Open Access

Expression profiling of genes in rheumatoid fibroblast‑like synoviocytes regulated by Fas ligand via cDNA microarray analysis

  • Authors:
    • Koji Fukuda
    • Yasushi Miura
    • Toshihisa Maeda
    • Shinya Hayashi
    • Tomoyuki Matsumoto
    • Ryosuke Kuroda
  • View Affiliations

  • Published online on: July 15, 2021     https://doi.org/10.3892/etm.2021.10432
  • Article Number: 1000
  • Copyright: © Fukuda et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Rheumatoid arthritis (RA) is an autoimmune disease that causes chronic inflammation in synovial tissues. Hyperplasia of synovial tissues leads to the formation of pannus that invades the joint cartilage and bone, resulting in joint destruction. Fas ligand (FasL), which is a member of the tumor necrosis factor superfamily, contributes to the pathogenesis of autoimmune diseases, including RA. The current study attempted to identify genes whose expressions in rheumatoid fibroblast‑like synoviocytes (RA‑FLS) were regulated by FasL, using cDNA microarray. A total of four individual lines of primary cultured RA‑FLS were incubated either with recombinant human FasL protein or PBS as an unstimulated control for 12 h. Gene expression was detected using a microarray assay. The results revealed the expression profiles of genes in RA‑FLS regulated by Fas and investigated the functions of the genes that were regulated. Among the genes in this profile, the mRNA expression changes of the following genes were indicated to be of note using RT‑qPCR: Dual specificity phosphatase 6, epiregulin, interleukin 11, angiopoietin‑like 7, protein inhibitor of activated STAT 2 and growth differentiation factor 5. These genes may affect the pathogenesis of RA by affecting apoptosis, proliferation, cytokine production, cytokine‑induced inflammation, intracellular signaling, angiogenesis, bone destruction and chondrogenesis. To the best of our knowledge, the current study is the first study to reveal the expression profile of genes in RA‑FLS regulated by FasL. The data demonstrated that FasL may regulate the expression of a number of key molecules in RA‑FLS, thus affecting RA pathogenesis. Further studies of the genes detected may improve the understanding of RA pathogenesis and provide novel treatment targets for RA.

Introduction

Rheumatoid arthritis (RA) is an autoimmune disease that causes chronic inflammation in synovial tissues. Hyperplasia of synovial tissues leads to the formation of pannus, which invades joint cartilage and bone, resulting in joint destruction. Previous reports have indicated that a number of features of transformed long-lived cells are observed in hyperplastic synovial tissues of patients with RA, such as oncogene expression, resistance to apoptosis, and the presence of somatic mutations (1-3). Several explanations for resistance to apoptosis of rheumatoid fibroblast-like synoviocytes (RA-FLS) have been proposed, including deregulation of the Bcl-2 family of proteins critical to the intrinsic apoptosis pathway, deregulation of NF-κB signaling, p53 mutations, and low expression of PUMA; these are all found in RA synovium and FLS, which provides an explanation for the lack of p53-induced FLS apoptosis (4). In addition, it has been reported that hyperproliferation of RA synovial cells involves the abnormal function of death receptors such as Fas and death receptor 3 (5,6).

Fas ligand (FasL)/TNFSF6, a member of the tumor necrosis factor (TNF) superfamily, is expressed by various cell types in arthritic synovium, including T cells, synovial fibroblasts, and macrophages (7), and can promote apoptosis in activated primary B cells, T cells, dendritic cells, and synovial fibroblasts through Fas (8,9). Inhibition of the Fas/FasL pathway contributes to synovial hyperplasia of RA (10-12). Apoptosis through the Fas/FasL pathway in RA synovial cells is inhibited by pro-inflammatory cytokines present within the synovium (8). Meanwhile, the Fas/FasL system may have a pro-inflammatory effect in RA (13,14). Audo et al demonstrated that membrane-bound FasL induces apoptosis as well as proliferation, whereas soluble FasL stimulates only proliferation (13). Moreover, soluble FasL activates several signaling pathways in RA-FLS, such as extracellular signal-regulated kinase (ERK)-1/2, phosphatidyl-inositol 3- kinase, caspase 8, and c-jun N-terminal kinase (13). However, the mechanisms and cell targets for these effects are still poorly understood.

Decoy receptor 3 (DcR3)/TR6/M68/TNFRSF6b, a member of the TNF receptor superfamily, binds to 3 ligands belonging to the TNF superfamily: FasL, LIGHT, and TL1A (15). Overexpression of DcR3 may benefit tumors by helping them avoid the cytotoxic and regulatory effects of FasL (16,17), LIGHT (18), and TL1A (19). In our previous studies, we demonstrated that DcR3 overexpressed in RA-FLS and stimulated by TNFα protects cells from Fas-induced apoptosis (20). We previously also reported that DcR3 could play a role as a ligand by binding to membrane-bound TL1A in the pathogenesis of RA (21-24).

Furthermore, the expression profiles of genes regulated by DcR3 and TL1A in RA-FLS have been revealed by the use of cDNA microarrays in our previous studies (25,26), suggesting that signaling through DcR3 and its ligands is involved in the pathogenesis of RA. However, the contribution of FasL, another ligand of DcR3, to the pathogenesis of RA remains to be fully elucidated.

In the current study, we searched for genes whose expressions in RA-FLS were regulated by FasL using a cDNA microarray. The gene expression profiles revealed a series of genes that may play a significant role in the pathogenesis of RA via the FasL-Fas signaling pathway. Further study is needed to reveal the difference of the gene expression profiles among the ligands, which might result in better understanding the role of the FasL/TL1A/DcR3 signaling system in the pathogenesis of RA.

Materials and methods

Isolation and culture of synovial fibroblasts

RA-FLS were obtained from ten patients (samples 1-10) with RA who fulfilled the 1987 criteria of the American College of Rheumatology (formerly, the American Rheumatism Association) (27) during total knee replacement surgery from September 2014 to April 2019. Patients included one male and nine females aged 70.4±8.5 years old. Their C-reactive protein levels and erythrocyte sedimentation rates were 1.4±2.6 mg/dl and 25.6±14.0 mm/h, respectively. As for the drug therapy for RA, five patients were administered oral methotrexate (MTX) (average MTX dose, 8.8±4.6 mg/week), two were administered tacrolimus (1.5±0.5 g/day), two were administered salazosulfapyridine (1.0±0.0 g/day), and two were administered bucillamine (150.0±50.0 mg/day). Prednisolone (PSL) was used to treat three patients (average PSL dose, 4.7±0.6 mg/day). None of the patients had been treated with biological disease-modifying anti-rheumatic drugs (bioDMARDs) or Janus kinase inhibitors.

Synovial samples were collected from the patients, all of whom provided informed written consent to participate in this study in accordance with the World Medical Association Declaration of Helsinki Ethical Principles for Medical Research Involving Human Subjects. The protocol, including consent procedures, was approved by the Kobe University Graduate School of Health Sciences Ethics Committee (approval no. 308). Tissue specimens were minced and digested in Dulbecco's modified Eagle's medium (DMEM; Merck KGaA) containing 0.2% collagenase (Merck KGaA) for 2 h at 37˚C with 5% CO2. Dissociated cells were cultured in DMEM supplemented with 10% fetal bovine serum (Merck KGaA) and 100 U/ml of penicillin/streptomycin (Meiji Seika Pharma Co., Ltd.). Following incubation overnight and the removal of non-adherent cells, adherent cells were further incubated in fresh medium. All experiments were conducted using cells from passages 3 to 4(20).

Gene expression profiling

Four individual cell lines (samples 1-4) of primary cultured RA-FLS (2x106 cells/well) were incubated with 1,000 ng/ml of recombinant human FasL protein (R&D Systems) or were left untreated with OPTI-MEM medium (Thermo Fisher Scientific, Inc.) as control for 12 h at 37˚C with 5% CO2. The concentration of FasL was determined by a preliminary experiment based of the previous reports using FasL (28,29). After incubation, RNA was extracted with QIAshredder (Qiagen GmbH) and an RNeasy Mini kit (Qiagen GmbH) according to the manufacturer's protocol. Extraction of total RNA was performed for each sample separately.

Gene expressions were detected by a microarray assay (Human Genome U133 Plus 2.0, GeneChip® 3' Expression Array; Thermo Fisher Scientific, Inc.). The labeling of RNA probes, hybridization, and washing were carried out according to the manufacturer's protocol.

RT-qPCR analysis for mRNA expression of genes regulated by FasL

RA-FLS (samples 5-10) were cultured in six-well plates at a density of 2x106 cells/well with 1,000 ng/ml of FasL or serum-free medium only as a control. RNA was extracted using the QIAshredder and RNeasy mini kits according to the manufacturer's protocols. Oligo (dT)-primed first-strand complementary DNA (cDNA) was synthesized (2 µg total RNA) using a High Capacity cDNA Transcription kit (Applied Biosystems; Thermo Fisher Scientific). Relative expression levels of mRNA encoding DUSP6, EREG, IL-11, ANGPTL7, PIAS2, and GDF5 were compared using TaqMan® real-time PCR on a StepOne™ real-time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). Pre-designed primers and probes for DUSP6 (Hs04329643_s1), EREG (Hs00914313_m1), IL-11 (Hs01055413_g1), ANGPTL7 (Hs00221727_m1), PIAS2 (Hs00915227_m1), GDF5 (Hs00167060_m1), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH; Hs99999905_m1) were obtained from Applied Biosystems (Thermo Fisher Scientific, Inc.). Comparative analysis of each of these genes in individual patients was performed using StepOne™ 2.1 software (Applied Biosystems; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. All amplifications were conducted in duplicate. The mRNA expression levels of each gene were calculated using the comparative threshold cycle (ΔΔCq) method as previously described (30).

Statistical analysis

Values are expressed as the mean ± standard deviation unless otherwise indicated. As for the data analysis of the microarray assay, Avadis 3.3 Prophetic software (Strand Life Sciences) was used for statistical analysis (31). Differentially expressed genes were extracted by a paired t-test, with P values <0.05 considered to indicate statistical significance and fold-change >2.0, and ordered into hierarchical clusters using the Euclidean algorithm as the distance measure and the complete algorithm as the linkage method.

The data analysis of the RT-qPCR assay was as follows. The Wilcoxon signed ranked test was used to evaluate the differences between mRNA expression levels of genes in the control group and FasL-stimulated group. Statistical analyses were conducted using Statcel (version 3; OMS Publishing, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Microarray analysis (gene expression profiling of RA-FLS stimulated by FasL)

The microarray analysis used in the current study (Human Genome U133 Plus 2.0, GeneChip® 3' Expression Array) was able to detect the expression of 27,420 genes.

The microarray analysis revealed that FasL upregulates or downregulates the expressions of various genes in RA-FLS. We used the NCBI's UniGene database (https://www.ncbi.nlm.nih.gov/UniGene/clust.cgi?ORG=Hs&CID=55682) to identify the genes. Among the 1039 genes differentially upregulated by FasL, 806 were annotated in the database. Twenty of the 806 genes upregulated by FasL are shown in Table I. Gene annotations of 1190 among the 1518 genes differentially downregulated by FasL were also in the database. Twenty of the 1190 downregulated genes by FasL are shown in Table II. Hierarchical clustering analysis was performed for genes for which expression changes were detected in at least 2 of the 4 samples, which was 247 genes. The results of hierarchical clustering analysis for these 247 genes are illustrated in Fig. 1.

Table I

The 20 genes most upregulated by FasL. P-values were detected by a paired t-test.

Table I

The 20 genes most upregulated by FasL. P-values were detected by a paired t-test.

Gene abbreviationP-valueFC (abs)Gene name
DUSP60.00001834.61Dual specificity phosphatase 6
EREG0.01962229.23Epiregulin
IL-110.00727525.28Interleukin 11
ANGPTL40.00285323.50Angiopoietin-like 4
SLCO4A10.00209420.39Solute carrier organic anion transporter family, member 4A1
TNFSF110.00623618.48Tumor necrosis factor (ligand) superfamily, member 11
BDKRB10.00000414.39Bradykinin receptor B1
OTTHUMG00000172357//RP11-475A13.20.00000214.12NULL//NULL
AREG//AREGB0.03053713.77 Amphiregulin//amphiregulin B
LIF0.00049813.53Leukemia inhibitory factor
IFNA80.00003912.09Interferon, α 8
OTTHUMG00000175763//RP11-744D14.20.00071611.73NULL//NULL
HBEGF0.01477311.34Heparin-binding EGF-like growth factor
PPP4R40.02373911.03Protein phosphatase 4, regulatory subunit 4
NDP0.00883210.67Norrie disease (pseudoglioma)
NR4A30.00043210.59Nuclear receptor subfamily 4, group A, member 3
EGLN30.0284589.95Egl nine homolog 3 (C. elegans)
BMP20.0000829.92Bone morphogenetic protein 2
UBR20.0078479.91Ubiquitin protein ligase E3 component n-recognin 2
SLC38A100.0014009.34Solute carrier family 38, member 10

[i] FasL, Fas ligand.

Table II

The 20 genes most downregulated by FasL. P-values were detected by a paired t-test.

Table II

The 20 genes most downregulated by FasL. P-values were detected by a paired t-test.

Gene abbreviationP-valueFC (abs)Gene name
ANGPTL70.00028311.61Angiopoietin-like 7
PIAS20.00109911.34Protein inhibitor of activated STAT, 2
LINC003100.00003811.30Long intergenic non-protein coding RNA 310
GDF50.00426011.12Growth differentiation factor 5
TBX220.00075511.11T-box 22
DCAF4L10.00073410.64DDB1 and CUL4 associated factor 4-like 1
KRT160.01333110.62Keratin 16
OTTHUMG00000180314//RP1-193H18.20.00372610.31NULL//NULL
TAS2R400.00020210.14Taste receptor, type 2, member 40
HEPACAM20.0016569.93HEPACAM family member 2
CSMD10.0000969.87CUB and Sushi multiple domains 1
IQCA10.0096929.63IQ motif containing with AAA domain 1
LOC100996810//LOC2838610.0031829.26Uncharacterized LOC100996810//uncharacterized LOC283861
FGFR20.0296999.25Fibroblast growth factor receptor 2
WDR650.0000459.21WD repeat domain 65
LOC2535730.0015569.18Uncharacterized LOC253573
PHLDB20.0010309.06Pleckstrin homology-like domain, family B, member 2
PCDHAC20.0177119.01Protocadherin alpha subfamily C, 2
LOC1005066290.0029368.72Uncharacterized LOC100506629
FAM66C0.0036478.68Family with sequence similarity 66, member C

[i] FasL, Fas ligand.

Functional annotation

The 246 genes regulated by FasL were classified into categories registered in the David Bioinformatics Database (https://david.ncifcrf.gov/) according to their biological functions. The most significant 10 functional categories were as follows: Transcriptional activator activity, positive regulation of metabolic process, positive regulation of cellular metabolic process, positive regulation of macromolecule metabolic process, positive regulation of nitrogen compound metabolic process, regulation of phosphorylation, positive regulation of biological process, regulation of phosphate metabolic process, regulation of MAPK cascade, regulation of multicellular organismal process (Table III).

Table III

The 10 most significant functional categories of the 246 genes most differentially expressed by FasL exposure in RA-FLS. P-values were detected by a paired t-test.

Table III

The 10 most significant functional categories of the 246 genes most differentially expressed by FasL exposure in RA-FLS. P-values were detected by a paired t-test.

GO AccessionGO TermCorrected P-value
GO:0001228Transcriptional activator activity, RNA polymerase II transcription Regulatory region sequence-specific DNA binding0.000028
GO:0009893| GO:0044253Positive regulation of metabolic process0.000028
GO:0031325Positive regulation of cellular metabolic process0.000028
GO:0010604Positive regulation of macromolecule metabolic process0.000028
GO:0051173Positive regulation of nitrogen compound metabolic process0.000028
GO:0042325Regulation of phosphorylation0.000066
GO:0048518| GO:0043119Positive regulation of biological process0.000066
GO:0019220Regulation of phosphate metabolic process0.000087
GO:0043408Regulation of MAPK cascade0.000087
GO:0051239Regulation of multicellular organismal process0.000087

[i] GO, gene ontology; FasL, Fas ligand; RA, rheumatoid arthritis.

mRNA expression detected by RT-qPCR

Based on the microarray assay, we confirmed the mRNA expressions of genes by real-time PCR. Fig. 2 shows the mRNA expression levels of the 3 most upregulated genes. DUSP6 was upregulated 21 times by FasL compared to the control, EREG was upregulated 24 times by FasL compared to the control, and IL-11 was upregulated 9 times by FasL compared to the control. Fig. 3 shows the mRNA expression levels of the 3 most downregulated genes. ANGPTL7 was downregulated 0.15 times by FasL compared to the control, PIAS2 was downregulated 0.58 times by FasL compared to the control, and GDF5 was downregulated 0.11 times by FasL compared to the control.

Discussion

Genome-wide gene expression cDNA microarrays provide a powerful technique to investigate the pathophysiology of a variety of diseases, including tumors (32-34), immune-mediated diseases (35,36), and inflammatory diseases (37-39). Using microarray assays, we previously revealed the expression profiles of genes in RA-FLS regulated by DcR3(25) and TL1A (26). Subsequently, based on the profile regulated by DcR3, we investigated the significance of IL-12B p40(22), tryptophan hydroxylase 1(24), and centrosomal protein 70 kDa (23) as regulated by DcR3 in RA-FLS in detail. The profile regulated by TL1A included the following noteworthy genes: Spectrin repeat-containing nuclear envelope 1, Fc receptor-like 2, PYD (pyrin domain)-containing 1, cell division cycle 45 homolog, signal transducer and activator of transcription 5B, and interferon regulatory factor 4(26).

To the best of our knowledge, this is the first study to reveal the expression profiles of genes in RA-FLS regulated by FasL. Among the genes in this profile, the following genes were of note: Dual specificity phosphatase 6 (DUSP6), epiregulin (EREG), interleukin 11 (IL-11), angiopoietin-like 7 (ANGPTL7), protein inhibitor of activated STAT 2 (PIAS2), and growth differentiation factor 5 (GDF5); these genes were all highly regulated by FasL.

DUSP6 regulates CD4+ T-cell activation and differentiation by inhibiting T-cell receptor dependent ERK 1/2 activation (40). It has been reported that DUSP6 promotes endothelial inflammation through the inducible expression of TNF-α-induced intercellular adhesion molecule-1 via nuclear factor-κB, which is independent of ERK signaling (41).

Epiregulin is a growth regulator that belongs to the epidermal growth factor (EGF) family and mediates the dose-dependent increase in proliferation of primary mouse keratinocytes (42). EREG is increased in patients with RA and is associated with the development of IL-6 amplifier activation (43). EREG triggers the temporal regulation of growth factors such as amphiregulin, betacellulin, transforming growth factor (TGF)-α, fibroblast growth factor 2, placental growth factor 2, and tenascin C, contributing to the early phase of inflammation; each growth factor reciprocally regulates EREG in affected tissue during the late phase of inflammatory disease development (44). Secretion of vascular endothelial growth factor-A and EREG from RA-FLS was inhibited upon treatment with the aryl hydrocarbon receptor antagonist GNF351, resulting in attenuation of RA-FLS cell migration, along with cytokine-induced RA-FLS cell proliferation (45).

IL-11 signaling appears to be initiated by the binding of IL-11 to IL-11 receptor α chain (IL-11Rα), which then binds gp130, the signaling unit of the IL-6 cytokine family (46). IL-11 attenuates the inflammatory response through downregulation of proinflammatory cytokine release and nitric oxide production (47,48). IL-11 contributes to RA angiogenesis directly and indirectly. IL-11 promotes endothelial cell migration and tube formation mediated through IL-11Rα ligation. Vascular endothelial growth factor and IL-8 produced from IL-11-treated RA-FLS contribute to the indirect effect of IL-11 on angiogenesis (49). In addition, IL-11 plays a key role in osteoclast formation via the gp130/Jak signaling pathway (50).

ANGPTL7 is a member of angiopoietin family that exerts pro-angiogenic activities on endothelial cells. ANGPTL7 expression has been identified in some cancer cells induced by hypoxia (51). ANGPTL7 induces proinflammatory responses in macrophages, including the induction of immune gene expression, the promotion of proinflammatory cytokine secretion, enhanced phagocytosis, and antagonized anti-inflammatory signaling through the P38 MAPK signaling pathway (52). Down-regulation of a positive regulator of inflammation might have a negative effect for inflammation in patients with RA.

PIAS proteins inhibit activated STAT and play important roles in regulating many important cellular events, such as cell survival, migration, and signal transduction in many cell types (53,54). PIAS proteins also modulate the activity of several transcription factors and act as E3 ubiquitin ligases in the sumoylation pathway (54-57). In a similar fashion, down-regulation of a negative regulator of inflammation might have a positive effect for inflammation in patients with RA. Lao et al reported that PIAS3 regulates migration and invasion through the Rac1/PAK1/JNK pathway in RA-FLSs (53).

GDF5 is a member of the TGF-β superfamily and is most closely related to the bone morphogenetic protein subfamily. GDF5 increases glycosaminoglycan synthesis (58) and cartilage and bone formation (59). GDF5 is present in the synovium membrane and cartilage of patients with RA and is actively involved in the regulation of cartilage maintenance and repair (60). GDF5 is associated with joint destruction in patients with osteoarthritis (61) and RA (62). GDF5 in RA-FLS was suppressed by IL-1β and had a strong chondrogenic-promoting effect on TGF-β3-induced chondrocyte differentiation in RA-FLS (60).

In the present study, at the first brush, we exhaustively investigated and revealed the gene expression profiles regulated by FasL in RA-FLS by the microarray assay. Secondly, we confirmed the universality of the gene expression pattern by a different method, RT-qPCR assay, using the different samples of RA-FLS from those used for the microarray assay. In order to obtain the pathological homogeneity among the samples as much as possible, the patients who underwent similar clinical features were recruited; who had been treated only with conventional DMARDs, not with biological DMARDs or targeted synthetic DMARDs, and who had their knee joint destructed severely resulting total knee replacement surgery. Therefore, we considered that there were no differences among the 10 samples. The samples 1-4 used for the microarray and 5-10 for the RT-qPCR assay were randomly selected.

The expression profiles of genes regulated by TL1A were elucidated by use of a microarray assay in our previous report (26). TL1A and FasL are bound and inhibited by the common decoy receptor, DcR3. Therefore, clarifying the relationship between the expression profiles of genes regulated by FasL and those regulated by TL1A might help us to better understand the role of the FasL/TL1A/DcR3 signaling system in the pathogenesis of RA. Further study is needed to reveal the relationship between these gene expression profiles.

The limitations of the current study include its small sample size and that it presents gene expression data only. The results of the current study revealed a series of genes whose expression is regulated by FasL in RA-FLS with microarray analysis, and the mRNA expression of some genes of note was confirmed by RT-qPCR assay. However, in addition to the expression analysis of each gene, how the genes regulated by FasL in RA-FLS are involved in the pathogenesis of RA also requires further investigation. In the current study, we aimed to analyze exhaustively the gene expression pattern regulated by FasL in RA-FLS. Therefore, the assay for expression of proteins coded by each gene should also be performed in the further studies.

In conclusion, the current study is the first, to the best of our knowledge, to report the expression profile of genes in RA-FLS regulated by FasL. These data demonstrate that FasL may regulate the gene expression of various key molecules in RA-FLS, thus affecting the pathogenesis of RA, including apoptosis, proliferation, cytokine production, cytokine-induced inflammation, intracellular signaling, angiogenesis, bone destruction, and chondrogenesis. FasL may have pleiotropic actions not only protectively but also detrimentally for RA. Further investigation of the genes detected in this profile may provide a deeper understanding of the pathogenesis of RA and new targets for its treatment.

Acknowledgements

The authors would like to thank Ms. Kyoko Tanaka, Ms. Minako Nagata, and Ms. Maya Yasuda (all from Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine) for their technical assistance.

Funding

Funding: The current study was supported by Grants-in-Aid for Scientific Research (KAKENHI) (grant nos. 15K10473 and 18K09106).

Availability of data and materials

The datasets generated and analyzed during the current study are available in the NCBI's Gene Expression Omnibus (GEO) repository, GEO series accession no. GSE153378 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE153378).

Authors' contributions

KF conceived and designed the current study, was involved in data collection and analysis, confirmed the authenticity of all the raw data, wrote and gave final approval of the manuscript. YM conceived and designed the current study, was involved in data collection and analysis, confirmed the authenticity of all the raw data and gave final approval of the manuscript. TosM and TomM collected the data and gave final approval of the manuscript. SH conceived and designed the current study, was involved in data collection and analysis and gave final approval of the manuscript. RK conceived and designed the current study and gave final approval of the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Kobe University Graduate School of Health Sciences Ethics Committee (approval no. 308). All the participants provided written informed consent to participate in the present study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chou CT, Yang JS and Lee MR: Apoptosis in rheumatoid arthritis-expression of Fas, Fas-L, p53, and Bcl-2 in rheumatoid synovial tissues. J Pathol. 193:110–116. 2001.PubMed/NCBI View Article : Google Scholar

2 

Tak PP, Zvaifler NJ, Green DR and Firestein GS: Rheumatoid arthritis and p53: How oxidative stress might alter the course of inflammatory diseases. Immunol Today. 21:78–82. 2000.PubMed/NCBI View Article : Google Scholar

3 

Yamanishi Y, Boyle DL, Rosengren S, Green DR, Zvaifler NJ and Firestein GS: Regional analysis of p53 mutations in rheumatoid arthritis synovium. Proc Natl Acad Sci USA. 99:10025–10030. 2002.PubMed/NCBI View Article : Google Scholar

4 

Bustamante MF, Garcia-Carbonell R, Whisenant KD and Guma M: Fibroblast-like synoviocyte metabolism in the pathogenesis of rheumatoid arthritis. Arthritis Res Ther. 19(110)2017.PubMed/NCBI View Article : Google Scholar

5 

Baier A, Meineckel I, Gay S and Pap T: Apoptosis in rheumatoid arthritis. Curr Opin Rheumatol. 15:274–279. 2003.PubMed/NCBI View Article : Google Scholar

6 

Takami N, Osawa K, Miura Y, Komai K, Taniguchi M, Shiraishi M, Sato K, Iguchi T, Shiozawa K, Hashiramoto A and Shiozawa S: Hypermethylated promoter region of DR3, the death receptor 3 gene, in rheumatoid arthritis synovial cells. Arthritis Rheum. 54:779–787. 2006.PubMed/NCBI View Article : Google Scholar

7 

Peng SL: Fas (CD95)-related apoptosis and rheumatoid arthritis. Rheumatology (Oxford). 45:26–30. 2006.PubMed/NCBI View Article : Google Scholar

8 

Wakisaka S, Suzuki N, Takeba Y, Shimoyama Y, Nagafuchi H, Takeno M, Saito N, Yokoe T, Kaneko A, Asai T and Sakane T: Modulation by proinflammatory cytokines of Fas/Fas ligand-mediated apoptotic cell death of synovial cells in patients with rheumatoid arthritis (RA). Clin Exp Immunol. 114:119–128. 1998.PubMed/NCBI View Article : Google Scholar

9 

Croft M and Siegel RM: Beyond TNF: TNF superfamily cytokines as targets for the treatment of rheumatic diseases. Nat Rev Rheumatol. 13:217–233. 2017.PubMed/NCBI View Article : Google Scholar

10 

Schedel J, Gay RE, Kuenzler P, Seemayer C, Simmen B, Michel BA and Gay S: FLICE-inhibitory protein expression in synovial fibroblasts and at sites of cartilage and bone erosion in rheumatoid arthritis. Arthritis Rheum. 46:1512–1518. 2002.PubMed/NCBI View Article : Google Scholar

11 

Kobayashi T, Okamoto K, Kobata T, Hasunuma T, Kato T, Hamada H and Nishioka K: Differential regulation of Fas-mediated apoptosis of rheumatoid synoviocytes by tumor necrosis factor alpha and basic fibroblast growth factor is associated with the expression of apoptosis-related molecules. Arthritis Rheum. 43:1106–1114. 2000.PubMed/NCBI View Article : Google Scholar

12 

Scaffidi C, Fulda S, Srinivasan A, Friesen C, Li F, Tomaselli KJ, Debatin KM, Krammer PH and Peter ME: Two CD95 (APO-1/Fas) signaling pathways. EMBO J. 17:1675–1687. 1998.PubMed/NCBI View Article : Google Scholar

13 

Audo R, Calmon-Hamaty F, Papon L, Combe B, Morel J and Hahne M: Distinct effects of soluble and membrane-bound fas ligand on fibroblast-like synoviocytes from rheumatoid arthritis patients. Arthritis Rheumatol. 66:3289–3299. 2014.PubMed/NCBI View Article : Google Scholar

14 

Guegan JP and Legembre P: Nonapoptotic functions of Fas/CD95 in the immune response. FEBS J. 285:809–827. 2018.PubMed/NCBI View Article : Google Scholar

15 

Shi G, Wu Y, Zhang J and Wu J: Death decoy receptor TR6/DcR3 inhibits T cell chemotaxis in vitro and in vivo. J Immunol. 171:3407–3414. 2003.PubMed/NCBI View Article : Google Scholar

16 

Pitti RM, Marsters SA, Lawrence DA, Roy M, Kischkel FC, Dowd P, Huang A, Donahue CJ, Sherwood SW, Baldwin DT, et al: Genomic amplification of a decoy receptor for Fas ligand in lung and colon cancer. Nature. 396:699–703. 1998.PubMed/NCBI View Article : Google Scholar

17 

Tsuji S, Hosotani R, Yonehara S, Masui T, Tulachan SS, Nakajima S, Kobayashi H, Koizumi M, Toyoda E, Ito D, et al: Endogenous decoy receptor 3 blocks the growth inhibition signals mediated by Fas ligand in human pancreatic adenocarcinoma. Int J Cancer. 106:17–25. 2003.PubMed/NCBI View Article : Google Scholar

18 

Yu KY, Kwon B, Ni J, Zhai Y, Ebner R and Kwon BS: A newly identified member of tumor necrosis factor receptor superfamily (TR6) suppresses LIGHT-mediated apoptosis. J Biol Chem. 274:13733–13736. 1999.PubMed/NCBI View Article : Google Scholar

19 

Migone TS, Zhang J, Luo X, Zhuang L, Chen C, Hu B, Hong JS, Perry JW, Chen SF, Zhou JX, et al: TL1A is a TNF-like ligand for DR3 and TR6/DcR3 and functions as a T cell costimulator. Immunity. 16:479–492. 2002.PubMed/NCBI View Article : Google Scholar

20 

Hayashi S, Miura Y, Nishiyama T, Mitani M, Tateishi K, Sakai Y, Hashiramoto A, Kurosaka M, Shiozawa S and Doita M: Decoy receptor 3 expressed in rheumatoid synovial fibroblasts protects the cells against Fas-induced apoptosis. Arthritis Rheum. 56:1067–1075. 2007.PubMed/NCBI View Article : Google Scholar

21 

Takahashi M, Miura Y, Hayashi S, Tateishi K, Fukuda K and Kurosaka M: DcR3-TL1A signalling inhibits cytokine-induced proliferation of rheumatoid synovial fibroblasts. Int J Mol Med. 28:423–427. 2011.PubMed/NCBI View Article : Google Scholar

22 

Fukuda K, Miura Y, Maeda T, Hayashi S and Kurosaka M: Interleukin12B is upregulated by decoy receptor 3 in rheumatoid synovial fibroblasts. Mol Med Rep. 13:3647–3652. 2016.PubMed/NCBI View Article : Google Scholar

23 

Fukuda K, Miura Y, Maeda T, Hayashi S and Kuroda R: Decoy receptor 3 down-regulates centrosomal protein 70 kDa specifically in rheumatoid synovial fibroblasts. Mod Rheumatol. 28:287–292. 2018.PubMed/NCBI View Article : Google Scholar

24 

Maeda T, Miura Y, Fukuda K, Hayashi S and Kurosaka M: Decoy receptor 3 regulates the expression of tryptophan hydroxylase 1 in rheumatoid synovial fibroblasts. Mol Med Rep. 12:5191–5196. 2015.PubMed/NCBI View Article : Google Scholar

25 

Fukuda K, Miura Y, Maeda T, Takahashi M, Hayashi S and Kurosaka M: Decoy receptor 3 regulates the expression of various genes in rheumatoid arthritis synovial fibroblasts. Int J Mol Med. 32:910–916. 2013.PubMed/NCBI View Article : Google Scholar

26 

Fukuda K, Miura Y, Maeda T, Hayashi S and Kuroda R: Expression profiling of genes in rheumatoid fibroblast-like synoviocytes regulated by tumor necrosis factor-like ligand 1A using cDNA microarray analysis. Biomed Rep. 1:1–5. 2019.PubMed/NCBI View Article : Google Scholar

27 

Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, Cooper NS, Healey LA, Kaplan SR, Liang MH, Luthra HS, et al: The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 31:315–324. 1988.PubMed/NCBI View Article : Google Scholar

28 

Chang Q, Peter ME and Grassi MA: Fas ligand-Fas signaling participates in light-induced apoptotic death in photoreceptor cells. Invest Ophthalmol Vis Sci. 53:3703–3716. 2012.PubMed/NCBI View Article : Google Scholar

29 

Nitobe J, Yamaguchi S, Okuyama M, Nozaki N, Sata M, Miyamoto T, Takeishi Y, Kubota I and Tomoike H: Reactive oxygen species regulate FLICE inhibitory protein (FLIP) and susceptibility to Fas-mediated apoptosis in cardiac myocytes. Cardiovasc Res. 57:119–128. 2003.PubMed/NCBI View Article : Google Scholar

30 

Thiel CT, Kraus C, Rauch A, Ekici AB, Rautenstrauss B and Reis A: A new quantitative PCR multiplex assay for rapid analysis of chromosome 17p11.2-12 duplications and deletions leading to HMSN/HNPP. Eur J Hum Genet. 11:170–178. 2003.PubMed/NCBI View Article : Google Scholar

31 

Choi YJ and Yun HK: Transcriptional profiles of Rhizobium vitis-inoculated and salicylic acid-treated ‘Tamnara’ grapevines based on microarray analysis. J Plant Biotechnol. 43:37–48. 2016.

32 

Chang YC, Chen TC, Lee CT, Yang CY, Wang HW, Wang CC and Hsieh SL: Epigenetic control of MHC class II expression in tumor-associated macrophages by decoy receptor 3. Blood. 111:5054–5063. 2008.PubMed/NCBI View Article : Google Scholar

33 

Espinosa I, Catasus L, Canet B, D'Angelo E, Munoz J and Prat J: Gene expression analysis identifies two groups of ovarian high-grade serous carcinomas with different prognosis. Mod Pathol. 24:846–854. 2011.PubMed/NCBI View Article : Google Scholar

34 

Khan J, Simon R, Bittner M, Chen Y, Leighton SB, Pohida T, Smith PD, Jiang Y, Gooden GC, Trent JM, et al: Gene expression profiling of alveolar rhabdomyosarcoma with cDNA microarrays. Cancer Res. 58:5009–5013. 1998.PubMed/NCBI

35 

Whitney LW, Becker KG, Tresser NJ, Caballero-Ramos CI, Munson PJ, Prabhu VV, Trent JM, McFarland HF and Biddison WE: Analysis of gene expression in mutiple sclerosis lesions using cDNA microarrays. Ann Neurol. 46:425–428. 1999.PubMed/NCBI View Article : Google Scholar

36 

Li J, Yang S, Lu S, Zhao H, Feng J, Li W, Ma F, Ren Q, Liu B, Zhang L, et al: Differential gene expression profile associated with the abnormality of bone marrow mesenchymal stem cells in aplastic anemia. PLoS One. 7(e47764)2012.PubMed/NCBI View Article : Google Scholar

37 

van der Pouw Kraan TC, van Gaalen FA, Kasperkovitz PV, Verbeet NL, Smeets TJ, Kraan MC, Fero M, Tak PP, Huizinga TW, Pieterman E, et al: Rheumatoid arthritis is a heterogeneous disease: Evidence for differences in the activation of the STAT-1 pathway between rheumatoid tissues. Arthritis Rheum. 48:2132–2145. 2003.PubMed/NCBI View Article : Google Scholar

38 

Lee SK, Jeon EK, Kim YJ, Seo SH, Kim CD, Lim JS and Lee JH: A global gene expression analysis of the peripheral blood mononuclear cells reveals the gene expression signature in psoriasis. Ann Dermatol. 21:237–242. 2009.PubMed/NCBI View Article : Google Scholar

39 

Heller RA, Schena M, Chai A, Shalon D, Bedilion T, Gilmore J, Woolley DE and Davis RW: Discovery and analysis of inflammatory disease-related genes using cDNA microarrays. Proc Natl Acad Sci USA. 94:2150–2155. 1997.PubMed/NCBI View Article : Google Scholar

40 

Bertin S, Lozano-Ruiz B, Bachiller V, Garcia-Martinez I, Herdman S, Zapater P, Frances R, Such J, Lee J, Raz E and González-Navajas JM: Dual-specificity phosphatase 6 regulates CD4+ T-cell functions and restrains spontaneous colitis in IL-10-deficient mice. Mucosal Immunol. 8:505–515. 2015.PubMed/NCBI View Article : Google Scholar

41 

Hsu SF, Lee YB, Lee YC, Chung AL, Apaya MK, Shyur LF, Cheng CF, Ho FM and Meng TC: Dual specificity phosphatase DUSP6 promotes endothelial inflammation through inducible expression of ICAM-1. FEBS J. 285:1593–1610. 2018.PubMed/NCBI View Article : Google Scholar

42 

Patel RD, Kim DJ, Peters JM and Perdew GH: The aryl hydrocarbon receptor directly regulates expression of the potent mitogen epiregulin. Toxicol Sci. 89:75–82. 2006.PubMed/NCBI View Article : Google Scholar

43 

Murakami M, Harada M, Kamimura D, Ogura H, Okuyama Y, Kumai N, Okuyama A, Singh R, Jiang JJ, Atsumi T, et al: Disease-association analysis of an inflammation-related feedback loop. Cell Rep. 3:946–959. 2013.PubMed/NCBI View Article : Google Scholar

44 

Harada M, Kamimura D, Arima Y, Kohsaka H, Nakatsuji Y, Nishida M, Atsumi T, Meng J, Bando H, Singh R, et al: Temporal expression of growth factors triggered by epiregulin regulates inflammation development. J Immunol. 194:1039–1046. 2015.PubMed/NCBI View Article : Google Scholar

45 

Lahoti TS, Hughes JM, Kusnadi A, John K, Zhu B, Murray IA, Gowda K, Peters JM, Amin SG and Perdew GH: Aryl hydrocarbon receptor antagonism attenuates growth factor expression, proliferation, and migration in fibroblast-like synoviocytes from patients with rheumatoid arthritis. J Pharmacol Exp Ther. 348:236–245. 2014.PubMed/NCBI View Article : Google Scholar

46 

Yin T, Taga T, Tsang ML, Yasukawa K, Kishimoto T and Yang YC: Involvement of IL-6 signal transducer gp130 in IL-11-mediated signal transduction. J Immunol. 151:2555–2561. 1993.PubMed/NCBI

47 

Trepicchio WL, Bozza M, Pedneault G and Dorner AJ: Recombinant human IL-11 attenuates the inflammatory response through down-regulation of proinflammatory cytokine release and nitric oxide production. J Immunol. 157:3627–3634. 1996.PubMed/NCBI

48 

Hermann JA, Hall MA, Maini RN, Feldmann M and Brennan FM: Important immunoregulatory role of interleukin-11 in the inflammatory process in rheumatoid arthritis. Arthritis Rheum. 41:1388–1397. 1998.PubMed/NCBI View Article : Google Scholar

49 

Elshabrawy HA, Volin MV, Essani AB, Chen Z, McInnes IB, Van Raemdonck K, Palasiewicz K, Arami S, Gonzalez M, Ashour HM, et al: IL-11 facilitates a novel connection between RA joint fibroblasts and endothelial cells. Angiogenesis. 21:215–228. 2018.PubMed/NCBI View Article : Google Scholar

50 

Murakami K, Kobayashi Y, Uehara S, Suzuki T, Koide M, Yamashita T, Nakamura M, Takahashi N, Kato H, Udagawa N and Nakamura Y: A Jak1/2 inhibitor, baricitinib, inhibits osteoclastogenesis by suppressing RANKL expression in osteoblasts in vitro. PLoS One. 12(e0181126)2017.PubMed/NCBI View Article : Google Scholar

51 

Parri M, Pietrovito L, Grandi A, Campagnoli S, De Camilli E, Bianchini F, Marchio S, Bussolino F, Jin B, Sarmientos P, et al: Angiopoietin-like 7, a novel pro-angiogenetic factor over-expressed in cancer. Angiogenesis. 17:881–896. 2014.PubMed/NCBI View Article : Google Scholar

52 

Qian T, Wang K, Cui J, He Y and Yang Z: Angiopoietin-Like Protein 7 promotes an inflammatory phenotype in RAW264.7 macrophages through the P38 MAPK signaling pathway. Inflammation. 39:974–985. 2016.PubMed/NCBI View Article : Google Scholar

53 

Lao M, Shi M, Zou Y, Huang M, Ye Y, Qiu Q, Xiao Y, Zeng S, Liang L, Yang X and Xu H: Protein inhibitor of activated STAT3 regulates migration, invasion, and activation of Fibroblast-like synoviocytes in rheumatoid arthritis. J Immunol. 196:596–606. 2016.PubMed/NCBI View Article : Google Scholar

54 

Muller S, Ledl A and Schmidt D: SUMO: A regulator of gene expression and genome integrity. Oncogene. 23:1998–2008. 2004.PubMed/NCBI View Article : Google Scholar

55 

Schmidt D and Muller S: PIAS/SUMO: New partners in transcriptional regulation. Cell Mol Life Sci. 60:2561–2574. 2003.PubMed/NCBI View Article : Google Scholar

56 

Wu Y, Guo Z, Wu H, Wang X, Yang L, Shi X, Du J, Tang B, Li W, Yang L and Zhang Y: SUMOylation represses Nanog expression via modulating transcription factors Oct4 and Sox2. PLoS One. 7(e39606)2012.PubMed/NCBI View Article : Google Scholar

57 

Chowdhury D, Singh A, Gupta A, Tulsawani R, Meena RC and Chakrabarti A: p38 MAPK pathway-dependent SUMOylation of Elk-1 and phosphorylation of PIAS2 correlate with the downregulation of Elk-1 activity in heat-stressed HeLa cells. Cell Stress Chaperones. 24:393–407. 2019.PubMed/NCBI View Article : Google Scholar

58 

Bobacz K, Gruber R, Soleiman A, Graninger WB, Luyten FP and Erlacher L: Cartilage-derived morphogenetic protein-1 and -2 are endogenously expressed in healthy and osteoarthritic human articular chondrocytes and stimulate matrix synthesis. Osteoarthritis Cartilage. 10:394–401. 2002.PubMed/NCBI View Article : Google Scholar

59 

Hotten GC, Matsumoto T, Kimura M, Bechtold RF, Kron R, Ohara T, Tanaka H, Satoh Y, Okazaki M, Shirai T, et al: Recombinant human growth/differentiation factor 5 stimulates mesenchyme aggregation and chondrogenesis responsible for the skeletal development of limbs. Growth Factors. 13:65–74. 1996.PubMed/NCBI View Article : Google Scholar

60 

Liu FL, Lin LH, Sytwu HK and Chang DM: GDF-5 is suppressed by IL-1beta and enhances TGF-beta3-mediated chondrogenic differentiation in human rheumatoid fibroblast-like synoviocytes. Exp Mol Pathol. 88:163–170. 2010.PubMed/NCBI View Article : Google Scholar

61 

Miyamoto Y, Mabuchi A, Shi D, Kubo T, Takatori Y, Saito S, Fujioka M, Sudo A, Uchida A, Yamamoto S, et al: A functional polymorphism in the 5' UTR of GDF5 is associated with susceptibility to osteoarthritis. Nat Genet. 39:529–533. 2007.PubMed/NCBI View Article : Google Scholar

62 

Martinez A, Varade J, Lamas JR, Fernandez-Arquero M, Jover JA, de la Concha EG, Fernandez-Gutierrez B and Urcelay E: GDF5 Polymorphism associated with osteoarthritis: Risk for rheumatoid arthritis. Ann Rheum Dis. 67:1352–1353. 2008.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

September-2021
Volume 22 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fukuda K, Miura Y, Maeda T, Hayashi S, Matsumoto T and Kuroda R: Expression profiling of genes in rheumatoid fibroblast‑like synoviocytes regulated by Fas ligand via cDNA microarray analysis. Exp Ther Med 22: 1000, 2021
APA
Fukuda, K., Miura, Y., Maeda, T., Hayashi, S., Matsumoto, T., & Kuroda, R. (2021). Expression profiling of genes in rheumatoid fibroblast‑like synoviocytes regulated by Fas ligand via cDNA microarray analysis. Experimental and Therapeutic Medicine, 22, 1000. https://doi.org/10.3892/etm.2021.10432
MLA
Fukuda, K., Miura, Y., Maeda, T., Hayashi, S., Matsumoto, T., Kuroda, R."Expression profiling of genes in rheumatoid fibroblast‑like synoviocytes regulated by Fas ligand via cDNA microarray analysis". Experimental and Therapeutic Medicine 22.3 (2021): 1000.
Chicago
Fukuda, K., Miura, Y., Maeda, T., Hayashi, S., Matsumoto, T., Kuroda, R."Expression profiling of genes in rheumatoid fibroblast‑like synoviocytes regulated by Fas ligand via cDNA microarray analysis". Experimental and Therapeutic Medicine 22, no. 3 (2021): 1000. https://doi.org/10.3892/etm.2021.10432