1
|
Shin JH, Lee CM, Song JJ. Transcutaneous auricular vagus nerve stimulation mitigates gouty inflammation by reducing neutrophil infiltration in BALB/c mice. Sci Rep 2024; 14:25630. [PMID: 39463429 PMCID: PMC11514149 DOI: 10.1038/s41598-024-77272-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
Gouty inflammation, caused by uric acid crystal deposition, primarily affects tissues around the toe joints and triggers potent inflammatory responses. Current treatments focus on alleviating inflammation and pain using pharmaceutical agents, which can lead to side effects and complications. This has generated interest in non-pharmacological interventions, such as non-invasive vagus nerve stimulation (VNS). In this study, we explored the anti-inflammatory mechanisms of transcutaneous auricular vagus nerve stimulation (taVNS) in a mouse model of acute gout. Gouty inflammation was induced by injecting monosodium urate (MSU) crystals into the ankle joints of BALB/c mice. The effects of taVNS on the expression of inflammatory cytokines and chemokines in the ankle joint tissue were assessed using real-time quantitative PCR (qPCR), western blotting, histological assessments (H&E staining), and immunohistochemistry (IHC). The role of α7 nicotinic acetylcholine receptors (α7nAChR) was also evaluated by signal blocking. Our findings revealed that MSU significantly elevated gout-associated inflammatory cascades and mediators in the ankle joint. Notably, taVNS at 200 µA and 25 Hz effectively reduced these inflammatory responses, decreasing neutrophil infiltration and chemoattraction within the tissue. taVNS showed significant anti-inflammatory properties by suppressing neutrophil activity, offering a novel therapeutic approach for gout beyond conventional pharmacological methods. Additionally, taVNS holds potential for managing various chronic joint diseases. These results highlight taVNS as a promising non-pharmacological therapy for chronic inflammation.
Collapse
Affiliation(s)
- Jae Hee Shin
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul, 08308, Korea
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul, 08308, Korea
| | - Chan Mi Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul, 08308, Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul, 08308, Korea.
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul, 08308, Korea.
- Neurive Institute, Neurive Co., Ltd., Seoul, 08308, Korea.
| |
Collapse
|
2
|
Shin SH, Kim YJ, Kim SJ, Kim GT, Lee H, Kim EY, Lee SH, Sohn KY, Kim JW, Lee JS. The therapeutic effect of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol on chemically induced atopic dermatitis. Sci Rep 2024; 14:23402. [PMID: 39379428 PMCID: PMC11461884 DOI: 10.1038/s41598-024-73951-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/23/2024] [Indexed: 10/10/2024] Open
Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disease worldwide. However, it is still urgent to develop innovative treatments that can effectively manage refractory patients with unpredictable chronic disease courses. In this study, we evaluated the therapeutic efficacy of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) as a novel agent for AD treatment using a human-like mouse model of AD. PLAG significantly improved 2,4-dinitrochlorobenzene (DNCB)-induced AD skin lesions compared to those in mice treated with DNCB alone. PLAG substantially modulated the AD-induced infiltration of monocytes and eosinophils into skin lesions and humoral systemic responses involving immunoglobulin E (IgE), interleukin (IL)-4, and IL-13, restoring them to a normal state. Next, we compared the therapeutic efficacy of PLAG and abrocitinib for severe AD treatment. PLAG exhibited a significant therapeutic effect on AD skin lesions compared to abrocitinib. Unlike abrocitinib, PLAG significantly reduced AD-induced eosinophil infiltration to a level similar to that observed in untreated negative controls. Notably, both PLAG and abrocitinib downregulated IgE, IL-4, and IL-13 in a similar pattern, reaching levels similar to those in the untreated negative controls. Our findings strongly suggest that PLAG may serve as a therapeutic agent for AD with an efficacy comparable to that of abrocitinib.
Collapse
Affiliation(s)
- Su-Hyun Shin
- R&D Institute, Enzychem Lifesciences, 107 Gwanggyo-ro, Suwon, South Korea
| | - Yu-Jin Kim
- R&D Institute, Enzychem Lifesciences, 107 Gwanggyo-ro, Suwon, South Korea
| | - Su-Jin Kim
- R&D Institute, Enzychem Lifesciences, 107 Gwanggyo-ro, Suwon, South Korea
| | - Guen Tae Kim
- R&D Institute, Enzychem Lifesciences, 107 Gwanggyo-ro, Suwon, South Korea
| | - Hyowon Lee
- R&D Institute, Enzychem Lifesciences, 107 Gwanggyo-ro, Suwon, South Korea
| | - Eun Young Kim
- R&D Institute, Enzychem Lifesciences, 107 Gwanggyo-ro, Suwon, South Korea
| | - Se Hee Lee
- R&D Institute, Enzychem Lifesciences, 107 Gwanggyo-ro, Suwon, South Korea
| | - Ki-Young Sohn
- R&D Institute, Enzychem Lifesciences, 107 Gwanggyo-ro, Suwon, South Korea
| | - Jae Wha Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea.
| | - Jae Sam Lee
- R&D Institute, Enzychem Lifesciences, 107 Gwanggyo-ro, Suwon, South Korea.
| |
Collapse
|
3
|
Tian Y, He X, Li R, Wu Y, Ren Q, Hou Y. Recent advances in the treatment of gout with NLRP3 inflammasome inhibitors. Bioorg Med Chem 2024; 112:117874. [PMID: 39167977 DOI: 10.1016/j.bmc.2024.117874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/04/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
Gout is an autoinflammatory disorder characterized by the accumulation of monosodium urate crystals in joints and other tissues, representing the predominant type of inflammatory arthritis with a notable prevalence and propensity for severe outcomes. The NLRP3 inflammasome, a member of the pyrin domain-containing NOD-like receptor family, exerts a substantial impact on both innate and adaptive immune responses and serves as a pivotal factor in the pathogenesis of gout. In recent years, there has been significant academic and industrial interest in the development of NLRP3-targeted small molecule inhibitors as a promising therapeutic approach for gout. To assess the advancements in NLRP3 inflammasome inhibitors for gout treatment, this review offers a comprehensive analysis and evaluation of current clinical candidates and other inhibitors targeting NLRP3 inflammasome from a chemical structure standpoint, with the goal of identifying more efficacious options for clinical management of gout.
Collapse
Affiliation(s)
- Ye Tian
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Xiaofang He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Ruping Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yanxin Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Qiang Ren
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Yusen Hou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| |
Collapse
|
4
|
Dundee JM, Brown GC. The microglial P2Y 6 receptor as a therapeutic target for neurodegenerative diseases. Transl Neurodegener 2024; 13:47. [PMID: 39243044 PMCID: PMC11380353 DOI: 10.1186/s40035-024-00438-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/20/2024] [Indexed: 09/09/2024] Open
Abstract
Neurodegenerative diseases are associated with chronic neuroinflammation in the brain, which can result in microglial phagocytosis of live synapses and neurons that may contribute to cognitive deficits and neuronal loss. The microglial P2Y6 receptor (P2Y6R) is a G-protein coupled receptor, which stimulates microglial phagocytosis when activated by extracellular uridine diphosphate, released by stressed neurons. Knockout or inhibition of P2Y6R can prevent neuronal loss in mouse models of Alzheimer's disease (AD), Parkinson's disease, epilepsy, neuroinflammation and aging, and prevent cognitive deficits in models of AD, epilepsy and aging. This review summarises the known roles of P2Y6R in the physiology and pathology of the brain, and its potential as a therapeutic target to prevent neurodegeneration and other brain pathologies.
Collapse
Affiliation(s)
- Jacob M Dundee
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
| |
Collapse
|
5
|
Li C, Huang Y, Wu C, Qiu Y, Zhang L, Xu J, Zheng J, Zhang X, Li F, Xia D. Astilbin inhibited neutrophil extracellular traps in gouty arthritis through suppression of purinergic P2Y6 receptor. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155754. [PMID: 38820662 DOI: 10.1016/j.phymed.2024.155754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 05/04/2024] [Accepted: 05/15/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND Gouty arthritis (GA), a common inflammatory condition triggered by monosodium urate crystal accumulation, often necessitates safer treatment alternatives due to the limitations of current therapies. Astilbin, a flavonoid from Smilax glabra Roxb, has demonstrated potential in traditional Chinese medicine for its anti-inflammatory properties. However, the anti-GA effect and its underlying mechanism have not been fully elucidated. PURPOSE This study aimed to investigate the therapeutic potential of astilbin in GA, focusing on its effects on neutrophil extracellular traps (NETs), as well as the potential molecular target of GA both in vitro and in vivo. STUDY DESIGN Firstly, astilbin inhibited the citrullinated histone H3 (Cit h3) protein levels and reduced the NETs formation in neutrophils stimulated by monosodium urate (MSU). Secondly, we wondered the effect of astilbin on migration of neutrophils and dimethyl-sulfoxide (DMSO)-differentiated HL-60 (dHL-60) cells under the stimulation of MSU. Then, the effect of astilbin on suppressing NETs through purinergic P2Y6 receptor (P2Y6R) and Interlukin-8 (IL-8)/ CXC chemokine receptor 2 (CXCR2) pathway was investigated. Also, the relationship between P2Y6R and IL-8/CXCR2 was explored in dHL-60 cells under stimulation of MSU. Finally, we testified the effect of astilbin on reducing NETs in GA through suppressing P2Y6R and then down-regulating IL-8/CXCR2 pathway. METHODS MSU was used to induce NETs in neutrophils and dHL-60 cells. Real-time formation of NETs and migration of neutrophils were monitored by cell living imaging with or without MSU. Then, the effect of astilbin on NETs formation, P2Y6R and IL-8/CXCR2 pathway were detected by immunofluorescence (IF) and western blotting. P2Y6R knockdown dHL-60 cells were established by small interfering RNA to investigate the association between P2Y6R and IL-8/CXCR2 pathway. Also, plasmid of P2Y6R was used to overexpress P2Y6R in dHL-60 cells, which was employed to explore the role of P2Y6R in astilbin inhibiting NETs. Within the conditions of knockdown and overexpression of P2Y6R, migration and NETs formation were assessed by transmigration assay and IF staining, respectively. In vivo, MSU-induced GA mice model was established to assess the effect of astilbin on inflammation by haematoxylin-eosin and ELISA. Additionally, the effects of astilbin on neutrophils infiltration, NETs, P2Y6R and IL-8/CXCR2 pathway were analyzed by IF, ELISA, immunohistochemistry (IHC) and western blotting. RESULTS Under MSU stimulation, astilbin significantly suppressed the level of Cit h3 and NETs formation including the fluorescent expressions of Cit h3, neutrophils elastase, myeloperoxidase, and intra/extracellular DNA. Also, results showed that MSU caused NETs release in neutrophils as well as a trend towards recruitment of dHL-60 cells to MSU. Astilbin could markedly decrease expressions of P2Y6R and IL-8/CXCR2 pathway which were upregulated by MSU. By silencing P2Y6R, the expression of IL-8/CXCR2 pathway and migration of dHL-60 cells were inhibited, leading to the suppression of NETs. These findings indicated the upstream role of P2Y6R in the IL-8/CXCR2 pathway. Moreover, overexpression of P2Y6R was evidently inhibited by astilbin, causing a downregulation in IL-8/CXCR2 pathway, migration of dHL-60 cells and NETs formation. These results emphasized that astilbin inhibited the IL-8/CXCR2 pathway primarily through P2Y6R. In vivo, astilbin administration led to marked reductions in ankle swelling, inflammatory infiltration as well as neutrophils infiltration. Expressions of P2Y6R and IL-8/CXCR2 pathway were evidently decreased by astilbin and P2Y6R inhibitor MRS2578 either alone or in combination. Also, astilbin and MRS2578 showed notable effect on reducing MSU-induced NETs formation and IL-8/CXCR2 pathway whether used alone or in combination, parallelly demonstrating that astilbin decreased NETs formation mainly through P2Y6R. CONCLUSION This study revealed that astilbin suppressed NETs formation via downregulating P2Y6R and subsequently the IL-8/CXCR2 pathway, which evidently mitigated GA induced by MSU. It also highlighted the potential of astilbin as a promising natural therapeutic for GA.
Collapse
Affiliation(s)
- Cantao Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Yan Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Chenxi Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Yu Qiu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Lu Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Jiaman Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Junna Zheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Xiaoxi Zhang
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Fenfen Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Daozong Xia
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, PR China.
| |
Collapse
|
6
|
Zhao X, Li M, Lu Y, Wang M, Xiao J, Xie Q, He X, Shuai S. Sirt1 inhibits macrophage polarization and inflammation in gouty arthritis by inhibiting the MAPK/NF-κB/AP-1 pathway and activating the Nrf2/HO-1 pathway. Inflamm Res 2024; 73:1173-1184. [PMID: 38739197 PMCID: PMC11214610 DOI: 10.1007/s00011-024-01890-9] [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: 03/11/2024] [Revised: 03/13/2024] [Accepted: 05/04/2024] [Indexed: 05/14/2024] Open
Abstract
OBJECTIVE AND DESIGN To elucidate Sirt1's role in gouty arthritis inflammation and its potential mechanisms. MATERIAL Constructed murine models of gouty arthritis and conducted THP-1 cell experiments. TREATMENT 1 mg of MSU crystals injected into mice ankle joints for a 72-h intervention. After a 3-h pre-treatment with Sirt1-specific inhibitor (EX527) and agonist (SRT2104), inflammation was induced for 21 h using lipopolysaccharide (LPS) plus MSU crystals. METHODS We assessed gouty arthritis severity through joint inflammation index, swelling, and hematoxylin and eosin (H&E) staining, and measured CD68 mononuclear macrophages and Sirt1 expression in synovial tissue via immunohistochemistry. ELISA, NO assay, RT-qPCR, Flow cytometry, and Western blot were utilized to examine macrophage inflammatory factors, polarization, reactive oxygen species(ROS), MAPK/NF-κB/AP-1 and Nrf2/HO-1 pathways proteins. RESULTS Significant joint swelling, synovial tissue edema, and inflammatory cell infiltration were observed. CD68 mononuclear macrophages and Sirt1 expression were elevated in synovium. Sirt1 activation decreased inflammatory factors, M1 polarization, and ROS generation. Sirt1 activation reduced p38/JNK phosphorylation, thereby inhibiting downstream NF-κB p65/AP-1 and enhancing Nrf2/HO-1, thus suppressing inflammation. CONCLUSIONS Sirt1 alleviates M1 macrophage polarization and inflammation in gouty arthritis by inhibiting the MAPK/NF-κB/AP-1 pathway and activating the Nrf2/HO-1 pathway. Thus, activating Sirt1 may provide a new therapeutic target for gouty arthritis.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Menglan Li
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Yiwei Lu
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Mi Wang
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Jiawei Xiao
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Qingqing Xie
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Xinyi He
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Shiquan Shuai
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China.
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| |
Collapse
|
7
|
Li C, Wu C, Li F, Xu W, Zhang X, Huang Y, Xia D. Targeting Neutrophil Extracellular Traps in Gouty Arthritis: Insights into Pathogenesis and Therapeutic Potential. J Inflamm Res 2024; 17:1735-1763. [PMID: 38523684 PMCID: PMC10960513 DOI: 10.2147/jir.s460333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/07/2024] [Indexed: 03/26/2024] Open
Abstract
Gouty arthritis (GA) is an immune-mediated disorder characterized by severe inflammation due to the deposition of monosodium urate (MSU) crystals in the joints. The pathophysiological mechanisms of GA are not yet fully understood, and therefore, the identification of effective therapeutic targets is of paramount importance. Neutrophil extracellular traps (NETs), an intricate structure of DNA scaffold, encompassing myeloperoxidase, histones, and elastases - have gained significant attention as a prospective therapeutic target for gouty arthritis, due to their innate antimicrobial and immunomodulatory properties. Hence, exploring the therapeutic potential of NETs in gouty arthritis remains an enticing avenue for further investigation. During the process of gouty arthritis, the formation of NETs triggers the release of inflammatory cytokines, thereby contributing to the inflammatory response, while MSU crystals and cytokines are sequestered and degraded by the aggregation of NETs. Here, we provide a concise summary of the inflammatory processes underlying the initiation and resolution of gouty arthritis mediated by NETs. Furthermore, this review presents an overview of the current pharmacological approaches for treating gouty arthritis and summarizes the potential of natural and synthetic product-based inhibitors that target NET formation as novel therapeutic options, alongside elucidating the intrinsic challenges of these inhibitors in NETs research. Lastly, the limitations of HL-60 cell as a suitable substitute of neutrophils in NETs research are summarized and discussed. Series of recommendations are provided, strategically oriented towards guiding future investigations to effectively address these concerns. These findings will contribute to an enhanced comprehension of the interplay between NETs and GA, facilitating the proposition of innovative therapeutic strategies and novel approaches for the management of GA.
Collapse
Affiliation(s)
- Cantao Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Chenxi Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Fenfen Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Wenjing Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Xiaoxi Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Yan Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Daozong Xia
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
8
|
Xu Y, Song D, Wang W, Li S, Yue T, Xia T, Shi Y. Clec12a inhibits MSU-induced immune activation through lipid raft expulsion. Life Sci Alliance 2023; 6:e202301938. [PMID: 37339805 PMCID: PMC10282328 DOI: 10.26508/lsa.202301938] [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: 01/20/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/22/2023] Open
Abstract
Monosodium uric acid (MSU) crystal, the etiological agent of gout, has been shown to trigger innate immune responses via multiple pathways. It is known that MSU-induced lipid sorting on plasma membrane promotes the phosphorylation of Syk and eventually leads to the activation of phagocytes. However, whether this membrane lipid-centric mechanism is regulated by other processes is unclear. Previous studies showed that Clec12a, a member of the C-type lectin receptor family, is reported to recognize MSU and suppresses this crystalline structure-induced immune activation. How this scenario is integrated into the lipid sorting-mediated inflammatory responses by MSU, and particularly, how Clec12a intercepts lipid raft-originated signaling cascade remains to be elucidated. Here, we found that the ITIM motif of Clec12a is dispensable for its inhibition of MSU-mediated signaling; instead, the transmembrane domain of Clec12a disrupts MSU-induced lipid raft recruitment and thus attenuates downstream signals. Single amino acid mutagenesis study showed the critical role of phenylalanine in the transmembrane region for the interactions between C-type lectin receptors and lipid rafts, which is critical for the regulation of MSU-mediated lipid sorting and phagocyte activation. Overall, our study provides new insights for the molecular mechanisms of solid particle-induced immune activation and may lead to new strategies in inflammation control.
Collapse
Affiliation(s)
- Ying Xu
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Dingka Song
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- Institute of Coastal Environmental Pollution Control, Key Laboratory of Marine Environment and Ecology, Ministry of Education, Ocean University of China, Qingdao, China
| | - Shixin Li
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, China
| | - Tongtao Yue
- Institute of Coastal Environmental Pollution Control, Key Laboratory of Marine Environment and Ecology, Ministry of Education, Ocean University of China, Qingdao, China
| | - Tie Xia
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Yan Shi
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
9
|
Zaninelli TH, Martelossi-Cebinelli G, Saraiva-Santos T, Borghi SM, Fattori V, Casagrande R, Verri WA. New drug targets for the treatment of gout arthritis: what's new? Expert Opin Ther Targets 2023; 27:679-703. [PMID: 37651647 DOI: 10.1080/14728222.2023.2247559] [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: 04/12/2023] [Revised: 06/14/2023] [Accepted: 08/09/2023] [Indexed: 09/02/2023]
Abstract
INTRODUCTION Gout arthritis (GA) is an intermittent inflammatory disease affecting approximately 10% of the worldwide population. Symptomatic phases (acute flares) are timely spaced by asymptomatic periods. During an acute attack, redness, joint swelling, limited movement, and excruciating pain are common symptoms. However, the current available therapies are not fully effective in reducing symptoms and offer numerous side effects. Therefore, unveiling new drug targets and effector molecules are required in developing novel GA therapeutics. AREAS COVERED This review discusses the pathophysiological mechanisms of GA and explores potential pharmacological targets to ameliorate disease outcome. In addition, we listed promising pre-clinical studies demonstrating effector molecules with therapeutical potential. Among those, we emphasized the importance of natural products, including traditional Chinese medicine formulas and their multitarget mechanisms of action. EXPERT OPINION In our search, we observed that there is a massive gap between pre-clinical and clinical knowledge. Only a minority (4.4%) of clinical trials aimed to intervene by applying natural products or current hot targets described herein. In this sense, we envisage four possibilities for GA therapeutics, which include the repurposing of existing therapies, ALX/FPR2 agonism for improvement in disease outcome, the use of multitarget drugs (e.g. natural products), and targeting the neuroinflammatory component of GA.
Collapse
Affiliation(s)
- Tiago H Zaninelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Geovana Martelossi-Cebinelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Telma Saraiva-Santos
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Sergio M Borghi
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
- Center for Research in Health Sciences, University of Northern Londrina, Londrina, Brazil
| | - Victor Fattori
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital-Harvard Medical School, Karp Research Building, Boston, MA, USA
| | - Rubia Casagrande
- Laboratory of Antioxidants and Inflammation, Department of Pharmaceutical Sciences, Centre of Health Sciences, Londrina State University, Londrina, Brazil
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| |
Collapse
|
10
|
Girard M, Bellefeuille SD, Eiselt É, Arguin G, Longpré JM, Sarret P, Gendron FP. Ligand-dependent intracellular trafficking of the G protein-coupled P2Y 6 receptor. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119476. [PMID: 37059189 DOI: 10.1016/j.bbamcr.2023.119476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 04/16/2023]
Abstract
Endosomal trafficking is intricately linked to G protein-coupled receptors (GPCR) fate and signaling. Extracellular uridine diphosphate (UDP) acts as a signaling molecule by selectively activating the GPCR P2Y6. Despite the recent interest for this receptor in pathologies, such as gastrointestinal and neurological diseases, there is sparse information on the endosomal trafficking of P2Y6 receptors in response to its endogenous agonist UDP and synthetic selective agonist 5-iodo-UDP (MRS2693). Confocal microscopy and cell surface ELISA revealed delayed internalization kinetics in response to MRS2693 vs. UDP stimulation in AD293 and HCT116 cells expressing human P2Y6. Interestingly, UDP induced clathrin-dependent P2Y6 internalization, whereas receptor stimulation by MRS2693 endocytosis appeared to be associated with a caveolin-dependent mechanism. Internalized P2Y6 was associated with Rab4, 5, and 7 positive vesicles independent of the agonist. We have measured a higher frequency of receptor expression co-occurrence with Rab11-vesicles, the trans-Golgi network, and lysosomes in response to MRS2693. Interestingly, a higher agonist concentration reversed the delayed P2Y6 internalization and recycling kinetics in the presence of MRS2693 stimulation without changing its caveolin-dependent internalization. This work showed a ligand-dependent effect affecting the P2Y6 receptor internalization and endosomal trafficking. These findings could guide the development of bias ligands that could influence P2Y6 signaling.
Collapse
Affiliation(s)
- Mélissa Girard
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Canada
| | - Steve Dagenais Bellefeuille
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Émilie Eiselt
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Canada
| | - Guillaume Arguin
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jean-Michel Longpré
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Canada
| | - Philippe Sarret
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Canada
| | - Fernand-Pierre Gendron
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Canada.
| |
Collapse
|
11
|
Kim GT, Kim EY, Shin SH, Lee H, Lee SH, Sohn KY, Kim JW. Improving anticancer effect of aPD-L1 through lowering neutrophil infiltration by PLAG in tumor implanted with MB49 mouse urothelial carcinoma. BMC Cancer 2022; 22:727. [PMID: 35787261 PMCID: PMC9251917 DOI: 10.1186/s12885-022-09815-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The PD-L1 antibody is an immune checkpoint inhibitor (ICI) attracting attention. The third-generation anticancer drug has been proven to be very effective due to fewer side effects and higher tumor-specific reactions than conventional anticancer drugs. However, as tumors produce additional resistance in the host immune system, the effectiveness of ICI is gradually weakening. Therefore, it is very important to develop a combination therapy that increases the anticancer effect of ICI by removing anticancer resistance factors present around the tumor. METHODS The syngeneic model was used (n = 6) to investigate the enhanced anti-tumor effect of PD-L1 antibody with the addition of PLAG. MB49 murine urothelial cancer cells were implanted into the C57BL/6 mice subcutaneously. PLAG at different dosages (50/100 mpk) was daily administered orally for another 4 weeks with or without 5 mpk PD-L1 antibody (10F.9G2). PD-L1 antibody was delivered via IP injection once a week. RESULTS The aPD-L1 monotherapy group inhibited tumor growth of 56% compared to the positive group, while the PLAG and aPD-L1 co-treatment inhibited by 89%. PLAG treatment effectively reduced neutrophils infiltrating localized in tumor and converted to a tumor microenvironment with anti-tumor effective T-cells. PLAG increased tumor infiltration of CD8 positive cytotoxic T-cell populations while effectively inhibiting the infiltration of neoplastic T-cells such as CD4/FoxP3. Eventually, neutrophil-induced tumor ICI resistance was resolved by restoring the neutrophil-to-lymphocyte ratio to the normal range. In addition, regulation of cytokine and chemokine factors that inhibit neutrophil infiltration and increase the killing activity of cytotoxic T cells was observed in the tumors of mice treated with PLAG + aPD-L1. CONCLUSIONS PLAG effectively turned the tumor-promoting microenvironment into a tumor-suppressing microenvironment. As a molecule that increases the anti-tumor effectiveness of aPD-L1, PLAG has the potential to be an essential and effective ICI co-therapeutic agent.
Collapse
Affiliation(s)
- Guen Tae Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Eun Young Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Su-Hyun Shin
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Hyowon Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Se Hee Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Ki-Young Sohn
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Jae Wha Kim
- grid.249967.70000 0004 0636 3099Division of Systems Biology and Bioengineering, Cell Factory Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea
| |
Collapse
|
12
|
Kim GT, Shin SH, Kim EY, Lee H, Lee SH, Sohn KY, Kim JW. PLAG co-treatment increases the anticancer effect of Adriamycin and cyclophosphamide in a triple-negative breast cancer xenograft mouse model. Biochem Biophys Res Commun 2022; 619:110-116. [PMID: 35753218 DOI: 10.1016/j.bbrc.2022.06.051] [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: 05/24/2022] [Revised: 06/09/2022] [Accepted: 06/16/2022] [Indexed: 11/02/2022]
Abstract
Chemotherapy induces tumor cell death and inhibits tumor progression, but the accompanying immune responses in the surrounding dying tissue cause significant inflammation. These responses, such as excessive neutrophil infiltration into tumor tissue, are the main causes of resistance to anticancer treatment. The development of drugs that reduce neutrophil infiltration into tumors is necessary to increase the anticancer effect of chemotherapy. Here, we show that the antitumor effect of the chemotherapy AC regimen (Adriamycin and cyclophosphamide) was increased by 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) cotreatment in the MDA-MB-231 triple-negative breast cancer xenograft mouse model. Tumor growth was inhibited up to 56% in mice treated with AC and inhibited up to 94% in mice cotreated with AC and PLAG. Side effects of chemotherapy, such as a reduction in body weight, were alleviated in mice cotreated with AC and PLAG. Excessive neutrophil infiltration caused by the AC regimen was successfully cleared in mice cotreated with AC and PLAG. We conclude that PLAG inhibits excessive neutrophil infiltration that aids tumor growth. Reduced neutrophils and increased lymphocytes in PLAG-treated mice can maximize the antitumor effect of the AC regimen and inhibit tumor growth.
Collapse
Affiliation(s)
- Guen Tae Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Su-Hyun Shin
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Eun Young Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Hyowon Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Se Hee Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Ki-Young Sohn
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Jae Wha Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea.
| |
Collapse
|
13
|
Kim GT, Kim EY, Shin SH, Lee H, Lee SH, Sohn KY, Kim JW. Suppression of tumor progression by thioredoxin-interacting protein-dependent adenosine 2B receptor degradation in a PLAG-treated Lewis lung carcinoma-1 model of non-small cell lung cancer. Neoplasia 2022; 31:100815. [PMID: 35728512 PMCID: PMC9209866 DOI: 10.1016/j.neo.2022.100815] [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: 05/17/2022] [Revised: 05/23/2022] [Accepted: 06/08/2022] [Indexed: 11/03/2022]
Abstract
PLAG effectively inhibited excessive growth of LLC1 cells in an NSCLC model. PLAG inhibited tumor growth by inducing adenosine 2B receptor (A2BR) degradation. Unlike antagonists, PLAG terminates rather than suppresses signaling pathways. A2BR degradation by PLAG occurs through expression and re-localization of TXNIP.
Extracellular adenosine in the tumor microenvironment plays a vital role in cancer development. Specifically, activation of adenosine receptors affects tumor cell growth and adenosine release. We examined the anti-tumor efficacy of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) in animal models, revealing the role of PLAG in inhibiting tumor progression by promoting the degradation of adenosine 2B receptors (A2BRs) in tumors. PLAG induced the expression of thioredoxin-interacting protein (TXNIP), a type of α-arrestin that accelerates A2BR internalization by interacting with A2BR complexes containing β-arrestin. Engulfed receptors bound to TXNIP were rapidly degraded after E3 ligase recruitment and ubiquitination, resulting in early termination of intracellular signals that promote tumor overgrowth. However, in control cancer cells, A2BRs bound to protein phosphatase 2A and were returned to the cell membrane instead of being degraded, resulting in continuous receptor-mediated signaling by pathways including the Raf-Erk axis, which promotes tumor proliferation. A TXNIP-silenced cell-implanted mouse model and TXNIP knockout (KO) mice were used to verify that PLAG-mediated suppression of tumor progression is dependent on TXNIP expression. Increased tumor growth was observed in TXNIP-silenced cell-implanted mice, and the anti-tumor effects of PLAG, including delayed tumor overgrowth, were greatly reduced. However, the anti-tumor effects of PLAG were observed in cancer cell-implanted TXNIP-KO mice, which indicates that PLAG produces anti-tumor effects by enhancing TXNIP expression in tumor cells. These essential functions of PLAG, including delaying tumor growth via A2BR degradation, suggest innovative directions for anticancer drug development.
Collapse
Affiliation(s)
- Guen Tae Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Eun Young Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Su-Hyun Shin
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Hyowon Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Se Hee Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Ki-Young Sohn
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Jae Wha Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea.
| |
Collapse
|
14
|
Kim GT, Kim EY, Shin SH, Lee H, Lee SH, Park K, Sohn KY, Yoon SY, Kim JW. PLAG alleviates cisplatin-induced cachexia in lung cancer implanted mice. Transl Oncol 2022; 20:101398. [PMID: 35339890 PMCID: PMC8957043 DOI: 10.1016/j.tranon.2022.101398] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 12/23/2022] Open
Abstract
PLAG effectively relieves cachexia symptoms caused by chemotherapy. PLAG restores leg muscles atrophy by cisplatin to normal levels. PLAG returns cachexia-related elements modulated by cisplatin to normal levels. PLAG did not downregulate the anti-tumor effect of cisplatin.
Chemotherapy-induced cachexia has been a significant challenge to the successful treatment of cancer patients. Chemotherapy leads to loss of muscle, loss of appetite, and excessive weight loss, which makes these necessary treatments intolerable for most patients. Therefore, it is necessary to alleviate cachexia to successfully treat cancer patients. In this study, tumor-implanted mouse models administered cisplatin showed rapid weight loss and reduced feeding rate by the second week of treatment, and 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) effectively alleviated cisplatin-induced cachexia. In mice treated with cisplatin on a sacrificial day after 6 weeks, the weight of the two major leg muscles (quadriceps femoris and gastrocnemius) were reduced by up to 70%, but this muscle reduction was successfully prevented in the PLAG co-treatment group. The distribution and size of muscle fibers that appear in small units in cisplatin-treated mice were restored to normal levels by PLAG co-treatment. Furthermore, myostatin expression levels were upregulated by cisplatin, whereas myostatin decreased to normal levels with muscle recovery in the PLAG co-treated group. Tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6), which are commonly expressed in cachexia, were significantly increased in cisplatin-treated mice but were reduced to normal levels in PLAG co-treated mice. Glucose absorption, an indicator of muscle tissue activity, decreased with cisplatin treatment and recovered to normal levels with PLAG co-treatment. Overall, PLAG effectively alleviated cisplatin-induced cachexia symptoms and reduced tumor growth in tumor-implanted mice. These findings suggest PLAG may be a promising drug to alleviate cachexia in cancer patients receiving chemotherapy.
Collapse
Affiliation(s)
- Guen Tae Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Eun Young Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Su-Hyun Shin
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Hyowon Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Se Hee Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Kaapjoo Park
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Ki-Young Sohn
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Sun Young Yoon
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea.
| | - Jae Wha Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea.
| |
Collapse
|
15
|
Li X, Gao J, Tao J. Purinergic Signaling in the Regulation of Gout Flare and Resolution. Front Immunol 2021; 12:785425. [PMID: 34925366 PMCID: PMC8671294 DOI: 10.3389/fimmu.2021.785425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/15/2021] [Indexed: 12/15/2022] Open
Abstract
Gout flares require monosodium urate (MSU) to activate the NLRP3 inflammasome and secrete sufficient IL-1β. However, MSU alone is not sufficient to cause a flare. This is supported by the evidence that most patients with hyperuricemia do not develop gout throughout their lives. Recent studies have shown that, besides MSU, various purine metabolites, including adenosine triphosphate, adenosine diphosphate, and adenosine bind to different purine receptors for regulating IL-1β secretion implicated in the pathogenesis of gout flares. Purine metabolites such as adenosine triphosphate mainly activate the NLRP3 inflammasome through P2X ion channel receptors, which stimulates IL-1β secretion and induces gout flares, while some purine metabolites such as adenosine diphosphate and adenosine mainly act on the G protein-coupled receptors exerting pro-inflammatory or anti-inflammatory effects to regulate the onset and resolution of a gout flare. Given that the purine signaling pathway exerts different regulatory effects on inflammation and that, during the inflammatory process of a gout flare, an altered expression of purine metabolites and their receptors was observed in response to the changes in the internal environment. Thus, the purine signaling pathway is involved in regulating gout flare and resolution. This study was conducted to review and elucidate the role of various purine metabolites and purinergic receptors during the process.
Collapse
Affiliation(s)
| | | | - Jinhui Tao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|