Functional role of PPAR-γ on the proliferation and migration of fibroblast-like synoviocytes in rheumatoid arthritis

Xiao-Feng Li, Ying-Yin Sun, Jing Bao, Xin Chen, Yu-Huan Li, Yang Yang, Lei Zhang, Cheng Huang, Bao-Ming Wu, Xiao-Ming Meng, Jun Li, Xiao-Feng Li, Ying-Yin Sun, Jing Bao, Xin Chen, Yu-Huan Li, Yang Yang, Lei Zhang, Cheng Huang, Bao-Ming Wu, Xiao-Ming Meng, Jun Li

Abstract

Peroxisome proliferator-activated receptor (PPAR)-γ is involved in both normal physiological processes and pathology of various diseases. The purpose of this study was to explore the function and underlying mechanisms of PPAR-γ in rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLSs) proliferation and migration. In the present study, we found PPAR-γ expression was remarkably reduced in RA synovium patient compare with OA and normal, as well as it was low-expression in Adjuvant-induced arthritis (AA). Moreover, inhibition PPAR-γ expression by T0070907 (12.5 μM) or PPAR-γ siRNA could promote FLSs proliferation and expressions of c-Myc, Cyclin D1, MMP-1, and MMP-9 in AA FLSs, except for TIPM-1. These date indicate that up-regulation of PPAR-γ may play a critical role in RA FLSs. Interestingly, co-incubation FLSs with Pioditazone (25 μM) and over expression vector with pEGFP-N1-PPAR-γ reduced proliferation and expressions of c-Myc, Cyclin D1, MMP-1, and MMP-9 in AA FLSs, besides TIMP-1. Further study indicates that PPAR-γ may induce activation Wnt/β-catenin signaling. In short, these results indicate that PPAR-γ may play a pivotal role during FLSs activation and activation of Wnt/β-catenin signaling pathway.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
The expression of PPAR-γ was down-regulated in RA FLSs. (a) Representative H&E staining of AA and normal synovial tissues in rat (original magnification, ×10). (b) Representative H&E staining of RA, OA and normal synovial tissues in human (original magnification, ×10). (c) The expression of PPAR-γ in RA, OA and normal synovial tissue was analyzed by IHC staining analysis in human. (d) The protein level of PPAR-γ was analyzed by Western blot in RA, OA and normal synovial tissue. (e) The expression of PPAR-γ and Vimentin were analyzed by double immunofluorescence staining analysis in rat AA and normal FLSs. (f) The protein level of PPAR-γ was analyzed by Western blot in AA and normal FLSs. (g) The mRNA level of PPAR-γ was analyzed by Q-PCR in AA and normal FLSs. All values were expressed as mean ± SEM. ##P < 0.01 vs normal group.
Figure 2
Figure 2
Effect of PPAR-γ inhibitor increases FLSs proliferation and migration. (a) Concentration-dependent inhibition expression of PPAR-γ mRNA by T0070907 in normal FLSs, tested by Q-PCR assays. (b) The mRNA levels of PPAR-γ, c-Myc and Cyclin D1 were analyzed by Q-PCR in FLSs with T0070907 (12.5 μM). (c) The protein levels of PPAR-γ, c-Myc and Cyclin D1 were analyzed by Western blot in FLSs with T0070907 (12.5 μM). (d) The mRNA levels of MMP-3, MMP-9 and TIMP-1 were analyzed by Q-PCR in FLSs with T0070907 (12.5 μM). (e) The protein levels of MMP-3, MMP-9 and TIMP-1 were analyzed by Western blot in FLSs with T0070907 (12.5 μM). (f) Cell cycle of FLSs were incubated with T0070907 (12.5 μM) for 48 h and then subjected to the FACS analysis. (g) After stained with CFDA-SE, FLSs were incubated with T0070907 (12.5 μM) for six days and then subjected to the FACS analysis. (h) BrdU proliferation assay were treated with T0070907 (12.5 μM) 48 h in FLSs. (i) FLSs were treated with T0070907 (12.5 μM), and migration into the wound-healing 24 h was photographed (original magnification, ×10). (j) FLSs were treated with T0070907 (12.5 μM), and transwell migration 48 h was photographed (original magnification, ×10). All values were expressed as mean ± SEM. #P < 0.05, ##P < 0.01 vs normal group. *P < 0.05, **P < 0.01 vs model group.
Figure 3
Figure 3
Effect of PPAR-γ siRNA silencing increases FLSs proliferation and migration. (a) The mRNA levels of PPAR-γ, c-Myc and Cyclin D1 were analyzed by Q-PCR in FLSs with PPAR-γ siRNA. (b) The protein levels of PPAR-γ, c-Myc and Cyclin D1 were analyzed by Western blot in FLSs with PPAR-γ siRNA. (c) The mRNA levels of MMP-3, MMP-9 and TIMP-1 were analyzed by Q-PCR in FLSs with PPAR-γ siRNA. (d) The protein levels of MMP-3, MMP-9 and TIMP-1 were analyzed by Western blot in FLSs with PPAR-γ siRNA. (e) Cell cycle of FLSs were incubated with PPAR-γ siRNA for 48 h and then subjected to the FACS analysis. (f) After stained with CFDA-SE, FLSs were incubated with PPAR-γ siRNA for six days and then subjected to the FACS analysis. (g) BrdU proliferation assay were treated with PPAR-γ siRNA 48 h in FLSs. (h) FLSs were treated with PPAR-γ siRNA, and migration into the wound-healing 24 h was photographed (original magnification, ×10). (i) FLSs were treated with PPAR-γ siRNA, and transwell migration 48 h was photographed (original magnification, ×10). All values were expressed as mean ± SEM. #P < 0.05, ##P < 0.01 vs normal group. *P < 0.05, **P < 0.01 vs model group.
Figure 4
Figure 4
Effect of PPAR-γ agonist inhibits FLSs proliferation and migration. (a) Concentration-dependent inhibition expression of PPAR-γ mRNA by Pioditazone in AA FLSs, tested by Q-PCR assays. (b) The mRNA levels of PPAR-γ, c-Myc and Cyclin D1 were analyzed by Q-PCR in FLSs with Pioditazone. (c) The protein levels of PPAR-γ, c-Myc and Cyclin D1 were analyzed by Western blot in FLSs with Pioditazone. (d) The mRNA levels of MMP-3, MMP-9 and TIMP-1 were analyzed by Q-PCR in FLSs with Pioditazone. (e) The protein levels of MMP-3, MMP-9 and TIMP-1 were analyzed by Western blot in FLSs with Pioditazone. (f) Cell cycle of FLSs were incubated with Pioditazone for 48 h and then subjected to the FACS analysis. (g) After stained with CFDA-SE, FLSs were incubated with Pioditazone for six days and then subjected to the FACS analysis. (h) BrdU proliferation assay were treated with Pioditazone 48 h in FLSs. (i) FLSs were treated with Pioditazone, and migration into the wound-healing 24 h was photographed (original magnification, ×10). (j) FLSs were treated with Pioditazone, and transwell migration 48 h was photographed (original magnification, ×10). All values were expressed as mean ± SEM. #P < 0.05, ##P < 0.01 vs normal group. *P < 0.05, **P < 0.01 vs model group.
Figure 5
Figure 5
Effect of over expression vector of PPAR-γ inhibits FLSs proliferation and migration. (a) The mRNA levels of PPAR-γ, c-Myc and Cyclin D1 were analyzed by Q-PCR in FLSs with pEGFP-N1-PPAR-γ. (b) The protein levels of PPAR-γ, c-Myc and Cyclin D1 were analyzed by Western blot in FLSs with pEGFP-N1-PPAR-γ. (c) The mRNA levels of MMP-3, MMP-9 and TIMP-1 were analyzed by Q-PCR in FLSs with pEGFP-N1-PPAR-γ. (d) The protein levels of MMP-3, MMP-9 and TIMP-1 were analyzed by Western blot in FLSs with pEGFP-N1-PPAR-γ. (e) Cell cycle of FLSs were incubated with pEGFP-N1-PPAR-γ for 48 h and then subjected to the FACS analysis. (f) After stained with CFDA-SE, FLSs were incubated with pEGFP-N1-PPAR-γ for six days and then subjected to the FACS analysis. (g) BrdU proliferation assay were treated with pEGFP-N1-PPAR-γ 48 h in FLSs. (h) FLSs were treated with pEGFP-N1-PPAR-γ, and migration into the wound-healing 24 h was photographed (original magnification, ×10). (i) FLSs were treated with pEGFP-N1-PPAR-γ, and transwell migration 48 h was photographed (original magnification, ×10). All values were expressed as mean ± SEM. #P < 0.05, ##P < 0.01 vs normal group. *P < 0.05, **P < 0.01 vs model group.
Figure 6
Figure 6
PPAR-γ may modulate FLSs proliferation and be closely associated with Wnt/β-catenin signaling pathway. (a) The protein level of β-catenin was analyzed by Western blot in FLSs with T0070907 (12.5 μM). (b) The protein level of β-catenin was analyzed by Western blot in FLSs with PPAR-γ siRNA. (c) The protein level of β-catenin was analyzed by Western blot in FLSs with Pioditazone (25 μM). (d) The protein level of β-catenin was analyzed by Western blot in FLSs with over expression vector pEGFP-N1-PPAR-γ. (e) The protein level of c-Myc, Cyclin D1, MMP-3, MMP-9 and β-catenin was analyzed by Western blot in AA FLSs. All values were expressed as mean ± SEM. #P < 0.05, ##P < 0.01 vs normal group. *P < 0.05, **P < 0.01 vs model group. (e) #P < 0.05, ##P < 0.01 vs model group; **P < 0.01 vs PPAR-γ siRNA group; &P < 0.05, &&P < 0.01 vs model group.

References

    1. Feldmann M, Brennan FM, Maini RN. Rheumatoid arthritis. Cell. 1996;85:307–310. doi: 10.1016/S0092-8674(00)81109-5.
    1. Elshabrawy, H. A. et al. The pathogenic role of angiogenesis in rheumatoid arthritis. Angiogenesis (2015).
    1. Dulos J, et al. p38 inhibition and not MK2 inhibition enhances the secretion of chemokines from TNF-alpha activated rheumatoid arthritis fibroblast-like synoviocytes. Clin Exp Rheumatol. 2013;31:515–525.
    1. Yeremenko N, et al. Brief Report: Tumor Necrosis Factor and Interleukin-6 Differentially Regulate Dkk-1 in the Inflamed Arthritic Joint. Arthritis Rheumatol. 2015;67:2071–2075. doi: 10.1002/art.39183.
    1. Koga T, et al. Post-transcriptional regulation of IL-6 production by Zc3h12a in fibroblast-like synovial cells. Clin Exp Rheumatol. 2011;29:906–912.
    1. Kontny, E., Janicka, I., Skalska, U. & Maslinski, W. The effect of multimeric adiponectin isoforms and leptin on the function of rheumatoid fibroblast-like synoviocytes. Scand J Rheumatol 1–6 (2015).
    1. Fava RA, et al. Vascular permeability factor/endothelial growth factor (VPF/VEGF): accumulation and expression in human synovial fluids and rheumatoid synovial tissue. J Exp Med. 1994;180:341–346. doi: 10.1084/jem.180.1.341.
    1. Pap T, et al. Modulation of fibroblast-mediated cartilage degradation by articular chondrocytes in rheumatoid arthritis. Arthritis Rheum. 2000;43:2531–2536. doi: 10.1002/1529-0131(200011)43:11<2531::AID-ANR21>;2-V.
    1. Zhu J, et al. Overexpression of Suprabasin is Associated with Proliferation and Tumorigenicity of Esophageal Squamous Cell Carcinoma. Sci Rep. 2016;6:21549. doi: 10.1038/srep21549.
    1. Martin G, et al. Role of plasma matrix-metalloproteases (MMPs) and their polymorphisms (SNPs) in sepsis development and outcome in ICU patients. Sci Rep. 2014;4:5002. doi: 10.1038/srep05002.
    1. Ormseth MJ, et al. Reversing vascular dysfunction in rheumatoid arthritis: improved augmentation index but not endothelial function with peroxisome proliferator-activated receptor gamma agonist therapy. Arthritis Rheumatol. 2014;66:2331–2338. doi: 10.1002/art.38686.
    1. Lee YH, Bae SC, Song GG. Meta-analysis of associations between the peroxisome proliferator-activated receptor-gamma Pro12Ala polymorphism and susceptibility to nonalcoholic fatty liver disease, rheumatoid arthritis, and psoriatic arthritis. Genet Test Mol Biomarkers. 2014;18:341–348. doi: 10.1089/gtmb.2013.0503.
    1. Moore-Carrasco R, et al. Peroxisome proliferator-activated receptors: Targets for the treatment of metabolic illnesses (Review) Mol Med Rep. 2008;1:317–324.
    1. Roberts-Thomson SJ. Peroxisome proliferator-activated receptors in tumorigenesis: targets of tumour promotion and treatment. Immunol Cell Biol. 2000;78:436–441. doi: 10.1046/j.1440-1711.2000.00921.x.
    1. Abbas A, Blandon J, Rude J, Elfar A, Mukherjee D. PPAR- gamma agonist in treatment of diabetes: cardiovascular safety considerations. Cardiovasc Hematol Agents Med Chem. 2012;10:124–134. doi: 10.2174/187152512800388948.
    1. Feng X, et al. Chrysin attenuates inflammation by regulating M1/M2 status via activating PPARgamma. Biochem Pharmacol. 2014;89:503–514. doi: 10.1016/j.bcp.2014.03.016.
    1. Koufany M, et al. The peroxisome proliferator-activated receptor gamma agonist pioglitazone preserves bone microarchitecture in experimental arthritis by reducing the interleukin-17-dependent osteoclastogenic pathway. Arthritis Rheum. 2013;65:3084–3095. doi: 10.1002/art.38130.
    1. Marder W, et al. The peroxisome proliferator activated receptor-gamma pioglitazone improves vascular function and decreases disease activity in patients with rheumatoid arthritis. J Am Heart Assoc. 2013;2:e000441. doi: 10.1161/JAHA.113.000441.
    1. Rabelo Fde S, et al. The Wnt signaling pathway and rheumatoid arthritis. Autoimmun Rev. 2010;9:207–210. doi: 10.1016/j.autrev.2009.08.003.
    1. Imai K, et al. Differential expression of WNTs and FRPs in the synovium of rheumatoid arthritis and osteoarthritis. Biochem Biophys Res Commun. 2006;345:1615–1620. doi: 10.1016/j.bbrc.2006.05.075.
    1. Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol. 2004;20:781–810. doi: 10.1146/annurev.cellbio.20.010403.113126.
    1. Moon RT, Brown JD, Torres M. WNTs modulate cell fate and behavior during vertebrate development. Trends Genet. 1997;13:157–162. doi: 10.1016/S0168-9525(97)01093-7.
    1. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature. 2015;523:231–235. doi: 10.1038/nature14404.
    1. Schneider, S. et al. The prognostic significance of beta-catenin, cyclin D1 and PIN1 in minor salivary gland carcinoma: beta-catenin predicts overall survival. Eur Arch Otorhinolaryngol (2015).
    1. Li X, Wang X, Wang Y, Huang C, Li J. Inhibition of transient receptor potential melastatin 7 (TRPM7) channel induces RA FLSs apoptosis through endoplasmic reticulum (ER) stress. Clin Rheumatol. 2014;33:1565–1574. doi: 10.1007/s10067-014-2599-x.
    1. Li, X. F. et al. MicroRNA-20a negatively regulates expression of NLRP3-inflammasome by targeting TXNIP in adjuvant-induced arthritis fibroblast-like synoviocytes. Joint Bone Spine (2016).
    1. Smeets TJ, et al. Analysis of the cell infiltrate and expression of proinflammatory cytokines and matrix metalloproteinases in arthroscopic synovial biopsies: comparison with synovial samples from patients with end stage, destructive rheumatoid arthritis. Ann Rheum Dis. 2003;62:635–638. doi: 10.1136/ard.62.7.635.
    1. Larsen H, Muz B, Khong TL, Feldmann M, Paleolog EM. Differential effects of Th1 versus Th2 cytokines in combination with hypoxia on HIFs and angiogenesis in RA. Arthritis Res Ther. 2012;14:R180. doi: 10.1186/ar3934.
    1. Lee DG, Woo JW, Kwok SK, Cho ML, Park SH. MRP8 promotes Th17 differentiation via upregulation of IL-6 production by fibroblast-like synoviocytes in rheumatoid arthritis. Exp Mol Med. 2013;45:e20. doi: 10.1038/emm.2013.39.
    1. Park SJ, Kim KJ, Kim WU, Cho CS. Interaction of mesenchymal stem cells with fibroblast-like synoviocytes via cadherin-11 promotes angiogenesis by enhanced secretion of placental growth factor. J Immunol. 2014;192:3003–3010. doi: 10.4049/jimmunol.1302177.
    1. Huh YH, et al. HIF-2alpha-induced chemokines stimulate motility of fibroblast-like synoviocytes and chondrocytes into the cartilage-pannus interface in experimental rheumatoid arthritis mouse models. Arthritis Res Ther. 2015;17:302. doi: 10.1186/s13075-015-0816-x.
    1. Sahebkar A, Chew GT, Watts GF. New peroxisome proliferator-activated receptor agonists: potential treatments for atherogenic dyslipidemia and non-alcoholic fatty liver disease. Expert Opin Pharmacother. 2014;15:493–503. doi: 10.1517/14656566.2014.876992.
    1. Zhang JG, et al. MiR-148b suppresses cell proliferation and invasion in hepatocellular carcinoma by targeting WNT1/beta-catenin pathway. Sci Rep. 2015;5:8087. doi: 10.1038/srep08087.
    1. Bae SY, Kim HJ, Lee KJ, Lee K. Translationally controlled tumor protein induces epithelial to mesenchymal transition and promotes cell migration, invasion and metastasis. Sci Rep. 2015;5:8061. doi: 10.1038/srep08061.
    1. Bao X, et al. The roles of endogenous reactive oxygen species and nitric oxide in triptolide-induced apoptotic cell death in macrophages. J Mol Med (Berl) 2007;85:85–98. doi: 10.1007/s00109-006-0113-x.
    1. Gu J, Hu W, Liu X. Pioglitazone improves potassium channel remodeling induced by angiotensin II in atrial myocytes. Med Sci Monit Basic Res. 2014;20:153–160.
    1. Saha L, Bhandari S, Bhatia A, Banerjee D, Chakrabarti A. Anti-kindling Effect of Bezafibrate, a Peroxisome Proliferator-activated Receptors Alpha Agonist, in Pentylenetetrazole Induced Kindling Seizure Model. J Epilepsy Res. 2014;4:45–54. doi: 10.14581/jer.14011.
    1. Laghezza A, et al. On the metabolically active form of metaglidasen: improved synthesis and investigation of its peculiar activity on peroxisome proliferator-activated receptors and skeletal muscles. ChemMedChem. 2015;10:555–565. doi: 10.1002/cmdc.201402462.

Source: PubMed

3
Sottoscrivi