang Y, Zhang X, Xie C. Study of kaempferol in the treatment of rheumatoid arthritis through modulation of the NLRP3/CASP1/GSDMD axis and T-cell activation: Based on network pharmacology, single-cell analysis, and experimental validation. Int Immunopharmacol 2024; 143:113357. [PMID: 39388897 DOI: 10.1016/j.intimp.2024.113357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Kaempferol (Kae) is a natural flavonol compound with excellent anti-inflammatory and immunomodulatory effects, which is of great importance in the treatment of inflammatory diseases. The efficacy of Kae in the treatment of rheumatoid arthritis (RA) has been demonstrated. However, its relevant pharmacodynamic mechanism requires further investigation. PURPOSES This study aimed to further explore the potential mechanism of action of Kae in the treatment of RA using network pharmacology, single-cell analysis, and animal experiments. METHODS Drug target genes were downloaded and screened from the Comparative Toxicogenomics Database (CTD), SwissTargetPrediction database, BindingDB database, and TargetNet database. Transcriptome data from GEO databases (GSE55235, GSE89408, and GSE200815) were selected for disease transcriptome analysis and single-cell matrix data. Network pharmacology and molecular docking were used to investigate the potential mechanism of action of Kae in treating RA. Single-cell analysis, immune infiltration co-expression analyses, and Mendelian-Randomization (MR) studies were conducted to explore the relationship between Kae's target genes and immune cells. Collagen-induced arthritis (CIA) was induced in DBA/1 mouse models through enhanced immunization. Therapeutic efficacy of Kae was assessed using arthritis score, paw swelling index, body weight monitoring, microCT, hematoxylin and eosin (HE) staining, Safranin O-Fast green staining, and Tartrate-resistant acid phosphatase (TRAP) staining. Tissue immunofluorescence and flow cytometry were used to detect expression levels of key genes and immune cell activation status. RESULTS In vivo experiments demonstrated the efficacy of Kae in treating CIA mice. Network pharmacology indicated that Kae might exert anti-inflammatory effects through the NLRP3/CASP1/GSDMD axis. Immune infiltration, single-cell, and MR analyses revealed close associations between Kae's target genes and CD4+, CD8+, and regulatory T cells. Kae inhibited cellular pyroptosis in joint tissues and down-regulated NLRP3, CASP1, and GSDMD expression. Flow cytometry results showed decreased CD4/CD8 ratio, reduced proportion of CD4+ effector memory T cells (Tem), and increased naïve and regulatory T cells (Treg). CONCLUSION Kae might exert anti-inflammatory effects by modulating the NLRP3/CASP1/GSDMD axis to inhibit pyroptosis and suppress overactive immune responses by regulating T-cell proliferation. In summary, Kae demonstrated significant therapeutic efficacy in treating RA.
Collapse
Affiliation(s)
- Xiaoyu He
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, Anhui, 233004, China; Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Tianyu Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, 21009, China.
| | - Haohua He
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, Anhui, 233004, China; Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu 233003, China.
| | - Lili Chen
- Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Ke Han
- Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Jiaqian Zheng
- Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Zheng Zhang
- Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Shuqi Yuan
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, Anhui, 233004, China; Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Yanxin Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, Anhui, 233004, China; Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu 233003, China.
| | - Yan Zhang
- Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Xiaonan Zhang
- Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Changhao Xie
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, Anhui, 233004, China; Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu 233003, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, 287 Changhuai Road, Bengbu, Anhui, 233004, China.
| |
Collapse
|
13
|
Shao Y, Zhou Y, Wan J, Zhu Z, Peng Y, Zhao C, Zhu Y, Tang W. Using terahertz spectroscopy to quantify bioactive flavonoids in Moxa Wool as predictor of rheumatoid arthritis treatment outcomes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155927. [PMID: 39096543 DOI: 10.1016/j.phymed.2024.155927] [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: 01/30/2024] [Revised: 06/12/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024]
Abstract
BACKGROUND Moxibustion, a traditional Chinese medicine practice, employs Moxa Wool, derived from Artemisia argyi. Flavonoids, the key pharmacological constituents in Moxa Wool, are known for their anti-inflammatory and analgesic properties. The purity of Moxa Wool, particularly its flavonoid content, directly influences the efficacy of moxibustion treatments. However, quantifying these bioactive flavonoids accurately and non-destructively has been a challenge. PURPOSE This study introduces terahertz spectroscopy as a non-destructive optical detection method for qualitative detection and quantitative analysis of flavonoids in Moxa Wool. By establishing a mathematical model between spectral signals and clinical efficacy, a reliable correlation between flavonoid concentration and the therapeutic effect of moxibustion can be established, providing a potential predictive model for the treatment outcomes of rheumatoid arthritis. STUDY DESIGN We adopted terahertz spectroscopy technology and combined it with terahertz metamaterial biosensors to achieve rapid, efficient, and non-destructive testing of the quality of Moxa Wool. This method reduces the detection time from hours to minutes while lowering the sample detection limit, overcoming the limitations of traditional detection methods in pharmacological research. METHODS Through terahertz metamaterial biosensors, rapid detection of the purity of Moxa Wool has been achieved. A combination of molecular simulation and terahertz spectroscopy was used to quantitatively analyze the flavonoid content in different purities of Moxa Wool. To ensure accuracy, the quantitative results of flavonoids obtained by terahertz spectroscopy were validated using high-performance liquid chromatography (HPLC). In addition, moxibustion treatment was performed on rats with rheumatoid arthritis using Moxa Wool, and medical indicator information was recorded. A mathematical analysis model was established to evaluate the correlation between flavonoid content and analgesic and anti-inflammatory effects. RESULTS Terahertz spectroscopy analysis shows that there is a direct correlation between the flavonoid content in moxibustion and the absorption peak intensity. The maximum R2 in the model analysis is 0.98, indicating a high accuracy in predicting the purity of Moxa Wool. These results were also validated by HPLC. In a rat model, the purity of 30:1 Moxa Wool samples showed a 50 % decrease in TNF-α, IL-1β, and IL-6 levels during treatment compared to low-purity samples, significantly reducing inflammation markers and pain symptoms. Meanwhile, The PLS prediction model established a correlation between terahertz-detected flavonoid levels and treatment outcomes (PWL and IL-1β). The maximum R2 in the model is 0.91, indicating a high correlation between flavonoid levels and the anti-inflammatory and analgesic effects of moxibustion treatment. CONCLUSION This study not only demonstrates the effectiveness of terahertz spectroscopy in the pharmacological quantification of bioactive compounds but also establishes a novel predictive model for the efficacy of moxibustion in rheumatoid arthritis treatment. It underscores the potential of integrating traditional medicine insights with advanced technology to enhance therapeutic strategies in pharmacology.
Collapse
Affiliation(s)
- Yongni Shao
- Terahertz Technology Innovation Research Institute, Terahertz Spectrum and Imaging Technology Cooperative Innovation Center, Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China; Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai 200092, China
| | - Yuxin Zhou
- Terahertz Technology Innovation Research Institute, Terahertz Spectrum and Imaging Technology Cooperative Innovation Center, Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jiayao Wan
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhi Zhu
- Terahertz Technology Innovation Research Institute, Terahertz Spectrum and Imaging Technology Cooperative Innovation Center, Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yan Peng
- Terahertz Technology Innovation Research Institute, Terahertz Spectrum and Imaging Technology Cooperative Innovation Center, Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China; Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai 200092, China
| | - Chen Zhao
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yiming Zhu
- Terahertz Technology Innovation Research Institute, Terahertz Spectrum and Imaging Technology Cooperative Innovation Center, Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China; Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai 200092, China.
| | - Wenchao Tang
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
14
|
Wu Y, Zhang Y, Wang Z, Lu Y, Wang Y, Pan J, Liu C, Zhu W, Wang Y. Bitongqing Attenuates CIA Rats by Suppressing Macrophage Pyroptosis and Modulating the NLRP3/Caspase-1/GSDMD Pathway. J Inflamm Res 2024; 17:5453-5469. [PMID: 39165322 PMCID: PMC11335010 DOI: 10.2147/jir.s466624] [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: 04/04/2024] [Accepted: 08/02/2024] [Indexed: 08/22/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovitis and inflammatory cell infiltration. The traditional Chinese medicine prescription, Bitongqing (BTQ) exhibited significant efficacy in the clinical treatment of RA. However, the potential therapeutic mechanisms of BTQ in treating RA have not been fully investigated. This study aims to elucidate the effect of BTQ on collagen-induced arthritis (CIA) rat macrophage pyroptosis, providing a theoretical basis for treating RA. Methods This research employed liquid chromatography-mass spectrometry (LC-MS) to identify the primary components of BTQ. The therapeutic effects of BTQ were evaluated in a rat model of CIA. In vivo experiments were conducted using pathohistological staining, immunofluorescence, micro-CT, and Western blotting. Next, Mouse leukemia cells of monocyte macrophage cells (RAW264.7) were induced to undergo pyroptosis using lipopolysaccharide (LPS) and adenosine triphosphate (ATP), and the impact of BTQ on RAW264.7 macrophages was assessed through cell viability, immunofluorescence analysis, lactate dehydrogenase (LDH) secretion measurement, and Western blotting. Results BTQ had a therapeutic effect on CIA rats, which was mainly manifested as a reduction in joint inflammation, foot swelling, bone erosion, and amelioration of pathological changes in these rats. Further studies revealed that BTQ inhibited the levels of cytokine production interleukin-18 (IL-18) and interleukin-1β (IL-1β), and likewise, it inhibited the expression of key proteins in the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) mediated pyroptosis in the synovial tissues of CIA rats. The results of in vitro experiments demonstrated that BTQ attenuated LDH secretion, decreased IL-18 and IL-1β cytokine production, and downregulated expression of key proteins involved in the NLRP3-mediated pyroptosis on RAW264.7 macrophages. Conclusion The therapeutic potential of BTQ in CIA lies in its ability to inhibit NLRP3-mediated macrophage pyroptosis, thereby suggesting a promising strategy for the treatment of RA.
Collapse
Affiliation(s)
- Yunxia Wu
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Yue Zhang
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Zishan Wang
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Yun Lu
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Yabei Wang
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Jie Pan
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Chenxi Liu
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Wen Zhu
- Department of Rheumatology & Immunology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Yue Wang
- Department of Rheumatology & Immunology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
15
|
Li Y, He Y, Yang F, Liang R, Xu W, Li Y, Cheng J, Liang B, Tang M, Shi X, Zhuang J, Luo M, Li L, Zhang R, Liu H, Jie H, Li X, Han X, Sun E, Zhai Z. Gasdermin E-mediated keratinocyte pyroptosis participates in the pathogenesis of psoriasis by promoting skin inflammation. Br J Dermatol 2024; 191:385-396. [PMID: 38655652 DOI: 10.1093/bjd/ljae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 02/29/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Psoriasis is a common chronic inflammatory disease with an unclear aetiology. Keratinocytes in psoriasis are susceptible to exogenous triggers that induce inflammatory cell death. OBJECTIVES To investigate whether gasdermin E (GSDME)-mediated pyroptosis in keratinocytes contributes to the pathogenesis of psoriasis. METHODS Skin samples from patients with psoriasis and from healthy controls were collected to evaluate the expression of GSDME, cleaved caspase-3 and inflammatory factors. We then analysed the data series GSE41662 to further compare the expression of GSDME between lesional and nonlesional skin samples in those with psoriasis. In vivo, a caspase-3 inhibitor and GSDME-deficient mice (Gsdme-/-) were used to block caspase-3/GSDME activation in an imiquimod-induced psoriasis model. Skin inflammation, disease severity and pyroptosis-related proteins were analysed. In vitro, tumour necrosis factor (TNF)-α-induced caspase-3/GSDME-mediated pyroptosis in the HACAT cell line was explored. RESULTS Our analysis of the GSE41662 data series found that GSDME was upregulated in psoriasis lesions vs. normal skin. High levels of inflammatory cytokines such as interleukin (IL)-1β, IL-6 and TNF-α were also found in psoriasis lesions. In mice in the Gsdme-/- and caspase-3 inhibitor groups, the severity of skin inflammation was attenuated and GSDME and cleaved caspase-3 levels decreased after imiquimod treatment. Similarly, IL-1β, IL-6 and TNF-α expression was decreased in the Gsdme-/- and caspase-3 inhibitor groups. In vitro, TNF-α induced HACAT cell pyroptosis through caspase-3/GSDME pathway activation, which was suppressed by blocking caspase-3 or silencing Gsdme. CONCLUSIONS Our study provides a novel explanation of TNF-α/caspase-3/GSDME-mediated keratinocyte pyroptosis in the initiation and -acceleration of skin inflammation and the progression of psoriasis.
Collapse
Affiliation(s)
- Yingfei Li
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yi He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Fangyuan Yang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Rongmei Liang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Wenchao Xu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yehao Li
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jingbo Cheng
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Baozhu Liang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ming Tang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xingliang Shi
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jian Zhuang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Minshuang Luo
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Liuying Li
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ruilin Zhang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Huijuan Liu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Hongyu Jie
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xing Li
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xinai Han
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Erwei Sun
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zeqing Zhai
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
Zhang S, Li D, Fan M, Yuan J, Xie C, Yuan H, Xie H, Gao H. Mechanism of Reactive Oxygen Species-Guided Immune Responses in Gouty Arthritis and Potential Therapeutic Targets. Biomolecules 2024; 14:978. [PMID: 39199366 PMCID: PMC11353092 DOI: 10.3390/biom14080978] [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: 07/04/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
Gouty arthritis (GA) is an inflammatory disease caused by monosodium urate (MSU) crystals deposited in the joint tissues causing severe pain. The disease can recur frequently and tends to form tophus in the joints. Current therapeutic drugs for the acute phase of GA have many side effects and limitations, are unable to prevent recurrent GA attacks and tophus formation, and overall efficacy is unsatisfactory. Therefore, we need to advance research on the microscopic mechanism of GA and seek safer and more effective drugs through relevant targets to block the GA disease process. Current research shows that the pathogenesis of GA is closely related to NLRP3 inflammation, oxidative stress, MAPK, NET, autophagy, and Ferroptosis. However, after synthesizing and sorting out the above mechanisms, it is found that the presence of ROS is throughout almost the entire spectrum of micro-mechanisms of the gout disease process, which combines multiple immune responses to form a large network diagram of complex and tight connections involved in the GA disease process. Current studies have shown that inflammation, oxidative stress, cell necrosis, and pathological signs of GA in GA joint tissues can be effectively suppressed by modulating ROS network-related targets. In this article, on the one hand, we investigated the generative mechanism of ROS network generation and its association with GA. On the other hand, we explored the potential of related targets for the treatment of gout and the prevention of tophus formation, which can provide effective reference ideas for the development of highly effective drugs for the treatment of GA.
Collapse
Affiliation(s)
- Sai Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Daocheng Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Mingyuan Fan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Jiushu Yuan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Haipo Yuan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Hongyan Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Hong Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| |
Collapse
|
17
|
He C, Liu J, Li J, Wu H, Jiao C, Ze X, Xu S, Zhu Z, Guo W, Xu J, Yao H. Hit-to-Lead Optimization of the Natural Product Oridonin as Novel NLRP3 Inflammasome Inhibitors with Potent Anti-Inflammation Activity. J Med Chem 2024; 67:9406-9430. [PMID: 38751194 DOI: 10.1021/acs.jmedchem.4c00504] [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: 06/14/2024]
Abstract
Targeting NLRP3 inflammasome with inhibitors is a novel strategy for NLRP3-driven diseases. Herein, hit compound 5 possessing an attractive skeleton was identified from our in-house database of oridonin, and then a potential lead compound 32 was obtained by optimization of 5, displaying two-digit nanomolar inhibition on NLRP3. Moreover, compound 32 showed enhanced safety index (SI) relative to oridonin (IC50 = 77.2 vs 780.4 nM, SI = 40.5 vs 8.5) and functioned through blocking ASC oligomerization and interaction of NLRP3-ASC/NEK7, thereby suppressing NLRP3 inflammasome assembly and activation. Furthermore, diverse agonists-induced activations of NLRP3 could be impeded by compound 32 without altering NLRC4 or AIM2 inflammasome. Crucially, compound 32 possessed tolerable pharmaceutical properties and significant anti-inflammatory activity in MSU-induced gouty arthritis model. Therefore, this work enriched the SAR of NLRP3 inflammasome inhibitors and provided a potential candidate for the treatment of NLRP3-associated diseases.
Collapse
Affiliation(s)
- Chen He
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Junkai Liu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Junda Li
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Hongyu Wu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Chenyang Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, P. R. China
| | - Xiaotong Ze
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Shengtao Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Zheying Zhu
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, U.K
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, P. R. China
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Hong Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| |
Collapse
|
18
|
Karakaya T, Slaufova M, Di Filippo M, Hennig P, Kündig T, Beer HD. CARD8: A Novel Inflammasome Sensor with Well-Known Anti-Inflammatory and Anti-Apoptotic Activity. Cells 2024; 13:1032. [PMID: 38920661 PMCID: PMC11202080 DOI: 10.3390/cells13121032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Inflammasomes comprise a group of protein complexes with fundamental roles in the induction of inflammation. Upon sensing stress factors, their assembly induces the activation and release of the pro-inflammatory cytokines interleukin (IL)-1β and -18 and a lytic type of cell death, termed pyroptosis. Recently, CARD8 has joined the group of inflammasome sensors. The carboxy-terminal part of CARD8, consisting of a function-to-find-domain (FIIND) and a caspase activation and recruitment domain (CARD), resembles that of NLR family pyrin domain containing 1 (NLRP1), which is recognized as the main inflammasome sensor in human keratinocytes. The interaction with dipeptidyl peptidases 8 and 9 (DPP8/9) represents an activation checkpoint for both sensors. CARD8 and NLRP1 are activated by viral protease activity targeting their amino-terminal region. However, CARD8 also has some unique features compared to the established inflammasome sensors. Activation of CARD8 occurs independently of the inflammasome adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC), leading mainly to pyroptosis rather than the activation and secretion of pro-inflammatory cytokines. CARD8 was also shown to have anti-inflammatory and anti-apoptotic activity. It interacts with, and inhibits, several proteins involved in inflammation and cell death, such as the inflammasome sensor NLRP3, CARD-containing proteins caspase-1 and -9, nucleotide-binding oligomerization domain containing 2 (NOD2), or nuclear factor kappa B (NF-κB). Single nucleotide polymorphisms (SNPs) of CARD8, some of them occurring at high frequencies, are associated with various inflammatory diseases. The molecular mechanisms underlying the different pro- and anti-inflammatory activities of CARD8 are incompletely understood. Alternative splicing leads to the generation of multiple CARD8 protein isoforms. Although the functional properties of these isoforms are poorly characterized, there is evidence that suggests isoform-specific roles. The characterization of the functions of these isoforms, together with their cell- and disease-specific expression, might be the key to a better understanding of CARD8's different roles in inflammation and inflammatory diseases.
Collapse
Affiliation(s)
- Tugay Karakaya
- Department of Dermatology, University Hospital Zurich, CH-8952 Schlieren, Switzerland; (T.K.); (M.S.); (M.D.F.); (P.H.); (T.K.)
| | - Marta Slaufova
- Department of Dermatology, University Hospital Zurich, CH-8952 Schlieren, Switzerland; (T.K.); (M.S.); (M.D.F.); (P.H.); (T.K.)
| | - Michela Di Filippo
- Department of Dermatology, University Hospital Zurich, CH-8952 Schlieren, Switzerland; (T.K.); (M.S.); (M.D.F.); (P.H.); (T.K.)
| | - Paulina Hennig
- Department of Dermatology, University Hospital Zurich, CH-8952 Schlieren, Switzerland; (T.K.); (M.S.); (M.D.F.); (P.H.); (T.K.)
| | - Thomas Kündig
- Department of Dermatology, University Hospital Zurich, CH-8952 Schlieren, Switzerland; (T.K.); (M.S.); (M.D.F.); (P.H.); (T.K.)
- Faculty of Medicine, University of Zurich, CH-8006 Zurich, Switzerland
| | - Hans-Dietmar Beer
- Department of Dermatology, University Hospital Zurich, CH-8952 Schlieren, Switzerland; (T.K.); (M.S.); (M.D.F.); (P.H.); (T.K.)
- Faculty of Medicine, University of Zurich, CH-8006 Zurich, Switzerland
| |
Collapse
|
19
|
Saadon SR, Allwsh TA. Lipocalin-2 as a marker of inflammation, bone density, and triglyceride-glucose index for new-onset arthritis patients in Mosul, Iraq. Qatar Med J 2024; 2024:23. [PMID: 38765271 PMCID: PMC11097680 DOI: 10.5339/qmj.2024.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 04/17/2024] [Indexed: 05/21/2024] Open
Abstract
OBJECTIVE Lipocalin-2 is an acute phase-associated adipokine that can serve as an inflammatory and biomarker indicator of cartilage deterioration in osteoarthritis. However, its role in the musculoskeletal system remains not fully understood. Hence, this study aimed to evaluate lipocalin-2 and its relationship with markers of inflammation (Interferon-gamma, ESR, and CRP), bone density (vitamin D3 and calcium), and the triglyceride-glucose index in new-onset arthritis patients in Mosul, Iraq. METHODS This study included 125 participants aged 20 to 65, divided into two groups. The Arthritis Patient Group comprised 70 participants (37 females and 33 males) attending the Bone Diseases Consultation Unit at the Ibn Sina Teaching Hospital in Mosul, Iraq. The Control Group comprised 31 females and 24 males. Ethical approval was obtained from the Iraqi Ministry of Health - Nineveh Health (No. 2022095). Commercial ELISA kits were used to measure serum lipocalin-2, Interferon-gamma, ESR, and CRP as inflammation markers, vitamin D3, and calcium as bone density markers. Moreover, the Triglyceride Glucose (TYG) Index was evaluated. RESULTS The findings revealed a significant increase in lipocalin-2 levels in males compared to females, with LCN-2 increasing with age. Arthritis patients showed a significant increase (72%) in lipocalin-2 levels. Inflammatory indicators (erythrocyte sedimentation rate, C-reactive protein, interferon-gamma) displayed significant increases (46%, 1200%, and 581%, respectively). Glucose (23%), triglycerides (71%), and TYG index (21%) also exhibited significant increases. Meanwhile, bone density indicators (vitamin D3 and calcium) found a significant decrease (53% and 20%, respectively) in arthritis patients. Linear correlation coefficient (R) analysis revealed a significant positive relationship between lipocalin-2 and indicators of inflammation, glucose, TG, and TYG index. CONCLUSION This study's findings suggest that LCN-2 serum levels were higher in patients with new-onset arthritis than in controls in Mosul, and LCN-2 serum increased in males compared with females and getting older serum LCN-2 increased for the patients and control groups. Furthermore, a significant correlation was found between the Triglyceride Glucose Index, which measures metabolic disorders, and serum LCN-2 levels and inflammatory indicators in new-onset arthritis patients in Mosul, Iraq.
Collapse
Affiliation(s)
- Safa Rabea Saadon
- Department of Chemistry, Collage of Science, University of Mosul, Mosul, Iraq *
| | - Thikra Ali Allwsh
- Department of Chemistry, Collage of Science, University of Mosul, Mosul, Iraq *
| |
Collapse
|
20
|
Shao JJ, Li WF, Sun JF, Zhuang ZS, Min JL, Long XH, Wu GJ, Xu HW, Liang G. Britannin as a novel NLRP3 inhibitor, suppresses inflammasome activation in macrophages and alleviates NLRP3-related diseases in mice. Acta Pharmacol Sin 2024; 45:803-814. [PMID: 38172305 PMCID: PMC10943196 DOI: 10.1038/s41401-023-01212-5] [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: 09/04/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024]
Abstract
Overactivation of the NLRP3 inflammasomes induces production of pro-inflammatory cytokines and drives pathological processes. Pharmacological inhibition of NLRP3 is an explicit strategy for the treatment of inflammatory diseases. Thus far no drug specifically targeting NLRP3 has been approved by the FDA for clinical use. This study was aimed to discover novel NLRP3 inhibitors that could suppress NLRP3-mediated pyroptosis. We screened 95 natural products from our in-house library for their inhibitory activity on IL-1β secretion in LPS + ATP-challenged BMDMs, found that Britannin exerted the most potent inhibitory effect with an IC50 value of 3.630 µM. We showed that Britannin (1, 5, 10 µM) dose-dependently inhibited secretion of the cleaved Caspase-1 (p20) and the mature IL-1β, and suppressed NLRP3-mediated pyroptosis in both murine and human macrophages. We demonstrated that Britannin specifically inhibited the activation step of NLRP3 inflammasome in BMDMs via interrupting the assembly step, especially the interaction between NLRP3 and NEK7. We revealed that Britannin directly bound to NLRP3 NACHT domain at Arg335 and Gly271. Moreover, Britannin suppressed NLRP3 activation in an ATPase-independent way, suggesting it as a lead compound for design and development of novel NLRP3 inhibitors. In mouse models of MSU-induced gouty arthritis and LPS-induced acute lung injury (ALI), administration of Britannin (20 mg/kg, i.p.) significantly alleviated NLRP3-mediated inflammation; the therapeutic effects of Britannin were dismissed by NLRP3 knockout. In conclusion, Britannin is an effective natural NLRP3 inhibitor and a potential lead compound for the development of drugs targeting NLRP3.
Collapse
Affiliation(s)
- Jing-Jing Shao
- Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wei-Feng Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jin-Feng Sun
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, School of Pharmaceutical Sciences, Yanbian University, Yanji, 133002, China
| | - Zai-Shou Zhuang
- Affiliated Cangnan Hospital, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ju-Lian Min
- Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiao-Hong Long
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Gao-Jun Wu
- Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Hao-Wen Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, China.
| | - Guang Liang
- Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, China.
| |
Collapse
|
21
|
Jiang Y, Zhong S, Tan H, Fu Y, Lai J, Liu L, Weng J, Chen H, He S. Study on the mechanism of action of Saposhnikovia divaricata and its key phytochemical on rheumatoid arthritis based on network pharmacology and bioinformatics. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117586. [PMID: 38104871 DOI: 10.1016/j.jep.2023.117586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Saposhnikovia divaricata (Turcz.) Schischk (SD; called "fangfeng" in China) has been widely used in the clinical treatment of rheumatoid arthritis (RA) and has shown well therapeutic effects, but the specific mechanisms of action of its bioactive phytochemicals remain unclear. AIM OF THE STUDY This study aimed to investigate the molecular biological mechanism of SD in treating RA through a pharmacology-based strategy. The SD-specific core ingredient Prangenidin was screened for further in-depth study. MATERIALS AND METHODS The bioactive phytochemicals of SD and potential targets for the treatment of RA were screened by network pharmacology, and phytochemicals-related parameters such as pharmacology, and toxicology were evaluated. The protein interaction network was established to screen the core targets, and the correlation between the core targets and RA was further validated by bioinformatics strategy. Finally, molecular docking of core components and corresponding targets was performed. The in vitro experiments were performed to elucidate the regulation of Prangenidin on MH7A cells and on the PI3K/AKT pathway, and the in vivo therapeutic effect of Prangenidin was validated in collagen-induced arthritis (CIA) mice. RESULTS A total of 18 bioactive phytochemicals and 66 potential target genes intersecting with the screened RA disease target genes were identified from SD. Finally, core ingredients such as wogonin, beta-sitosterol, 5-O-Methylvisamminol, and prangenidin and core targets such as PTGS2, RELA, and AKT1 were obtained. The underlying mechanism of SD in treating RA might be achieved by regulating pathways such as PI3K/AKT, IL-17 pathway, apoptosis, and multiple biological processes to exert anti-inflammatory and immunomodulatory effects. Molecular docking confirmed that all core ingredients and key targets had great docking activity. Prangenidin inhibited viability, migration, and invasion, and induced apoptosis in MH7A cells. Prangenidin also reduced the production of IL-1β, IL-6, IL-8, MMP-1, and MMP-3. Molecular analysis showed that Prangenidin exerts its regulatory effect on MH7A cells by inhibiting PI3K/AKT pathway. Treatment with Prangenidin ameliorated synovial inflammation in the joints of mice with CIA. CONCLUSION Our findings provide insights into the therapeutic effects of SD on RA, successfully predicting the effective ingredients and potential targets, which could suggest a novel theoretical basis for further exploration of its molecular mechanisms. It also revealed that Prangenidin inhibited viability, migration, invasion, cytokine, and MMPs expression, and induced apoptosis in RA FLSs via the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Yong Jiang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Shuxin Zhong
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Huangsheng Tan
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Yuanfei Fu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Juyi Lai
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Lijin Liu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Juanling Weng
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Hanwei Chen
- Department of Radiology, Panyu Health Management Center (Panyu Rehabilitation Hospital), Guangzhou, 511495, China.
| | - Shenghua He
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China.
| |
Collapse
|
22
|
Ji R, Wu Y, Ye Y, Li Y, Li Y, Zhong G, Fan W, Feng C, Chen H, Teng X, Wu Y, Xu J. Stimulation of PSTPIP1 to trigger proinflammatory responses in asymptomatic SARS-CoV-2 infections. Heliyon 2024; 10:e26886. [PMID: 38463809 PMCID: PMC10920375 DOI: 10.1016/j.heliyon.2024.e26886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/12/2024] Open
Abstract
Background A hyperinflammatory response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection gravely worsens the clinical progression of coronavirus disease 2019 (COVID-19). Although the undesirable effects of inflammasome activation have been correlated to the severity of COVID-19, the mechanisms of this process in the asymptomatic infection and disease progression have not yet been clearly elucidated. Methods We performed strand-specific RNA sequencing in 39 peripheral blood mononuclear cell (PBMC) samples from asymptomatic individuals(n = 10), symptomatic patients(n = 16) and healthy donors(n = 13). Results Dysregulation of pyrin inflammasomes along with the proline-serine-threonine phosphatase-interacting protein 1 (PSTPIP1) gene was identified in SARS-COV-2 infection. Notably, the PSTPIP1 expression level showed a significant negative correlation with an adjacent long-noncoding RNA (lncRNA) RP11-797A18.6 in the asymptomatic individuals compared with the healthy controls. In addition, a decline in the nuclear factor kappa B subunit 1 (NFKB1) gene expression was observed in asymptomatic infection, followed by a rise in the mild and moderate disease stages, suggesting that altered NFKB1 expression and associated proinflammatory signals may trigger a disease progression. Conclusions Overall, our results indicate that PSTPIP1-dependent pyrin inflammasomes-mediated pyroptosis and NF-κB activation might be potential preventive targets for COVID-19 disease development and progression.
Collapse
Affiliation(s)
- Ruili Ji
- Department of Laboratory Medicine, Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, China
| | - Yue Wu
- Department of Laboratory Medicine, Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, China
| | - Yuhua Ye
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yanling Li
- Guangzhou Huayin Medical Laboratory Center Ltd., Guangzhou, Guangdong, China
| | - Yizhe Li
- Department of Laboratory Science, West China TianFu Hospital, Sichuan University, Sichuan, China
| | - Guojiu Zhong
- Department of Respiratory, Maoming Hospital of Guangzhou University of Chinese Medicine, Maoming 525000, Guangdong, China
| | - Wentao Fan
- Guangzhou Huayin Medical Laboratory Center Ltd., Guangzhou, Guangdong, China
| | - Chengjuan Feng
- Department of Clinical Laboratory, Maoming Hospital of Guangzhou University of Chinese Medicine, Maoming 525000, Guangdong, China
| | - Hui Chen
- Guangzhou Huayin Medical Laboratory Center Ltd., Guangzhou, Guangdong, China
| | - Xiangyun Teng
- Department of Clinical Laboratory, Maoming Hospital of Guangzhou University of Chinese Medicine, Maoming 525000, Guangdong, China
| | - Yunli Wu
- Guangzhou Huayin Medical Laboratory Center Ltd., Guangzhou, Guangdong, China
| | - Jianhua Xu
- Department of Laboratory Medicine, Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, China
- Department of Clinical Laboratory, Maoming Hospital of Guangzhou University of Chinese Medicine, Maoming 525000, Guangdong, China
| |
Collapse
|
23
|
Chen P, Luo Z, Lu C, Jian G, Qi X, Xiong H. Gut-immunity-joint axis: a new therapeutic target for gouty arthritis. Front Pharmacol 2024; 15:1353615. [PMID: 38464719 PMCID: PMC10920255 DOI: 10.3389/fphar.2024.1353615] [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: 12/11/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Gouty arthritis (GA) is an inflammatory disease characterized by pain. The primary goal of current treatment strategies during GA flares remains the reduction of inflammation and pain. Research suggests that the gut microbiota and microbial metabolites contribute to the modulation of the inflammatory mechanism associated with GA, particularly through their effect on macrophage polarization. The increasing understanding of the gut-joint axis emphasizes the importance of this interaction. The primary objective of this review is to summarize existing research on the gut-immune-joint axis in GA, aiming to enhance understanding of the intricate processes and pathogenic pathways associated with pain and inflammation in GA, as documented in the published literature. The refined comprehension of the gut-joint axis may potentially contribute to the future development of analgesic drugs targeting gut microbes for GA.
Collapse
Affiliation(s)
- Pei Chen
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- The First Hospital of Hunan University Chinese Medicine, Changsha, Hunan, China
| | - Zhiqiang Luo
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chengyin Lu
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gonghui Jian
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- College of Integrative Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xinyu Qi
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hui Xiong
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- The First Hospital of Hunan University Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
24
|
Ma N, Lu H, Li N, Ni W, Zhang W, Liu Q, Wu W, Xia S, Wen J, Zhang T. CHOP-mediated Gasdermin E expression promotes pyroptosis, inflammation, and mitochondrial damage in renal ischemia-reperfusion injury. Cell Death Dis 2024; 15:163. [PMID: 38388468 PMCID: PMC10883957 DOI: 10.1038/s41419-024-06525-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/24/2024]
Abstract
In clinical practice, renal ischemia-reperfusion injury (IRI) is a common cause of acute kidney injury (AKI), often leading to acute renal failure or end-stage renal disease (ESRD). The current understanding of renal IRI mechanisms remains unclear, and effective therapeutic strategies and clear targets are lacking. Therefore, the need to find explicit and effective ways to reduce renal IRI remains a scientific challenge. The current study explored pyroptosis, a type of inflammation-regulated programmed cell death, and the role of Gasdermins E (GSDME)-mediated pyroptosis, mitochondrial damage, and inflammation in renal IRI. The analysis of human samples showed that the expression levels of GSDME in normal human renal tissues were higher than those of GSDMD. Moreover, our study demonstrated that GSDME played an important role in mediating pyroptosis, inflammation, and mitochondrial damage in renal IRI. Subsequently, GSDME-N accumulated in the mitochondrial membrane, leading to mitochondrial damage and activation of caspase3, which generated a feed-forward loop of self-amplification injury. However, GSDME knockout resulted in the amelioration of renal IRI. Moreover, the current study found that the transcription factor CHOP was activated much earlier in renal IRI. Inhibition of BCL-2 by CHOP leaded to casapse3 activation, resulting in mitochondrial damage and apoptosis; not only that, but CHOP positively regulated GSDME thereby causing pyroptosis. Therefore, this study explored the transcriptional mechanisms of GSDME during IRI development and the important role of CHOP/Caspase3/GSDME mechanistic axis in regulating pyroptosis in renal IRI. This axis might serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Nannan Ma
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Hao Lu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Ning Li
- Department of Nephropathy, The Zhongda Affilicated Hospital of Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Weijian Ni
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Department of Pharmacy, Centre for Leading Medicine and Advanced Technologies of IHM, Anhui Provincial Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, Anhui, People's Republic of China
| | - Wenbo Zhang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Qiang Liu
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Wenzheng Wu
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Shichao Xia
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Jiagen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China.
| | - Tao Zhang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.
| |
Collapse
|
25
|
Yi YS. Roles of the Caspase-11 Non-Canonical Inflammasome in Rheumatic Diseases. Int J Mol Sci 2024; 25:2091. [PMID: 38396768 PMCID: PMC10888639 DOI: 10.3390/ijms25042091] [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: 01/13/2024] [Revised: 02/07/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Inflammasomes are intracellular multiprotein complexes that activate inflammatory signaling pathways. Inflammasomes comprise two major classes: canonical inflammasomes, which were discovered first and are activated in response to a variety of pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), and non-canonical inflammasomes, which were discovered recently and are only activated in response to intracellular lipopolysaccharide (LPS). Although a larger number of studies have successfully demonstrated that canonical inflammasomes, particularly the NLRP3 inflammasome, play roles in various rheumatic diseases, including rheumatoid arthritis (RA), infectious arthritis (IR), gouty arthritis (GA), osteoarthritis (OA), systemic lupus erythematosus (SLE), psoriatic arthritis (PA), ankylosing spondylitis (AS), and Sjögren's syndrome (SjS), the regulatory roles of non-canonical inflammasomes, such as mouse caspase-11 and human caspase-4 non-canonical inflammasomes, in these diseases are still largely unknown. Interestingly, an increasing number of studies have reported possible roles for non-canonical inflammasomes in the pathogenesis of various mouse models of rheumatic disease. This review comprehensively summarizes and discusses recent emerging studies demonstrating the regulatory roles of non-canonical inflammasomes, particularly focusing on the caspase-11 non-canonical inflammasome, in the pathogenesis and progression of various types of rheumatic diseases and provides new insights into strategies for developing potential therapeutics to prevent and treat rheumatic diseases as well as associated diseases by targeting non-canonical inflammasomes.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, Republic of Korea
| |
Collapse
|
26
|
Madahar SS, Gideon A, Abdul-Sater AA. Nod-like receptors in inflammatory arthritis. Biomed J 2024; 47:100655. [PMID: 37598797 PMCID: PMC10825342 DOI: 10.1016/j.bj.2023.100655] [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: 06/22/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023] Open
Abstract
Nod-like receptors (NLRs) are innate immune receptors that play a key role in sensing components from pathogens and from damaged cells or organelles. NLRs form signaling complexes that can lead to activation of transcription factors or effector caspases - by means of inflammasome activation -Inflammatory arthritis (IA) culminating in promoting inflammation. An increasing body of research supports the role of NLRs in driving pathogenesis of IA, a collection of diseases that include rheumatoid arthritis (RA), psoriatic arthritis (PsA), ankylosing spondylitis, and pediatric arthritis. In this review, we briefly discuss the main drivers of IA diseases and dive into the evidence for - and against - various NLRs in driving these diseases. We also review the studies examining the use of NLR and inflammasome inhibitors as potential therapies for IA.
Collapse
Affiliation(s)
- Sahib Singh Madahar
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada; Department of Biology, York University, Toronto, Ontario, Canada
| | - Alita Gideon
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Ali A Abdul-Sater
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada.
| |
Collapse
|
27
|
Zhang J, Cai H, Sun W, Wu W, Nan Y, Ni Y, Wu X, Chen M, Xu H, Wang Y. Endoplasmic reticulum aminopeptidase 2 regulates CD4 + T cells pyroptosis in rheumatoid arthritis. Arthritis Res Ther 2024; 26:36. [PMID: 38273310 PMCID: PMC10810225 DOI: 10.1186/s13075-024-03271-3] [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: 07/29/2023] [Accepted: 01/14/2024] [Indexed: 01/27/2024] Open
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is a chronic, progressive autoimmune disease with a complex pathogenesis that has not yet been fully elucidated, and T-cell pyroptosis is an important pathogenetic factor in RA. This study aimed to investigate the role of endoplasmic reticulum aminopeptidase 2 (ERAP2) in the pyroptosis of CD4+ T cells in RA and the specific molecular mechanism. METHODS Peripheral venous blood was collected from human subjects, and CD4+ T cells were isolated and activated to measure the level of pyroptosis and ERAP2 expression. Pyroptosis levels were assessed using immunofluorescence, flow cytometry, qRT-PCR, and Western blotting. Changes in pyroptosis levels were observed upon knockdown or overexpression of ERAP2. To detect activated Caspase-1 in tissues, chimeric mice were engrafted with human synovial tissue and reconstituted with human CD4+ T cells. CD4 + T cells were treated with GLI1 antagonists and SMO receptor agonists to detect changes in pyroptosis levels. RESULTS CD4+ T cell levels undergoing pyroptosis were found to be elevated in the blood and synovium of RA patients. The gene and protein expression of ERAP2 were significantly higher in CD4+ T cells from RA patients. Deletion of ERAP2 suppressed pyroptosis of these cells, attenuated the activation of Caspase-1 in tissue T cells, and reduced tissue inflammatory responses. Reciprocally, overexpression of ERAP2 triggered inflammasome assembly, activated Caspase-1, and induced pyroptosis in CD4+ T cells. Mechanistically, ERAP2 inhibits the Hedgehog signaling pathway and upregulates the expression of nucleotide-binding oligomerization segment-like receptor family 3(NLRP3), cleaved Caspase-1, and Gasdermin D to promote pyroptosis in CD4+ T cells. CONCLUSIONS Taken together, our results identify a novel mechanism by which ERAP2 regulates RA development and document the effect of the ERAP2/Hedgehog signaling axis on pyroptosis of CD4+ T cells from RA patients.
Collapse
Affiliation(s)
- Jianhua Zhang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Hao Cai
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Weiwei Sun
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Weijie Wu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yunyi Nan
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yingchen Ni
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xinyuan Wu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Minhao Chen
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| | - Hua Xu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| | - Youhua Wang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
28
|
Maarouf RE, Abdel-Rafei MK, Thabet NM, Azab KS, Rashed L, El Bakary NM. Ondansetron or beta-sitosterol antagonizes inflammatory responses in liver, kidney, lung and heart tissues of irradiated arthritic rats model. Int J Immunopathol Pharmacol 2024; 38:3946320241260635. [PMID: 38831558 PMCID: PMC11149447 DOI: 10.1177/03946320241260635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 05/22/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disorder mainly affecting joints, yet the systemic inflammation can influence other organs and tissues. The objective of this study was to unravel the ameliorative capability of Ondansetron (O) or β-sitosterol (BS) against inflammatory reactions and oxidative stress that complicates Extra-articular manifestations (EAM) in liver, kidney, lung, and heart of arthritic and arthritic irradiated rats. METHODS This was accomplished by exposing adjuvant-induced arthritis (AIA) rats to successive weekly fractions of total body γ-irradiation (2 Gray (Gy)/fraction once per week for four weeks, up to a total dose of 8 Gy). Arthritic and/or arthritic irradiated rats were either treated with BS (40 mg/kg b.wt. /day, orally) or O (2 mg/kg) was given ip) or were kept untreated as model groups. RESULTS Body weight changes, paw circumference, oxidative stress indices, inflammatory response biomarkers, expression of Janus kinase-2 (JAK-2), Signal transducer and activator of transcription 3 (STAT3), high mobility group box1 (HMGB1), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), as well as pro- and anti-inflammatory mediators in the target organs, besides histopathological examination of ankle joints and extra-articular tissues. Treatment of arthritic and/or arthritic irradiated rats with BS or O powerfully alleviated changes in body weight gain, paw swelling, oxidative stress, inflammatory reactions, and histopathological degenerative alterations in articular and non-articular tissues. CONCLUSION The obtained data imply that BS or O improved the articular and EAM by regulating oxidative and inflammatory indices in arthritic and arthritic irradiated rats.
Collapse
Affiliation(s)
- Rokaya E Maarouf
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Mohamed K Abdel-Rafei
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Noura M Thabet
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Khaled S Azab
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Laila Rashed
- Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nermeen M El Bakary
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| |
Collapse
|
29
|
Zhu M, Liu Y, Chen C, Chen H, Ni W, Song Y, Lv B, Hua F, Cui G, Zhang Z. TLR4/Rac1/NLRP3 Pathway Mediates Amyloid-β-Induced Neuroinflammation in Alzheimer's Disease. J Alzheimers Dis 2024; 99:911-925. [PMID: 38728187 DOI: 10.3233/jad-240012] [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] [Indexed: 05/12/2024]
Abstract
Background Neuroinflammation plays a crucial part in the initial onset and progression of Alzheimer's disease (AD). NLRP3 inflammasome was demonstrated to get involved in amyloid-β (Aβ)-induced neuroinflammation. However, the mechanism of Aβ-triggered activation of NLRP3 inflammasome remains poorly understood. Objective Based on our previous data, the study aimed to identify the downstream signals that bridge the activation of TLR4 and NLRP3 inflammasome associated with Aβ. Methods BV-2 cells were transfected with TLR4siRNA or pretreated with a CLI-095 or NSC23766, followed by Aβ1-42 treatment. APP/PS1 mice were injected intraperitoneally with CLI-095 or NSC23766. NLRP3 inflammasome and microglia activation was detected with immunostaining and western blot. G-LISA and Rac1 pull-down activation test were performed to investigate the activation of Rac1. Real-time PCR and ELISA were used to detect the inflammatory cytokines. Aβ plaques were assessed by western blotting and immunofluorescence staining. Morris water maze test was conducted to determine the spatial memory in mice. Results Rac1 and NLRP3 inflammasome were activated by Aβ in both in vitro and in vivo experiments. Inhibition of TLR4 reduced the activity of Rac1 and NLRP3 inflammasome induced by Aβ1-42. Furthermore, inhibition of Rac1 blocked NLRP3 inflammasome activation mediated by TLR4. Blocking the pathway by CLI095 or NSC23766 suppressed Aβ1-42-triggered activation of microglia, reduced the expression of pro-inflammatory mediators and ameliorated the cognition deficits in APP/PS1 mice. Conclusions Our study demonstrated that TLR4/Rac1/NLRP3 pathway mediated Aβ-induced neuroinflammation, which unveiled a novel pathway and key contributors underlying the pathogenic mechanism of Aβ.
Collapse
Affiliation(s)
- Mengxin Zhu
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yang Liu
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, Xi'an People's hospital, Xi'an, China
| | - Chen Chen
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hao Chen
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wanyan Ni
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuanjian Song
- Department of Genetics, Xuzhou Medical University, Xuzhou, China
| | - Bingchen Lv
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Fang Hua
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Guiyun Cui
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zuohui Zhang
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
30
|
Zheng J, Hu J, Yang Y, Xiong L, Yang H, Zhang Z, Jiang N, Liu H. Suppressive effect of Tripterygium hypoglaucum (Levl.) Hutch extract on rheumatoid arthritis in mice by modulating inflammasome and bile acid metabolism. Biomed Pharmacother 2023; 167:115494. [PMID: 37734264 DOI: 10.1016/j.biopha.2023.115494] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023] Open
Abstract
Tripterygium hypoglaucum (Levl.) Hutch (THH) has long been used as a remedy for rheumatoid arthritis (RA) in China. However, it is unclear whether the anti-RA mechanism of THH is associated with inflammasome or gut-joint axis. In this study, we aimed to explore the critical role of NOD-like receptor thermal protein domain associated protein 3 (NLRP3) and bile acid (BA) in the anti-RA mechanism. Complete Freund's adjuvant (CFA)-injected mice were treated with THH extract (250 mg/kg/d) for 35 days, and joint swelling and disease scores were measured. After THH treatment, the joint swelling and RA disease score in CFA-treated mice significantly subsided. The increased ratios of lymphocytes, monocytes, and white blood cells were attenuated by THH treatment. Notably, THH treatment blocked the inflammation in both joints and colons by suppressing the NLRP3-mediated inflammasome, as indicated by NLRP3, interleukin 1beta (IL-1β), and Caspase-1. Meanwhile, THH significantly remodeled the bile acid (BA) profiles in RA mice. Spearman's analysis shed light on the close link between BAs, NLRP3 inflammasome, and RA indicators. However, THH treatment failed to improve inflammasome activation, snoptivis, and joint swelling in RA mice with gut microbiota depletion. In summary, we revealed the pivotal role of BA-mediated gut-joint axis and inflammasome in THH's RA amelioration. In the future, more work should be done to explain the in-depth mechanism between altered BAs and inflammasome.
Collapse
Affiliation(s)
- Junping Zheng
- College of Life Sciences, Wuchang University of Technology, No. 16 Jiangxia Road, Jiangxia District, Wuhan 430223, China; School of Basic Medical Sciences, Hubei University of Chinese Medicine, No. 16 Huangjiahu Road, Hongshan District, Wuhan 430065, China
| | - Jianghui Hu
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, No. 16 Huangjiahu Road, Hongshan District, Wuhan 430065, China
| | - Yong Yang
- Chongqing Academy of Chinese Materia Medica, No. 34 Nanshan Road, Nan'an District, Chongqing 400065, China
| | - Lei Xiong
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, No. 16 Huangjiahu Road, Hongshan District, Wuhan 430065, China
| | - Huabing Yang
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, No. 16 Huangjiahu Road, Hongshan District, Wuhan 430065, China
| | - Zhigang Zhang
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, No. 16 Huangjiahu Road, Hongshan District, Wuhan 430065, China
| | - Nan Jiang
- Affiliated Hospital of Hubei University of Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Hubei Provincial Institute of Traditional Chinese Medicine, Wuhan 430074, China.
| | - Hongtao Liu
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, No. 16 Huangjiahu Road, Hongshan District, Wuhan 430065, China.
| |
Collapse
|
31
|
Xiao Y, Zhang L. Mechanistic and therapeutic insights into the function of NLRP3 inflammasome in sterile arthritis. Front Immunol 2023; 14:1273174. [PMID: 37954594 PMCID: PMC10634342 DOI: 10.3389/fimmu.2023.1273174] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/17/2023] [Indexed: 11/14/2023] Open
Abstract
The NLRP3 inflammasome, which belongs to the pyrin domain containing 3 family of NOD-like receptors, has a significant impact on both the innate and adaptive immune responses. Regulating host immune function and protecting against microbial invasion and cell damage, the NLRP3 inflammasome plays a crucial role. By triggering caspase-1, it facilitates the development of the inflammatory cytokines IL-1β and IL-18, and triggers cell pyroptosis, resulting in cell lysis and demise. Common sterile arthritis includes osteoarthritis (OA), rheumatoid arthritis (RA) and gouty arthritis (GA), all of which manifest as bone destruction and synovial inflammation in a complex inflammatory state, placing a significant medical burden on the families of patients and government agencies. In the past few years, there has been a growing interest in investigating the impact of cell pyroptosis on arthritis development, particularly the widespread occurrence of pyroptosis mediated by the NLRP3 inflammasome. The NLRP3 inflammasome's biological properties are briefly described in this review, along with the presentation of the fundamental processes of pyroptosis resulting from its activation. Furthermore, we provide a summary of the advancements made in studying the NLRP3 inflammasome in various forms of arthritis and enumerate the intervention approaches that target the NLRP3-mediated pyroptosis, either directly or indirectly. These discoveries lay the groundwork for future investigations on medications for arthritis, offering fresh approaches for the clinical identification and treatment of this condition.
Collapse
Affiliation(s)
- Yi Xiao
- Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Zhang
- Department of Orthopedics, Hangzhou Medical College, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
32
|
Kong X, Zhao Y, Wang X, Yu Y, Meng Y, Yan G, Yu M, Jiang L, Song W, Wang B, Wang X. Loganin reduces diabetic kidney injury by inhibiting the activation of NLRP3 inflammasome-mediated pyroptosis. Chem Biol Interact 2023; 382:110640. [PMID: 37473909 DOI: 10.1016/j.cbi.2023.110640] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/19/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Diabetic kidney disease (DKD) is an essential cause of end-stage renal disease. The ongoing inflammatory response in the proximal tubule promotes the progression of DKD. Timely and effective blockade of the inflammatory process to protect the kidney during DKD progression is a proven strategy. The purpose of this study was to investigate the protective effect of loganin on diabetic nephropathy in vivo and in vitro and whether this effect was related to the inhibition of pyroptosis. The results indicated that loganin reduced fasting blood glucose, blood urea nitrogen and serum creatinine concentrations, and alleviated renal pathological changes in DKD mice. In parallel, loganin downregulated the expression of pyroptosis related proteins in the renal tubules of DKD mice and decreased serum levels of interleukin-1beta (IL-1β) and interleukin-18 (IL-18). Furthermore, in vitro experiments showed that loganin attenuated high glucose-induced HK-2 cell injury by reducing the expression of pyroptosis-related proteins, and cytokine levels were also decreased. These fundings were also confirmed in the polyphyllin VI (PPVI) -induced HK-2 cell pyroptosis model. Loganin reduces high glucose induced HK-2 cells pyroptosis by inhibiting reactive oxygen species (ROS) production and NOD-like receptor protein 3 (NLRP3) inflammasome activation. In conclusion, the inhibition of pyroptosis via inhibition of the NLRP3/Caspase-1/Gasdermin D (GSDMD) pathway might be an essential mechanism for loganin treatment of DKD.
Collapse
Affiliation(s)
- Xiangri Kong
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China; Endocrinology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Yunyun Zhao
- Endocrinology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Xingye Wang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China; Department of Cardiovascular Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Yongjiang Yu
- Endocrinology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Ying Meng
- College of Clinical Medical, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Guanchi Yan
- Endocrinology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Miao Yu
- Endocrinology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Lihong Jiang
- Department of Cardiovascular Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Wu Song
- College of Clinical Medical, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China.
| | - Bingmei Wang
- College of Clinical Medical, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China.
| | - Xiuge Wang
- Endocrinology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China.
| |
Collapse
|
33
|
Wang Y, Huang Y, Cheng C, Xue Q, Chang J, Wang X, Duan Q, Miao C. Dysregulation of circRNAs in rheumatoid arthritis, with special emphasis on circRNAs secreted by exosomes and the crosstalk between circRNAs and RNA methylations. Int Immunopharmacol 2023; 122:110549. [PMID: 37421778 DOI: 10.1016/j.intimp.2023.110549] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/09/2023] [Accepted: 06/18/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease caused by a variety of unknown factors. It mainly occurs in the small joints of hands and feet, leading to cartilage destruction and bone erosion. Various pathologic mechanisms such as exosomes and RNA methylations are involved in the pathogenesis of RA. METHODS This work searches PubMed, Web of Science (SCIE) and Science Direct Online (SDOL) databases, it role of abnormally expressed circulating RNAs (circRNAs) in the pathogenesis of RA was summarized. And the relationship between circRNAs and exosomes and methylations. RESULTS Both the abnormal expression of circRNAs and the sponge effect of circRNAs on microRNAs (miRNAs) affect the pathogenesis of RA by regulating target genes. CircRNAs affect the proliferation, migration and inflammatory reaction of RA-fibroblast-like synovial cells (FLSs), circRNAs in peripheral blood mononuclear cells (PBMCs) and macrophages also participate in the pathological mechanism of RA (Fig. 1). CircRNAs in exosomes are closely related to the pathogenesis of RA. In addition, exosomal circRNAs and the relationship between circRNAs and RNA methylations are closely related to the pathogenesis of RA. CONCLUSION CircRNAs play an important role in the pathogenesis of RA and have the potential to be a new target for the diagnosis and treatment of RA. However, the development of mature circRNAs for clinical application is not a small challenge.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yurong Huang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Chenglong Cheng
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Qiuyun Xue
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jun Chang
- Department of Orthopaedics, The First Affiliated Hospital, Anhui Medical University, Hefei 230032, China; Anhui Public Health Clinical Center, Hefei, China.
| | - Xiao Wang
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China.
| | - Qiangjun Duan
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China.
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
34
|
Yan Y, Zhou L, La R, Jiang M, Jiang D, Huang L, Xu W, Wu Q. The association between triglyceride glucose index and arthritis: a population-based study. Lipids Health Dis 2023; 22:132. [PMID: 37608322 PMCID: PMC10463930 DOI: 10.1186/s12944-023-01899-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/07/2023] [Indexed: 08/24/2023] Open
Abstract
OBJECTIVES Insulin resistance is a well-established contributor to inflammation; however, the specific association between the triglyceride glucose (TyG) index, a biomarker reflecting insulin resistance, and arthritis remains unexplored. As a result, the main aim of this study was to examine the correlation between the TyG index and arthritis. METHODS This observational study used data from the National Health and Nutrition Examination Survey (NHANES), which was conducted between 2007 and 2018. To investigate the relationship between the TyG index and arthritis, various statistical analyses were employed, including weighted multivariable logistic regression analysis, subgroup analysis, curve fit analysis, and threshold effect analysis. RESULTS In total, 14,817 patients were enrolled in the trial, with 4,191 individuals (28.29%) diagnosed with arthritis. An increased risk of arthritis was found to be significantly correlated with higher TyG index values (odds ratio OR = 1.15, 95% confidence interval CI: 1.07-1.23), according to the results of multivariable logistic regression analysis after full adjustment. Subgroup analysis and interaction tests further indicated that the TyG index exhibited an additive effect when combined with other established risk factors, including age (OR = 1.29; 95% CI: 1.17-1.41), body mass index (BMI) (OR = 1.43; 95% CI: 1.24-1.69), and diabetes (OR = 1.20; 95% CI: 1.11-1.31). Additionally, curve fit analysis and threshold effect analysis demonstrated a nonlinear relationship with a breakpoint identified at 8.08 µmol/L. CONCLUSION The TyG index was positively correlated with arthritis in adults under 60 years of age in the United States who had normal weight and no diabetes. Further large-scale prospective studies are warranted for a comprehensive analysis of the role of the TyG index in arthritis.
Collapse
Affiliation(s)
- Yuxin Yan
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Liyu Zhou
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Rui La
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Ming Jiang
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Dinghua Jiang
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Lixin Huang
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Wu Xu
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Qian Wu
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
35
|
Lv D, Cao X, Zhong L, Dong Y, Xu Z, Rong Y, Xu H, Wang Z, Yang H, Yin R, Chen M, Ke C, Hu Z, Deng W, Tang B. Targeting phenylpyruvate restrains excessive NLRP3 inflammasome activation and pathological inflammation in diabetic wound healing. Cell Rep Med 2023; 4:101129. [PMID: 37480849 PMCID: PMC10439185 DOI: 10.1016/j.xcrm.2023.101129] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/30/2023] [Accepted: 06/27/2023] [Indexed: 07/24/2023]
Abstract
Moderate inflammation is essential for standard wound healing. In pathological conditions, such as diabetes, protracted and refractory wounds are associated with excessive inflammation, manifested by persistent proinflammatory macrophage states. However, the mechanisms are still unclear. Herein, we perform a metabolomic profile and find a significant phenylpyruvate accumulation in diabetic foot ulcers. Increased phenylpyruvate impairs wound healing and augments inflammatory responses, whereas reducing phenylpyruvate via dietary phenylalanine restriction relieves uncontrolled inflammation and benefits diabetic wounds. Mechanistically, phenylpyruvate is ingested into macrophages in a scavenger receptor CD36-dependent manner, binds to PPT1, and inhibits depalmitoylase activity, thus increasing palmitoylation of the NLRP3 protein. Increased NLRP3 palmitoylation is found to enhance NLRP3 protein stability, decrease lysosome degradation, and promote NLRP3 inflammasome activation and the release of inflammatory factors, such as interleukin (IL)-1β, finally triggering the proinflammatory macrophage phenotype. Our study suggests a potential strategy of targeting phenylpyruvate to prevent excessive inflammation in diabetic wounds.
Collapse
Affiliation(s)
- Dongming Lv
- Department of Burns, Wound Repair and Reconstruction, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xiaoling Cao
- Department of Burns, Wound Repair and Reconstruction, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Li Zhong
- Center of Digestive Diseases, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 517108, China
| | - Yunxian Dong
- Department of Plastic Surgery, Guangdong Second Provincial General Hospital, Southern Medical University, Guangzhou, Guangdong 510317, China
| | - Zhongye Xu
- Department of Burns, Wound Repair and Reconstruction, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Yanchao Rong
- Department of Burns, Wound Repair and Reconstruction, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Hailin Xu
- Department of Burns, Wound Repair and Reconstruction, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Zhiyong Wang
- Department of Burns, Wound Repair and Reconstruction, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Hao Yang
- Department of Burns, Wound Repair and Reconstruction, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Rong Yin
- Department of Dermatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Miao Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong 510080, China
| | - Chao Ke
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong 510080, China
| | - Zhicheng Hu
- Department of Burns, Wound Repair and Reconstruction, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong 510080, China.
| | - Bing Tang
- Department of Burns, Wound Repair and Reconstruction, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
36
|
Hu Q, Zhang F, Wei Y, Liu J, Nie Y, Xie J, Yang L, Luo R, Shen B, Wang Y. Drug-Embedded Nanovesicles Assembled from Peptide-Decorated Hyaluronic Acid for Rheumatoid Arthritis Synergistic Therapy. Biomacromolecules 2023; 24:3532-3544. [PMID: 37417966 DOI: 10.1021/acs.biomac.3c00294] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that causes endless pain and poor quality of life in patients. Usage of a lubricant combined with anti-inflammatory therapy is considered a reasonable and effective approach for the treatment of RA. Herein, inspired by glycopeptides, a peptide-decorated hyaluronic acid was synthesized, and the grafted Fmoc-phenylalanine-phenylalanine-COOH (FmocFF) peptide self-assembled with β-sheet conformations could induce the folding of polymer molecular chains to form a vesicle structure in aqueous solution. The hydrophobic anti-inflammatory drug curcumin (Cur) could be embedded in the vesicle walls through π-π interactions with the FmocFF peptide. Furthermore, the inflammation suppression function of the Cur-loaded vesicles both in vitro and in vivo was demonstrated to be an effective treatment for RA therapy. This work proposes new insights into the folding and hierarchical assembly of glycopeptide mimics, providing an efficient approach for constructing intelligent platforms for drug delivery, disease therapy, and diagnostic applications.
Collapse
Affiliation(s)
- Qinsheng Hu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Orthopedic Surgery, Yaan People's Hospital, Yaan 625000, China
| | - Fanjun Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, China
| | - Yuan Wei
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, China
| | - Jingze Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, China
| | - Yong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinwei Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, China
| | - Bin Shen
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, China
| |
Collapse
|
37
|
Liu P, Ye L, Ren Y, Zhao G, Zhang Y, Lu S, Li Q, Wu C, Bai L, Zhang Z, Zhao Z, Shi Z, Yin S, Liao M, Lan Z, Feng J, Chen L. Chemotherapy-induced phlebitis via the GBP5/NLRP3 inflammasome axis and the therapeutic effect of aescin. Br J Pharmacol 2023; 180:1132-1147. [PMID: 36479683 DOI: 10.1111/bph.16002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 10/03/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Intravenous infusion of chemotherapy drugs can cause severe chemotherapy-induced phlebitis (CIP) in patients. However, the underlying mechanism of CIP development remains unclear. EXPERIMENTAL APPROACH RNA-sequencing analysis was used to identify potential disease targets in CIP. Guanylate binding protein-5 (GBP5) genetic deletion approaches also were used to investigate the role of GBP5 in NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in lipopolysaccharide (LPS) primed murine bone-marrow-derived macrophages (BMDMs) induced by vinorelbine (VIN) in vitro and in mouse models of VIN-induced CIP in vivo. The anti-CIP effect of aescin was evaluated, both in vivo and in vivo. KEY RESULTS Here, we show that the expression of GBP5 was upregulated in human peripheral blood mononuclear cells from CIP patients. Genetic ablation of GBP5 in murine macrophages significantly alleviated VIN-induced CIP in the experimental mouse model. Mechanistically, GBP5 contributed to the inflammatory responses through activating NLRP3 inflammasome and driving the production of the inflammatory cytokine IL-1β. Moreover, aescin, a mixture of triterpene saponins extracted from horse chestnut seed, can alleviate CIP by inhibiting the GBP5/NLRP3 axis. CONCLUSION AND IMPLICATIONS These findings suggest that GBP5 is an important regulator of NLRP3 inflammasome in CIP mouse model. Our work further reveals that aescin may serve as a promising candidate in the clinical treatment of CIP.
Collapse
Affiliation(s)
- Peng Liu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Lichun Ye
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Yongshen Ren
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Guodun Zhao
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Zhang
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shaojuan Lu
- School of Medicine, Tongji University, Shanghai, China
| | - Qiang Li
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Chen Wu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Lijie Bai
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Zhongyun Zhang
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Zhongqiu Zhao
- Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.,Barnes-Jewish Hospital, St. Louis, Missouri, USA
| | - Zhaohua Shi
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Shijin Yin
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Maochuan Liao
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Zhou Lan
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Jing Feng
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lvyi Chen
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| |
Collapse
|
38
|
de Lima JD, de Paula AGP, Yuasa BS, de Souza Smanioto CC, da Cruz Silva MC, Dos Santos PI, Prado KB, Winter Boldt AB, Braga TT. Genetic and Epigenetic Regulation of the Innate Immune Response to Gout. Immunol Invest 2023; 52:364-397. [PMID: 36745138 DOI: 10.1080/08820139.2023.2168554] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gout is a disease caused by uric acid (UA) accumulation in the joints, causing inflammation. Two UA forms - monosodium urate (MSU) and soluble uric acid (sUA) have been shown to interact physically with inflammasomes, especially with the nod-like receptor (NLR) family pyrin domain containing 3 (NLRP3), albeit the role of the immune response to UA is poorly understood, given that asymptomatic hyperuricemia does also exist. Macrophage phagocytosis of UA activate NLRP3, lead to cytokines release, and ultimately, lead to chemoattract neutrophils and lymphocytes to the gout flare joint spot. Genetic variants of inflammasome genes and of genes encoding their molecular partners may influence hyperuricemia and gout susceptibility, while also influencing other comorbidities such as metabolic syndrome and cardiovascular diseases. In this review, we summarize the inflammatory responses in acute and chronic gout, specifically focusing on innate immune cell mechanisms and genetic and epigenetic characteristics of participating molecules. Unprecedently, a novel UA binding protein - the neuronal apoptosis inhibitor protein (NAIP) - is suggested as responsible for the asymptomatic hyperuricemia paradox.Abbreviation: β2-integrins: leukocyte-specific adhesion molecules; ABCG2: ATP-binding cassete family/breast cancer-resistant protein; ACR: American college of rheumatology; AIM2: absent in melanoma 2, type of pattern recognition receptor; ALPK1: alpha-protein kinase 1; ANGPTL2: angiopoietin-like protein 2; ASC: apoptosis-associated speck-like protein; BIR: baculovirus inhibitor of apoptosis protein repeat; BIRC1: baculovirus IAP repeat-containing protein 1; BIRC2: baculoviral IAP repeat-containing protein 2; C5a: complement anaphylatoxin; cAMP: cyclic adenosine monophosphate; CARD: caspase activation and recruitment domains; CARD8: caspase recruitment domain-containing protein 8; CASP1: caspase 1; CCL3: chemokine (C-C motif) ligand 3; CD14: cluster of differentiation 14; CD44: cluster of differentiation 44; Cg05102552: DNA-methylation site, usually cytosine followed by guanine nucleotides; contains arbitrary identification code; CIDEC: cell death-inducing DNA fragmentation factor-like effector family; CKD: chronic kidney disease; CNV: copy number variation; CPT1A: carnitine palmitoyl transferase - type 1a; CXCL1: chemokine (CXC motif) ligand 1; DAMPs: damage associated molecular patterns; DC: dendritic cells; DNMT(1): maintenance DNA methyltransferase; eQTL: expression quantitative trait loci; ERK1: extracellular signal-regulated kinase 1; ERK2: extracellular signal-regulated kinase 2; EULAR: European league against rheumatism; GMCSF: granulocyte-macrophage colony-stimulating factor; GWAS: global wide association studies; H3K27me3: tri-methylation at the 27th lysine residue of the histone h3 protein; H3K4me1: mono-methylation at the 4th lysine residue of the histone h3 protein; H3K4me3: tri-methylation at the 4th lysine residue of the histone h3 protein; HOTAIR: human gene located between hoxc11 and hoxc12 on chromosome 12; IκBα: cytoplasmatic protein/Nf-κb transcription inhibitor; IAP: inhibitory apoptosis protein; IFNγ: interferon gamma; IL-1β: interleukin 1 beta; IL-12: interleukin 12; IL-17: interleukin 17; IL18: interleukin 18; IL1R1: interleukin-1 receptor; IL-1Ra: interleukin-1 receptor antagonist; IL-22: interleukin 22; IL-23: interleukin 23; IL23R: interleukin 23 receptor; IL-33: interleukin 33; IL-6: interleukin 6; IMP: inosine monophosphate; INSIG1: insulin-induced gene 1; JNK1: c-jun n-terminal kinase 1; lncRNA: long non-coding ribonucleic acid; LRR: leucine-rich repeats; miR: mature non-coding microRNAs measuring from 20 to 24 nucleotides, animal origin; miR-1: miR followed by arbitrary identification code; miR-145: miR followed by arbitrary identification code; miR-146a: miR followed by arbitrary identification code, "a" stands for mir family; "a" family presents similar mir sequence to "b" family, but different precursors; miR-20b: miR followed by arbitrary identification code; "b" stands for mir family; "b" family presents similar mir sequence to "a" family, but different precursors; miR-221: miR - followed by arbitrary identification code; miR-221-5p: miR followed by arbitrary identification code; "5p" indicates different mature miRNAs generated from the 5' arm of the pre-miRNA hairpin; miR-223: miR followed by arbitrary identification code; miR-223-3p: mir followed by arbitrary identification code; "3p" indicates different mature miRNAs generated from the 3' arm of the pre-miRNA hairpin; miR-22-3p: miR followed by arbitrary identification code, "3p" indicates different mature miRNAs generated from the 3' arm of the pre-miRNA hairpin; MLKL: mixed lineage kinase domain-like pseudo kinase; MM2P: inductor of m2-macrophage polarization; MSU: monosodium urate; mTOR: mammalian target of rapamycin; MyD88: myeloid differentiation primary response 88; n-3-PUFAs: n-3-polyunsaturated fatty-acids; NACHT: acronym for NAIP (neuronal apoptosis inhibitor protein), C2TA (MHC class 2 transcription activator), HET-E (incompatibility locus protein from podospora anserina) and TP1 (telomerase-associated protein); NAIP: neuronal apoptosis inhibitory protein (human); Naip1: neuronal apoptosis inhibitory protein type 1 (murine); Naip5: neuronal apoptosis inhibitory protein type 5 (murine); Naip6: neuronal apoptosis inhibitory protein type 6 (murine); NBD: nucleotide-binding domain; Nek7: smallest NIMA-related kinase; NET: neutrophil extracellular traps; Nf-κB: nuclear factor kappa-light-chain-enhancer of activated b cells; NFIL3: nuclear-factor, interleukin 3 regulated protein; NIIMA: network of immunity in infection, malignancy, and autoimmunity; NLR: nod-like receptor; NLRA: nod-like receptor NLRA containing acidic domain; NLRB: nod-like receptor NLRA containing BIR domain; NLRC: nod-like receptor NLRA containing CARD domain; NLRC4: nod-like receptor family CARD domain containing 4; NLRP: nod-like receptor NLRA containing PYD domain; NLRP1: nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 1; NLRP12: nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 12; NLRP3: nod-like receptor family pyrin domain containing 3; NOD2: nucleotide-binding oligomerization domain; NRBP1: nuclear receptor-binding protein; Nrf2: nuclear factor erythroid 2-related factor 2; OR: odds ratio; P2X: group of membrane ion channels activated by the binding of extracellular; P2X7: p2x purinoceptor 7 gene; p38: member of the mitogen-activated protein kinase family; PAMPs: pathogen associated molecular patters; PBMC: peripheral blood mononuclear cells; PGGT1B: geranylgeranyl transferase type-1 subunit beta; PHGDH: phosphoglycerate dehydrogenase; PI3-K: phospho-inositol; PPARγ: peroxisome proliferator-activated receptor gamma; PPARGC1B: peroxisome proliferative activated receptor, gamma, coactivator 1 beta; PR3: proteinase 3 antigen; Pro-CASP1: inactive precursor of caspase 1; Pro-IL1β: inactive precursor of interleukin 1 beta; PRR: pattern recognition receptors; PYD: pyrin domain; RAPTOR: regulatory associated protein of mTOR complex 1; RAS: renin-angiotensin system; REDD1: regulated in DNA damage and development 1; ROS: reactive oxygen species; rs000*G: single nuclear polymorphism, "*G" is related to snp where replaced nucleotide is guanine, usually preceded by an id number; SLC2A9: solute carrier family 2, member 9; SLC7A11: solute carrier family 7, member 11; SMA: smooth muscular atrophy; Smac: second mitochondrial-derived activator of caspases; SNP: single nuclear polymorphism; Sp3: specificity protein 3; ST2: serum stimulation-2; STK11: serine/threonine kinase 11; sUA: soluble uric acid; Syk: spleen tyrosine kinase; TAK1: transforming growth factor beta activated kinase; Th1: type 1 helper T cells; Th17: type 17 helper T cells; Th2: type 2 helper T cells; Th22: type 22 helper T cells; TLR: tool-like receptor; TLR2: toll-like receptor 2; TLR4: toll-like receptor 4; TNFα: tumor necrosis factor alpha; TNFR1: tumor necrosis factor receptor 1; TNFR2: tumor necrosis factor receptor 2; UA: uric acid; UBAP1: ubiquitin associated protein; ULT: urate-lowering therapy; URAT1: urate transporter 1; VDAC1: voltage-dependent anion-selective channel 1.
Collapse
Affiliation(s)
- Jordana Dinorá de Lima
- Microbiology, Parasitology and Pathology Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | | | - Bruna Sadae Yuasa
- Microbiology, Parasitology and Pathology Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | | | - Maria Clara da Cruz Silva
- Microbiology, Parasitology and Pathology Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | | | - Karin Braun Prado
- Genetics Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | - Angelica Beate Winter Boldt
- Program of Internal Medicine, Universidade Federal do Parana (UFPR), Curitiba, Brazil
- Genetics Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | - Tárcio Teodoro Braga
- Microbiology, Parasitology and Pathology Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
- Biosciences and Biotechnology Program, Instituto Carlos Chagas (ICC), Fiocruz-Parana, Brazil
| |
Collapse
|
39
|
Shahi A, Afzali S, Firoozi Z, Mohaghegh P, Moravej A, Hosseinipour A, Bahmanyar M, Mansoori Y. Potential roles of NLRP3 inflammasome in the pathogenesis of Kawasaki disease. J Cell Physiol 2023; 238:513-532. [PMID: 36649375 DOI: 10.1002/jcp.30948] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/12/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023]
Abstract
There is a heterogeneous group of rare illnesses that fall into the vasculitis category and are characterized mostly by blood vessel inflammation. Ischemia and disrupted blood flow will cause harm to the organs whose blood arteries become inflamed. Kawasaki disease (KD) is the most prevalent kind of vasculitis in children aged 5 years or younger. Because KD's cardiovascular problems might persist into adulthood, it is no longer thought of as a self-limiting disease. KD is a systemic vasculitis with unknown initiating factors. Numerous factors, such as genetic predisposition and infectious pathogens, are implicated in the etiology of KD. As endothelial cell damage and inflammation can lead to coronary endothelial dysfunction in KD, some studies hypothesized the crucial role of pyroptosis in the pathogenesis of KD. Additionally, pyroptosis-related proteins like caspase-1, apoptosis-associated speck-like protein containing a CARD (ASC), proinflammatory cytokines like IL-1 and IL-18, lactic dehydrogenase, and Gasdermin D (GSDMD) have been found to be overexpressed in KD patients when compared to healthy controls. These occurrences may point to an involvement of inflammasomes and pyroptotic cell death in the etiology of KD and suggest potential treatment targets. Based on these shreds of evidence, in this review, we aim to focus on one of the well-defined inflammasomes, NLRP3, and its role in the pathophysiology of KD.
Collapse
Affiliation(s)
- Abbas Shahi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Afzali
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Firoozi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| | - Poopak Mohaghegh
- Pediatrics Department, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Ali Moravej
- Department of Immunology, Fasa University of Medical Sciences, Fasa, Iran
| | - Ali Hosseinipour
- Department of Internal Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Maryam Bahmanyar
- Pediatrics Department, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
40
|
Zhang F, Yan Y, Cai Y, Liang Q, Liu Y, Peng B, Xu Z, Liu W. Current insights into the functional roles of ferroptosis in musculoskeletal diseases and therapeutic implications. Front Cell Dev Biol 2023; 11:1112751. [PMID: 36819098 PMCID: PMC9936329 DOI: 10.3389/fcell.2023.1112751] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Ferroptosis is a novel type of cell death associated with iron accumulation and excessive lipid peroxidation. Elucidating the underlying molecular mechanisms of ferroptosis is intensively related to the development and treatment of multiple diseases, including musculoskeletal disorders. Moreover, in vitro and in vivo studies have shown the importance of oxidative stress in musculoskeletal conditions such as osteoporosis, osteoarthritis, rheumatoid arthritis, and osteosarcoma. Ferroptosis-derived clinical management of musculoskeletal diseases offers tremendous and attractive opportunities. Notably, ferroptosis agonists have been proven to enhance the sensitivity of osteosarcoma cells to conventional therapeutic strategies. In this review, we have mainly focused on the implications of ferroptosis regulation in the pathophysiology and therapeutic response of musculoskeletal disorders. Understanding roles of ferroptosis for controlling musculoskeletal diseases might provide directions for ferroptosis-driven therapies, which could be promising for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Cai
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiuju Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanhong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Bi Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Liu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Orthopedic Surgery, The Second Hospital University of South China, Hengyang, China
| |
Collapse
|
41
|
Ding L, Yin J, Xu X, Xie D, Xiang D, Tong P, Liu S, Yang X. Bufalin alleviates acute kidney injury by regulating NLRP3 inflammasome-mediated pyroptosis. Apoptosis 2023; 28:539-548. [PMID: 36652129 DOI: 10.1007/s10495-023-01815-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2023] [Indexed: 01/19/2023]
Abstract
BACKGROUND Recently, there has been an increasing clinical incidence of acute kidney injury (AKI), which rapidly declines renal function and leads to massive tubular cell necrosis. Pyroptosis is an inflammatory process of cell death that is more rapid than apoptosis, which is accompanied by a massive release of inflammasome activation. In the study, we aim to explore whether Bufalin regulates the AKI through the pyroptosis pathway. METHODS We have established gentamicin (GM)-induced AKI animal and cell models to simulate the pathological conditions of kidney injury. The expression of renal injury and pyroptosis-related indicators were detected by western blot. PAS staining and IHC staining were used to analyze renal function. CCK-8 assay was performed to detect cell viability after AKI with different treatments. TUNEL staining, flow cytometry and immunofluorescence assays were performed to measure pyroptosis. RESULTS After intraperitoneal injection of GM in rats, renal function was significantly decreased, along with a significant increase of damaged and necrotic cells as suggested by renal tubular epithelial tissue sections. In addition, there was an increase in the pyroptosis-related markers expression and pyroptosis-induced cell death. Consistently, studies in vitro found that GM significantly induced pyroptosis and its associated protein expression in NRK52e cells. Whereas, the administration of Bufalin reversed these effects of GM in vivo and in vitro. Further, we found that Nigericin (NLRP3 agonist) could reversed the effects of bufalin on GM-induced pyroptosis. CONCLUSION Bufalin attenuates pyroptosis generated AKI by inhibiting NLRP3 inflammasome.
Collapse
Affiliation(s)
- Ling Ding
- Department of Pediatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Yin
- Infectious Department, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xueping Xu
- Department of Pediatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dan Xie
- Department of Pediatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dongxiao Xiang
- Department of Pediatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pingfan Tong
- Department of Pediatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuyu Liu
- Department of General Practice, The Fourth Affiliated Hospital of Nanjing Medical University, 298 Nanpu road, Jiangbei New District, Nanjing, Jiangsu, China
| | - Xilan Yang
- Department of General Practice, The Fourth Affiliated Hospital of Nanjing Medical University, 298 Nanpu road, Jiangbei New District, Nanjing, Jiangsu, China.
| |
Collapse
|
42
|
Theodoropoulou K, Spel L, Zaffalon L, Delacrétaz M, Hofer M, Martinon F. NLRP3 leucine-rich repeats control induced and spontaneous inflammasome activation in cryopyrin-associated periodic syndrome. J Allergy Clin Immunol 2023; 151:222-232.e9. [PMID: 36075321 DOI: 10.1016/j.jaci.2022.08.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/15/2022] [Accepted: 08/11/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND The cryopyrin-associated periodic syndromes (CAPS) comprise a group of rare autoinflammatory diseases caused by gain-of-function mutations in the NLRP3 gene. NLRP3 contains a leucine-rich repeats (LRR) domain with a highly conserved exonic organization that is subjected to extensive alternative splicing. Aberrant NLRP3 inflammasome assembly in CAPS causes chronic inflammation; however, the mechanisms regulating inflammasome function remain unclear. OBJECTIVE We aimed to elucidate the mechanisms regulating NLRP3-mediated autoinflammation in human disease, characterizing the role of LRR in inflammasome activation. METHODS We analyzed sequence read archive data to characterize the pattern of NLRP3 splicing in human monocytes and investigated the role of each LRR-coding exon in inflammasome regulation in genetically modified U937 cells representing CAPS and healthy conditions. RESULTS We detected a range of NLRP3 splice variants in human primary cells and monocytic cell lines, including 2 yet-undescribed splice variants. We observe that lipopolysaccharides affect the abundance of certain splice variants, suggesting that they may regulate NLRP3 activation by affecting alternative splicing. We showed that exons 4, 5, 7, and 9 are essential for inflammasome function, both in the context of wild-type NLRP3 activation by the agonist molecule nigericin and in a model of CAPS-mediated NLRP3 inflammasome assembly. Moreover, the SGT1-NLRP3 interaction is decreased in nonfunctional variants, suggesting that alternative splicing may regulate the recruitment of proteins that facilitate inflammasome assembly. CONCLUSION These findings demonstrate the contribution of the LRR domain in inflammasome function and suggest that navigating LRR exon usage within NLRP3 is sufficient to dampen inflammasome assembly in CAPS.
Collapse
Affiliation(s)
- Katerina Theodoropoulou
- Department of Immunobiology, University of Lausanne; Pediatric Unit of Immunology, Allergology and Rheumatology, University Hospital of Lausanne, Lausanne
| | - Lotte Spel
- Department of Immunobiology, University of Lausanne
| | - Léa Zaffalon
- Department of Immunobiology, University of Lausanne
| | | | - Michaël Hofer
- Pediatric Unit of Immunology, Allergology and Rheumatology, University Hospital of Lausanne, Lausanne
| | | |
Collapse
|
43
|
Ren X, Tao M, Liu X, Zhang L, Li M, Hai Z. Caspase-1-responsive fluorescence biosensors for monitoring endogenous inflammasome activation. Biosens Bioelectron 2023; 219:114812. [PMID: 36272346 DOI: 10.1016/j.bios.2022.114812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/27/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022]
Abstract
The activation of inflammasome leads to secretion of inflammatory factors and cell pyroptosis that are critical in the pathogenesis of various chronic and acute inflammatory diseases. Recruitment and activation of caspase-1 is a marker of inflammasome activation. However, there is still lack of real-time and efficient methods to detect the activation of inflammasome, especially in vivo. Herein, we developed two activatable caspase-1-responsive fluorescence biosensors, WEHD-HCy and YVAD-HCy, to specifically monitor the activation of inflammasome in vivo. Our in vitro study demonstrated that WEHD-HCy and YVAD-HCy can sensitively and specifically respond to caspase-1 activation. Moreover, these biosensors can efficiency and specifically activated in the common inflammatory disease model, including inflammatory bowel disease, Salmonella infection, and acute arthritis. In particular, WEHD-HCy is more advantageous than YVAD-HCy to specifically image of caspase-1 activity both in vitro and in vivo. These caspase-1-responsive fluorescence biosensors provide an efficient, rapid, and in situ tool for monitoring inflammasome activation, and have the potential to be suitable for clinical diagnosis of various inflammatory diseases associated with inflammasome activation.
Collapse
Affiliation(s)
- Xingxing Ren
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510145, China
| | - Menglin Tao
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials, Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Xiaoming Liu
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510145, China
| | - Lele Zhang
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials, Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Mingsong Li
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510145, China.
| | - Zijuan Hai
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials, Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China.
| |
Collapse
|
44
|
Fang Y, Liu J. Novel regulatory role of non-coding RNAs in ankylosing spondylitis. Front Immunol 2023; 14:1131355. [PMID: 36911689 PMCID: PMC9998703 DOI: 10.3389/fimmu.2023.1131355] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Ankylosing spondylitis (AS) is a type of arthritis that primarily affects the spine and involves disorders of the immune and skeletal systems. However, the exact pathogenesis of AS is not fully understood. Non-coding RNAs (ncRNAs), particularly, long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and micro RNAs (miRNAs) and their interactions have been shown to influence many biological processes such as inflammatory responses, osteogenic differentiation and apoptosis, pyroptosis, and proliferation. In addition, ncRNAs reflect the disease activity of AS. In this review, we discuss the regulatory roles of ncRNAs in AS cell functions (inflammatory responses, cellular osteogenic differentiation and apoptosis, pyroptosis, and proliferation) and their potential applications in AS diagnosis and treatment. Understanding the role of ncRNAs in the pathogenesis of AS will lay the foundation for exploring potential new therapeutic approaches for AS.
Collapse
Affiliation(s)
- Yanyan Fang
- The First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, China.,Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Modern Chinese Medicine Department of Internal Medicine Application Foundation Research and Development, Hefei, China
| | - Jian Liu
- The First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Modern Chinese Medicine Department of Internal Medicine Application Foundation Research and Development, Hefei, China.,Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, China
| |
Collapse
|
45
|
Ramirez-Perez S, Reyes-Perez IV, Martinez-Fernandez DE, Hernandez-Palma LA, Bhattaram P. Targeting inflammasome-dependent mechanisms as an emerging pharmacological approach for osteoarthritis therapy. iScience 2022; 25:105548. [PMID: 36465135 PMCID: PMC9708800 DOI: 10.1016/j.isci.2022.105548] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Arthritic diseases have attracted enormous scientific interest because of increased worldwide prevalence and represent a significant socioeconomic burden. Osteoarthritis (OA) is the most prevalent form of arthritis. It is a disorder of the diarthrodial joints, characterized by degeneration and loss of articular cartilage associated with adjacent subchondral bone changes. Chronic and unresolving inflammation has been identified as a critical factor driving joint degeneration and pain in OA. Despite numerous attempts at therapeutic intervention, no effective disease-modifying agents targeting OA inflammation are available to the patients. Inflammasomes are protein complexes known to play a critical role in the inflammatory pathology of several diseases, and their roles in OA pathogenesis have become evident over the last decade. In this sense, it is relevant to evaluate the vital role of inflammasomes as potential modulators of pathogenic features in OA. This review will provide an overview and perspectives on why understanding inflammasome activation is critical for identifying effective OA therapies. We elaborate on the contribution of extracellular mediators from the circulatory system and synovial fluid as well as intracellular activators within the synovial fibroblasts and articular chondrocytes toward invoking the inflammasome in OA. We further discuss the merits of emerging inflammasome targeting therapies and speculate on the potential strategies for inflammasome blockade for OA therapy.
Collapse
Affiliation(s)
- Sergio Ramirez-Perez
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory Musculoskeletal Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Itzel Viridiana Reyes-Perez
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco 44340, México
| | - Diana Emilia Martinez-Fernandez
- Departamento de Farmacobiología, Centro Universitario de Ciencias Exactas e ingenierías, Universidad de Guadalajara, Guadalajara, Jalisco 44430, México
| | - Luis Alexis Hernandez-Palma
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición (IICAN), Centro Universitario del Sur, Universidad de Guadalajara, Guadalajara, Jalisco 49000, México
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco 44340, México
| | - Pallavi Bhattaram
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory Musculoskeletal Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
46
|
Wang S, Wang H, Feng C, Li C, Li Z, He J, Tu C. The regulatory role and therapeutic application of pyroptosis in musculoskeletal diseases. Cell Death Discov 2022; 8:492. [PMID: 36522335 PMCID: PMC9755533 DOI: 10.1038/s41420-022-01282-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Pyroptosis is a controlled form of inflammatory cell death characterized by inflammasome activation, pore formation, and cell lysis. According to different caspases, pyroptosis can be divided into canonical, non-canonical, and other pathways. The role of pyroptosis in disease development has been paid more attention in recent years. The trigger factors of pyroptosis are often related to oxidative stress and proinflammatory substances, which coincide with the pathological mechanism of some diseases. Pyroptosis directly leads to cell lysis and death, and the release of cytosolic components and proinflammatory cytokines affects cell activity and amplifies the inflammatory response. All the above are involved in a series of basic pathological processes, such as matrix degradation, fibrosis, and angiogenesis. Since these pathological changes are also common in musculoskeletal diseases (MSDs), emerging studies have focused on the correlations between pyroptosis and MSDs in recent years. In this review, we first summarized the molecular mechanism of pyroptosis and extensively discussed the differences and crosstalk between pyroptosis, apoptosis, and necrosis. Next, we elaborated on the role of pyroptosis in some MSDs, including osteoarthritis, rheumatoid arthritis, osteoporosis, gout arthritis, ankylosing spondylitis, intervertebral disc degeneration, and several muscle disorders. The regulation of pyroptosis could offer potential therapeutic targets in MSDs treatment. Herein, the existing drugs and therapeutic strategies that directly or indirectly target pyroptosis pathway components have been discussed in order to shed light on the novel treatment for MSDs.
Collapse
Affiliation(s)
- Siyu Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hua Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Chengyao Feng
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Chenbei Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jieyu He
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
47
|
Anand S, Azam Ansari M, Kumaraswamy Sukrutha S, Alomary MN, Anwar Khan A, Elderdery AY. Resolvins Lipid Mediators: Potential Therapeutic Targets in Alzheimer and Parkinson Disease. Neuroscience 2022; 507:139-148. [PMID: 36372297 DOI: 10.1016/j.neuroscience.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/12/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
Abstract
Inflammation and resolution are highly programmed processes involving a plethora of immune cells. Lipid mediators synthesized from arachidonic acid metabolism play a pivotal role in orchestrating the signaling cascades in the game of inflammation. The majority of the studies carried out so far on inflammation were aimed at inhibiting the generation of inflammatory molecules, whereas recent research has shifted more towards understanding the resolution of inflammation. Owing to chronic inflammation as evident in neuropathophysiology, the resolution of inflammation together with the class of lipid mediators actively involved in its regulation has attracted the attention of the scientific community as therapeutic targets. Both omega-three polyunsaturated fatty acids, eicosapentaenoic acid and docosahexaenoic acid, orchestrate a vital regulatory role in inflammation development. Resolvins derived from these fatty acids comprise the D-and E-series resolvins. A growing body of evidence using in vitro and in vivo models has revealed the pro-resolving and anti-inflammatory potential of resolvins. This systematic review sheds light on the synthesis, specialized receptors, and resolution of inflammation mediated by resolvins in Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Santosh Anand
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka, India
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institutes for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia.
| | - Sambamurthy Kumaraswamy Sukrutha
- Department of Microbiology, Biotechnology and Food Technology, Jnana Bharathi Campus, Bangalore University, Bengaluru, Karnataka, India
| | - Mohammad N Alomary
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Anmar Anwar Khan
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abozer Y Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Saudi Arabia
| |
Collapse
|
48
|
Mourmoura E, Papathanasiou I, Trachana V, Konteles V, Tsoumpou A, Goutas A, Papageorgiou AA, Stefanou N, Tsezou A. Leptin-depended NLRP3 inflammasome activation in osteoarthritic chondrocytes is mediated by ROS. Mech Ageing Dev 2022; 208:111730. [PMID: 36087742 DOI: 10.1016/j.mad.2022.111730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 12/30/2022]
Abstract
Leptin and ROS are implicated in the regulation of inflammatory pathways including NLRP3-inflammasome. We investigated the functional link between leptin, ROS and NLRP3-inflammasome formation/activation in osteoarthritis (OA), an age-related disease. We found that inflammasome components' (NLRP3, ASC, Caspase-1 and cleaved Caspase-1) protein expression were increased in OA cartilage biopsies and chondrocytes compared to healthy cartilage and chondrocytes. Immunofluorescence showed increased co-localization of NLRP3/ASC and NLRP3/Caspase-1, ASC-specks formation and ROS levels in OA compared to normal chondrocytes. NOX4 mRNA expression and IL-1β/IL-18 secretion levels were also elevated in OA chondrocytes. Furthermore, NLRP3-siRNA in OA chondrocytes revealed significant MMP-9/MMP-13 downregulation. To elucidate leptin/ROS/NLRP3-inflammasome interactions, OA chondrocytes were treated with ROS-inhibitor NAC, NOXs-inhibitor DPI, NOX4-inhibitor GLX351322 and leptin-siRNA, while normal chondrocytes were incubated with leptin with or without DPI or GLX351322. We observed attenuated ROS levels and NLRP3-inflammasome formation/activation in NAC-, DPI- or GLX351322-treated OA chondrocytes, while the same effect was shown after transfection with leptin-siRNA. Furthermore, incubation of normal chondrocytes with leptin enhanced ROS production and inflammasome formation/activation, while pretreatment with DPI or GLX351322 abolished leptin's stimulatory effects confirming leptin-NOX4-ROS-inflammasome regulatory axis. Overall, our findings provide novel evidence indicating that leptin-induced NLRP3-inflammasome formation/activation in OA chondrocytes is mediated by NOX4-dependent ROS production.
Collapse
Affiliation(s)
- Evanthia Mourmoura
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Ioanna Papathanasiou
- Department of Biology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Varvara Trachana
- Department of Biology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Vasilis Konteles
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Alexandra Tsoumpou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Andreas Goutas
- Department of Biology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | | | - Nikolaos Stefanou
- Department of Orthopaedics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Aspasia Tsezou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece.
| |
Collapse
|
49
|
Zheng Q, Lin R, Chen Y, Lv Q, Zhang J, Zhai J, Xu W, Wang W. SARS-CoV-2 induces "cytokine storm" hyperinflammatory responses in RA patients through pyroptosis. Front Immunol 2022; 13:1058884. [PMID: 36532040 PMCID: PMC9751040 DOI: 10.3389/fimmu.2022.1058884] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
Background The coronavirus disease (COVID-19) is a pandemic disease that threatens worldwide public health, and rheumatoid arthritis (RA) is the most common autoimmune disease. COVID-19 and RA are each strong risk factors for the other, but their molecular mechanisms are unclear. This study aims to investigate the biomarkers between COVID-19 and RA from the mechanism of pyroptosis and find effective disease-targeting drugs. Methods We obtained the common gene shared by COVID-19, RA (GSE55235), and pyroptosis using bioinformatics analysis and then did the principal component analysis(PCA). The Co-genes were evaluated by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and ClueGO for functional enrichment, the protein-protein interaction (PPI) network was built by STRING, and the k-means machine learning algorithm was employed for cluster analysis. Modular analysis utilizing Cytoscape to identify hub genes, functional enrichment analysis with Metascape and GeneMANIA, and NetworkAnalyst for gene-drug prediction. Network pharmacology analysis was performed to identify target drug-related genes intersecting with COVID-19, RA, and pyroptosis to acquire Co-hub genes and construct transcription factor (TF)-hub genes and miRNA-hub genes networks by NetworkAnalyst. The Co-hub genes were validated using GSE55457 and GSE93272 to acquire the Key gene, and their efficacy was assessed using receiver operating curves (ROC); SPEED2 was then used to determine the upstream pathway. Immune cell infiltration was analyzed using CIBERSORT and validated by the HPA database. Molecular docking, molecular dynamics simulation, and molecular mechanics-generalized born surface area (MM-GBSA) were used to explore and validate drug-gene relationships through computer-aided drug design. Results COVID-19, RA, and pyroptosis-related genes were enriched in pyroptosis and pro-inflammatory pathways(the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome complex, death-inducing signaling complex, regulation of interleukin production), natural immune pathways (Network map of SARS-CoV-2 signaling pathway, activation of NLRP3 inflammasome by SARS-CoV-2) and COVID-19-and RA-related cytokine storm pathways (IL, nuclear factor-kappa B (NF-κB), TNF signaling pathway and regulation of cytokine-mediated signaling). Of these, CASP1 is the most involved pathway and is closely related to minocycline. YY1, hsa-mir-429, and hsa-mir-34a-5p play an important role in the expression of CASP1. Monocytes are high-caspase-1-expressing sentinel cells. Minocycline can generate a highly stable state for biochemical activity by docking closely with the active region of caspase-1. Conclusions Caspase-1 is a common biomarker for COVID-19, RA, and pyroptosis, and it may be an important mediator of the excessive inflammatory response induced by SARS-CoV-2 in RA patients through pyroptosis. Minocycline may counteract cytokine storm inflammation in patients with COVID-19 combined with RA by inhibiting caspase-1 expression.
Collapse
Affiliation(s)
- Qingcong Zheng
- Department of Orthopedics, 900th Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Rongjie Lin
- Department of Orthopedics, 900th Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Yuchao Chen
- Department of Paediatrics, Fujian Provincial Hospital South Branch, Fuzhou, China
| | - Qi Lv
- Department of Orthopedics, 900th Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Jin Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, China
| | - Weihong Xu
- Department of Orthopedics, First Affiliated Hospital of Fujian Medical University, Fuzhou, China,*Correspondence: Weihong Xu, ; Wanming Wang,
| | - Wanming Wang
- Department of Orthopedics, 900th Hospital of Joint Logistics Support Force, Fuzhou, China,*Correspondence: Weihong Xu, ; Wanming Wang,
| |
Collapse
|
50
|
Jiao C, Liang H, Liu L, Li S, Chen J, Xie Y. Transcriptomic analysis of the anti-inflammatory effect of Cordyceps militaris extract on acute gouty arthritis. Front Pharmacol 2022; 13:1035101. [PMID: 36313318 PMCID: PMC9614083 DOI: 10.3389/fphar.2022.1035101] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Gouty arthritis (GA) is a common inflammatory disease that causes pain due to the deposition of monosodium urate (MSU) crystals into joints and surrounding tissues. Anti-inflammatory drugs have significant clinical anti-inflammatory and analgesic effects, but they have many side effects. Cordyceps militaris is an edible and medicinal fungus, and its extract (CME) has good anti-inflammatory and analgesic effects. This study aimed to investigate the anti-inflammatory effect of CME on GA and its underlying mechanism. Methods: The effect of CME on the expression of related inflammatory factors and histopathological changes in the MSU-induced acute inflammatory gout model in rats was studied by ELISA and HE, and its anti-inflammatory mechanism was analyzed by transcriptome combined with RT-qPCR. Results: CME significantly improved gait scores and joint swelling in GA rats, and reduced MSU-induced inflammatory cell infiltration. CME inhibited MSU-induced inflammatory responses by reducing the levels of pro-inflammatory factors TNF-α, IL-1β, IL-6, and Caspase-1 and increasing the anti-inflammatory factor IL-10. Transcriptome analysis showed that CME significantly altered inflammation-related cytokine pathways, and identified four major genes involved in regulation of inflammation, CCL7, CSF2RB, LIF, and IL-1β. In addition, RT-qPCR was performed to verify these differential genes. Conclusion: CME significantly alleviated the inflammatory progression of GA and ameliorated the onset of GA. The underlying mechanism may be related to triggering the cytokine-cytokine receptor interaction signaling pathway to inhibit the activation of the inflammasome and regulate the immune system. And it regulates the inflammatory response induced by MSU crystals through the genes CCL7, CSF2RB, and IL-1β.
Collapse
Affiliation(s)
- Chunwei Jiao
- Guangdong Yuewei Edible Fungi Technology Co, Ltd., Guangzhou, China
- Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing, China
| | - Huijia Liang
- Guangdong Yuewei Edible Fungi Technology Co, Ltd., Guangzhou, China
| | - Li Liu
- Guangdong Yuewei Edible Fungi Technology Co, Ltd., Guangzhou, China
| | - Shunxian Li
- Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing, China
| | - Jiaming Chen
- Guangdong Yuewei Edible Fungi Technology Co, Ltd., Guangzhou, China
| | - Yizhen Xie
- Guangdong Yuewei Edible Fungi Technology Co, Ltd., Guangzhou, China
- Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing, China
- *Correspondence: Yizhen Xie,
| |
Collapse
|