51
|
Xiao WZ, Zhao L, Cao L, Zhu XX, Zou HJ. Melatonin Alleviates Acute Gouty Inflammation In Vivo and In Vitro. Curr Med Sci 2021; 41:757-763. [PMID: 34047943 DOI: 10.1007/s11596-021-2362-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/02/2021] [Indexed: 12/17/2022]
Abstract
The aim of this study was to identify the effects of melatonin on acute gouty inflammation and to investigate the underlying mechanisms. We found significantly lower serum melatonin levels in gout patients in the acute phase than in those in the remission phase or in normal individuals. The mRNA expression of melatonin receptor 2 (MT2) was also lower in gout patients than in normal individuals. To verify the in-vivo role of melatonin, a gouty arthritis model was established by intraarticular injection of monosodium urate (MSU, 1 mg) crystals into the paws of C57BL/6 mice. Joint inflammation in the mouse model was evaluated by measuring the thickness of the right paw/left paw, and the inflammation index was determined by examining infiltrating neutrophils with haematoxylin and eosin (H&E) staining. Melatonin was found to reduce both paw thickness and the inflammation index in the mouse model, and melatonin also reduced the mRNA levels of interleukin-1 beta (IL-1β), IL-6 and NLR family pyrin domain containing 3 (NLRP3) inflammasome. To mimic gouty inflammation in vitro, mouse peritoneal macrophages were stimulated with lipopolysaccharides (LPS) plus MSU. Melatonin was revealed to reduce IL-1β secretion by stimulated macrophages. The mRNA expression levels of IL-1β and IL-6 were also inhibited by melatonin. Western blot analysis showed that the expression of NLRP3, caspase-1 and pro-IL-1β was also inhibited by melatonin. In conclusion, our study demonstrated that melatonin alleviated gouty inflammation in vivo and in vitro, and the underlying mechanism may involve inhibiting the assembly of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Wen-Ze Xiao
- Department of Rheumatology and Immunology, Huashan Hospital Affiliated to Fudan University, Shanghai, 200040, China
- Department of Rheumatology and Immunology, Fudan University Pudong Medical Center, Shanghai, 201300, China
| | - Li Zhao
- Department of Rheumatology and Immunology, Huashan Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - Lin Cao
- Department of Rheumatology and Immunology, Huashan Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - Xiao-Xia Zhu
- Department of Rheumatology and Immunology, Huashan Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - He-Jian Zou
- Department of Rheumatology and Immunology, Huashan Hospital Affiliated to Fudan University, Shanghai, 200040, China.
| |
Collapse
|
52
|
Yang QB, Li LQ, Zhang QB, He YL, Mi QS, Zhou JG. microRNA-223 Deficiency Exacerbates Acute Inflammatory Response to Monosodium Urate Crystals by Targeting NLRP3. J Inflamm Res 2021; 14:1845-1858. [PMID: 34007200 PMCID: PMC8123978 DOI: 10.2147/jir.s307796] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/14/2021] [Indexed: 01/13/2023] Open
Abstract
Objective MicroRNAs were identified as master-switch molecules limiting acute inflammatory response. This study investigated the potential role of microRNA (miR)-223 in the mechanism of gout. Methods Wild-type (WT) and miR-223 knock-out (KO) mice were used to evaluate the phenotypes of gout models. Inflammatory cytokines were measured in air pouch and peritoneal cavity lavage fluid. In addition to miR-223 level in gout patients, miR-223 and pro-inflammatory genes were examined in bone marrow-derived macrophages (BMDMs) from mice as well as peripheral blood mononuclear cells from healthy controls (HC) treated with monosodium urate (MSU) crystals in vitro. Results MiR-223 was up-regulated in the early phase in BMDMs from WT mice after MSU challenge and decreased rapidly, and this was not observed in miR-223 KO mice in vitro. In addition, miR-223 was required for macrophages homeostasis. In comparison with WT mice in vivo, miR-223 deficiency exacerbated swelling index of MSU-induced inflammation in foot pad and ankle joint models. MiR-223 deficiency also markedly aggravated inflammatory cells infiltration and cytokines release including interleukin (IL)-1β, IL-6 and monocyte chemotactic protein-1 (MCP-1) in the air pouch and peritonitis models. In the in vitro experiments, miR-223 deficiency promoted the inflammatory response by targeting NLR family pyrin domain containing protein 3 (NLRP3). Besides, miR-223 level was down-regulated in gout patients and in HC exposed to MSU in vitro. Conclusion MiR-223 was down-regulated in gout patients and miR-223 deficiency exacerbated inflammatory response in diverse murine models, suggesting that up-regulation of miR-223 could be a potential therapeutic strategy for alleviating gouty inflammation.
Collapse
Affiliation(s)
- Qi-Bin Yang
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan Province, People's Republic of China.,Henry Ford Immunology Program, Departments of Dermatology and Internal Medicine, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Ling-Qin Li
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan Province, People's Republic of China
| | - Quan-Bo Zhang
- Henry Ford Immunology Program, Departments of Dermatology and Internal Medicine, Henry Ford Health System, Detroit, MI, 48202, USA.,Department of Gerontology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan Province, People's Republic of China
| | - Yong-Long He
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan Province, People's Republic of China
| | - Qing-Sheng Mi
- Henry Ford Immunology Program, Departments of Dermatology and Internal Medicine, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Jing-Guo Zhou
- Department of Rheumatology and Immunology, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan Province, People's Republic of China
| |
Collapse
|
53
|
Jeong JH, Choi SJ, Ahn SM, Oh JS, Kim YG, Lee CK, Yoo B, Hong S. Neutrophil extracellular trap clearance by synovial macrophages in gout. Arthritis Res Ther 2021; 23:88. [PMID: 33741037 PMCID: PMC7977263 DOI: 10.1186/s13075-021-02472-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/02/2021] [Indexed: 01/27/2023] Open
Abstract
Background Monosodium urate (MSU) crystals, i.e., the central etiological factors in gouty arthritis, induce the formation of neutrophil extracellular traps (NETs). We investigated whether synovial macrophages could clear NETs as a self-resolution mechanism in acute gouty arthritis. Methods Synovial fluid mononuclear cells (SFMCs) were incubated with NETs induced by MSU crystals. NET engulfment was determined based on neutrophil elastase (NE), myeloperoxidase (MPO), and SYTOX Green signals within synovial fluid CD14+ cells. In addition, the correlations between CD14+ cells, MPO-dsDNA complexes, and expression of pro- and anti-inflammatory cytokines were analyzed in the synovial fluid CD14+ macrophages of patients with gouty arthritis. Results Synovial fluid CD14+ macrophages significantly engulfed the MSU crystal-induced NETs, as evidenced by the alteration in SYTOX Green intensity or the presence of NE and MPO in the cytoplasm of CD14+ cells. The proportion of CD14+ macrophages was significantly and inversely correlated with levels of MPO-dsDNA complex in the synovial fluid of gout patients. Synovial fluid CD14+ macrophages cultured with NETs did not show a significant induction in pro- and anti-inflammatory cytokines. Conclusion Synovial fluid macrophages may play an important role in the resolution of MSU crystal-induced gouty inflammation by clearing NETs without causing any significant immunological response. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02472-4.
Collapse
Affiliation(s)
- Ji Hye Jeong
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea.,Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Su Jin Choi
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Soo Min Ahn
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Ji Seon Oh
- Clinical Research Center, Asan Medical Center, Seoul, South Korea
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Chang-Keun Lee
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Bin Yoo
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Seokchan Hong
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea.
| |
Collapse
|
54
|
Zhang X, Ding H, Hu G, Lu Y. Simiaosan alleviates the symptoms of gouty arthritis via the NALP3/IL‑1β pathway. Mol Med Rep 2021; 23:223. [PMID: 33655332 PMCID: PMC7851828 DOI: 10.3892/mmr.2021.11862] [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: 03/02/2020] [Accepted: 09/28/2020] [Indexed: 12/17/2022] Open
Abstract
Previous studies have suggested that the herbal medicine simiaosan has beneficial effects on gouty arthritis (GA), for which conventional Western medicines are insufficient (particularly in cases of multiple episodes). The objective of the present study was to investigate the mechanism by which simiaosan alleviated the symptoms of GA. Sprague-Dawley rat models of acute GA were successfully established, as verified by pathological analyses. Additionally, an NLR family pyrin domain containing 3 (NLRP3) overexpression vector was constructed and a high transfection efficiency was confirmed by reverse transcription PCR. The following five treatment groups were established: i) Normal control; ii) model + saline; iii) model + simiaosan; iv) model + NALP3-overexpressing adenovirus + simiaosan; and v) model + empty vector adenovirus + simiaosan. The samples from mice in each group were subjected to hematoxylin and eosin (H&E) staining for assessing the histopathological changes, enzyme-linked immunosorbent assays for determining IL-1β and TGF-β1 levels and western blotting for evaluating NALP3 expression. H&E staining indicated that simiaosan could reduce the infiltration of inflammatory cells, while NALP3 overexpression aggravated the inflammatory response in tissues. Expression levels of IL-1β, TGF-β1 and NALP3 were significantly higher in the model and the model + NALP3-overexpressing adenovirus + simiaosan groups compared with the normal control group. Levels of IL-1β, TGF-β1 and NALP3 were significantly lower in the model + simiaosan and model + empty vector adenovirus + simiaosan groups compared with the model group. These results indicated that the effects of simiaosan were mediated through NALP3 inhibition. Therefore, the herbal medicine simiaosan was revealed to possess an ability to alleviate the symptoms of GA by regulating the NALP3/IL-1β signaling pathway.
Collapse
Affiliation(s)
- Xianzhen Zhang
- Department of Rheumatology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Huideng Ding
- Department of Rheumatology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Gang Hu
- Department of Rheumatology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Ying Lu
- Department of Rheumatology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| |
Collapse
|
55
|
Huang Y, Wu X, Gui L, Jiang Y, Tu L, Li X, Jiang B, Wang Y, Zheng X, Wei Q, Li Q, Ou J, Chen Z, Xie Y, Lin Z, Liao Z, Fang L, Qiu M, Cao S, Gu J. Age-Specific Imbalance of Circulating Tfh Cell Subsets and Its Association With Gout-Targeted Kidney Impairment. Front Immunol 2021; 11:625458. [PMID: 33505406 PMCID: PMC7829215 DOI: 10.3389/fimmu.2020.625458] [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: 11/03/2020] [Accepted: 11/16/2020] [Indexed: 11/16/2022] Open
Abstract
Objective Gout is a chronic disease characterized by the deposition of monosodium urate (MSU) crystals in tissue. Study with a focus on adaptive immune response remains to be understood although innate immune response has been reported extensively in gout etiology. Our study attempted to investigate the association of gout-related immune cell imbalance with clinical features and comorbidity with renal impairment and the implicated pathogenesis via the assessment of T and B cell subsets in different activity phases or with immune effects combined with the analyses of clinical parameters. Methods Fifty-eight gout patients and 56 age- and sex-matched healthy individuals were enrolled. To learn the roles of circulating T cells, a lymphocyte profile incorporating 32 T cell subsets was tested from isolated freshly peripheral blood monocyte cells (PBMCs) with multiple-color flow cytometry. Furthermore, the collected clinical features of participants were used to analyze the characteristics of these differential cell subsets. Stratified on the basis of the level of creatinine (Cr, enzymatic method), all patients were categorized into Crlow (Cr ≤ 116 μmol/L) and Crhi (Cr > 116 μmol/L) groups to exploit whether these gout-associated T cell subsets were functional in gout-targeted kidney dysfunction. The differentiation of B cells was investigated in gout patients. Results Our results show that CD 4+ T cells, Th2 cells, and Tc2 cells were upregulated, whereas Tc17 cells were downregulated. Tfh cells skewed toward the polarization of Tfh2 cells. Specifically, Tfh2 cells increased, but Tfh1 cells decreased, accompanied with aging for gout patients, suggesting that age might trigger the skewing of Tfh1/Tfh2 cell subsets to influence gout development. Moreover, Tfh2 cells were connected to renal dysfunction as well. No alterations of B cell subsets were observed in patients when compared to controls. Conclusions Our data demonstrate age-specific dysfunctions of Tfh1/2 cells in gout occurrence, and Tfh2 cell upregulation is associated with gout-targeted renal dysfunction. However, Tfh2 cells may function in auto-inflammatory gout independent of helping B differentiation, and an in-depth study remains to be conducted.
Collapse
Affiliation(s)
- Yefei Huang
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xinyu Wu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Lian Gui
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yutong Jiang
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Liudan Tu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaomin Li
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Boxiong Jiang
- Medical Examination Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yina Wang
- VIP Medical Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xuqi Zheng
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qiujing Wei
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qiuxia Li
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jiayong Ou
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zena Chen
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ya Xie
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhiming Lin
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zetao Liao
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Linkai Fang
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Minli Qiu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shuangyan Cao
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jieruo Gu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
56
|
Qadri M, ElSayed S, Elsaid KA. Fingolimod Phosphate (FTY720-P) Activates Protein Phosphatase 2A in Human Monocytes and Inhibits Monosodium Urate Crystal-Induced Interleukin-1 β Production. J Pharmacol Exp Ther 2020; 376:222-230. [PMID: 33239408 DOI: 10.1124/jpet.120.000321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/20/2020] [Indexed: 12/27/2022] Open
Abstract
Gout is a chronic inflammatory arthritis caused by monosodium urate monohydrate (MSU) crystal deposits in joints of lower limbs. Phagocytic uptake of MSU crystals by joint-resident macrophages and recruited circulating monocytes results in IL-1β expression and production. Current acute gout treatments have serious toxicities and suffer suboptimal clinical outcomes. Protein phosphatase 2A (PP2A) plays an important role in regulating signaling pathways relevant to inflammation. We hypothesized that innate immune danger signals, e.g., lipopolysaccharide (LPS) and soluble uric acid (sUA), prime human monocytes toward MSU crystal phagocytosis and that increased IL-1β production mediated by a reduction in PP2A activity and restoring PP2A activity exerts an anti-inflammatory effect in this setting. Priming monocytes with LPS + sUA increased cytosolic pro-IL-1β and mature IL-1β and enhanced MSU crystal phagocytosis and its downstream IL-1β expression (P < 0.001). A combination of LPS + sUA priming and MSU crystals reduced PP2A activity in monocytes by 60% (P = 0.013). PP2A catalytic subunit gene knockdown reduced PP2A activity and exacerbated MSU crystal-induced IL-1β expression and secretion (P < 0.0001). Fingolimod (FTY720) and its active metabolite, fingolimod phosphate (FTY720-P), were evaluated for their ability to activate PP2A in human monocytes over 24 hours. FTY720 and FTY720-P activated PP2A to a similar extent, and maximal enzyme activity occurred at 24 hours for FTY720 and at 6 hours for FTY720-P. FTY720-P (2.5 μM) reduced pro-IL-1β production and IL-1β secretion in primed and MSU crystal-stimulated monocytes (P < 0.0001) without changing the magnitude of crystal phagocytosis. We conclude that PP2A is a promising new target in acute gout. SIGNIFICANCE STATEMENT: The activity of protein phosphatase 2A (PP2A) is implicated in the enhanced expression and production of IL-1β by human monocytes in response to priming with soluble uric acid and lipopolysaccharide and phagocytosis of monosodium urate monohydrate (MSU) crystals. Fingolimod phosphate activates PP2A in human monocytes and reduces cytosolic pro-IL-1β content and its conversion to biologically active IL-1β in human monocytes exposed to MSU crystals.
Collapse
Affiliation(s)
- Marwa Qadri
- Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia (M.Q.) and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Sciences Campus, Irvine, California (S.E., K.A.E.)
| | - Sandy ElSayed
- Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia (M.Q.) and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Sciences Campus, Irvine, California (S.E., K.A.E.)
| | - Khaled A Elsaid
- Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia (M.Q.) and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Sciences Campus, Irvine, California (S.E., K.A.E.)
| |
Collapse
|
57
|
Fine N, Gracey E, Dimitriou I, La Rose J, Glogauer M, Rottapel R. GEF-H1 Is Required for Colchicine Inhibition of Neutrophil Rolling and Recruitment in Mouse Models of Gout. THE JOURNAL OF IMMUNOLOGY 2020; 205:3300-3310. [PMID: 33199537 DOI: 10.4049/jimmunol.1900783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/20/2020] [Indexed: 11/19/2022]
Abstract
Gout is a painful arthritic inflammatory disease caused by buildup of monosodium urate (MSU) crystals in the joints. Colchicine, a microtubule-depolymerizing agent that is used in prophylaxis and treatment of acute gout flare, alleviates the painful inflammatory response to MSU crystals. Using i.p. and intra-articular mouse models of gout-like inflammation, we found that GEF-H1/GEF-H1/AHRGEF2, a microtubule-associated Rho-GEF, was necessary for the inhibitory effect of colchicine on neutrophil recruitment. GEF-H1 was required for neutrophil polarization in response to colchicine, characterized by uropod formation, accumulation of F-actin and myosin L chain at the leading edge, and accumulation of phosphorylated myosin L chain, flotillin-2, and P-selectin glycoprotein ligand-1 (PSGL-1) in the uropod. Wild-type neutrophils that were pre-exposed to colchicine failed to roll or accumulate on activated endothelial monolayers, whereas GEF-H1 knockout (GEF-H1-/-) neutrophils were unaffected by treatment with colchicine. In vivo, colchicine blocked MSU-induced recruitment of neutrophils to the peritoneum and the synovium in wild-type mice, but not in GEF-H1-/- mice. Inhibition of macrophage IL-1β production by colchicine was independent of GEF-H1, supporting a neutrophil-intrinsic mode of action. Our results suggest that the anti-inflammatory effects of colchicine in acute gout-like inflammation can be accounted for by inhibition of neutrophil-rolling interactions with the inflamed vasculature and occurs through GEF-H1-dependent neutrophil stimulation by colchicine. These results contribute to our understanding of the therapeutic action of colchicine, and could inform the application of this drug in other conditions.
Collapse
Affiliation(s)
- Noah Fine
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada
| | - Eric Gracey
- Vlaams Institute for Biotechnology Centre for Inflammation Research, 9052 Ghent, Belgium.,Department of Internal Medicine and Pediatrics, University of Ghent, 9000 Ghent, Belgium
| | - Ioannis Dimitriou
- Department of Immunology, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - José La Rose
- Department of Immunology, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada
| | - Robert Rottapel
- Department of Immunology, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario M5G 1L7, Canada; .,Department of Medicine, Ontario Institute for Cancer Research, University of Toronto, Toronto, Ontario M5G 1L7, Canada; and.,Division of Rheumatology, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| |
Collapse
|
58
|
Renaudin F, Orliaguet L, Castelli F, Fenaille F, Prignon A, Alzaid F, Combes C, Delvaux A, Adimy Y, Cohen-Solal M, Richette P, Bardin T, Riveline JP, Venteclef N, Lioté F, Campillo-Gimenez L, Ea HK. Gout and pseudo-gout-related crystals promote GLUT1-mediated glycolysis that governs NLRP3 and interleukin-1β activation on macrophages. Ann Rheum Dis 2020; 79:1506-1514. [PMID: 32699039 DOI: 10.1136/annrheumdis-2020-217342] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/25/2020] [Accepted: 06/25/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Macrophage activation by monosodium urate (MSU) and calcium pyrophosphate (CPP) crystals mediates an interleukin (IL)-1β-dependent inflammation during gout and pseudo-gout flare, respectively. Since metabolic reprogramming of macrophages goes along with inflammatory responses dependently on stimuli and tissue environment, we aimed to decipher the role of glycolysis and oxidative phosphorylation in the IL-1β-induced microcrystal response. METHODS Briefly, an in vitro study (metabolomics and real-time extracellular flux analysis) on MSU and CPP crystal-stimulated macrophages was performed to demonstrate the metabolic phenotype of macrophages. Then, the role of aerobic glycolysis in IL-1β production was evaluated, as well in vitro as in vivo using 18F-fluorodeoxyglucose positron emission tomography imaging and glucose uptake assay, and molecular approach of glucose transporter 1 (GLUT1) inhibition. RESULTS We observed that MSU and CPP crystals led to a metabolic rewiring toward the aerobic glycolysis pathway explained by an increase in GLUT1 plasma membrane expression and glucose uptake on macrophages. Also, neutrophils isolated from human synovial fluid during gout flare expressed GLUT1 at their plasma membrane more frequently than neutrophils isolated from bloodstream. Both glucose deprivation and treatment with either 2-deoxyglucose or GLUT1 inhibitor suppressed crystal-induced NLRP3 activation and IL-1β production, and microcrystal inflammation in vivo. CONCLUSION In conclusion, we demonstrated that GLUT1-mediated glucose uptake is instrumental during the inflammatory IL-1β response induced by MSU and CPP crystals. These findings open new therapeutic paths to modulate crystal-related inflammation.
Collapse
Affiliation(s)
- Felix Renaudin
- Universite de Paris, Paris, France.,INSERM, UMR-S 1132, F-75010, Paris, France
| | - Lucie Orliaguet
- Universite de Paris, Paris, France.,INSERM, Immunity and Metabolism in Diabetes Laboratory, Centre de Recherche des Cordelier, Paris, France
| | - Florence Castelli
- Service de Pharmacologie et immunoanalyse (SPI), Laboratoire d'Etude du Métabolisme des Médicaments, CEA, INRAE, Université Paris Saclay, Gif-Sur-Yvette, France
| | - François Fenaille
- Service de Pharmacologie et immunoanalyse (SPI), Laboratoire d'Etude du Métabolisme des Médicaments, CEA, INRAE, Université Paris Saclay, Gif-Sur-Yvette, France
| | - Aurelie Prignon
- UMS28 Phénotypage du Petit Animal, Laboratoire d'Imagerie Moléculaire Positonique (LIMP), F-75020, Sorbonne Université, Paris, France
| | - Fawaz Alzaid
- Universite de Paris, Paris, France.,INSERM, Immunity and Metabolism in Diabetes Laboratory, Centre de Recherche des Cordelier, Paris, France
| | - Christele Combes
- UMR 5085 INPT-UPS-CNRS, Université de Toulouse, ENSIACET, F-31000, Toulouse, France
| | - Aurélie Delvaux
- Service de Pharmacologie et immunoanalyse (SPI), Laboratoire d'Etude du Métabolisme des Médicaments, CEA, INRAE, Université Paris Saclay, Gif-Sur-Yvette, France
| | - Yasmina Adimy
- Service de Pharmacologie et immunoanalyse (SPI), Laboratoire d'Etude du Métabolisme des Médicaments, CEA, INRAE, Université Paris Saclay, Gif-Sur-Yvette, France
| | - Martine Cohen-Solal
- Universite de Paris, Paris, France.,Bone and Joint Laboratory, INSERM U1132, Paris, France
| | - Pascal Richette
- Universite de Paris, Paris, France.,INSERM, UMR-S 1132, F-75010, Paris, France
| | - Thomas Bardin
- Universite de Paris, Paris, France.,INSERM, UMR-S 1132, F-75010, Paris, France
| | - Jean-Pierre Riveline
- Universite de Paris, Paris, France.,INSERM, Immunity and Metabolism in Diabetes Laboratory, Centre de Recherche des Cordelier, Paris, France
| | - Nicolas Venteclef
- Universite de Paris, Paris, France.,INSERM, Immunity and Metabolism in Diabetes Laboratory, Centre de Recherche des Cordelier, Paris, France
| | - Frédéric Lioté
- Universite de Paris, Paris, France.,INSERM, UMR-S 1132, F-75010, Paris, France
| | | | - Hang-Korng Ea
- Universite de Paris, Paris, France .,INSERM, UMR-S 1132, F-75010, Paris, France
| |
Collapse
|
59
|
Wu M, Tian Y, Wang Q, Guo C. Gout: a disease involved with complicated immunoinflammatory responses: a narrative review. Clin Rheumatol 2020; 39:2849-2859. [PMID: 32382830 DOI: 10.1007/s10067-020-05090-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/26/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
Gout is a disease with acute and/or chronic inflammation and tissue damage induced by the precipitation of monosodium urate crystal (MSU) crystals in bone joints, kidneys, and subcutaneous sites. In recent years, with the continuous research on gout animal models and patient clinical investigations, the mechanism of inflammation activation of gout has been further discovered. Studies have shown that pro-inflammatory factors such as interleukin (IL)-1β, IL-8 and IL-17, NLRP3 inflammasome, and tumor necrosis factor alpha (TNF-α), anti-inflammatory factors such as IL-10, IL-37 are all involved in the MSU-induced gout inflammatory process. And the immune cells in gout, including neutrophils, monocytes/macrophages, and lymphocytes, all play important roles in the pathogenesis of gout. In this review, we mainly emphasize the understanding of various cytokines, inflammasome, and immune cells involved in the onset of gout, in order to provide a systematic and theoretical basis for the novel exploration of inflammatory therapeutic targets for gout.
Collapse
Affiliation(s)
- Meimei Wu
- Southern Medical University Second Clinical College, Shenzhen, 518101, China
- Department of Rheumatology and Immunology, Southern Medical University Affiliated Shenzhen Baoan Hospital, Shenzhen, 518101, China
| | - Ye Tian
- Department of Rheumatology and Immunology, Southern Medical University Affiliated Shenzhen Baoan Hospital, Shenzhen, 518101, China
- Department of Rheumatology and Immunology, Guangdong Medical University Shenzhen Baoan Clinical College, Shenzhen, 518101, China
- Department of Rheumatology and Immunology, Shenzhen University Second affiliated Hospital, Shenzhen, 518101, China
| | - Qianqian Wang
- Department of Rheumatology and Immunology, Southern Medical University Affiliated Shenzhen Baoan Hospital, Shenzhen, 518101, China
- Department of Rheumatology and Immunology, Guangdong Medical University Shenzhen Baoan Clinical College, Shenzhen, 518101, China
- Department of Rheumatology and Immunology, Shenzhen University Second affiliated Hospital, Shenzhen, 518101, China
| | - Chengshan Guo
- Department of Rheumatology and Immunology, Southern Medical University Affiliated Shenzhen Baoan Hospital, Shenzhen, 518101, China.
- Department of Rheumatology and Immunology, Guangdong Medical University Shenzhen Baoan Clinical College, Shenzhen, 518101, China.
- Department of Rheumatology and Immunology, Shenzhen University Second affiliated Hospital, Shenzhen, 518101, China.
| |
Collapse
|
60
|
Zhou M, Ze K, Wang Y, Li X, Hua L, Lu Y, Chen X, Ding X, Chen S, Ru Y, Zhang M, Li B. Huzhang Tongfeng Granule Improves Monosodium Urate-Induced Inflammation of Gouty Arthritis Rat Model by Downregulation of Cyr61 and Related Cytokines. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:9238797. [PMID: 32419834 PMCID: PMC7206887 DOI: 10.1155/2020/9238797] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/18/2020] [Accepted: 04/06/2020] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Gouty arthritis (GA) is a noninfectious inflammatory disease characterized by self-limited and severe pain. Huzhang Tongfeng granule is one of the most effective traditional Chinese medicines in the treatment of acute GA. However, its effects on the inflammatory factors in the process of acute gout inflammation remain unknown. In the present study, we aimed to evaluate the effect of Huzhang Tongfeng granule on the expressions of Cyr61 and related inflammatory factors in both experimental gout models in vivo and in vitro. METHODS Huzhang Tongfeng granule was provided by the pharmaceutical preparation room of Yueyang Hospital of Integrated Traditional Chinese and Western Medicine. The expressions of Cyr61, IL-1β, TNF-α, and IL-6 in monosodium urate- (MSU-) induced rat models and fibroblast-like synoviocytes (FLSs) were determined by RT-PCR, Western blotting analysis, ELISA, immunohistochemistry, and hematoxylin and eosin staining. RESULTS Huzhang Tongfeng granule could downregulate the expressions of IL-1β, TNF-α, and IL-6 to some extent by inhibiting the expression of Cyr61. CONCLUSIONS Collectively, our findings indicated that Cyr61 was highly expressed in rat models of gout. By inhibiting the expression of Cyr61, Huzhang Tongfeng granule could partially attenuate the inflammation induced by MSU crystal.
Collapse
Affiliation(s)
- Mi Zhou
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| | - Kan Ze
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yifei Wang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| | - Liang Hua
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi Lu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xi Chen
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaojie Ding
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| | - Siting Chen
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi Ru
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ming Zhang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
61
|
Activation Status of NLRP3 Inflammasome in Peripheral Blood Mononuclear Cells From Patients With Gout Flare. J Clin Rheumatol 2020; 26:S208-S212. [PMID: 32332275 DOI: 10.1097/rhu.0000000000001394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND/OBJECTIVE Although gout flares are featured by systemic signs of inflammation, cellular sources of inflammatory mediators are not yet properly characterized. Our objective was to evaluate serum levels and gene expression in peripheral blood mononuclear cells (PBMCs) of several molecules associated with the activation of NLRP3 inflammasome. METHODS Fifteen patients with gout flare and 15 individuals with asymptomatic hyperuricemia were cross-sectionally studied. Serum levels of interleukin 1β (IL-1β), IL-18, monocyte chemoattractant protein 1/chemokine (C-C motif) ligand 2 (CCL2), and vascular cell adhesion molecule 1 were measured as a reflection of systemic inflammation, whereas the expression of NLRP3, CASP1, IL18, and CCL2 genes was measured to assess the inflammatory characteristics of PBMCs. RESULTS Serum levels of IL-1β (1.27 [0.07-1.99] pg/mL vs. 0 [0-0.82] pg/mL, p = 0.032) and vascular cell adhesion molecule 1 (606 [435-748] pg/mL vs. 349 [305-422] pg/mL, p = 0.014) were significantly higher in patients with gout flare than in individuals with asymptomatic hyperuricemia, whereas differences in IL-18 and monocyte chemoattractant protein 1/CCL2 were not found. Notably, no differences were observed in the expression of NLRP3, CASP1, IL18, or CCL2 in PBMCs from individuals of one or another group. CONCLUSIONS Systemic inflammation during gout flares does not appear to be associated with NLRP3 inflammasome activation in PBMCs, suggesting that it may represent the systemic spread of local (synovial) inflammation to monosodium urate crystals, which provides a rationale for redirecting anti-inflammatory therapy from a systemic approach to one centered on the inflamed joint.
Collapse
|
62
|
Kodar K, McConnell MJ, Harper JL, Timmer MSM, Stocker BL. The coadministration of trehalose dibehenate and monosodium urate crystals promotes an antitumor phenotype in human‐derived myeloid cells. Immunol Cell Biol 2020; 98:411-422. [DOI: 10.1111/imcb.12329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/12/2020] [Accepted: 03/15/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Kristel Kodar
- School of Chemical and Physical Sciences Victoria University of Wellington Wellington New Zealand
- Centre for Biodiscovery Victoria University of Wellington Wellington New Zealand
| | - Melanie J McConnell
- Centre for Biodiscovery Victoria University of Wellington Wellington New Zealand
- School of Biological Sciences Victoria University of Wellington Wellington New Zealand
| | - Jacquie L Harper
- School of Chemical and Physical Sciences Victoria University of Wellington Wellington New Zealand
| | - Mattie SM Timmer
- School of Chemical and Physical Sciences Victoria University of Wellington Wellington New Zealand
- Centre for Biodiscovery Victoria University of Wellington Wellington New Zealand
| | - Bridget L Stocker
- School of Chemical and Physical Sciences Victoria University of Wellington Wellington New Zealand
- Centre for Biodiscovery Victoria University of Wellington Wellington New Zealand
| |
Collapse
|
63
|
Rossato MF, Hoffmeister C, Trevisan G, Bezerra F, Cunha TM, Ferreira J, Silva CR. Monosodium urate crystal interleukin-1β release is dependent on Toll-like receptor 4 and transient receptor potential V1 activation. Rheumatology (Oxford) 2020; 59:233-242. [PMID: 31298290 DOI: 10.1093/rheumatology/kez259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 05/03/2019] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVE The present study aimed to elucidate the mechanisms involved in MSU-induced IL-1β release in a rodent animal model of acute gout arthritis. METHODS Painful (mechanical and thermal hypersensitivity, ongoing pain and arthritis score) and inflammatory (oedema, plasma extravasation, cell infiltration and IL-1β release) parameters were assessed several hours after intra-articular injection of MSU (100 µg/articulation) in wild-type or knockout mice for Toll-like receptor 4 (TLR4), inducible nitric oxide synthase (iNOS), transient receptor potential (TRP) V1 and the IL-1 receptor (IL-1R). Also, wild-type animals were treated with clodronate, lipopolysaccharide from Rhodobacter sphaeroides (LPS-RS) (TLR4 antagonist), spleen tyrosine kinase (SYK) inhibitor (iSYK), aminoguanidine (AMG, an iNOS inhibitor) or SB366791 (TRPV1 antagonist). Nitrite/nitrate and IL-1β levels were measured on the synovial fluid of wild-type mice, 2 h after intra-articular MSU injections, or medium from macrophages stimulated for MSU (1000 μg) for 2 h. RESULTS Intra-articular MSU injection caused robust nociception and severe inflammation from 2 up to 6 h after injection, which were prevented by the pre-treatment with clodronate, LPS-RS, iSYK, AMG and SB366791, or the genetic ablation of TLR4, iNOS, TRPV1 or IL-1R. MSU also increased nitrite/nitrate and IL-1β levels in the synovial fluid, which was prevented by clodronate, LPS-RS, iSYK and AMG, but not by SB366791. Similarly, MSU-stimulated peritoneal macrophages released nitric oxide, which was prevented by LPS-RS, iSYK and AMG, but not by SB366791, and released IL-1β, which was prevented by LPS-RS, iSYK, AMG and SB366791. CONCLUSION Our data indicate that MSU may activate TLR4, SYK, iNOS and TRPV1 to induce the release of IL-1β by macrophages, triggering nociception and inflammation during acute gout attack.
Collapse
Affiliation(s)
- Mateus F Rossato
- Graduated Program in Pharmacology, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carin Hoffmeister
- Graduated Program in Pharmacology, Biochemistry and Molecular Biology Department, Federal University of Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - Gabriela Trevisan
- Graduated Program in Pharmacology, Biochemistry and Molecular Biology Department, Federal University of Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - Fabio Bezerra
- Graduated Program in Pharmacology, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thiago M Cunha
- Graduated Program in Pharmacology, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Juliano Ferreira
- Graduated Program in Pharmacology, Pharmacology Department, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
| | - Cassia R Silva
- Graduated Program in Genetics and Biochemistry, Biotechnology Institute, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| |
Collapse
|
64
|
Wang L, Zhu L, Duan C, Li L, Chen G. Total saponin of Dioscorea collettii attenuates MSU crystal‑induced inflammation via inhibiting the activation of the NALP3 inflammasome and caspase‑1 in THP‑1 macrophages. Mol Med Rep 2020; 21:2466-2474. [PMID: 32236574 PMCID: PMC7185280 DOI: 10.3892/mmr.2020.11035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 01/30/2020] [Indexed: 01/13/2023] Open
Abstract
Total saponins extracted from Dioscorea collettii (TSD), extracts of the Chinese herb Dioscorea, are thought to exhibit therapeutic benefit in gouty arthritis. However, its exact mechanism remains unclear. The current study aimed to elucidate the underlying mechanisms by investigating the effects of TSD on the inflammation induced by monosodium urate (MSU) crystals in THP-1 macrophages. The viability of THP-1 macrophages was examined using the MTT assay and the levels of inflammatory cytokines, including interleukin (IL)-1β, IL-18 and tumor necrosis factor (TNF)-α, released by the cells were quantitatively measured using ELISA kits. The results revealed that the protein level of cluster of differentiation 11b increased in THP-1 cells treated with 100 ng/ml phorbol ester, suggesting that monocytic THP-1 cells were successfully differentiated into macrophages. TSD decreased the levels of inflammatory cytokines, including TNF-α, IL-18 and IL-1β, secreted by THP-1 macrophages. As the release of IL-1β and IL-18 is dependent on the NLR family pyrin domain containing 3 (NALP3) inflammasome and caspase-1, the current study investigated the effect of TSD on the aforementioned proteins. The results revealed that TSD decreased the protein levels of NALP3 and apoptosis-associated speck-like, which serve important roles in the assembly of the NALP3 inflammasome. Furthermore, NALP3 inflammasome-related proteins were also decreased by TSD in rotenone induced THP-1 macrophages, TSD inhibited the activation of caspase-1 and rotenone-induced NALP3 inflammasome activation in THP-1 macrophages. The results obtained in the current study revealed that TSD attenuated MSU crystal-induced inflammation by inhibiting rotenone-induced activation of the NALP3 inflammasome and caspase-1, suggesting that these two proteins may be novel targets for the treatment of gouty arthritis.
Collapse
Affiliation(s)
- Lu Wang
- College of Integrative Medicine, Anhui University of Chinese Medicine, Hefei, Anhui 230012, P.R. China
| | - Liran Zhu
- Anhui Institute of Pediatric Research, Anhui Provincial Children's Hospital, Hefei, Anhui 230051, P.R. China
| | - Chenfangyuan Duan
- School of Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, P.R. China
| | - Lu Li
- College of Integrative Medicine, Anhui University of Chinese Medicine, Hefei, Anhui 230012, P.R. China
| | - Guangliang Chen
- College of Integrative Medicine, Anhui University of Chinese Medicine, Hefei, Anhui 230012, P.R. China
| |
Collapse
|
65
|
Bousoik E, Qadri M, Elsaid KA. CD44 Receptor Mediates Urate Crystal Phagocytosis by Macrophages and Regulates Inflammation in A Murine Peritoneal Model of Acute Gout. Sci Rep 2020; 10:5748. [PMID: 32238827 PMCID: PMC7113258 DOI: 10.1038/s41598-020-62727-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 03/16/2020] [Indexed: 02/04/2023] Open
Abstract
Gout is a chronic arthritis caused by the deposition of poorly soluble monosodium urate monohydrate (MSU) crystals in peripheral joints. Resident macrophages initiate inflammation in response to MSU mediated by NF-κB nuclear translocation and NLRP3 inflammasome activation. We investigated the role of CD44, a transmembrane receptor, in mediating MSU phagocytosis by macrophages. We used an antibody that sheds the extracellular domain (ECD) of CD44 to study the role of the receptor and its associated protein phosphatase 2A (PP2A) in macrophage activation. We also studied the significance of CD44 in mediating MSU inflammation in-vivo. Cd44−/− BMDMs showed reduced MSU phagocytosis, LDH release, IL-1β expression and production compared to Cd44+/+ BMDMs. Elevated CD44 staining was detected intracellularly and CD44 colocalized with α-tubulin as a result of MSU exposure and ECD-shedding reduced MSU phagocytosis in murine and human macrophages. Anti-CD44 antibody treatment reduced NF-κB p65 subunit nuclear levels, IL-1β expression, pro-IL-1β and IL-8 production in MSU stimulated THP-1 macrophages (p < 0.01). The effect of the antibody was mediated by an enhancement in PP2A activity. CD44 ECD-shedding reduced the conversion of procaspase-1 to active caspase-1, caspase-1 activity and resultant generation of mature IL-1β in macrophages. Neutrophil and monocyte influx and upregulated production of IL-1β was evident in wildtype mice. MSU failed to trigger neutrophil and monocyte recruitment in Cd44−/− mice and lower IL-1β levels were detected in peritoneal lavages from Cd44−/− mice (p < 0.01). Anti-CD44 antibody treatment reduced neutrophil and monocyte recruitment and resulted in reduced lavage IL-1β levels in the same model. CD44 plays a biologically significant role in mediating phagocytosis of MSU and downstream inflammation and is a novel target in gout treatment.
Collapse
Affiliation(s)
- Emira Bousoik
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Chapman University, Irvine, CA, USA.,School of Pharmacy, Omar-Al-Mukhtar University, Derna, Libya
| | - Marwa Qadri
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Chapman University, Irvine, CA, USA.,Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, 82826, Saudi Arabia
| | - Khaled A Elsaid
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Chapman University, Irvine, CA, USA.
| |
Collapse
|
66
|
Liu L, Zhu X, Zhao T, Yu Y, Xue Y, Zou H. Sirt1 ameliorates monosodium urate crystal-induced inflammation by altering macrophage polarization via the PI3K/Akt/STAT6 pathway. Rheumatology (Oxford) 2020; 58:1674-1683. [PMID: 31106362 DOI: 10.1093/rheumatology/kez165] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/02/2019] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES Acute gout is an inflammatory response to MSU crystals. In our previous research, Sirt1 was shown to have an effect in preventing acute gouty inflammation. In the current study, we aimed to investigate the underlying mechanism involving Sirt1 in acute gout. METHODS The cytological changes and Sirt1 expression in the synovium were observed in patients with acute or intermittent gout. The effect of Sirt1 and its mechanism in gout were studied in macrophages, C57BL/6 mice and Sirt1+/- mice. RESULTS Sirt1 expression was increased in the peripheral blood mononuclear cells (PBMCs) of patients with acute gout but not in the chronic tophus tissue. The arthritis score and numbers of inflammatory cells in injured paw tissue from murine gout models were upregulated in Sirt1+/- mice compared with wild-type mice. A PCR array of the paw tissue from murine gout models indicated that Sirt1 activation might attenuate MSU-induced inflammation by altering the polarization state of macrophages. Furthermore, in patients with acute gout, the phagocytosis of MSU crystals by a macrophage was found in a smear of the joint fluid and large amounts of macrophages were also found in the synovium. The activation of Sirt1 in gouty mice actually decreased the tendency toward M1 polarization. The inhibition of PI3K/Akt partially blocked the anti-inflammatory effect of Sirt1 and the translocation of STAT6, and phosphorylated STAT6 expression was decreased in RAW 264.7 cells treated with MSU crystals. CONCLUSION Our studies revealed that Sirt1 ameliorates MSU-induced inflammation by altering macrophage polarization via the PI3K/Akt/STAT6 pathway.
Collapse
Affiliation(s)
- Lei Liu
- Division of Rheumatology, Huashan Hospital
| | - Xiaoxia Zhu
- Division of Rheumatology, Huashan Hospital.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, P.R. China
| | - Tianyi Zhao
- Division of Rheumatology, Huashan Hospital.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, P.R. China
| | - Yiyun Yu
- Division of Rheumatology, Huashan Hospital.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, P.R. China
| | - Yu Xue
- Division of Rheumatology, Huashan Hospital.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, P.R. China
| | - Hejian Zou
- Division of Rheumatology, Huashan Hospital.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, P.R. China
| |
Collapse
|
67
|
Cabău G, Crișan TO, Klück V, Popp RA, Joosten LAB. Urate-induced immune programming: Consequences for gouty arthritis and hyperuricemia. Immunol Rev 2020; 294:92-105. [PMID: 31853991 PMCID: PMC7065123 DOI: 10.1111/imr.12833] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022]
Abstract
Trained immunity is a process in which innate immune cells undergo functional reprogramming in response to pathogens or damage-associated molecules leading to an enhanced non-specific immune response to subsequent stimulation. While this capacity to respond more strongly to stimuli is beneficial for host defense, in some circumstances it can lead to maladaptive programming and chronic inflammation. Gout is characterized by persistent low-grade inflammation and is associated with an increased number of comorbidities. Hyperuricemia is the main risk factor for gout and is linked to the development of comorbidities. Several experimental studies have shown that urate can mechanistically alter the inflammatory capacity of myeloid cells, while observational studies have indicated an association of hyperuricemia to a wide spectrum of common adult inflammatory diseases. In this review, we argue that hyperuricemia is a main culprit in the development of the long-term systemic inflammation seen in gout. We revisit existing evidence for urate-induced transcriptional and epigenetic reprogramming that could lead to an altered functional state of circulating monocytes consisting in enhanced responsiveness and maladaptive immune responses. By discussing specific functional adaptations of monocytes and macrophages induced by soluble urate or monosodium urate crystals and their contribution to inflammation in vitro and in vivo, we further enforce that urate is a metabolite that can induce innate immune memory and we discuss future research and possible new therapeutic approaches for gout and its comorbidities.
Collapse
Affiliation(s)
- Georgiana Cabău
- Department of Medical GeneticsIuliu Haţieganu” University of Medicine and PharmacyCluj‐NapocaRomania
| | - Tania O. Crișan
- Department of Medical GeneticsIuliu Haţieganu” University of Medicine and PharmacyCluj‐NapocaRomania
| | - Viola Klück
- Department of Internal MedicineRadboud Institute of Molecular Life Sciences (RIMLS)Radboud University Medical CenterNijmegenThe Netherlands
| | - Radu A. Popp
- Department of Medical GeneticsIuliu Haţieganu” University of Medicine and PharmacyCluj‐NapocaRomania
| | - Leo A. B. Joosten
- Department of Medical GeneticsIuliu Haţieganu” University of Medicine and PharmacyCluj‐NapocaRomania
- Department of Internal MedicineRadboud Institute of Molecular Life Sciences (RIMLS)Radboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
68
|
Hsieh CY, Li LH, Lam Y, Fang Z, Gan CH, Rao YK, Chiu HW, Wong WT, Ju TC, Chen FH, Chernikov OV, Liu ML, Hsu CH, Hua KF. Synthetic 4-Hydroxy Auxarconjugatin B, a Novel Autophagy Inducer, Attenuates Gouty Inflammation by Inhibiting the NLRP3 Inflammasome. Cells 2020; 9:cells9020279. [PMID: 31979265 PMCID: PMC7072356 DOI: 10.3390/cells9020279] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/29/2022] Open
Abstract
Gouty arthritis results from the generation of uric acid crystals within the joints. These uric acid crystals activate the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome, which is involved in chronic inflammatory diseases, including gouty arthritis. This study identified the polyenylpyrrole derivative 4-hydroxy auxarconjugatin B (4-HAB), a novel autophagy inducer, which attenuated uric acid crystals-mediated activation of the NLRP3 inflammasome in vitro and in vivo. 4-HAB dose-dependently reduced the release of interleukin (IL)-1β, IL-18, active caspase-1 and apoptosis-associated speck-like protein (ASC) in uric acid crystals-activated macrophages. In a mechanistic study, 4-HAB was shown to inhibit uric acid crystals-induced mitochondrial damage, lysosomal rupture and ASC oligomerization. Additionally, 4-HAB inhibited the NLRP3 inflammasome through Sirt1-dependent autophagy induction. Furthermore, the anti-inflammatory properties of 4-HAB were confirmed in a mouse model of uric acid crystals-mediated peritonitis by the reduced levels of neutrophil influx, IL-1β, active caspase-1, IL-6 and MCP-1 in lavage fluids. In conclusion, 4-HAB attenuates gouty inflammation, in part by attenuating activation of the NLRP3 inflammasome through the Sirt1/autophagy induction pathway.
Collapse
Affiliation(s)
- Chih-Yu Hsieh
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260, Taiwan; (C.-Y.H.); (Y.K.R.); (H.-W.C.); (W.-T.W.)
| | - Lan-Hui Li
- Department of Laboratory Medicine, Linsen, Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei 10844, Taiwan;
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11483, Taiwan
| | - Yulin Lam
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore; (Y.L.); (Z.F.); (C.H.G.)
| | - Zhanxiong Fang
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore; (Y.L.); (Z.F.); (C.H.G.)
| | - Chin Heng Gan
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore; (Y.L.); (Z.F.); (C.H.G.)
| | - Yerra Koteswara Rao
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260, Taiwan; (C.-Y.H.); (Y.K.R.); (H.-W.C.); (W.-T.W.)
| | - Hsiao-Wen Chiu
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260, Taiwan; (C.-Y.H.); (Y.K.R.); (H.-W.C.); (W.-T.W.)
| | - Wei-Ting Wong
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260, Taiwan; (C.-Y.H.); (Y.K.R.); (H.-W.C.); (W.-T.W.)
| | - Tz-Chuen Ju
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407, Taiwan;
| | - Fang-Hsin Chen
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Oleg V. Chernikov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, Vladivostok 690022, Russia;
| | - May-Lan Liu
- Department of Nutritional Science, Toko University, Chiayi 61363, Taiwan;
| | - Chung-Hua Hsu
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Chinese Medicine, Linsen, Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei 10844, Taiwan
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260, Taiwan; (C.-Y.H.); (Y.K.R.); (H.-W.C.); (W.-T.W.)
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11483, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
- Correspondence: ; Tel.: +886-3931-7630
| |
Collapse
|
69
|
Zhang Y, Liu L, Sun D, He Y, Jiang Y, Cheng KW, Chen F. DHA protects against monosodium urate-induced inflammation through modulation of oxidative stress. Food Funct 2020; 10:4010-4021. [PMID: 31214670 DOI: 10.1039/c9fo00573k] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Acute gouty inflammation could be triggered by phagocytosis of monosodium urate (MSU) by immune cells. This study investigated the protective effect and underlying mechanism of docosahexaenoic acid (DHA) on MSU-induced inflammation in vitro and in vivo. Results showed that DHA effectively inhibited MSU-induced expression and secretion of interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) in THP-1 cells. Intracellular reactive oxygen species (ROS) production triggered by MSU was alleviated by DHA treatment. Furthermore, DHA promoted the nuclear translocation of nuclear factor E2-related factor 2 (Nrf2), wherein Nrf2 further mediated the expression of multiple antioxidant enzymes such as, heme oxygenase-1 (HO-1), NAD(P)H: quinone oxidoreductase-1 (NQO1) and catalase, which are closely related with redox homeostasis. DHA treatment also restored MSU-induced impairment of mitochondrial transmembrane potential. In addition, oral administration of DHA-rich microalgal oil to C57BL/6 mice effectively reduced the infiltration of neutrophils, and decreased the expression and secretion of inflammatory cytokines. Altogether, our results suggest that DHA or DHA-rich microalgal oil may be a promising natural agent for the prevention of MSU-induced inflammation and potentially acute gout at least partly by attenuating oxidative stress.
Collapse
Affiliation(s)
- Yue Zhang
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518000, China. and Institute for Food & Bioresource Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Lu Liu
- Institute for Food & Bioresource Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Dongzhe Sun
- Nutrition & Health Research Institute, China National Cereals, Oils and Foodstuffs Corporation (COFCO), Beijing 102209, P. R. China
| | - Yongjing He
- Institute for Food & Bioresource Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Yue Jiang
- RunkeBioengn Co Ltd, Zhangzhou, Fujian, People's Republic of China
| | - Ka-Wing Cheng
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518000, China. and Institute for Food & Bioresource Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Feng Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518000, China. and Institute for Food & Bioresource Engineering, College of Engineering, Peking University, Beijing, 100871, China
| |
Collapse
|
70
|
Zoghebi KA, Bousoik E, Parang K, Elsaid KA. Design and Biological Evaluation of Colchicine-CD44-Targeted Peptide Conjugate in an In Vitro Model of Crystal Induced Inflammation. Molecules 2019; 25:E46. [PMID: 31877739 PMCID: PMC6982808 DOI: 10.3390/molecules25010046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/15/2019] [Accepted: 12/20/2019] [Indexed: 01/23/2023] Open
Abstract
Gout is an inflammatory arthritis due to the joint deposition of monosodium urate (MSU) crystals. Phagocytosis of MSU crystals by tissue macrophages results in the generation of reactive oxygen species (ROS) and production of inflammatory cytokines and chemokines. Colchicine use in gout is limited by severe toxicity. CD44 is a transmembrane glycoprotein that is highly expressed in tissue macrophages and may be involved in gout pathogenesis. The P6 peptide is a 20-amino acid residue peptide that binds to CD44. We hypothesized that the conjugation of colchicine to the P6 peptide would reduce its off-target cytotoxicity while preserving its anti-inflammatory effect. A modified version of P6 peptide and colchicine-P6 peptide conjugate were synthesized using Fmoc/tBu solid-phase and solution-phase chemistry, respectively. A glutaryl amide was used as a linker. The P6 peptide was evaluated for its binding to CD44, association, and internalization by macrophages. Cytotoxic effects of P6 peptide, colchicine, and colchicine-P6 peptide on macrophages were compared and the inhibition of ROS generation and interleukin-8 (IL-8) secretion in MSU-stimulated macrophages treated with P6 peptide, colchicine, or colchicine-P6 peptide was studied. We confirmed that the P6 peptide binds to CD44 and its association and internalization by macrophages were CD44-dependent. Colchicine (1, 10, and 25 μM) demonstrated a significant cytotoxic effect on macrophages while the P6 peptide and colchicine-P6 peptide conjugate (1, 10 and 25 μM) did not alter the viability of the macrophages. The P6 peptide (10 and 25 μM) reduced ROS generation and IL-8 secretion mediated by a reduction in MSU phagocytosis by macrophages. The colchicine-P6 peptide significantly reduced ROS generation and IL-8 secretion compared to the P6 peptide alone at 1 and 10 μM concentrations. Conjugation of colchicine to the P6 peptide reduced the cytotoxic effect of colchicine while preserving its anti-inflammatory activity.
Collapse
Affiliation(s)
- Khalid A. Zoghebi
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA; (K.A.Z.); (E.B.)
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 82826, Saudi Arabia
| | - Emira Bousoik
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA; (K.A.Z.); (E.B.)
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA; (K.A.Z.); (E.B.)
| | - Khaled A. Elsaid
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA; (K.A.Z.); (E.B.)
| |
Collapse
|
71
|
Wahi K, Kodar K, McConnell MJ, Harper JL, Timmer MSM, Stocker BL. MSU Crystals Enhance TDB-Mediated Inflammatory Macrophage IL-1β Secretion. Inflammation 2019; 42:1129-1136. [PMID: 30806957 DOI: 10.1007/s10753-019-00976-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The tumour microenvironment predominantly consists of macrophages with phenotypes ranging from pro-inflammatory (M1-like) to anti-inflammatory (M2-like). Trehalose-6,6'-dibehenate (TDB) displays moderate anti-tumour activity and stimulates M1-like macrophages via the macrophage inducible C-type lectin (Mincle) resulting in IL-1β production. In this study, we examined if monosodium urate (MSU), a known vaccine adjuvant, can boost IL-1β production by TDB-stimulated macrophages. We investigated the effect of MSU/TDB co-treatment on IL-1β production by GM-CSF (M1-like) and M-CSF/IL-4 (M2-like) differentiated mouse bone marrow macrophages (BMMs) and found that MSU/TDB co-treatment of GM-CSF BMMs significantly enhanced IL-1β production in a Mincle-dependent manner. Western blot analysis showed that increased IL-1β production by GM-CSF BMMs was associated with the induction of pro-IL-1β expression by TDB rather than MSU. Flow cytometry analysis showed that MSU/TDB co-stimulation of GM-CSF BMMs led to greater expansion of CD86high/MHC IIhigh and CD86low/MHC IIlow subpopulations; however, only the latter showed increased production of IL-1β. Together, these findings provide evidence of the potential to use MSU/TDB co-treatment to boost IL-1β-mediated anti-tumour activity in M1-like tumour-associated macrophages.
Collapse
Affiliation(s)
- Kanu Wahi
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.,Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Kristel Kodar
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.,Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Melanie J McConnell
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.,School of Biological Sciences, PO Box 600, Wellington, New Zealand
| | - Jacquie L Harper
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Mattie S M Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand. .,Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| | - Bridget L Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand. .,Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| |
Collapse
|
72
|
Pharmacological inhibition of the NLRP3 inflammasome as a potential target for cancer-induced bone pain. Pharmacol Res 2019; 147:104339. [DOI: 10.1016/j.phrs.2019.104339] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/30/2019] [Accepted: 07/01/2019] [Indexed: 12/27/2022]
|
73
|
Raucci F, Iqbal AJ, Saviano A, Minosi P, Piccolo M, Irace C, Caso F, Scarpa R, Pieretti S, Mascolo N, Maione F. IL-17A neutralizing antibody regulates monosodium urate crystal-induced gouty inflammation. Pharmacol Res 2019; 147:104351. [DOI: 10.1016/j.phrs.2019.104351] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/10/2019] [Accepted: 07/10/2019] [Indexed: 01/17/2023]
|
74
|
Sirt1 inhibits gouty arthritis via activating PPARγ. Clin Rheumatol 2019; 38:3235-3242. [DOI: 10.1007/s10067-019-04697-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/25/2019] [Accepted: 07/15/2019] [Indexed: 12/16/2022]
|
75
|
Li XD, Hong MN, Chen J, Lu YY, Ye MQ, Ma Y, Zhu DL, Gao PJ. Adventitial fibroblast-derived vascular endothelial growth factor promotes vasa vasorum-associated neointima formation and macrophage recruitment. Cardiovasc Res 2019; 116:708-720. [DOI: 10.1093/cvr/cvz159] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/10/2019] [Accepted: 06/24/2019] [Indexed: 12/12/2022] Open
Abstract
Abstract
Aims
Adventitial vasa vasorum provides oxygen and nourishment to the vascular wall, but whether it regulates vascular disease remains unclear. We have previously shown that an increased expression of VEGF (vascular endothelial growth factor) is associated with macrophage infiltration. This study aims to determine whether adventitial fibroblast (AF)-derived VEGF increases the number of vasa vasorum contributing to neointima formation through macrophage recruitment.
Methods and results
In rat balloon injury model, vasa vasorum count was increased particularly in the adventitia accompanied by cell proliferation and VEGF expression. Both endogenous and PKH26-labelled exogenous macrophages were mainly distributed in adventitia around vasa vasorum. Interestingly, perivascular delivery of Ranibizumab preferentially concentrated in adventitia resulted in a decrease of neointima formation with concurrent reduction of vasa vasorum count and macrophage infiltration. AFs with adenovirus-mediated VEGF over-expression delivered to the adventitia significantly enhanced these pathological changes after injury. In Tie2-cre/Rosa-LoxP-RFP mice, endothelial cells were increased in the adventitia after wire injury. By using multiphoton laser scanning microscopy, macrophage rolling, adhesion and transmigration were observed in vasa vasorum. Moreover, adoptive transfer of macrophages accelerated injury-induced neointima formation. VEGF-neutralizing antibody administration also attenuated wire injury-induced neointima formation and macrophage infiltration. In primary cultured AFs, exogenous VEGF increased VEGF expression and secretion in a time- and dose-dependent manner. AF-conditioned medium promoted endothelial cell angiogenesis, vascular cell adhesion molecule-1 expression and macrophage adhesion was blocked by VEGF-neutralizing antibody and VEGFR2 inhibitor ZM323881, which also inhibited activation of VEGFR2/ERK1/2 pathway.
Conclusion
These results demonstrate that AF-derived VEGF plays a significant role in the increase of vasa vasorum count which is involved in macrophage recruitment and neointima formation.
Collapse
Affiliation(s)
- Xiao-Dong Li
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Mo-Na Hong
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Jing Chen
- Department of Hypertension, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, 999 Xiwang Road, Shanghai 201801, China
| | - Yuan-Yuan Lu
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Mao-Qing Ye
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Yu Ma
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Ding-Liang Zhu
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Ping-Jin Gao
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| |
Collapse
|
76
|
Mei J, Zhou F, Qiao H, Li H, Tang T. Nerve modulation therapy in gouty arthritis: targeting increased sFRP2 expression in dorsal root ganglion regulates macrophage polarization and alleviates endothelial damage. Am J Cancer Res 2019; 9:3707-3722. [PMID: 31281508 PMCID: PMC6587340 DOI: 10.7150/thno.33908] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/17/2019] [Indexed: 12/16/2022] Open
Abstract
Gouty arthritis (GA) is a form of arthritis caused by uric acid deposition in the joints that result in intense inflammation and pain. Accumulating evidence showed the importance of the sensory neurons signal upon immune cells by releasing neuropeptides and chemokines to regulate associated immune-inflammatory response. In this study, we investigated the significance of sensory neuron neuropeptides and chemokine signals on inflammation-induced macrophages polarization during GA. Methods: We screened the mRNA expression profile during GA in dorsal root ganglion (DRG) neurons to identify the most likely candidate that mediates the neuro-immune communication. Then, we silenced specific gene expression in the DRG by lentiviral vectors in the monosodium urate (MSU)-induced ankle GA mouse model and evaluated alterations in the inflammatory response. In vitro, primary macrophages were used to investigate the neural impact on M1/M2 subtype polarization, proinflammatory cytokine production and downstream endothelial damage. Mechanism by which macrophage inflammation is induced in the DRG was evaluated by Western blot, immunofluorescence, and immunoprecipitation. Results: We found that secreted frizzled-related protein 2 (sFRP2) was the most upregulated gene in dorsal root ganglion (DRG) neurons in response to monosodium urate (MSU) deposition. Injection of LV-sFRP2-shRNA into the L4 and L5 DRG significantly suppressed inflammatory cell infiltration and M1 polarization in the synovial membrane, attenuating hyperalgesia and ankle swelling in the GA mouse model. In vitro, DRG neurons-derived sFRP2 promoted M1 polarization and macrophage migration, thereby upregulating the production of proinflammatory cytokines and preventing endothelial apoptosis. Furthermore, DRG-derived sFRP2 activated the nuclear factor (NF)-κB pathway by destabilizing the β-catenin and p65 complex. Conclusion: We demonstrated the involvement of a sensory neuron-macrophage axis in GA pathology that was regulated by sFRP2 expression in a paracrine manner. Targeting increased sFRP2 expressions in DRG provide novel insights for future GA research in both pain alleviation and treatment of gout inflammation.
Collapse
|
77
|
Yang QB, He YL, Zhang QB, Mi QS, Zhou JG. Downregulation of Transcription Factor T-Bet as a Protective Strategy in Monosodium Urate-Induced Gouty Inflammation. Front Immunol 2019; 10:1199. [PMID: 31231373 PMCID: PMC6558421 DOI: 10.3389/fimmu.2019.01199] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/13/2019] [Indexed: 12/25/2022] Open
Abstract
Gout is sterile joint inflammation triggered by the damaging effects of monosodium urate (MSU) crystals accumulation. Previous studies suggest transcription factor T-bet plays an important role in inflammatory arthritis. Notably, mice lacking T-bet markedly reduced joint inflammation of rheumatoid arthritis models, however, the involvement of T-bet in gouty inflammation has yet to be clarified. Here, we took advantage of T-bet knockout (KO) mice to investigate the role of T-bet in the pathogenesis of MSU-induced gout inflammation. T-bet KO and wild type (WT) mice were used for models of acute inflammation induced with MSU crystals, including footpad, air pouch and peritonitis models. Inflammatory cytokines and phagocytosis were detected in bone-marrow-derived macrophages (BMDMs) from T-bet KO and WT mice treated with MSU crystals in vitro. In addition, T-bet expression in peripheral blood mononuclear cells (PBMCs) from gout patients was measured, as well as plasma inflammatory cytokines. We found that the levels of interleukin (IL)-17, IL-23, and interferon-γ were reduced, but tumor necrosis factor-α was not, in BMDMs from T-bet KO compared with WT mice after MSU challenge in vitro, as well as MSU phagocytosis. In comparison with WT mice in vivo, the swelling index of T-bet KO mice was significantly decreased in the footpad model. T-bet deficiency also dramatically relieved MSU-induced inflammatory cell infiltration in peritonitis and air pouch models in vivo, and as well as the IL-1β levels of air pouch lavage fluid (APLF). In addition, plasma IL-17 and IL-23 levels were elevated in acute gout, whereas protein levels of T-bet were downregulated in PBMCs from acute gout patients and intercritical gout treated with MSU crystals in vitro as well. Transcription factor T-bet deficiency protects against MSU-induced gouty inflammation, suggesting that downregulation of T-bet could be a protective strategy and contribute to spontaneous remission of inflammation in acute gout.
Collapse
Affiliation(s)
- Qi-Bin Yang
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yong-Long He
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Quan-Bo Zhang
- Department of Gerontology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qing-Sheng Mi
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, United States
| | - Jing-Guo Zhou
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
78
|
Renaudin F, Sarda S, Campillo-Gimenez L, Séverac C, Léger T, Charvillat C, Rey C, Lioté F, Camadro JM, Ea HK, Combes C. Adsorption of Proteins on m-CPPD and Urate Crystals Inhibits Crystal-induced Cell Responses: Study on Albumin-crystal Interaction. J Funct Biomater 2019; 10:E18. [PMID: 31027151 PMCID: PMC6616386 DOI: 10.3390/jfb10020018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 04/21/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
The biological effects and cellular activations triggered by monosodium urate (MSU) and calcium pyrophosphate dihydrate (monoclinic: m-CPPD) crystals might be modulated by protein coating on the crystal surface. This study is aimed at: (i) Identifying proteins adsorbed on m-CPPD crystals, and the underlying mechanisms of protein adsorption, and (ii) to understand how protein coating did modulate the inflammatory properties of m-CPPD crystals. The effects of protein coating were assessed in vitro using primary macrophages and THP1 monocytes. Physico-chemical studies on the adsorption of bovine serum albumin (BSA) upon m-CPPD crystals were performed. Adsorption of serum proteins, and BSA on MSU, as well as upon m-CPPD crystals, inhibited their capacity to induce interleukin-1-β secretions, along with a decreased ATP secretion, and a disturbance of mitochondrial membrane depolarization, suggesting an alteration of NLRP3 inflammasome activation. Proteomic analysis identified numerous m-CPPD-associated proteins including hemoglobin, complement, albumin, apolipoproteins and coagulation factors. BSA adsorption on m-CPPD crystals followed a Langmuir-Freundlich isotherm, suggesting that it could modulate m-CPPD crystal-induced cell responses through crystal/cell-membrane interaction. BSA is adsorbed on m-CPPD crystals with weak interactions, confirmed by the preliminary AFM study, but strong interactions of BSA molecules with each other occurred favoring crystal agglomeration, which might contribute to a decrease in the inflammatory properties of m-CPPD crystals. These findings give new insights into the pathogenesis of crystal-related rheumatic diseases and subsequently may open the way for new therapeutic approaches.
Collapse
Affiliation(s)
- Felix Renaudin
- Université Paris 7 Denis Diderot, Inserm UMR 1132 Bioscar, Hôpital Lariboisière, Centre Viggo Petersen, Paris 75010, France.
| | - Stéphanie Sarda
- CIRIMAT, Université de Toulouse, CNRS, Université Toulouse 3, Toulouse INP - ENSIACET, Toulouse 31030, France.
| | - Laure Campillo-Gimenez
- Université Paris 7 Denis Diderot, Inserm UMR 1132 Bioscar, Hôpital Lariboisière, Centre Viggo Petersen, Paris 75010, France.
| | | | - Thibaut Léger
- Institut Jacques Monod, UMR7592 CNRS, Université Paris Diderot, Paris 75013, France.
| | - Cédric Charvillat
- CIRIMAT, Université de Toulouse, CNRS, Université Toulouse 3, Toulouse INP - ENSIACET, Toulouse 31030, France.
| | - Christian Rey
- CIRIMAT, Université de Toulouse, CNRS, Université Toulouse 3, Toulouse INP - ENSIACET, Toulouse 31030, France.
| | - Frédéric Lioté
- Université Paris 7 Denis Diderot, Inserm UMR 1132 Bioscar, Hôpital Lariboisière, Centre Viggo Petersen, Paris 75010, France.
| | - Jean-Michel Camadro
- Institut Jacques Monod, UMR7592 CNRS, Université Paris Diderot, Paris 75013, France.
| | - Hang-Korng Ea
- Université Paris 7 Denis Diderot, Inserm UMR 1132 Bioscar, Hôpital Lariboisière, Centre Viggo Petersen, Paris 75010, France.
| | - Christèle Combes
- CIRIMAT, Université de Toulouse, CNRS, Université Toulouse 3, Toulouse INP - ENSIACET, Toulouse 31030, France.
| |
Collapse
|
79
|
Fine N, Sheikh Z, Al‐Jaf F, Oveisi M, Borenstein A, Hu Y, Pilliar R, Grynpas M, Glogauer M. Differential response of human blood leukocytes to brushite, monetite, and calcium polyphosphate biomaterials. J Biomed Mater Res B Appl Biomater 2019; 108:253-262. [DOI: 10.1002/jbm.b.34385] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 03/30/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Noah Fine
- Faculty of DentistryUniversity of Toronto Toronto Ontario Canada
| | - Zeeshan Sheikh
- Faculty of DentistryUniversity of Toronto Toronto Ontario Canada
- Lunenfeld‐Tanenbaum Research Institute (LTRI), Mount Sinai Hospital Toronto Ontario Canada
- Department of Laboratory Medicine and Pathobiology (LMP)University of Toronto Toronto Ontario Canada
| | - Faik Al‐Jaf
- Faculty of DentistryUniversity of Toronto Toronto Ontario Canada
| | - Morvarid Oveisi
- Faculty of DentistryUniversity of Toronto Toronto Ontario Canada
| | - Alon Borenstein
- Faculty of DentistryUniversity of Toronto Toronto Ontario Canada
| | - Youxin Hu
- Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto Toronto Ontario Canada
| | - Robert Pilliar
- Faculty of DentistryUniversity of Toronto Toronto Ontario Canada
- Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto Toronto Ontario Canada
| | - Marc Grynpas
- Lunenfeld‐Tanenbaum Research Institute (LTRI), Mount Sinai Hospital Toronto Ontario Canada
- Department of Laboratory Medicine and Pathobiology (LMP)University of Toronto Toronto Ontario Canada
- Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto Toronto Ontario Canada
| | - Michael Glogauer
- Faculty of DentistryUniversity of Toronto Toronto Ontario Canada
- Princess Margaret Cancer Centre, Department of Dental Oncology and Maxillofacial Prosthetics Toronto Ontario Canada
| |
Collapse
|
80
|
Jeong JH, Jung JH, Lee JS, Oh JS, Kim YG, Lee CK, Yoo B, Hong S. Prominent Inflammatory Features of Monocytes/Macrophages in Acute Calcium Pyrophosphate Crystal Arthritis: a Comparison with Acute Gouty Arthritis. Immune Netw 2019; 19:e21. [PMID: 31281718 PMCID: PMC6597439 DOI: 10.4110/in.2019.19.e21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/17/2019] [Accepted: 03/13/2019] [Indexed: 12/01/2022] Open
Abstract
Calcium pyrophosphate (CPP) crystals can present as acute inflammatory arthritis which is known as an acute CPP crystal arthritis. Although monocytes/macrophages have been shown to play a role in the initiation of crystal-mediated inflammatory responses, differences in their phenotypes between acute CPP crystal arthritis and acute gouty arthritis have not yet been investigated. We examined the immunological characteristics of synovial monocytes/macrophages in patients with acute CPP crystal and acute gouty arthritis. CD14+CD3−CD19−CD56− cell frequencies in synovial fluid mononuclear cells (SFMCs) were measured. Expression of pro- and anti-inflammatory cytokines and markers was determined. The SFMCs were dominated by a population of monocytes/macrophages in acute CPP crystal arthritis similar to that in acute gout. Synovial monocytes/macrophages showed the phenotypes of infiltrated monocytes as shown by expression of CD88, C-C chemokine receptor type 2, myeloid-related protein (MRP)8 and MRP14 but not proto-oncogene tyrosine-protein kinase MER. Comparatively, the CD14+ cells from patients with acute CPP crystal arthritis had similar high levels of IL-1β and TNF-α production but significantly lower expression of IL-10 and M2 marker (CD163). The monocytes/macrophages had the capacity to produce IL-8 in response to CPP crystals. Proinflammatory features were more dominant in monocytes/macrophages during acute CPP crystal arthritis than those during acute gouty arthritis.
Collapse
Affiliation(s)
- Ji Hye Jeong
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea.,Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Korea
| | - Jae Hyung Jung
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea.,Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Korea
| | - Jung Sun Lee
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Ji Seon Oh
- Clinical Research Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Chang-Keun Lee
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Bin Yoo
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Seokchan Hong
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| |
Collapse
|
81
|
Wu Z, Koh B, Lawrence LM, Kanamala M, Pool B, Svirskis D, Dalbeth N, Astin JW, Crosier KE, Crosier PS, Hall CJ. Liposome-Mediated Drug Delivery in Larval Zebrafish to Manipulate Macrophage Function. Zebrafish 2019; 16:171-181. [PMID: 30724716 DOI: 10.1089/zeb.2018.1681] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Chemical interventions are regularly used to examine and manipulate macrophage function in larval zebrafish. Given chemicals are typically administered by simple immersion or injection, it is not possible to resolve whether their impact on macrophage function is direct or indirect. Liposomes provide an attractive strategy to target drugs to specific cellular compartments, including macrophages. As an example, injecting liposomal clodronate into animal models, including zebrafish, is routinely used to deliver toxic levels of clodronate specifically to macrophages for targeted cell ablation. Here we show that liposomes can also target the delivery of drugs to zebrafish macrophages to selectively manipulate their function. We utilized the drugs etomoxir (a fatty acid oxidation inhibitor) and MitoTEMPO (a scavenger of mitochondrial reactive oxygen species [mROS]), that we have previously shown, through free drug delivery, suppress monosodium urate (MSU) crystal-driven macrophage activation. We generated poloxamer 188 modified liposomes that were readily phagocytosed by macrophages, but not by neutrophils. Loading these liposomes with etomoxir or MitoTEMPO and injecting into larvae suppressed macrophage activation in response to MSU crystals, as evidenced by proinflammatory cytokine expression and macrophage-driven neutrophil recruitment. This work reveals the utility of packaging drugs into liposomes as a strategy to selectively manipulate macrophage function.
Collapse
Affiliation(s)
- Zimei Wu
- 1 School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Ben Koh
- 1 School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Lisa M Lawrence
- 2 Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Manju Kanamala
- 1 School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Bregina Pool
- 3 Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Darren Svirskis
- 1 School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Nicola Dalbeth
- 3 Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jonathan W Astin
- 2 Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kathryn E Crosier
- 2 Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Philip S Crosier
- 2 Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Christopher J Hall
- 2 Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
82
|
Chen Y, Chen X. Ancient herbal component may be a novel therapeutic for gouty arthritis. J Leukoc Biol 2018; 105:7-9. [PMID: 30517770 DOI: 10.1002/jlb.3ce1018-384r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 11/02/2018] [Accepted: 11/13/2018] [Indexed: 11/08/2022] Open
Affiliation(s)
- Yibo Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
83
|
Hsieh CY, Li LH, Rao YK, Ju TC, Nai YS, Chen YW, Hua KF. Mechanistic insight into the attenuation of gouty inflammation by Taiwanese green propolis via inhibition of the NLRP3 inflammasome. J Cell Physiol 2018; 234:4081-4094. [PMID: 30370562 DOI: 10.1002/jcp.27204] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/16/2018] [Indexed: 12/21/2022]
Abstract
Dysregulation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome is involved in many chronic inflammatory diseases, including gouty arthritis. Activation of the NLRP3 inflammasome requires priming and activation signals: the priming signal controls the expression of NLRP3 and interleukin (IL)-1β precursor (proIL-1β), while the activation signal leads to the assembly of the NLRP3 inflammasome and to caspase-1 activation. Here, we reported the effects of the alcoholic extract of Taiwanese green propolis (TGP) on the NLRP3 inflammasome in vitro and in vivo. TGP inhibited proIL-1β expression by reducing nuclear factor kappa B activation and reactive oxygen species (ROS) production in lipopolysaccharide-activated macrophages. Additionally, TGP also suppressed the activation signal by reducing mitochondrial damage, ROS production, lysosomal rupture, c-Jun N-terminal kinases 1/2 phosphorylation and apoptosis-associated speck-like protein oligomerization. Furthermore, we found that TGP inhibited the NLRP3 inflammasome partially via autophagy induction. In the in vivo mouse model of uric acid crystal-induced peritonitis, TGP attenuated the peritoneal recruitment of neutrophils, and the levels of IL-1β, active caspase-1, IL-6 and monocyte chemoattractant protein-1 in lavage fluids. As a proof of principle, in this study, we purified a known compound, propolin G, from TGP and identified this compound as a potential inhibitor of the NLRP3 inflammasome. Our results indicated that TGP might be useful for ameliorating gouty inflammation via inhibition of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Chih-Yu Hsieh
- Department of Biotechnology and Animal Science, National Ilan University, Yilan, Taiwan
| | - Lan-Hui Li
- Department of Laboratory Medicine, Linsen, Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei, Taiwan
| | - Yerra Koteswara Rao
- Department of Biotechnology and Animal Science, National Ilan University, Yilan, Taiwan
| | - Tz-Chuen Ju
- Department of Animal Science and Biotechnology, Tunghai University, Taichung, Taiwan
| | - Yu-Shin Nai
- Department of Biotechnology and Animal Science, National Ilan University, Yilan, Taiwan
| | - Yue-Wen Chen
- Department of Biotechnology and Animal Science, National Ilan University, Yilan, Taiwan
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Yilan, Taiwan.,Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
84
|
Campillo-Gimenez L, Renaudin F, Jalabert M, Gras P, Gosset M, Rey C, Sarda S, Collet C, Cohen-Solal M, Combes C, Lioté F, Ea HK. Inflammatory Potential of Four Different Phases of Calcium Pyrophosphate Relies on NF-κB Activation and MAPK Pathways. Front Immunol 2018; 9:2248. [PMID: 30356764 PMCID: PMC6189479 DOI: 10.3389/fimmu.2018.02248] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 09/10/2018] [Indexed: 12/20/2022] Open
Abstract
Background: Calcium pyrophosphate (CPP) microcrystal deposition is associated with wide clinical phenotypes, including acute and chronic arthritis, that are interleukin 1β (IL-1β)-driven. Two CPP microcrystals, namely monoclinic and triclinic CPP dihydrates (m- and t-CPPD), have been identified in human tissues in different proportions according to clinical features. m-CPP tetrahydrate beta (m-CPPTβ) and amorphous CPP (a-CPP) phases are considered as m- and t-CPPD crystal precursors in vitro. Objectives: We aimed to decipher the inflammatory properties of the three crystalline phases and one amorphous CPP phase and the intracellular pathways involved. Methods: The four synthesized CPP phases and monosodium urate crystals (MSU, as a control) were used in vitro to stimulate the human monocytic leukemia THP-1 cell line or bone marrow-derived macrophages (BMDM) isolated from WT or NLRP3 KO mice. The gene expression of pro- and anti-inflammatory cytokines was evaluated by quantitative PCR; IL-1β, IL-6 and IL-8 production by ELISA; and mitogen-activated protein kinase (MAPK) activation by immunoblot analysis. NF-κB activation was determined in THP-1 cells containing a reporter plasmid. In vivo, the inflammatory potential of CPP phases was assessed with the murine air pouch model via cell analysis and production of IL-1β and CXCL1 in the exudate. The role of NF-κB was determined by a pharmacological approach, both in vivo and in vitro. Results:In vitro, IL-1β production induced by m- and t-CPPD and m-CPPTβ crystals was NLRP3 inflammasome dependent. m-CPPD crystals were the most inflammatory by inducing a faster and higher production and gene expression of IL-1β, IL-6, and IL-8 than t-CPPD, m-CPPTβ and MSU crystals. The a-CPP phase did not show an inflammatory property. Accordingly, m-CPPD crystals led to stronger activation of NF-κB, p38, extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK) MAPKs. Inhibition of NF-κB completely abrogated IL-1β and IL-8 synthesis and secretion induced by all CPP crystals. Also, inhibition of JNK and ERK1/2 MAPKs decreased both IL-1β secretion and NF-κB activation induced by CPP crystals. In vivo, IL-1β and CXCL1 production and neutrophil infiltration induced by m-CPPD crystals were greatly decreased by NF-κB inhibitor treatment. Conclusion: Our results suggest that the inflammatory potential of different CPP crystals relies on their ability to activate the MAPK-dependent NF-κB pathway. Studies are ongoing to investigate the underlying mechanisms.
Collapse
Affiliation(s)
- Laure Campillo-Gimenez
- INSERM, UMR-S 1132, Université Paris Diderot (UFR Médecine), Sorbonne Paris Cité, Paris, France
| | - Félix Renaudin
- INSERM, UMR-S 1132, Université Paris Diderot (UFR Médecine), Sorbonne Paris Cité, Paris, France
| | - Maud Jalabert
- INSERM, UMR-S 1132, Université Paris Diderot (UFR Médecine), Sorbonne Paris Cité, Paris, France
| | - Pierre Gras
- CIRIMAT, Université de Toulouse, CNRS, INPT-ENSIACET, Toulouse, France
| | - Marjolaine Gosset
- EA2496 Orofacial Pathologies, Imaging and Biotherapies, Dental School Faculty, Université Paris Descartes PRES Sorbonne Paris Cité, Montrouge, France
| | - Christian Rey
- CIRIMAT, Université de Toulouse, CNRS, INPT-ENSIACET, Toulouse, France
| | - Stéphanie Sarda
- CIRIMAT, Université de Toulouse, CNRS, Université Paul Sabatier, Toulouse, France
| | - Corinne Collet
- INSERM, UMR-S 1132, Université Paris Diderot (UFR Médecine), Sorbonne Paris Cité, Paris, France.,Service de Biochimie, AP-HP, Hôpital Lariboisière, Paris, France
| | - Martine Cohen-Solal
- INSERM, UMR-S 1132, Université Paris Diderot (UFR Médecine), Sorbonne Paris Cité, Paris, France.,Service de Rhumatologie, AP-HP, Hôpital Lariboisière, Paris, France
| | - Christèle Combes
- CIRIMAT, Université de Toulouse, CNRS, INPT-ENSIACET, Toulouse, France
| | - Frédéric Lioté
- INSERM, UMR-S 1132, Université Paris Diderot (UFR Médecine), Sorbonne Paris Cité, Paris, France.,Service de Biochimie, AP-HP, Hôpital Lariboisière, Paris, France
| | - Hang-Korng Ea
- INSERM, UMR-S 1132, Université Paris Diderot (UFR Médecine), Sorbonne Paris Cité, Paris, France.,Service de Rhumatologie, AP-HP, Hôpital Lariboisière, Paris, France
| |
Collapse
|
85
|
Qadri M, Jay GD, Zhang LX, Wong W, Reginato AM, Sun C, Schmidt TA, Elsaid KA. Recombinant human proteoglycan-4 reduces phagocytosis of urate crystals and downstream nuclear factor kappa B and inflammasome activation and production of cytokines and chemokines in human and murine macrophages. Arthritis Res Ther 2018; 20:192. [PMID: 30157934 PMCID: PMC6116363 DOI: 10.1186/s13075-018-1693-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 08/01/2018] [Indexed: 01/29/2023] Open
Abstract
Background Gout is an inflammatory arthritis caused by monosodium urate monohydrate (MSU) crystals’ joint deposition. MSU phagocytosis by resident macrophages is a key step in gout pathogenesis. MSU phagocytosis triggers nuclear factor kappa B (NFκB) activation and production of cytokines and chemokines. Proteoglycan-4 (PRG4) is a glycoprotein produced by synovial fibroblasts and exerts an anti-inflammatory effect in the joint mediated by its interaction with cell surface receptor CD44. PRG4 also binds and antagonizes TLR2 and TLR4. The objective of this study is to evaluate the efficacy of recombinant human PRG4 (rhPRG4) in suppressing MSU-induced inflammation and mechanical allodynia in vitro and in vivo. Methods THP-1 macrophages were incubated with MSU crystals ± rhPRG4 or bovine submaxillary mucin (BSM), and crystal phagocytosis, cytokines and chemokines expression and production were determined. NFκB p65 subunit nuclear translocation, NLRP3 induction, caspase-1 activation and conversion of proIL-1β to mature IL-1β were studied. MSU phagocytosis by Prg4+/+ and Prg4−/− peritoneal macrophages was determined in the absence or presence of rhPRG4, BSM, anti-CD44, anti-TLR2, anti-TLR4 and isotype control antibodies. Rhodamine-labeled rhPRG4 was incubated with murine macrophages and receptor colocalization studies were performed. Lewis rats underwent intra-articular injection of MSU crystals followed by intra-articular treatment with PBS or rhPRG4. Weight bearing and SF myeloperoxidase activities were determined. Results rhPRG4 reduced MSU crystal phagocytosis at 4 h (p < 0.01) and IL-1β, TNF-α, IL-8 and MCP-1 expression and production at 6 h (p < 0.05). BSM did not alter MSU phagocytosis or IL-1β production in human and murine macrophages. rhPRG4 treatment reduced NFκB nuclear translocation, NLRP3 expression, caspase-1 activation and generation of mature IL-1β (p < 0.05). MSU-stimulated IL-1β production was higher in Prg4−/− macrophages compared to Prg4+/+ macrophages (p < 0.001). rhPRG4, anti-CD44, anti-TLR2 and anti-TLR4 antibody treatments reduced MSU phagocytosis and IL-1β production in murine macrophages (p < 0.05). rhPRG4 preferentially colocalized with CD44 on Prg4−/− peritoneal macrophages compared to TLR2 or TLR4 (p < 0.01). rhPRG4 normalized weight bearing and reduced SF myeloperoxidase activity compared to PBS in vivo. Conclusion rhPRG4 inhibits MSU crystal phagocytosis and exhibits an anti-inflammatory and anti-nociceptive activity in vitro and in vivo. rhPRG4’s anti-inflammatory mechanism may be due to targeting CD44 on macrophages.
Collapse
Affiliation(s)
- Marwa Qadri
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Sciences Campus, 9401 Jeronimo Road, Irvine, CA, 92618, USA
| | - Gregory D Jay
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI, USA.,Department of Biomedical Engineering, Brown University, Providence, RI, USA
| | - Ling X Zhang
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI, USA
| | - Wendy Wong
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI, USA
| | - Anthony M Reginato
- Division of Rheumatology and Department of Dermatology, Rhode Island Hospital, Providence, RI, USA
| | - Changqi Sun
- Division of Rheumatology and Department of Dermatology, Rhode Island Hospital, Providence, RI, USA
| | - Tannin A Schmidt
- Biomedical Engineering Department, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Khaled A Elsaid
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Sciences Campus, 9401 Jeronimo Road, Irvine, CA, 92618, USA.
| |
Collapse
|
86
|
Fernandes MJ, Naccache PH. The Role of Inhibitory Receptors in Monosodium Urate Crystal-Induced Inflammation. Front Immunol 2018; 9:1883. [PMID: 30177932 PMCID: PMC6109781 DOI: 10.3389/fimmu.2018.01883] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/30/2018] [Indexed: 11/13/2022] Open
Abstract
Inhibitory receptors are key regulators of immune responses. Aberrant inhibitory receptor function can either lead to an exacerbated or defective immune response. Several regulatory mechanisms involved in the inflammatory reaction induced by monosodium urate crystals (MSU) during acute gout have been identified. One of these mechanisms involves inhibitory receptors. The engagement of the inhibitory receptors Clec12A and SIRL-1 has opposing effects on the responses of neutrophils to MSU. We review the general concepts of inhibitory receptor biology and apply them to understand and compare the modulation of MSU-induced inflammation by Clec12A and SIRL-1. We also discuss gaps in our knowledge of the contribution of inhibitory receptors to the pathogenesis of gout and propose future avenues of research.
Collapse
Affiliation(s)
- Maria J Fernandes
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, CHU de Québec Research Center, Québec, QC, Canada
| | - Paul H Naccache
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, CHU de Québec Research Center, Québec, QC, Canada
| |
Collapse
|
87
|
Zhang H, Li F, Li WW, Stary C, Clark JD, Xu S, Xiong X. The inflammasome as a target for pain therapy. Br J Anaesth 2018; 117:693-707. [PMID: 27956668 DOI: 10.1093/bja/aew376] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The interleukin-1 family of cytokines are potent inducers of inflammation and pain. Proteolytic activation of this family of cytokines is under the control of several innate immune receptors that coordinate to form large multiprotein signalling platforms, termed inflammasomes. Recent evidence suggests that a wide range of inflammatory diseases, cancers, and metabolic and autoimmune disorders, in which pain is a common complaint, may be coordinated by inflammasomes. Activation of inflammasomes results in cleavage of caspase-1, which subsequently induces downstream initiation of several potent pro-inflammatory cascades. Therefore, it has been proposed that targeting inflammasome activity may be a novel and effective therapeutic strategy for these pain-related diseases. The purpose of this narrative review article is to provide the reader with an overview of the activation and regulation of inflammasomes and to investigate the potential therapeutic role of inflammasome inhibition in the treatment of diseases characterized by pain, including the following: complex regional pain syndrome, gout, rheumatoid arthritis, inflammatory pain, neuropathic pain, chronic prostatitis, chronic pelvic pain syndrome, and fibromyalgia. We conclude that the role of the inflammasome in pain-associated diseases is likely to be inflammasome subtype and disease specific. The currently available evidence suggests that disease-specific targeting of the assembly and activity of the inflammasome complex may be a novel therapeutic opportunity for the treatment of refractory pain in many settings.
Collapse
Affiliation(s)
- H Zhang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, PR China
| | - F Li
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, PR China
| | - W-W Li
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - C Stary
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - J D Clark
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - S Xu
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, PR China
| | - X Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| |
Collapse
|
88
|
Bhattacharya S, Aggarwal A. M2 macrophages and their role in rheumatic diseases. Rheumatol Int 2018; 39:769-780. [DOI: 10.1007/s00296-018-4120-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 08/02/2018] [Indexed: 12/21/2022]
|
89
|
Yang Q, Zhang Q, Qing Y, Zhou L, Mi Q, Zhou J. miR-155 is dispensable in monosodium urate-induced gouty inflammation in mice. Arthritis Res Ther 2018; 20:144. [PMID: 29996893 PMCID: PMC6042462 DOI: 10.1186/s13075-018-1550-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/21/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The findings of a previous study by Jin et al. have shown that microRNA (miR)-155 was upregulated in patients with acute gouty arthritis and enhanced the proinflammatory cytokines. There is no direct evidence to support that miR-155 is indeed involved in monosodium urate (MSU)-induced inflammatory responses in vivo. The aim of this study was to investigate the role of miR-155 knock-out (KO) or knock-in (KI) mice in MSU-induced animal models to mimic acute gout. METHODS MiR-155 expression in cultured bone marrow-derived macrophages (BMDMs) from miR-155 KO, miR-155 KI, and wild-type (WT) mice treated with MSU crystals in vitro was detected by real-time quantitative polymerase chain reaction (qPCR). MiR-155 KO and WT mice were used to induce an acute gouty inflammatory response with MSU crystals including models of foot pad inflammation, ankle arthritis, air pouch inflammation, and peritonitis. Furthermore, the proinflammatory interleukin (IL)-1β levels in lavage fluids from air pouch and peritoneal cavity models were measured by enzyme-linked immunosorbent assay (ELISA), and tumor necrosis factor (TNF)-α production from BMDMs of miR-155 KI mice treated with MSU were measured by flow cytometry. RESULTS MiR-155 expression was quickly upregulated in BMDMs from WT mice following MSU treatment in vitro. In comparison with WT mice in vivo, the swelling index of miR-155 KO mice showed no significant difference in the murine foot pad and ankle arthritis models for the indicated different time points. There were similar changes in total cell numbers of lavage fluids in the air pouch and peritoneal cavity models between miR-155 KO and WT mice following MSU crystal injection. Moreover, the IL-1β levels of lavage fluids in the air pouch and peritonitis models from miR-155 KO mice were almost the same as those from WT mice. TNF-α levels were comparable from BMDMs treated with MSU crystals in vitro between miR-155 KI mice and WT mice. CONCLUSIONS MiR-155 is dispensable in MSU-induced gouty inflammation in mice. Deletion of miR-155 might not be an effective therapeutic approach to relieve the inflammation in acute gout.
Collapse
Affiliation(s)
- Qibin Yang
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Sichuan Province, Nanchong, 637000, China.,Henry Ford Immunology Program, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA.,Department of Dermatology, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA
| | - Quanbo Zhang
- Department of Gerontology, Affiliated Hospital of North Sichuan Medical College, Sichuan Province, Nanchong, 637000, China.,Henry Ford Immunology Program, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA.,Department of Dermatology, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA
| | - Yufeng Qing
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Sichuan Province, Nanchong, 637000, China
| | - Li Zhou
- Henry Ford Immunology Program, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA.,Department of Dermatology, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA.,Department of Internal Medicine, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA
| | - Qingsheng Mi
- Henry Ford Immunology Program, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA. .,Department of Dermatology, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA. .,Department of Internal Medicine, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA.
| | - Jingguo Zhou
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Chengdu Medical College, Sichuan Province, Chengdu, 610000, China.
| |
Collapse
|
90
|
Mahon OR, Dunne A. Disease-Associated Particulates and Joint Inflammation; Mechanistic Insights and Potential Therapeutic Targets. Front Immunol 2018; 9:1145. [PMID: 29892292 PMCID: PMC5985611 DOI: 10.3389/fimmu.2018.01145] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/07/2018] [Indexed: 12/27/2022] Open
Abstract
It is now well established that intra-articular deposition of endogenous particulates, such as osteoarthritis-associated basic calcium phosphate crystals, gout-associated monosodium urate crystals, and calcium deposition disease-associated calcium pyrophosphate crystals, contributes to joint destruction through the production of cartilage-degrading enzymes and pro-inflammatory cytokines. Furthermore, exogenous wear-debris particles, generated from prosthetic implants, drive periprosthetic osteolysis which impacts on the longevity of total joint replacements. Over the last few years, significant insight has been gained into the mechanisms through which these particulates exert their effects. Not only has this increased our understanding of the pathological processes associated with crystal deposition but it has also led to the identification of a number of therapeutic targets to treat particulate-associated disease. In this review, we discuss recent developments regarding the cellular events triggered by joint-associated particulates, as well as future directions in therapy for particulate-related arthropathies.
Collapse
Affiliation(s)
- Olwyn R Mahon
- School of Biochemistry and Immunology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, The University of Dublin, Dublin, Ireland
| | - Aisling Dunne
- School of Biochemistry and Immunology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, The University of Dublin, Dublin, Ireland
| |
Collapse
|
91
|
Zhou H, Xu J, Wang S, Peng J. Role of cantharidin in the activation of IKKα/IκBα/NF‑κB pathway by inhibiting PP2A activity in cholangiocarcinoma cell lines. Mol Med Rep 2018; 17:7672-7682. [PMID: 29620225 PMCID: PMC5983964 DOI: 10.3892/mmr.2018.8860] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 02/08/2018] [Indexed: 12/15/2022] Open
Abstract
Cantharidin (CAN), a potent inhibitor of serine/threonine‑protein phosphatase 2A (PP2A), is widely used in clinical practice, particularly in the treatment of advanced cancer due to its specific action on these types of cancer. In the present study, the inhibitory effect of CAN was examined in two cholangiocarcinoma cell lines (QBC939 and Hucc‑t1). Following treatment with CAN, cell viability was effectively reduced in QBC939 and Hucc‑t1 cells and normal human intrahepatic biliary epithelial cells. However, a slight increase in reactive oxygen species levels in QBC939 cells treated with CAN was observed post‑treatment. CAN significantly inhibited cell migration and invasion in a dose‑dependent manner. Western blot analysis demonstrated that the nuclear factor‑κB (NF‑κB) pathway was stimulated by CAN, which was confirmed by the upregulated phosphorylation levels of inhibitor of NF‑κB kinase subunit α (IKKα) and NF‑κB inhibitor α (IκBα) in cells, and an increased NF‑κB p65 subunit level in the nucleus. The expression levels of 72 kDa type IV collagenase (MMP2) and matrix metalloproteinase 9 (MMP9) were downregulated by CAN. Notably, there was a negative association between MMP2 and MMP9 expression levels, and NF‑κB p65, although NF‑κB p65 regulates the expression of MMP2 and MMP9 and has a positive association with these proteins in various types of cancer. Notably, it was observed that CAN exerted specific inhibition on PP2A activity and thereby resulted in the activation of the IKKα/IκBα/NF‑κB pathway. Therefore, CAN‑induced cell inhibition maybe partially dependent on the activation of the IKKα/IκBα/NF‑κB pathway. In conclusion, it was demonstrated that CAN selectively and effectively inhibited cholangiocarcinoma cell migration and invasion. The present study may provide a novel insight into the use of CAN as a therapeutic candidate in the treatment of cholangiocarcinoma.
Collapse
Affiliation(s)
- Huijiang Zhou
- Department of General Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, P.R. China
| | - Jiangfeng Xu
- Department of General Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, P.R. China
| | - Shuai Wang
- Department of General Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, P.R. China
| | - Jinfeng Peng
- Department of General Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, P.R. China
| |
Collapse
|
92
|
Yang W, Hu P. Skeletal muscle regeneration is modulated by inflammation. J Orthop Translat 2018; 13:25-32. [PMID: 29662788 PMCID: PMC5892385 DOI: 10.1016/j.jot.2018.01.002] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 01/01/2023] Open
Abstract
Skeletal muscle regeneration is a complex process orchestrated by multiple steps. Recent findings indicate that inflammatory responses could play central roles in bridging initial muscle injury responses and timely muscle injury reparation. The various types of immune cells and cytokines have crucial roles in muscle regeneration process. In this review, we briefly summarise the functions of acute inflammation in muscle regeneration. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE Immune system is closely relevant to the muscle regeneration. Understanding the mechanisms of inflammation in muscle regeneration is therefore critical for the development of effective regenerative, and therapeutic strategies in muscular disorders. This review provides information for muscle regeneration research regarding the effects of inflammation on muscle regeneration.
Collapse
Affiliation(s)
| | - Ping Hu
- State Key Laboratory of Cell Biology, Center of Excellence in Molecular and Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| |
Collapse
|
93
|
Hall CJ, Sanderson LE, Lawrence LM, Pool B, van der Kroef M, Ashimbayeva E, Britto D, Harper JL, Lieschke GJ, Astin JW, Crosier KE, Dalbeth N, Crosier PS. Blocking fatty acid-fueled mROS production within macrophages alleviates acute gouty inflammation. J Clin Invest 2018; 128:1752-1771. [PMID: 29584621 DOI: 10.1172/jci94584] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 02/07/2018] [Indexed: 12/17/2022] Open
Abstract
Gout is the most common inflammatory arthritis affecting men. Acute gouty inflammation is triggered by monosodium urate (MSU) crystal deposition in and around joints that activates macrophages into a proinflammatory state, resulting in neutrophil recruitment. A complete understanding of how MSU crystals activate macrophages in vivo has been difficult because of limitations of live imaging this process in traditional animal models. By live imaging the macrophage and neutrophil response to MSU crystals within an intact host (larval zebrafish), we reveal that macrophage activation requires mitochondrial ROS (mROS) generated through fatty acid oxidation. This mitochondrial source of ROS contributes to NF-κB-driven production of IL-1β and TNF-α, which promote neutrophil recruitment. We demonstrate the therapeutic utility of this discovery by showing that this mechanism is conserved in human macrophages and, via pharmacologic blockade, that it contributes to neutrophil recruitment in a mouse model of acute gouty inflammation. To our knowledge, this study is the first to uncover an immunometabolic mechanism of macrophage activation that operates during acute gouty inflammation. Targeting this pathway holds promise in the management of gout and, potentially, other macrophage-driven diseases.
Collapse
Affiliation(s)
| | | | | | - Bregina Pool
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | | | | | - Jacquie L Harper
- Malaghan Institute for Medical Research, Wellington, New Zealand
| | - Graham J Lieschke
- Australian Regenerative Medicine Institute, Monash University, Victoria, Australia
| | | | | | - Nicola Dalbeth
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
94
|
Ma T, Liu X, Cen Z, Xin C, Guo M, Zou C, Song W, Xie R, Wang K, Zhou H, Zhang J, Wang Z, Bian C, Cui K, Li J, Wei YQ, Li J, Zhou X. MicroRNA-302b negatively regulates IL-1β production in response to MSU crystals by targeting IRAK4 and EphA2. Arthritis Res Ther 2018; 20:34. [PMID: 29482609 PMCID: PMC5828083 DOI: 10.1186/s13075-018-1528-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 01/26/2018] [Indexed: 02/05/2023] Open
Abstract
Background Interleukin-1β (IL-1β) is a pivotal proinflammatory cytokine that is strongly associated with the inflammation of gout. However, the underlying mechanism through which the production of IL-1β is regulated has not been fully elucidated. Our previous work identified that miR-302b had an important immune regulatory role in bacterial lung infections. This study was conducted to evaluate the function of miR-302b on monosodium urate (MSU) crystal-induced inflammation and its mechanism. Methods The expression pattern and the immune-regulatory role of miR-302b were evaluated both in vitro and in vivo. The functional targets of miR-302b were predicted by bioinformatics, and then validated by genetic approaches. In addition, the clinical feature of miR-302b was analyzed using serum samples of patients with gouty arthritis. Results The extremely high expression of miR-302b was observed in both macrophages and mouse air membranes treated with MSU. Intriguingly, overexpression of miR-302b regulated NF-κB and caspase-1 signaling, leading to significantly attenuate MSU-induced IL-1β. By genetic analysis, miR-302b exhibited inhibitory function on IRAK4 and EphA2 by binding to their 3′-UTR regions. Corporately silencing IRAK4 and EphA2 largely impaired MSU-induced IL-1β protein production. Moreover, it was also found that miR-302b and EphA2 suppressed the migration of macrophages. Finally, it was observed that high expression of miR-302b was a general feature in patients with gouty arthritis. Conclusions These results suggest that miR-302b can regulate IL-1β production in MSU-induced inflammation by targeting NF-κB and caspase-1 signaling, and may be a potential therapeutic target for gouty arthritis. Electronic supplementary material The online version of this article (10.1186/s13075-018-1528-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Teng Ma
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Xiao Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Zhifu Cen
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Xin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Mingfeng Guo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Chaoyu Zou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Wenpeng Song
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Rou Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Kailun Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Hong Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Jun Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Zhen Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Ce Bian
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, 610041, China
| | - Kaijun Cui
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Yu-Quan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Xikun Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
95
|
Karasawa T, Kawashima A, Usui-Kawanishi F, Watanabe S, Kimura H, Kamata R, Shirasuna K, Koyama Y, Sato-Tomita A, Matsuzaka T, Tomoda H, Park SY, Shibayama N, Shimano H, Kasahara T, Takahashi M. Saturated Fatty Acids Undergo Intracellular Crystallization and Activate the NLRP3 Inflammasome in Macrophages. Arterioscler Thromb Vasc Biol 2018; 38:744-756. [PMID: 29437575 DOI: 10.1161/atvbaha.117.310581] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 01/25/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Inflammation provoked by the imbalance of fatty acid composition, such as excess saturated fatty acids (SFAs), is implicated in the development of metabolic diseases. Recent investigations suggest the possible role of the NLRP3 (nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain containing 3) inflammasome, which regulates IL-1β (interleukin 1β) release and leads to inflammation, in this process. Therefore, we investigated the underlying mechanism by which SFAs trigger NLRP3 inflammasome activation. APPROACH AND RESULTS The treatment with SFAs, such as palmitic acid and stearic acid, promoted IL-1β release in murine primary macrophages while treatment with oleic acid inhibited SFA-induced IL-1β release in a dose-dependent manner. Analyses using polarized light microscopy revealed that intracellular crystallization was provoked in SFA-treated macrophages. As well as IL-1β release, the intracellular crystallization and lysosomal dysfunction were inhibited in the presence of oleic acid. These results suggest that SFAs activate NLRP3 inflammasome through intracellular crystallization. Indeed, SFA-derived crystals activated NLRP3 inflammasome and subsequent IL-1β release via lysosomal dysfunction. Excess SFAs also induced crystallization and IL-1β release in vivo. Furthermore, SFA-derived crystals provoked acute inflammation, which was impaired in IL-1β-deficient mice. CONCLUSIONS These findings demonstrate that excess SFAs cause intracellular crystallization and subsequent lysosomal dysfunction, leading to the activation of the NLRP3 inflammasome, and provide novel insights into the pathogenesis of metabolic diseases.
Collapse
Affiliation(s)
- Tadayoshi Karasawa
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.).
| | - Akira Kawashima
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Fumitake Usui-Kawanishi
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Sachiko Watanabe
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Hiroaki Kimura
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Ryo Kamata
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Koumei Shirasuna
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Yutaro Koyama
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Ayana Sato-Tomita
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Takashi Matsuzaka
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Hiroshi Tomoda
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Sam-Yong Park
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Naoya Shibayama
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Hitoshi Shimano
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Tadashi Kasahara
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Masafumi Takahashi
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.).
| |
Collapse
|
96
|
Liu T, Liu F, Peng LW, Chang L, Jiang YM. The Peritoneal Macrophages in Inflammatory Diseases and Abdominal Cancers. Oncol Res 2017; 26:817-826. [PMID: 29237519 PMCID: PMC7844755 DOI: 10.3727/096504017x15130753659625] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Peritoneal macrophages (PMs) are the major cell type of peritoneal cells that participate in multiple aspects of innate and acquired immunity in the peritoneal cavity. PMs have an ability to release a large amount of proinflammatory and anti-inflammatory cytokines and therefore play a critical role in regulating the differentiation of innate immune cells and inflammatory T cells. Accumulating studies demonstrate that the immunological reactions and inflammatory responses of PMs are strongly related to the pathogenic processes of various inflammatory diseases and abdominal cancers. Consequently, the regulation of PM activation has gradually emerged as a promising target for immunotherapy, and better understanding of the distinctly biological function of PMs in individual diseases is crucial for designing specific and effective therapeutic agents. This review covers the characterization and immunological function of PMs in hosts with inflammatory diseases and abdominal cancers.
Collapse
Affiliation(s)
- Ting Liu
- Department of Laboratory Medicine, West China Second University Hospital, and Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Sichuan University, Chengdu, P.R. China
| | - Fang Liu
- Department of Laboratory Medicine, West China Second University Hospital, and Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Sichuan University, Chengdu, P.R. China
| | - Lei-Wen Peng
- Department of Laboratory Medicine, West China Second University Hospital, and Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Sichuan University, Chengdu, P.R. China
| | - Li Chang
- Department of Laboratory Medicine, West China Second University Hospital, and Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Sichuan University, Chengdu, P.R. China
| | - Yong-Mei Jiang
- Department of Laboratory Medicine, West China Second University Hospital, and Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
97
|
Wang J, Yang Q, Zhang Q, Yin C, Zhou L, Zhou J, Wang Y, Mi QS. Invariant Natural Killer T Cells Ameliorate Monosodium Urate Crystal-Induced Gouty Inflammation in Mice. Front Immunol 2017; 8:1710. [PMID: 29312287 PMCID: PMC5733058 DOI: 10.3389/fimmu.2017.01710] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/20/2017] [Indexed: 12/31/2022] Open
Abstract
Gout is an inflammatory arthritis caused by deposition of intra-articular monosodium urate (MSU) crystal. Previous studies have focused on resident macrophage, infiltrating monocyte, and neutrophil responses to MSU crystal; yet the mechanisms of cellular changes and the potential involvement of other regulatory immune cells remain largely unknown. Invariant natural killer T (iNKT) cells, an innate type of T cell, are involved in the development of various inflammatory diseases. Here, we investigate the role of iNKT cells in MSU crystal-induced gouty inflammation. MSU crystal-induced inflammatory profiles in an air-pouch model were examined in iNKT-deficient CD1d knockout (KO) and wild-type (WT) control mice. To explore potential mechanisms of iNKT cell regulation of gouty inflammation, we cocultured CD4+ or CD4−iNKT cells with bone marrow-derived macrophages (BMDMs). We found that iNKT cells quickly migrated to the site of inflammation upon MSU crystal stimulation in WT mice. The total number of infiltrating cells in CD1d KO mice, especially neutrophils, was dramatically increased at 6 and 12 h (P < 0.01) post-MSU crystal challenge, compared with WT controls. BMDMs cocultured with CD4+iNKT cells produced less tumor necrosis factor-α and expressed higher levels of M2 macrophage markers, including Clec7a, Pdcd1Ig2, and interleukin-4 (P < 0.01), compared with BMDMs cocultured with CD4−iNKT cells or conventional CD4+ T cells. CD4+iNKT cells are one of the key regulators of MSU crystal-induced gouty inflammation through the control of macrophage polarization. iNKT cells may serve as a new therapeutic target for gout.
Collapse
Affiliation(s)
- Jie Wang
- Department of Endocrinology, Affiliated Hospital of Qingdao University, Qingdao, China.,Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States.,Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States
| | - Qibin Yang
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States.,Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Department of Rheumatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Quanbo Zhang
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States.,Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Department of Gerontology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Congcong Yin
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States.,Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States
| | - Li Zhou
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States.,Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Department of Internal Medicine, Henry Ford Health System, Detroit, MI, United States
| | - Jingguo Zhou
- Department of Rheumatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yangang Wang
- Department of Endocrinology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qing-Sheng Mi
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States.,Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Department of Internal Medicine, Henry Ford Health System, Detroit, MI, United States
| |
Collapse
|
98
|
Ligand-mediated cytoplasmic retention of the Ah receptor inhibits macrophage-mediated acute inflammatory responses. J Transl Med 2017; 97:1471-1487. [PMID: 28892097 PMCID: PMC5711556 DOI: 10.1038/labinvest.2017.92] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 07/12/2017] [Accepted: 07/18/2017] [Indexed: 12/25/2022] Open
Abstract
The Ah receptor (AHR) has been shown to exhibit both inflammatory and anti-inflammatory activity in a context-specific manner. In vivo macrophage-driven acute inflammation models were utilized here to test whether the selective Ah receptor modulator 1-allyl-7-trifluoromethyl-1H-indazol-3-yl]-4-methoxyphenol (SGA360) would reduce inflammation. Exposure to SGA360 was capable of significantly inhibiting lipopolysaccharide (LPS)-mediated endotoxic shock in a mouse model, both in terms of lethality and attenuating inflammatory signaling in tissues. Topical exposure to SGA360 was also able to mitigate joint edema in a monosodium urate (MSU) crystal gout mouse model. Inhibition was dependent on the expression of the high-affinity allelic AHR variant in both acute inflammation models. Upon peritoneal MSU crystal exposure SGA360 pretreatment inhibited neutrophil and macrophage migration into the peritoneum. RNA-seq analysis revealed that SGA360 attenuated the expression of numerous inflammatory genes and genes known to be directly regulated by AHR in thioglycolate-elicited primary peritoneal macrophages treated with LPS. In addition, expression of the high-affinity allelic AHR variant in cultured macrophages was necessary for SGA360-mediated repression of inflammatory gene expression. Mechanistic studies revealed that SGA360 failed to induce nuclear translocation of the AHR and actually enhanced cytoplasmic localization. LPS treatment of macrophages enhanced the occupancy of the AHR and p65 to the Ptgs2 promoter, whereas SGA360 attenuated occupancy. AHR ligand activity was detected in peritoneal exudates isolated from MSU-treated mice, thus suggesting that the anti-inflammatory activity of SGA360 is mediated at least in part through AHR antagonism of endogenous agonist activity. These results underscore an important role of the AHR in participating in acute inflammatory signaling and warrants further investigations into possible clinical applications.
Collapse
|
99
|
Jeong JH, Hong S, Kwon OC, Ghang B, Hwang I, Kim YG, Lee CK, Yoo B. CD14 + Cells with the Phenotype of Infiltrated Monocytes Consist of Distinct Populations Characterized by Anti-inflammatory as well as Pro-inflammatory Activity in Gouty Arthritis. Front Immunol 2017; 8:1260. [PMID: 29056937 PMCID: PMC5635328 DOI: 10.3389/fimmu.2017.01260] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/21/2017] [Indexed: 12/11/2022] Open
Abstract
It has been suggested that inflammasome-mediated IL-1β production in monocytic cells is responsible for the acute inflammatory response in gouty arthritis. However, phenotypical and functional analyses of monocytes during gouty arthritis have yet to be conducted. Therefore, we investigated the characteristics of monocytes/macrophages in the synovial fluid cells of patients with acute gout. The number and frequency of monocytes/macrophages in the synovial fluid mononuclear cells (SFMCs) of patients was examined. The expression of markers for monocyte recruitment and tissue-resident macrophages, the production of pro-inflammatory and anti-inflammatory cytokines, and phagocytosis were analyzed in the monocytes/macrophages of patients with acute gout attacks. The number and frequency of CD14+CD3−CD19−CD56− monocytes/macrophages was markedly increased in the SFMCs of patients with gout compared to those of patients with rheumatoid arthritis (RA). CD14+ cells showed the phenotypes of infiltrated monocytes rather than tissue-resident macrophages, characterized by a high expression of CCR2, MRP8, and MRP14, but a low expression of MERTK and 25F9. These cells had the capacity to produce pro-inflammatory cytokines such as TNF-α and IL-1β after stimulation with lipopolysaccharides. In addition, anti-inflammatory features, including CD163 expression and IL-10 production from CD14+ cells, were significantly higher in patients with gout than in those with RA. CD14+ cells with phenotype of M2 macrophages had high phagocytic activity for monosodium urate crystals. Thus, our results indicate that monocytes/macrophages from patients with gout have the phenotype of infiltrated monocytes, and these cells consist of different populations characterized by anti-inflammatory activities as well as pro-inflammatory functions.
Collapse
Affiliation(s)
- Ji Hye Jeong
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea.,Asan Institute for Life Science, Asan Medical Center, Seoul, South Korea
| | - Seokchan Hong
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Oh Chan Kwon
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Byeongzu Ghang
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Inseok Hwang
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea.,Asan Institute for Life Science, Asan Medical Center, Seoul, South Korea
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Chang-Keun Lee
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Bin Yoo
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| |
Collapse
|
100
|
Anti-Gouty Arthritis and Antihyperuricemia Effects of Sunflower (Helianthus annuus) Head Extract in Gouty and Hyperuricemia Animal Models. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5852076. [PMID: 28929115 PMCID: PMC5591994 DOI: 10.1155/2017/5852076] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 07/08/2017] [Accepted: 07/19/2017] [Indexed: 12/26/2022]
Abstract
This study was performed to investigate the therapeutic effects and possible mechanisms of sunflower (Helianthus annuus) head extract (SHE) on gout. First, the components of sunflower head powder and SHE were analyzed systematically. SHE, especially SHEB (extracted with 20% ethanol and 80% double-distilled water), strongly suppressed the swelling of the ankles in rats with acute gout induced by monosodium urate (MSU) crystals and reduced the levels of uric acid and xanthine oxidase (XO) in mice with hyperuricemia induced by oteracil potassium and yeast extract powder. Hematoxylin and eosin staining indicated that SHEB reduced inflammation cells and increased the joint space in the ankle compared with the control rats with MSU-induced gout. In the rats with acute gout, among 13 detected inflammatory cytokines, SHEB significantly enhanced the serum levels of interleukin-10 and the monocyte chemoattractant protein 1α. In the mice with hyperuricemia, SHEB reduced the levels of glutathione peroxidase, superoxide dismutase, malondialdehyde, and nitrogen monoxide in liver tissues. The potential therapeutic effects of SHE on gout are probably due to the production of anti-inflammatory cytokines and the suppression of XO activity via the modulation of oxidative stress status.
Collapse
|