scholarly journals KDM4B Overexpression Promotes the Growth, Migration, and Invasion of Rheumatoid Arthritis Fibroblast-Like Synoviocytes by Activating STAT3 Pathway

Author(s):  
Xin Zhang ◽  
He Nan ◽  
Jialong Guo ◽  
Jinyu Liu

AbstractIn rheumatoid arthritis (RA), fibroblast-like synoviocytes (FLS) present a unique aggressive phenotype and have a passive response to the inflammatory microenvironment, which are critical for the disease’s progression. KDM4B, as a histone demethylase, functions as an oncogenic factor in many cancers and is implicated in osteoclastogenesis as well as pro-inflammatory cytokine release in inflammatory diseases. However, the effects of KDM4B on RA FLS have not been reported. To investigate this issue, our study determined the expression of KDM4B in RA FLS using RT-qPCR and western blot. The effects of KDM4B on RA FLS viability, apoptosis, migration, and invasion were detected by MTT, flow cytometry, transwell migration, and invasion assays. Furthermore, the interaction of KDM4B with STAT3 signaling was studied by western blot, MTT, flow cytometry, transwell migration, and invasion assays. The experimental results showed that KDM4B expression was upregulated in RA synovial tissues and FLS as compared to healthy control tissues and normal FLS. Knockdown of KDM4B obviously suppressed RA FLS viability, migration and invasion, and induced apoptosis. In addition, knockdown of KDM4B in RA FLS decreased the expression of p-STAT3 and MMP-9 but increased cleaved caspase-3 expression compared with the control group. Moreover, KDM4B overexpression could promote cell growth, migration and invasion, and suppress apoptosis in RA FLS by activating STAT3 signaling. Therefore, these findings provide new insight for understanding the pathogenesis of RA and indicate that KDM4B may have a potential to be an effective therapeutic target for RA.

2020 ◽  
Author(s):  
Jia Liu ◽  
Qing Zhang ◽  
Ruo-Lan Li ◽  
Shu-Jun Wei ◽  
Yong-Xiang Gao ◽  
...  

Abstract Background: Rheumatoid arthritis (RA) is a complex chronic inflammatory disease that is associated with the aberrant activation of fibroblast-like synoviocytes (FLS). The extract of Cinnamomi ramulus has been reported to exert alleviates pain, anti-tumor and anti-inflammatory effects. The present study was designed to investigate the effects of Cinnamomi ramulus on RA and explore the underlying mechanisms. Material/methods: TNF-α induced human synoviocyte MH7A cells was performed to evaluate the anti-proliferative and anti-migration effect of Cinnamomi ramulus. The anti-proliferative effect of Cinnamomi ramulus was determined by CCK-8 assay and colony formation assay. Apoptosis was measured by AnnexinV FITC/PI staining and flow cytometry. Cell cycle was evaluated by flow cytometry. The expressions of mitochondrial apoptosis and cell cycle-related molecules, including Bcl-2, Bax, C-Caspase-3, CDC2 and Cycylin B1 were determined by Western blotting. Furthermore, the migration and invasion abilities of MH7A cells were determined using scratch wound healing assay and transwell assay. mRNA expressions of (MMP)-1, -2, & -3, P53, P21 and Cyclin D were determined using qRT-PCR analysis. For qualitative analysis on its chemical components, an ultra-high performance liquid chromatography (UPLC) coupled with Q-Exactive MS (QE-MS) was established for rapid separation and structural identification of the constituents in Cinnamomi ramulus. The further computationally study on the relationships between the 9 compounds and the potential target proteins of RA were carried out with molecular docking strategy. Results: Our data demonstrate that Cinnamomi ramulus inhibited proliferation of MH7A cells, induced cell apoptosis, blocked the cell cycle in the G2/M phase and regulated the protein expression of Bcl-2, Bax, C-Caspase-3, CDCD2 and Cyclin B1. Moreover, Cinnamomi ramulus was proven to significantly inhabited migration and invasion of MH7A cells through inhibition of levels of matrix metalloproteinase (MMP)-1, -2, & -3.Cinnamomi ramulus reduced mRNA levels of CDK4 whereas increased the expression of P53, P21 and CyclinD, implying its regulation effects on apoptosis and cell cycle distribution in MH7A cells. The chromatographic profiling of the extract by UPLC-QE-MS/MS analysis showed 9 compounds are the main components. And the molecular docking strategy results showed that the compounds in Cinnamomi ramulus have good affinity with protein crystal, and benzyl cinnamate may be the main active component of Cinnamomi ramulus to induce cell apoptosis and cycle resistance. Conclusions: Cinnamomi ramulus exhibits anti-proliferative and anti-migration effects on MH7A rheumatoid arthritis-derived fibroblast-like synoviocytes through induction of apoptosis & cell arrest and suppression of matrix metalloproteinase.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Ying-Jie Chen ◽  
Yu-Xi Liu ◽  
Jia-Ying Wu ◽  
Chun-Yu Li ◽  
Min-Min Tang ◽  
...  

AbstractFibroblast-like synoviocytes (FLS) play a pathogenic role in rheumatoid arthritis (RA). STAT3 signaling is activated in FLS of RA patients (RA-FLS), which in turn causes RA-FLS hyperproliferation. RL is a traditional remedy for treating inflammatory diseases in China. It comprises Rosae Multiflorae Fructus and Lonicerae Japonicae Flos. A standardized ethanolic extract of RL (RLE) has been shown to exert anti-arthritic effects in collagen-induced arthritis (CIA) rats. Some constituents of RLE were reported to inhibit JAK2/STAT3 signaling in rat FLS. Here, we determined whether RLE inhibits FLS hyperproliferation, and explored the involvement of STAT3 signaling in this inhibition. In joints of CIA rats, RLE increased apoptotic FLS. In IL-6/sIL-6R-stimulated RA-FLS, RLE reduced cell viability and evoked cell apoptosis. In synovial tissues of CIA rats, RLE lowered the protein level of phospho-STAT3. In IL-6/sIL-6R-stimulated RA-FLS, RLE inhibited activation/phosphorylation of STAT3 and JAK2, decreased the nuclear localization of STAT3, and downregulated protein levels of Bcl-2 and Mcl-1. Over-activation of STAT3 diminished RLE’s anti-proliferative effects in IL-6/sIL-6R-stimulated RA-FLS. In summary, RLE inhibits hyperproliferation of FLS in rat and cell models, and suppression of STAT3 signaling contributes to the underlying mechanisms. This study provides further pharmacological groundwork for developing RLE as a modern anti-arthritic drug.


2021 ◽  
Vol 16 (1) ◽  
Author(s):  
Xia Li ◽  
Meiting Qu ◽  
Jie Zhang ◽  
Kuanyin Chen ◽  
Xianghui Ma

Abstract Background Previous study showed that circular RNA Absent-Small-Homeotic-2--Like protein (circASH2L) was higher in rheumatoid arthritis (RA) patients. However, the roles and mechanisms of circASH2L in RA progression remain unclear. Methods Levels analysis was conducted using western blot and qRT-PCR. The proliferation, apoptosis, cell cycle progression, migration, invasiveness, and inflammation of RA fibroblast-like synoviocytes (RA-FLSs) were determined via MTT, flow cytometry, western blot, transwell, and ELISA assays. Results CircASH2L knockdown in RA-FLSs suppressed cell proliferative, migratory, and invasive capacities, triggered cell cycle arrest, promoted apoptosis, and inhibited inflammation. Mechanistically, circASH2L targeted miR-129-5p, and repression of miR-129-5p abolished the functions of circASH2L silencing on the growth, motility, and inflammation of RA-FLSs. Besides, miR-129-5p was found to directly target HIPK2, and suppressed the tumor-like biologic behaviors and inflammation of RA-FLSs via regulating HIPK2. Importantly, we proved that circASH2L could modulate HIPK2 expression via miR-129-5p. Conclusion CircASH2L promoted RA-FLS growth, motility, and inflammation through miR-129-5p/HIPK2 axis.


2021 ◽  
Vol 16 (1) ◽  
Author(s):  
Zhihuan Luo ◽  
Shaojian Chen ◽  
Xiaguang Chen

Abstract Background Rheumatoid arthritis (RA) is a chronic inflammatory joint disease, and fibroblast-like synoviocytes (FLSs) are key effector cells in RA development. Mounting evidence indicates that circular RNAs (circRNAs) participate in the occurrence and development of RA. However, the precise mechanism of circRNA mitogen-activated protein kinase (circMAPK9) in the cell processes of FLSs has not been reported. Methods The expression levels of circMAPK9, microRNA-140-3p (miR-140-3p), and protein phosphatase magnesium-dependent 1A (PPM1A) were determined by quantitative real-time polymerase chain reaction (qRT-PCR) or western blot assay. Cell proliferation was examined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cell apoptosis and cycle distribution were assessed by flow cytometry. Cell migration and invasion were tested by transwell assay. All the proteins were inspected by western blot assay. Inflammatory response was evaluated by enzyme-linked immunosorbent assay (ELISA). The interaction between miR-140-3p and circMAPK9 or PPM1A was verified by dual-luciferase reporter assay. Results CircMAPK9 and PPM1A were upregulated and miR-140-3p was downregulated in RA patients and FLSs from RA patients (RA-FLSs). CircMAPK9 silence suppressed cell proliferation, migration, invasion, inflammatory response, and promoted apoptosis in RA-FLSs. MiR-140-3p was a target of circMAPK9, and miR-140-3p downregulation attenuated the effects of circMAPK9 knockdown on cell progression and inflammatory response in RA-FLSs. PPM1A was targeted by miR-140-3p, and circMAPK9 could regulate PPM1A expression by sponging miR-140-3p. Furthermore, miR-140-3p could impede cell biological behaviors in RA-FLSs via targeting PPM1A. Conclusion CircMAPK9 knockdown might inhibit cell proliferation, migration, invasion, inflammatory response, and facilitate apoptosis in RA-FLSs via regulating miR-140-3p/PPM1A axis, offering a new mechanism for the comprehension of RA development and a new insight into the potential application of circMAPK9 in RA treatment.


2020 ◽  
Vol 79 (Suppl 1) ◽  
pp. 1097.1-1097
Author(s):  
F. Zhu ◽  
X. Zhang

Background:Connective tissue disease-associated interstitial lung disease (CTD-ILD) is a class of refractory diseases.Non-specific treatment with hormone and immunosuppressive agents is mostly used at present, but the effect is limited and the long-term survival rate is not improved [1],while anti-fibrosis treatments (such as Pirfenidone and Nintedanib) have only recently been approved, the long-term efficacy is still unknown.Tofacitinib(TOFA), a JAK inhibitor, has recently been used to treat patients with severe dermatomyositis related interstitial pulmonary disease, with significantly improved survival rate [2-4].A basic study showed that TOFA improved interstitial pulmonary disease in mice by promoting the proliferation of myelogenic inhibitory cells [5].However, whether TOFA can affect the migration and invasion of human lung fibroblasts and further research to reveal the mechanism of its inhibition of pulmonary fibrosis has not been reported.Objectives:To investigate the anti - fibrosis effect of TOFA in CTD-ILD.Methods:Cell migration and invasion AssaysHLFs were incubated with TOFA for 72h, followed by TGF- β1 for 24h.DMEM serum-free medium was used to determine the cell density to 5. 0 × 107/L, 600 uL medium containing 10% fetal bovine serum was added to the lower compartment of Transwell chamber, and 200 uL cell suspension was added to the upper compartment.Incubate in incubator for 12 h.After fixation, staining and sealing, the cells were observed and counted under a microscope. At least 5 random field transmembrane cells were counted in each hole, and the mean value was taken.For the invasion assays, Transwell chamber coated with matrigel was used, and the cell incubation time was 16 h.Results:1. Effect of TOFA on HLFs migration function (Figure 1)Figure 1.Effect of TOFA on HLFs migration function(×200).Mean ± SEM. n = 5.The number of cells passing through the biofilm in the three groups was counted.It can be seen that TGF-β1 group significantly increased compared with control group (*P < 0.0001), and TOFA group significantly decreased compared with TGF- β1 group (#P < 0.0001), suggesting that TOFA can significantly inhibit TGF-β1- induced HLFs migration.2. Effect of TOFA on HLFs invasion function (Figure 2)Figure 2.Effect of TOFA on HLFs invasion function(×200).Mean ± SEM. n = 5.The number of cells passing through the matrigel in the three groups was counted.It can be seen that TGF-β1 group was significantly higher than the control group (*P < 0.0001), and TOFA group was significantly lower than TGF-β1 group(#P < 0.001), suggesting that TOFA can significantly inhibit the invasion function of HLFs induced by TGF-β1.Conclusion:TOFA can effectively inhibit the function of HLFs migration and invasion. Although further studies are needed to elucidate the mechanism by which TOFA inhibit the function of HLFs migration and invasion, our study suggests that TOFA has a potential therapeutic effect for CTD-ILD.References:[1]Aparicio, I.J. and J.S. Lee, Connective Tissue Disease-Associated Interstitial Lung Diseases: Unresolved Issues. Semin Respir Crit Care Med, 2016. 37(3): p. 468-76.[2]Kato, M., et al., Successful Treatment for Refractory Interstitial Lung Disease and Pneumomediastinum With Multidisciplinary Therapy Including Tofacitinib in a Patient With Anti-MDA5 Antibody-Positive Dermatomyositis. J Clin Rheumatol, 2019.[3]Kurasawa, K., et al., Tofacitinib for refractory interstitial lung diseases in anti-melanoma differentiation-associated 5 gene antibody-positive dermatomyositis. Rheumatology (Oxford), 2018. 57(12): p. 2114-2119.[4]Chen, Z., X. Wang, and S. Ye, Tofacitinib in Amyopathic Dermatomyositis-Associated Interstitial Lung Disease. N Engl J Med, 2019. 381(3): p. 291-293.[5]Sendo, S., et al., Tofacitinib facilitates the expansion of myeloid-derived suppressor cells and ameliorates interstitial lung disease in SKG mice. Arthritis Res Ther, 2019. 21(1): p. 184Disclosure of Interests:None declared


2020 ◽  
Author(s):  
fujuan qiu ◽  
Chen Yong ◽  
Qiu Fujuan ◽  
Zhao Xiaofeng ◽  
Xiao Changhong

Abstract Background To determine whether any differences of AIM2 inflammasome expression levels between rheumatoid arthritis (RA) and osteoarthritis (OA) and investigate the effects of AIM2 when transferred into RA fibroblast-like synoviocytes (RA-FLS).Methods Serum AIM2 levels between OA and RA patients were compared by ELISA. Different expression levels of AIM2, ASC, Caspase-1 and IL-1β between RA and OA synovium were semi-quantified by RT-qPCR and immunohistochemical (IHC) staining. IHC staining were recorded by H scores, and determine the correlation with ESR and CRP levels of RA patients. SiRNA AIM2 was transferred to RA-FLS and observe its effects on proliferation and migration by MTT assay and transwell test respectively.Results In RA sera, no significant difference was observed between OA and RA patients. However, in affected knee synovium, AIM2, ASC, Caspase-1 and IL-1β were expressed higher in RA than that of OA. Plus, H score of AIM2, ASC, and IL-1β were positively correlated to ESR and CRP levels in RA patients. After transferred AIM2 siRNA to FLS and incubation for 48 hours, the proliferation of FLS were significantly inhibited, and the apoptosis rate were significantly increased compared to FLS in control group. However, no effect on migration was detected.Conclusions AIM2 participated in the proliferation of FLS, and might be a potential target for therapy.


2018 ◽  
Vol 2018 ◽  
pp. 1-7 ◽  
Author(s):  
Gyu-Un Jung ◽  
Ji-Young Han ◽  
Kyung-Gyun Hwang ◽  
Chang-Joo Park ◽  
Panagiota G. Stathopoulou ◽  
...  

Rheumatoid arthritis (RA) and periodontitis are common chronic inflammatory diseases and periodontitis is known to be more common and more severe in patients with RA. Based on a paucity of studies about the relationship between common conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) and periodontitis, this prospective study aimed to evaluate the adjunctive effect of csDMARDs on response to nonsurgical periodontal treatment in patients with RA. Thirty-two patients with RA (RA group) and 32 systemically healthy patients (control group) with periodontitis were included in this study. The RA group patients were treated with csDMARDs, such as methotrexate, hydroxychloroquine, and sulfasalazine. Conventional nonsurgical periodontal treatment with scaling and root planing was performed in both groups. The extent and severity of periodontitis were evaluated by plaque index (PI), gingival index (GI), probing depth (PD), clinical attachment level (CAL), and bleeding on probing (BOP) at baseline and 4 weeks after periodontal treatment. There was no statistically significant difference of periodontal parameters between the RA and control groups at baseline. Four weeks after scaling and root planing, PD reduction, and CAL gain were higher in the RA group treated with csDMARDs compared to the control group, and the difference was statistically significant (P = 0.006 and 0.003, respectively). A post hoc analysis of the RA group showed no statistically significant difference on the response to nonsurgical periodontal treatment in multiple csDMARDs therapy and addition of NSAIDs and/or steroids to csDMARDs. In patients with RA, csDMARDs showed beneficial effect on periodontal clinical parameters following the nonsurgical periodontal treatment.


2021 ◽  
Vol 11 (3) ◽  
pp. 407-411
Author(s):  
Shenhua Zhang ◽  
Ting Yu

We investigated the effects of silencing the regulator of ribosome synthesis 1 (RRS1) gene on the proliferation, migration, and invasion of ovarian carcinoma cells, and its possible role in modulating signal transduction in these cells. Normal ovarian epithelial cell line IOSE80 was used as a control. We examined the mRNA and protein level of RRS1 using qRT-PCR and western blot in control and ovarian carcinoma cells (SKOV-3, SW626, and CAOV3). RNA interference technology was used to knockdown RRS1 expression in CAOV3 cells. MTT was used to examine the proliferation of these cells, while a Transwell assay was used to assay the cells’ migration and invasion abilities. Western blot was used to measure the levels of CyclinD1, P21, MMP-2, MMP-9, p-JAK2 and p-STAT3 proteins. In comparison with normal ovarian epithelial cells (IOSE80), RRS1 mRNA and protein levels were increased in ovarian carcinoma cells (SKOV-3, SW626 and CAOV3) (P < 0.05). Because RRS1 levels were highest in CAOV3 cells, these cells were used for subsequent experiments. RRS1 gene expression was knocked down in CAOV3 cells, and in comparison with the negative control group, siRNA-RRS1 cells exhibited decreased proliferation in the MTT assay after 48 h and 72 h (P < 0.05). These cells also exhibited reduced migration and invasion (P < 0.05). Further, siRNA-RRS1 cells exhibited reduced expression of CyclinD1, MMP-2, MMP-9, P-JAK2 and P-STAT3 proteins (P < 0.05), while P21 protein levels were increased (P < 0.05). Silencing RRS1 expression inhibits the proliferation, migration, and invasion of ovarian carcinoma cells. This effect may be mediated by the inhibition of the STAT3 signaling pathway in these cells.


Sign in / Sign up

Export Citation Format

Share Document