ASIC1a induces synovial inflammation via the Ca2+/NFATc3/ RANTES pathway

Yihao Zhang, Xuewen Qian, Xiaojuan Yang, Ruowen Niu, Sujing Song, Fei Zhu, Chuanjun Zhu, Xiaoqing Peng, Feihu Chen, Yihao Zhang, Xuewen Qian, Xiaojuan Yang, Ruowen Niu, Sujing Song, Fei Zhu, Chuanjun Zhu, Xiaoqing Peng, Feihu Chen

Abstract

Rationale: Synovial inflammation is one of the main pathological features of rheumatoid arthritis (RA) and is a key factor leading to the progression of RA. Understanding the regulatory mechanism of synovial inflammation is crucial for the treatment of RA. Acid-sensing ion channel 1a (ASIC1a) is an H+-gated cation channel that promotes the progression of RA, but the role of ASIC1a in synovial inflammation is unclear. This study aimed to investigate whether ASIC1a is involved in the synovial inflammation and explore the underlying mechanisms in vitro and in vivo. Methods: The expression of ASIC1a and nuclear factor of activated T cells (NFATs) were analyzed by Western blotting, immunofluorescence, and immunohistochemistry both in vitro and in vivo. The Ca2+ influx mediated by ASIC1a was detected by calcium imaging and flow cytometry. The role of ASIC1a in inflammation was studied in rats with adjuvant-induced arthritis (AA). Inflammatory cytokine profile was analyzed by protein chip in RA synovial fibroblasts (RASF) and verified by a magnetic multi-cytokine assay and ELISA. The NFATc3-regulated RANTES (Regulated upon activation, normal T cell expressed and secreted) gene transcription was investigated by ChIP-qPCR and dual-luciferase reporter assay. Results: The expression of ASIC1a was significantly increased in human RA synovial tissues and primary human RASF as well as in ankle synovium of AA rats. Activated ASIC1a mediated Ca2+ influx to increase [Ca2+]i in RASF. The activation/overexpression of ASIC1a in RASF up-regulated the expression of inflammatory cytokines RANTES, sTNF RI, MIP-1a, IL-8, sTNF RII, and ICAM-1 among which RANTES was increased most remarkably. In vivo, ASIC1a promoted inflammation, synovial hyperplasia, articular cartilage, and bone destruction, leading to the progression of AA. Furthermore, activation of ASIC1a upregulated the nuclear translocation of NFATc3, which bound to RANTES promoter and directly regulated gene transcription to enhance RANTES expression. Conclusion: ASIC1a induces synovial inflammation, which leads to the progression of RA. Our study reveals a novel RA inflammation regulatory mechanism and indicates that ASIC1a might be a potential therapeutic target for RA.

Keywords: ASIC1a; NFATc3; inflammation; rheumatoid arthritis.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interest exists.

© The author(s).

Figures

Figure 1
Figure 1
ASIC1a is highly expressed in human RA synovial tissues and primary human RASF. (A) Immunohistochemistry analysis of ASIC1a expression in human normal and RA synovial tissues. (B) Immunofluorescence analysis of ASIC1a expression in human normal and RA synovial tissues. The scale bars are 100 μm. (C) Cells identified by flow cytometry. (D) Western blot analysis of total ASIC1a expression in NSF and RASF. (E) Western blot analysis of membrane ASIC1a expression in NSF and RASF. (F) Membrane ASIC1a expression detected by flow cytometry. MFI, is mean fluorescence intensity. (G) Immunofluorescence analysis of ASIC1a expression in NSF and RASF. The scale bars are 20 μm. Student´s t-test or one-way ANOVA was used for statistical analysis, and data are expressed as mean ± SEM for three separate experiments.
Figure 2
Figure 2
ASIC1a has an ion channel activity that mediates Ca2+ influx. (A) Total ASIC1a protein was detected by Western blotting in RASF-ASIC1a transfectants and ASIC1a shRNA transfectants. (B) Membrane ASIC1a protein was detected by Western blotting in RASF-ASIC1a transfectants and ASIC1a shRNA transfectants. (C) Immunofluorescence analysis of ASIC1a expression in RASF-ASIC1a transfectants and ASIC1a shRNA transfectants. The scale bars are 20 μm. (D) Calcium imaging detected the effect of ASIC1a on [Ca2+]i in RASF with 100 nM PcTx-1, 1 mM EGTA, 10 μM BAPTA-AM, 5 μM verapamil and HMA (E) Calcium imaging detected ASIC1a-mediated Ca2+ influx in RASF with 100 nM PcTx-1, 1 mM EGTA, 10 μM BAPTA-AM, 5 μM verapamil and HMA. Student´s t-test or one-way ANOVA was used for statistical analysis, and data are expressed as mean ± SEM for three separate experiments.
Figure 3
Figure 3
ASIC1a upregulates the expression of inflammatory cytokines by mediating Ca2+ influx. (A) 40 inflammatory cytokines were analyzed by KEGG pathway enrichment analysis. (B) An inflammatory cytokines antibody array was used to examine the expression of 40 inflammatory cytokines in RASF with 100 nM PcTx-1. (C) Semi-quantitative analysis of the expressions of RANTES, sTNF RI, MIP-1a, IL-8, sTNF RII, and ICAM-1 by fluorescence intensity presented by heat map. (D) RANTES, sTNF RI, MIP-1a, IL-8, sTNF RII, and ICAM-1 were analyzed by KEGG pathway enrichment analysis. (E) The expression of RANTES was determined by magnetic multi-cytokine assay. Cells were treated for 6 h with100 nM PcTx-1. (F) Magnetic multi-cytokine assay detected the increase of RANTES expression mediated by ASIC1a in RASF treated for 6 h with 1 mM EGTA, 10 μM BAPTA-AM, 5 μM verapamil and HMA. Student´s t-test or one-way ANOVA was used for statistical analysis, and data are expressed as mean ± SEM for three separate experiments.
Figure 4
Figure 4
ASIC1a promotes inflammation by mediating the expression of inflammatory cytokines in vivo. (A) Representative image of right hind paw edema in rats with adjuvant-induced arthritis (AA) on day 20. (B) Normal and AA rat right hind ankle joint sections stained with hematoxylin and eosin. Histological section showing subchondral bone (B), bone erosion (BE), articular cartilage (C), cartilage erosion (CE), inflammatory cell infiltration (I), normal synovium (NS), pannus formation (P), synovium (S), and synovial hyperplasia (SH). (C) Immunohistochemistry analysis of ASIC1a expression in rat ankle synovium (n = 8). (D) The right hind paws of rats were measured by toe volume measuring instrument on day 0, 20, 23, 26, 29, 32, 35, 38, and 41. (E) Rat arthritic severity in each paw was evaluated by using a scoring system. (F) Representative image of right hind paw edema in rats treated with 0.5, 1, and 2 μg/kg PcTx-1. (G) Semi-quantitative analysis of RANTES expression in rat ankle synovium by integral optical density (IOD). Student´s t-test or one-way ANOVA was used for statistical analysis, and data are expressed as mean ± SEM for three separate experiments.
Figure 5
Figure 5
ASIC1a induces nuclear translocation of NFATc3 by mediating Ca2+ influx. (A) Semi-quantitative analysis of NFATc1-4 and NFAT5 expressions in human normal and RA synovial tissues by integral optical density (IOD). (B) Semi-quantitative analysis of NFATc1-4 and NFAT5 expressions in rat ankle synovium by integral optical density (IOD). (C) Western blot analysis of total, nuclear, and cytoplasmic NFATc1-4 and NFAT5 expressions in NSF and RASF. (D) Western blot analysis of total, nuclear, and cytoplasmic NFATc1-4 expressions in RASF in an acidic environment (pH 6.0). (E) Western blot analysis of total NFATc3 (tNFATc3), nuclear NFATc3 (nNFATc3), and cytoplasmic NFATc3 (cNFATc3) expression in RASF-ASIC1a transfectants treated with pH 6.0 for 6 h. (F) Western blot analysis of total NFATc3 (tNFATc3), nuclear NFATc3 (nNFATc3) and cytoplasmic NFATc3 (cNFATc3) expression in ASIC1a shRNA transfectants treated with pH 6.0 for 6 h. (G) Western blot analysis of Nuclear NFATc3 (nNFATc3) proteins in RASF with 1 mM EGTA, 10 μM BAPTA-AM, 5 μM verapamil and HMA. (H) Western blot analysis of total, nuclear and cytoplasmic NFATc1-4 proteins in with 100 nM PcTx-1 were used in the experiment. (I) Immunofluorescence analysis of NFATc1-4 expressions in RASF treated with pH 6.0 + 100 nM PcTx-1. The scale bars are 20 μm. (J) Immunofluorescence analysis of NFATc3 expression in RASF with 1 mM EGTA, 10 μM BAPTA-AM, 5 μM verapamil and HMA. The scale bars are 20 μm. (K) Immunohistochemistry analysis of NFATc3 expression in rat ankle synovium on day 41 (n = 8). Student´s t-test or one-way ANOVA was used for statistical analysis, and data are expressed as mean ± SEM for three separate experiments.
Figure 5
Figure 5
ASIC1a induces nuclear translocation of NFATc3 by mediating Ca2+ influx. (A) Semi-quantitative analysis of NFATc1-4 and NFAT5 expressions in human normal and RA synovial tissues by integral optical density (IOD). (B) Semi-quantitative analysis of NFATc1-4 and NFAT5 expressions in rat ankle synovium by integral optical density (IOD). (C) Western blot analysis of total, nuclear, and cytoplasmic NFATc1-4 and NFAT5 expressions in NSF and RASF. (D) Western blot analysis of total, nuclear, and cytoplasmic NFATc1-4 expressions in RASF in an acidic environment (pH 6.0). (E) Western blot analysis of total NFATc3 (tNFATc3), nuclear NFATc3 (nNFATc3), and cytoplasmic NFATc3 (cNFATc3) expression in RASF-ASIC1a transfectants treated with pH 6.0 for 6 h. (F) Western blot analysis of total NFATc3 (tNFATc3), nuclear NFATc3 (nNFATc3) and cytoplasmic NFATc3 (cNFATc3) expression in ASIC1a shRNA transfectants treated with pH 6.0 for 6 h. (G) Western blot analysis of Nuclear NFATc3 (nNFATc3) proteins in RASF with 1 mM EGTA, 10 μM BAPTA-AM, 5 μM verapamil and HMA. (H) Western blot analysis of total, nuclear and cytoplasmic NFATc1-4 proteins in with 100 nM PcTx-1 were used in the experiment. (I) Immunofluorescence analysis of NFATc1-4 expressions in RASF treated with pH 6.0 + 100 nM PcTx-1. The scale bars are 20 μm. (J) Immunofluorescence analysis of NFATc3 expression in RASF with 1 mM EGTA, 10 μM BAPTA-AM, 5 μM verapamil and HMA. The scale bars are 20 μm. (K) Immunohistochemistry analysis of NFATc3 expression in rat ankle synovium on day 41 (n = 8). Student´s t-test or one-way ANOVA was used for statistical analysis, and data are expressed as mean ± SEM for three separate experiments.
Figure 6
Figure 6
ASIC1a induces nuclear translocation of NFATc3 to regulate transcription of the RANTES gene. (A) Western blot analysis of total, nuclear, and cytoplasmic NFATc3 in NFATc3 shRNA transfectants. (B) Immunofluorescence analysis of NFATc3 expression in NFATc3 shRNA transfectants. The scale bars are 20 μm. (C) Western blot analysis of nuclear NFATc3 in RASF treated with 5μM ionomycin. (D) Immunofluorescence analysis of NFATc3 expression in RASF treated with 5μM ionomycin. The scale bars are 20 μm. (E) Schematic diagram of the primer 2-guided amplified DNA fragments (1005, 902) and predicted binding site for NFATc3 in the promoter region of the human RANTES gene. Transcription start site (TSS) of the RANTES promoter is +1. (F) Primer 1, 2, and 3 were used to detect the binding of NFATc3 to RANTES promoter region in untreated RASF by ChIP-qPCR assay. (G) Primer 2 was used to detect the binding of NFATc3 to RANTES promoter region in treated RASF by ChIP-qPCR assay in the presence of 5 μM ionomycin. (H) Schematic diagram of the promoter region of the human RANTES gene used for dual-luciferase reporter assay. Transcription start site (TSS) of the RANTES promoter is +1. (I) Dual-luciferase reporter assay showing regulation of RANTES transcription by direct binding of NFATc3 to the RANTES promoter region in RASF. pGL3 basic group and pGL3 promoter group served as negative and positive controls, respectively. (J) The expression of RANTES was determined in RASF treated with 5 μM ionomycin and NFATc3 shRNA transfectants by magnetic multi-cytokine assay. Student´s t-test or one-way ANOVA was used for statistical analysis, and data are expressed as mean ± SEM for three separate experiments.
Figure 7
Figure 7
Schematic diagram of the molecular mechanism of ASIC1a-induced synovial inflammation. ASIC1a mediates Ca2+ influx to enhance nuclear translocation of NFATc3, which binds to the RANTES promoter to directly regulate RANTES gene transcription and protein expression.

References

    1. Aletaha D, Smolen JS. Diagnosis and Management of Rheumatoid Arthritis: A Review. Jama. 2018;320:1360–72.
    1. Van der Woude D, van der Helm-van Mil AHM. Update on the epidemiology, risk factors, and disease outcomes of rheumatoid arthritis. Best Pract Res Clin Rheumatol. 2018;32:174–87.
    1. Guo Q, Wang Y, Xu D, Nossent J, Pavlos NJ, Xu J. Rheumatoid arthritis: pathological mechanisms and modern pharmacologic therapies. Bone Res. 2018;6:15.
    1. Ito H, Moritoshi F, Hashimoto M, Tanaka M, Matsuda S. Control of articular synovitis for bone and cartilage regeneration in rheumatoid arthritis. Inflamm Regen. 2018;38:7.
    1. Boissier MC, Semerano L, Challal S. et al. Rheumatoid arthritis: from autoimmunity to synovitis and joint destruction. J Autoimmun. 2012;39:222–28.
    1. Townsend MJ. Molecular and cellular heterogeneity in the Rheumatoid Arthritis synovium: clinical correlates of synovitis. Best Pract Res Clin Rheumatol. 2014;28:539–49.
    1. Smolen JS, Aletaha D. Rheumatoid arthritis therapy reappraisal: strategies, opportunities and challenges. Nat Rev Rheumatoly. 2015;11:276–89.
    1. Taylor PC, Alten R, Gomez-Reino JJ, Caporali R, Bertin P, Sullivan E. et al. Clinical characteristics and patient-reported outcomes in patients with inadequately controlled rheumatoid arthritis despite ongoing treatment. RMD open. 2018;4:e000615.
    1. Winthrop KL, Strand V, van der Heijde D. et al. The unmet need in rheumatology: Reports from the targeted therapies meeting 2017. Clin Immunol. 2018;186:87–93.
    1. Arnett TR. Extracellular pH regulates bone cell function. J Nutr. 2008;138:415S–8S.
    1. Arnett TR. Acidosis, hypoxia and bone. Arch Biochem Biophys. 2010;503:103–9.
    1. Erra Diaz F, Dantas E, Geffner J. Unravelling the Interplay between Extracellular Acidosis and Immune Cells. Mediators Inflamm. 2018;2018:1218297.
    1. Trevani AS, Andonegui G, Giordano M. et al. Extracellular acidification induces human neutrophil activation. J Immunol. 1999;162:4849–57.
    1. Martínez D, Vermeulen M, Trevani A. et al. Extracellular acidosis induces neutrophil activation by a mechanism dependent on activation of phosphatidylinositol 3-kinase/Akt and ERK pathways. J Immunol. 2006;176:1163–71.
    1. Vermeulen M, Giordano M, Trevani AS. et al. Acidosis improves uptake of antigens and MHC class I-restricted presentation by dendritic cells. J Immunol. 2004;172:3196–3204.
    1. Cummings NA. Measurement of synovial fluid pH in normal and arthritic knees. Arthritis Rheum. 1966;9:47–56.
    1. Farr M, Garvey K, Bold AM. et al. Significance of the hydrogen ion concentration in synovial fluid in rheumatoid arthritis. Clin Exp Rheumatol. 1985;3:99–104.
    1. Goldie I. Synovial pH in rheumatoid knee-joints. I. The effect of synovectomy. Acta Orthop Scand. 1969;40:634–41.
    1. Falchuk KH, Goetzl EJ. Respiratory gases of synovial fluids. An approach to synovial tissue circulatory-metabolic imbalance in rheumatoid arthritis. Am J Med. 1970;49:223–31.
    1. Geborek P, Saxne T, Pettersson H. Synovial fluid acidosis correlates with radiological joint destruction in rheumatoid arthritis knee joints. J Rheumatol. 1989;16:468–72.
    1. Waldmann R, Champigny G, Bassilana F. et al. A proton-gated cation channel involved in acid-sensing. Nature. 1997;386:173–7.
    1. Lingueglia E. Acid-sensing ion channels in sensory perception. J Biol Chem. 2007;282:17325–9.
    1. Yoder N, Yoshioka C. Gating mechanisms of acid-sensing ion channels. Nature. 2018;555:397–401.
    1. Gründer S. Biophysical properties of acid-sensing ion channels (ASICs) Neuropharmacology. 2015;94:9–18.
    1. Xu Y, Jiang YQ, Li C. et al. Human ASIC1a mediates stronger acid-induced responses as compared with mouse ASIC1a. FASEB J. 2018;32:3832–43.
    1. Pathak T. Mitochondrial Ca2+ signaling. Pharmacol Ther. 2018;192:112–23.
    1. Santella L, Ercolano E. The cell cycle: a new entry in the field of Ca2+ signaling. Cell Mol Life Sci. 2005;62:2405–13.
    1. Wu X, Ren G, Zhou R. et al. The role of Ca in acid-sensing ion channel 1a-mediated chondrocyte pyroptosis in rat adjuvant arthritis. Lab Invest. 2019;99:499–513.
    1. Sun C, Wang S. Acid-sensing ion channel 1a mediates acid-induced inhibition of matrix metabolism of rat articular chondrocytes via the MAPK signaling pathway. Mol Cell Biochem. 2018;443:81–91.
    1. Zhou RP, Dai BB, Xie YY. et al. Interleukin-1β and tumor necrosis factor-α augment acidosis-induced rat articular chondrocyte apoptosis via nuclear factor-kappaB-dependent upregulation of ASIC1a channel. Biochim Biophys Acta Mol Basis Dis. 2018;1864:162–77.
    1. Tong J, Wu WN, Kong X. et al. Acid-sensing ion channels contribute to the effect of acidosis on the function of dendritic cells. J Immunol. 2011;186:3686–92.
    1. Lynagh T, Romero-Rojo JL, Lund C. Molecular Basis for Allosteric Inhibition of Acid-Sensing Ion Channel 1a by Ibuprofen. J Med Chem. 2017;60:8192–200.
    1. Yen YT, Tu PH, Chen CJ. et al. Role of acid-sensing ion channel 3 in sub-acute-phase inflammation. Mol Pain. 2009;5:1.
    1. Friese MA, Craner MJ, Etzensperger R. et al. Acid-sensing ion channel-1 contributes to axonal degeneration in autoimmune inflammation of the central nervous system. Nat Med. 2007;13:1483–9.
    1. Hogan PG, Chen L, Nardone J. Transcriptional regulation by calcium, calcineurin, and NFAT. Genes Dev. 2003;17:2205–32.
    1. Chuvpilo S, Avots A, Berberich-Siebelt F. et al. Multiple NF-ATc isoforms with individual transcriptional properties are synthesized in T lymphocytes. J Immunol. 1999;162:7294–301.
    1. Aramburu J, Drews-Elger K, Estrada-Gelonch A. et al. Regulation of the hypertonic stress response and other cellular functions by the Rel-like transcription factor NFAT5. Biochem Pharmacol. 2006;72:1597–1604.
    1. Hogan PG. Calcium-NFAT transcriptional signalling in T cell activation and T cell exhaustion. Cell calcium. 2017;63:66–9.
    1. Gallo EM, Winslow MM, Canté-Barrett K. et al. Calcineurin sets the bandwidth for discrimination of signals during thymocyte development. Nature. 2007;450:731–5.
    1. Brunvand MW, Schmidt A. Nuclear factors interacting with the mitogen-responsive regulatory region of the interleukin-2 gene. J Biol Chem. 1988;263:18904–10.
    1. Köck J, Kreher S, Lehmann K. et al. Nuclear factor of activated T cells regulates the expression of interleukin-4 in Th2 cells in an all-or-none fashion. J Biol Chem. 2014;289:26752–61.
    1. Kiani A, Habermann I, Haase M. et al. Expression and regulation of NFAT (nuclear factors of activated T cells) in human CD34+ cells: down-regulation upon myeloid differentiation. J Leukoc Biol. 2004;76:1057–65.
    1. Ogawa K, Kaminuma O, Okudaira H. et al. Transcriptional regulation of the IL-5 gene in peripheral T cells of asthmatic patients. Clin Exp Immunol. 2002;130:475–83.
    1. Liu XK, Lin X. Crucial role for nuclear factor of activated T cells in T cell receptor-mediated regulation of human interleukin-17. J Biol Chem. 2004;279:52762–71.
    1. Choi S, You S, Kim D. et al. Transcription factor NFAT5 promotes macrophage survival in rheumatoid arthritis. J Clin Invest. 2017;127:954–69.
    1. Zhou R, Wu X, Wang Z. et al. Interleukin-6 enhances acid-induced apoptosis via upregulating acid-sensing ion channel 1a expression and function in rat articular chondrocytes. Int Immunopharmacol. 2015;29:748–60.
    1. Kaufmann U, Kahlfuss S, Yang J, Ivanova E, Koralov SB, Feske S. Calcium Signaling Controls Pathogenic Th17 Cell-Mediated Inflammation by Regulating Mitochondrial Function. Cell metabolism; 2019.
    1. Tauseef M, Knezevic N, Chava KR. et al. TLR4 activation of TRPC6-dependent calcium signaling mediates endotoxin-induced lung vascular permeability and inflammation. J Exp Med. 2012;209:1953–68.
    1. van Eden W, Wagenaar-Hilbers JP, Wauben MH. Current protocols in immunology. New York, USA: Greene Pub. Associates and Wiley-Interscience Publisher; 2001.
    1. Hamada T, Arima N, Shindo M. et al. Suppression of adjuvant arthritis of rats by a novel matrix metalloproteinase-inhibitor. Br J Pharmacol. 2000;131:1513–20.
    1. Huang Y, Zheng S, Wang R, CCL5 and related genes might be the potential diagnostic biomarkers for the therapeutic strategies of rheumatoid arthritis. Clin Rheumatol; 2019.
    1. Agere SA, Akhtar N, Watson JM, Ahmed S. RANTES/CCL5 Induces Collagen Degradation by Activating MMP-1 and MMP-13 Expression in Human Rheumatoid Arthritis Synovial Fibroblasts. Frontiers in immunology. 2017;8:1341.
    1. Haringman JJ, Smeets TJ, Reinders-Blankert P. Chemokine and chemokine receptor expression in paired peripheral blood mononuclear cells and synovial tissue of patients with rheumatoid arthritis, osteoarthritis, and reactive arthritis. Ann Rheum Dis. 2006;65:294–300.
    1. Hirano F, Kobayashi A, Hirano Y. et al. Thrombin-induced expression of RANTES mRNA through protease activated receptor-1 in human synovial fibroblasts. Ann Rheum Dis. 2002;61:834–7.
    1. Hiyama A, Gajghate S, Sakai D. et al. Activation of TonEBP by calcium controls β1,3-glucuronosyltransferase-I expression, a key regulator of glycosaminoglycan synthesis in cells of the intervertebral disc. J Biol Chem. 2009;284:9824–34.
    1. Boissier MC. Cell and cytokine imbalances in rheumatoid synovitis. Jt Bone Spine. 2011;78:230–4.
    1. Harre U. Cellular and molecular pathways of structural damage in rheumatoid arthritis. Semin Immunopathol. 2017;39:355–63.
    1. Obry A, Hardouin J, Lequerré T. et al. Identification of 7 Proteins in Sera of RA Patients with Potential to Predict ETA/MTX Treatment Response. Theranostics. 2015;5:1214–24.
    1. Hao F, Lee RJ, Zhong L. et al. Hybrid micelles containing methotrexate-conjugated polymer and co-loaded with microRNA-124 for rheumatoid arthritis therapy. Theranostics. 2019;9:5282–97.
    1. Yang M, Ding J, Feng X. et al. Scavenger Receptor-Mediated Targeted Treatment of Collagen-Induced Arthritis by Dextran Sulfate-Methotrexate Prodrug. Theranostics. 2017;7:97–105.
    1. Smolen JS, Aletaha D. Rheumatoid arthritis. Lancet. 2016;388:2023–38.
    1. Wang Y, Liu Z, Li T. et al. Enhanced Therapeutic Effect of RGD-Modified Polymeric Micelles Loaded With Low-Dose Methotrexate and Nimesulide on Rheumatoid Arthritis. Theranostics. 2019;9:708–20.
    1. Konttinen YT, Bergroth V, Nordström D. et al. Cellular immunohistopathology of acute, subacute, and chronic synovitis in rheumatoid arthritis. Ann Rheum Dis. 1985;44:549–55.
    1. Kaul A, Blake DR. Vascular endothelium, cytokines, and the pathogenesis of inflammatory synovitis. Ann Rheum Dis. 1991;50:828–32.
    1. Bhattaram P. The joint synovium: A critical determinant of articular cartilage fate in inflammatory joint diseases. Semin Cell Dev Biol. 2017;62:86–93.
    1. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med. 2011;365:2205–19.
    1. Ziff M. Role of endothelium in the pathogenesis of rheumatoid synovitis. Int J Tissue React. 1993;15:135–7.
    1. Smolen JS, Aletaha D, Koeller M, Weisman M, Emery P. New therapies for the treatment of rheumatoid arthritis. Lancet. 2007;370:1861–74.
    1. Swenson ER. Metabolic acidosis. Respir Care. 2001;46:342–53.
    1. Feldman JL, Mitchell GS, Nattie EE. Breathing: rhythmicity, plasticity, chemosensitivity. Annu Rev Neurosci. 2003;26:239–66.
    1. Simmen HP, Blaser J. Analysis of pH and pO2 in abscesses, peritoneal fluid, and drainage fluid in the presence or absence of bacterial infection during and after abdominal surgery. Am J Surg. 1993;166:24–7.
    1. Martínez D, Vermeulen M, von Euw E, Sabatté J, Maggíni J, Ceballos A. et al. Extracellular acidosis triggers the maturation of human dendritic cells and the production of IL-12. J Immunol. 2007;179:1950–9.
    1. Lardner A. The effects of extracellular pH on immune function. J Leukoc Biol. 2001;69:522–30.
    1. Riemann A, Wußling H, Loppnow H. et al. Acidosis differently modulates the inflammatory program in monocytes and macrophages. Biochim Biophys Acta. 2016;1862:72–81.
    1. Kong X, Tang X, Du W. et al. Extracellular acidosis modulates the endocytosis and maturation of macrophages. Cell Immunol. 2013;281:44–50.
    1. Klein-Hessling S, Muhammad K, Klein M. et al. NFATc1 controls the cytotoxicity of CD8 T cells. Nat Commun. 2017;8:511.
    1. Yamanaka Y, Abu-Amer W, Foglia D. et al. NFAT2 is an essential mediator of orthopedic particle-induced osteoclastogenesis. J Orthop Res. 2008;26:1577–84.
    1. Ishida N, Hayashi K, Hoshijima M. et al. Large scale gene expression analysis of osteoclastogenesis in vitro and elucidation of NFAT2 as a key regulator. J Biol Chem. 2002;277:41147–56.
    1. Gonzalez Bosc LV, Plomaritas DR, Herbert LM. et al. ASIC1-mediated calcium entry stimulates NFATc3 nuclear translocation via PICK1 coupling in pulmonary arterial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2016;311:L48–58.
    1. Li X, Xu RS, Jiang DL. et al. Acid-sensing ion channel 1a is involved in acid-induced osteoclastogenesis by regulating activation of the transcription factor NFATc1. FEBS Lett. 2013;587:3236–42.
    1. Gerard C. Inflammatory chemokines: tuned in, turned on, dropped out. Nat Immunol. 2005;6:366–8.
    1. Gura T. Chemokines take center stage in inflammatory ills. Science. 1996;272:954–6.
    1. Okamoto H. Molecular aspects of rheumatoid arthritis: chemokines, environmental factors and transcription factors. FEBS J. 2008;275:4447.
    1. Szekanecz Z, Kim J. Chemokines and chemokine receptors in rheumatoid arthritis. Semin Immunol. 2003;15:15–21.
    1. Schall TJ. Biology of the RANTES/SIS cytokine family. Cytokine. 1991;3:165–83.
    1. Whyte A. RANTES and chemokines in rheumatoid arthritis. Lancet. 1994;343:1291–2.
    1. Appay V. RANTES: a versatile and controversial chemokine. Trends Immunol. 2001;22:83–7.
    1. Haringman JJ, Smeets TJ, Reinders-Blankert P. Chemokine and chemokine receptor expression in paired peripheral blood mononuclear cells and synovial tissue of patients with rheumatoid arthritis, osteoarthritis, and reactive arthritis. Ann Rheum Dis. 2006;65:294–300.
    1. Stanczyk J, Kowalski ML, Grzegorczyk J. et al. RANTES and chemotactic activity in synovial fluids from patients with rheumatoid arthritis and osteoarthritis. Mediators Inflamm. 2005;2005:343–8.
    1. Shadidi KR, Aarvak T, Henriksen JE. et al. The chemokines CCL5, CCL2 and CXCL12 play significant roles in the migration of Th1 cells into rheumatoid synovial tissue. Scand J Immunol. 2003;57:192–8.
    1. Shahrara S, Proudfoot AE, Park CC. et al. Inhibition of monocyte chemoattractant protein-1 ameliorates rat adjuvant-induced arthritis. J Immunol. 2008;180:3447–56.
    1. Ferrandi C, Ardissone V, Ferro P. et al. Phosphoinositide 3-kinase gamma inhibition plays a crucial role in early steps of inflammation by blocking neutrophil recruitment. J Pharmacol Exp Ther. 2007;322:923–30.
    1. Rathanaswami P, Hachicha M, Sadick M, Schall TJ, McColl SR. Expression of the cytokine RANTES in human rheumatoid synovial fibroblasts. Differential regulation of RANTES and interleukin-8 genes by inflammatory cytokines. J Biol Chem. 1993;268:5834–9.
    1. Pierer M, Rethage J, Seibl R. et al. Chemokine secretion of rheumatoid arthritis synovial fibroblasts stimulated by Toll-like receptor 2 ligands. J Immunol. 2004;172:1256–65.

Source: PubMed

3
Předplatit