RNA‑seq analysis of synovial fibroblasts in human rheumatoid arthritis

  • Authors:
    • Xiuhui Wang
    • Shengli Xia
    • Beigang Fu
  • View Affiliations

  • Published online on: April 24, 2014     https://doi.org/10.3892/mmr.2014.2182
  • Pages: 241-247
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to identify differentially expressed genes (DEGs) between individuals with rheumatoid arthritis (RA) and healthy controls, in order to provide a theoretical foundation for RA diagnosis and targeted gene therapy. Illumina mRNA sequence data (RNA‑Seq) corresponding to RA and control samples were downloaded from the Sequence Read Archive (SRA) database. Gene Ontology (GO) enrichment analysis was performed with the GOstat tool in order to identify over‑represented biological functions among DEGs, and the related Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were identified using the KEGG Automatic Annotation Server (KAAS). A total of 293 DEGs were identified, among which 16 DEGs have been previously shown to associate with RA, such as those encoding matrix metalloproteinase‑1 (MMP‑1), interleukin‑1 receptor type 1 (IL1R1), and chemokine (C-X3-C motif) ligand 1 (CX3CL1). GO functional annotation and enrichment analysis showed that the DEGs are enriched for 309 GO terms, mainly protein‑protein interactions, membrane formation and stability. KEGG pathway analysis demonstrated that these DEGs are involved in 131 pathways, including Wnt and calcium signaling, and cell adhesion molecule (CAM)-related pathways. In conclusion, the results provide both expansive and detailed insights into the molecular pathogenesis of RA, particularly with regards to the development of therapeutic targets, and may inspire further experimentation aiming to identify new strategies for RA treatment.

Introduction

Rheumatoid arthritis (RA) is a chronic inflammatory disease, characterized by synovial cell proliferation and excessive production of pro-inflammatory cytokines, leading to the destruction of diarthrodial joints cartilage and bones (1,2). RA is considered an autoimmune disease (3) that can cause severe disability and premature mortality (4), with ~1% of individuals afflicted worldwide (5). Although the etiology of RA has not been fully elucidated, numerous studies have demonstrated that it is a multifactorial disease that results from interactions between genetic and environmental factors (6).

Over the past 15 years, rheumatologists have developed a number of therapeutic strategies aiming to treat RA (7). Moreover, researchers have shown that the characterization of the cytokine signaling pathways involved in RA provides important opportunities for identifying pro-inflammatory cytokines that can be targeted in the context of novel therapeutic interventions (810). However, these agents are not effective in all patients (11). Furthermore, it has long been recognized that patients with RA have an increased risk for certain types of cancer (12,13). For example, it was demonstrated that RA-associated interstitial lung disease accounts for mortality of up to 20% of RA patients (14). Therefore, there is an urgent need to investigate the molecular mechanisms underlying the processes involved in RA.

High-throughput mRNA sequencing (RNA-seq), which allows simultaneous identification of transcripts and estimation of their abundance (15,16), is a recently developed transcriptome profiling approach that uses deep-sequencing technologies (17). It has fostered numerous advances in the characterization and quantification of transcripts, since it allows a nearly complete characterization of transcriptomes (18). A recent study making use of RNA-seq showed that the activation and proliferation of RA synovial fibroblasts relate to the pathogenesis of RA (19). Here, we performed a comprehensive meta-analysis of the transcriptome data from based on the data of Heruth et al (19), which were derived from RNA of healthy and RA patients. Our findings may enhance the understanding of the mechanisms underlying the process of joint destruction, and allow a more selective and specific application of therapeutic agents that target pro-inflammatory cytokines and thus, a more effective treatment of patients with RA and other inflammatory disorders.

Materials and methods

RNA-seq data

The RNA-seq data were downloaded from the Sequence Read Archive (SRA). Two samples of synovial fibroblasts were available for meta-analysis: One was from individuals with RA, and the control sample (CT) was from healthy individuals (Table I). The downloaded data come from Illumina RNA-seq (19) of paired-end (2 × 100 bp) cDNA libraries prepared from each RNA sample, with sequencing performed as in (20,21).

Table I

Information on raw RNA-seq data from rheumatoid arthritis (RA) and control (CT) samples, downloaded from the Sequence Read Archive (SRA).

Table I

Information on raw RNA-seq data from rheumatoid arthritis (RA) and control (CT) samples, downloaded from the Sequence Read Archive (SRA).

Sample nameTypeLibraryData size (bp)SRA no.
RARheumatoid arthritisPaired-end9.1 GSRR364313
CTHealthyPaired-end8.2 GSRR364315
Quality analysis of raw RNA-seq reads

SolexaQA is a software that allows investigation and trimming of sequences based on their base quality scores. To exclude analytical error from sequencing errors and low data quality, the SolexaQA software (22) was used to process the raw data, based on the read-level quality of each sequence. Raw sequences with quality scores <20 and a length <25 bp were removed from the final dataset that was used for downstream analyses.

Calculation of expression values and identification of differentially expressed genes (DEGs)

First, the TopHat software version 2.0.8 (23) was used to map the sequencing reads to the reference human genome (version hg19, UCSC Genome Browser). TopHat is an efficient read-mapping algorithm designed to align reads from an RNA-seq experiment to a reference genome; we used the following settings: maximum 20 multiple hits per read and maximum 2 mismatches allowed. Second, Cufflinks software package (version 2.0.2) was used to assemble transcripts and calculate their relative expression levels, expressed as fragments per kilobase of exon per million fragments mapped (FPKM) using the default parameters and an average insert size of 200 bp (24). Finally, the Cuffdiff module in Cufflinks was used to estimate differential expression, expressed as a ratio of RA to control expression for each transcript, along with the statistical significance of the observed differences (25). Genes with log2 fold change (FC) ≥1 and p-value ≤0.05 were selected as DEGs. In addition, the expression values of the differentially expressed genes were normalized by z-score transformation prior to visualization with the heatmap function available in the R statistical package (26).

Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis

The GOstat software (27) was used to conduct GO functional annotation and enrichment analysis of DEGs (significantly at P<0.05). The KAAS annotation server (28) was used to identify the KEGG metabolic pathways in which the identified DEGs are involved. Finally, the KOBAS server 2.0 (29), integrating data from a number of human disease databases, was used to identify DEGs related to RA.

Results

Identification of DEGs

Quality control statistics on the RNA-seq are shown in Table II. A total of 293 genes were identified as differentially expressed between the RA and CT samples based on our criteria (log2 FC ≥1; p-value ≤0.05). Fig. 1 shows the expression patterns of DEGs in the RA and CT samples. The relative expression (FKPM), log2 FC, and associated p-values of the top 10 DEGs (in terms of FKPM at the RA sample) are shown in Table III.

Table II

Quality control statistics of mRNA sequence reads.

Table II

Quality control statistics of mRNA sequence reads.

ReadsHealthy controlPercentageRheumatoid arthritisPercentage
Original80,782,262-89,757,726-
Low-quality9,605,51811.896,707,3747.47
Remaining71,176,74488.1183,050,35292.53

Table III

The top 10 significantly differentially expressed genes.

Table III

The top 10 significantly differentially expressed genes.

GeneChrRA FPKMCT FPKMLog2 FCp-value
CHI3L1Chr15,847.7200.211−14.7533.02E-14
MMP-1Chr1178.2710.017−12.1669.28E-11
SMOC2Chr636.8020.010−11.7191.95E-07
Ror2Chr98.9130.009−9.9951.23E-05
VITChr29.3130.013−9.5221.34E-04
HEYLChr10.04328.4599.3551.29E-14
MCAMChr110.155153.6749.9550
EFHD1Chr20.054103.31710.8935.70E-12
FOXE1Chr90.00918.42911.0692.92E-12
WFDC1Chr160.02076.09511.8825.09E-13

[i] Chr, human chromosome; RA, rheumatoid arthritis; FPKM, fragments per kilobase of exon per million fragments mapped; FC, fold change; CHI3L1, chitinase 3-like 1; MMP-1, matrix metalloproteinase-1; SMOC2, SPARC related modular calcium binding 2; Ror2, receptor tyrosine kinase-like orphan receptor 2; VIT, vitrin; HEYL, hes-related family bHLH transcription factor with YRPW motif-like; MCAM, melanoma cell adhesion molecule; EFHD1, EF-hand domain family, member D1; FOXE1, forkhead box E1; WFDC1, WAP four-disulfide core domain 1.

GO analysis

The GOstat tool retrieves GO annotations and investigates over-representation of these in a given gene list (27). We used GOstat to functionally annotate the identified DEGs based on the cut-off point of P<0.05. The 293 DEGs were enriched for 309 GO terms. The top 10 GO terms in terms of p-value are shown in Table IV. This analysis indicated that the identified DEGs are mainly involved in anatomical structure development, cell membrane formation and stability, and biological adhesion.

Table IV

The top 10 significantly enriched Gene Ontology (GO) terms among differentially expressed genes.

Table IV

The top 10 significantly enriched Gene Ontology (GO) terms among differentially expressed genes.

GO IDNameDescriptionp-value
0048856Anatomical structure developmentThe biological process whose specific outcome is the progression of an anatomical structure from an initial condition to its mature state. This process begins with the formation of the structure and ends with the mature structure, whatever form that may be including its natural destruction. An anatomical structure is any biological entity that occupies space and is distinguished from its surroundings. Anatomical structures can be macroscopic such as a carpel, or microscopic such as an acrosome.8.76e-44
0032502Developmental processA biological process whose specific outcome is the progression of an integrated living unit: An anatomical structure (which may be a subcellular structure, cell, tissue, or organ), or organism over time from an initial condition to a later condition.1.19e-36
0007275Multicellular organismal developmentThe biological process whose specific outcome is the progression of a multicellular organism over time from an initial condition (e.g. a zygote or a young adult) to a later condition (e.g. a multicellular animal or an aged adult)1.36e-35
0048731System developmentThe process whose specific outcome is the progression of an organismal system over time, from its formation to the mature structure. A system is a regularly interacting or interdependent group of organs or tissues that work together to carry out a given biological process.2.12e-33
0032501Multicellular organismal processAny biological process, occurring at the level of a multicellular organism, pertinent to its function.3.89e-32
0009653Anatomical structure morphogenesisThe process in which anatomical structures are generated and organized. Morphogenesis pertains to the creation of form.1.62e-28
0005886Plasma membraneThe membrane surrounding a cell that separates the cell from its external environment. It consists of a phospholipid bilayer and associated proteins.2.66e-28
0031226Intrinsic component of plasma membraneThe component of the plasma membrane consisting of gene products and protein complexes that have some covalently attached part (e.g. peptide sequence or GPI anchor), which spans or is embedded in one or both leaflets the membrane.8.71e-25
0005887Integral component of plasma membraneThe component of the plasma membrane consisting of gene products and protein complexes that have some part that penetrates at least one leaflet of the membrane bilayer. This component includes gene products that are buried in the bilayer with no exposure outside the bilayer.8.01e-24
0022610Biological adhesionThe attachment of a cell or organism to a substrate or other organism.1.01e-21
KEGG pathway analysis

The 293 DEGs were found to be involved in 131 pathways, including Wnt signaling, cell adhesion molecules (CAMs), neuroactive ligand-receptor interaction, PI3K-Akt signaling, cytokine-cytokine receptors interactions, calcium signaling, regulation of actin cytoskeleton and focal adhesion (Table V). By combining the functional annotation of DEGs with data from disease databases, we found that 16 genes among the DEGs have been previously associated with the occurrence of RA (Table VI).

Table V

KEGG pathways that DEGs are mainly involved in.

Table V

KEGG pathways that DEGs are mainly involved in.

Pathway idDescriptionCount
4080Neuroactive ligand-receptor interaction10
4151PI3K-Akt signaling pathway9
4060Cytokine-cytokine receptor interaction9
4020Calcium signaling pathway7
4512ECM-receptor interaction7
4514Cell adhesion molecules7
4810Regulation of actin cytoskeleton7
4510Focal adhesion7
4974Protein digestion and absorption7
5410Hypertrophic cardiomyopathy7

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes; DEGs, differentially expressed genes; PI3K, phosphoinositide-3-kinase; ECM, extracellular matrix.

Table VI

Genbank information on differentially expressed genes (n=16) that have been associated with rheumatoid arthritis.

Table VI

Genbank information on differentially expressed genes (n=16) that have been associated with rheumatoid arthritis.

Acc. no.Gene symbolNameExpression
NM_003881WISP2WNT1 inducible signaling pathway protein 2Down
NM_001066 TNFRSF1BTumor necrosis factor receptor superfamily, member 1BDown
NM_000877IL1R1Interleukin 1 receptor, type 1Down
NM_002996CX3CL1Chemokine (C-X3-C motif) ligand 1Up
NM_007365PADI2Peptidyl arginine deiminase, type IIUp
NM_001276CHI3L1Chitinase 3-like 1Down
NM_004864GDF15Growth differentiation factor 15Down
NM_000214JAG1Jagged 1Up
NM_000612IGF2Insulin-like growth factor 2Up
NM_004878PTGESProstaglandin E synthaseDown
NM_001145938MMP-1Matrix metalloproteinase-1Down
NM_019111HLA-DRAMajor histocompatibility complex, class II, DR αUp
NM_000396CTSKCathepsin KDown
NM_005118TNFSF15Tumor necrosis factor superfamily, member 15 transcript variant 1Up
NM_006379SEMA3CSema domain, Ig, short basic domain, secreted, semaphorin 3CDown
NM_000692ALDH1B1Aldehyde dehydrogenase 1 family, member B1Up

Discussion

RA is a systemic inflammatory disorder that commonly affects the diarthrodial joints (30). The pathogenesis of RA is characterized by the influx of immune system cells (31), which induce the production of pro-inflammatory cytokines, decreased synthesis of anti-inflammatory cytokines and the subsequent activation and proliferation of synovial fibroblasts (32). Remission in RA is an increasingly attainable goal, but there is no widely used definition of remission that is stringent yet achievable, and that could be uniformly applied as a criterion of clinical outcome (33). Therefore, the identification and characterization of genes related to RA is important for the understanding of the pathogenesis of this disease and the identification of novel anti-inflammatory therapeutic targets.

In the present study, we identified genes differentially expressed between RA and healthy individuals. A total of 293 genes were identified as DEGs, including these encoding the receptor tyrosine kinase-like orphan receptor 2 (Ror2), chitinase-3-like 1 (CHI3L1), matrix metalloproteinases (MMPs), interleukin (IL)-26, and v-maf musculoaponeurotic fibrosarcoma oncogene homolog B (MafB). Ror2 has been associated with RA. A previous study indicated that the Wnt5a-Ror2 pathway is crucial for osteoclastogenesis in physiological and pathological environments, and may represent a therapeutic target for bone diseases, including RA (34). Sonomoto et al (35) demonstrated that IL-1β can effectively and rapidly induce the differentiation of human mesenchymal stem cells into osteoblasts, as well as mineralization, mainly through the non-canonical Wnt5a-Ror2 pathway.

The CHI3L1 or YKL40 gene encodes for the human cartilage glycoprotein 39 (HC-gp39), which is secreted by synovial fibroblasts, macrophages, neutrophil granulocytes, and chondrocytes. Its expression is regulated by NF-κB (36). It was recently suggested that the YKL-40 protein might be implicated in the pathogenesis of RA and that its level may indicate the degree of joint inflammation (37). The carbohydrate-binding motif in YKL-40 specifically activates the Akt signaling pathway in colonic epithelial cells (38). The level of YKL-40 in the serum varies depending on the RA status of patients, and is increased in 54% of patients with clinically active disease (39). YKL-40 may thus be suitable for assessing the disease activity and pathophysiology of RA.

MMPs are members of an enzyme family that contain a zinc ion on their active site, which is required for their catalytic activity. MMPs are critical for maintaining tissue allostasis. These enzymes are active at neutral pH, and can therefore catalyze the physiological turnover of extracellular matrix (ECM) macromolecules (40). A previous study showed that the levels of MMP-8 and -9 in the systemic circulation are representative of the levels of these enzymes in the inflamed joint, and suggested that MMP-9 may be involved in degradation of the joint collagen. The study further confirmed the hypothesis of an MMP/TIMP imbalance in RA (41). In addition, inhibition of MMP-13 was shown to reduce cartilage erosions in two out of three tested animal models of RA, strongly supporting the development of drugs targeting MMPs in order to reduce or halt joint destruction in patients with RA (42).

IL-26, a member of the IL-10 cytokine family, induces the production of pro-inflammatory cytokines by epithelial cells. It was recently demonstrated that IL-26 is constitutively produced by fibroblast-like cells known as synoviocytes in RA, and that it induces the secretion of pro-inflammatory cytokines by myeloid cells and favors the formation of T helper cells producing interleukin 17 (Th17). The authors suggested that IL-26 is a pro-inflammatory cytokine located upstream of the pro-inflammatory cascade, and may constitute a promising target to treat RA and chronic inflammatory disorders (43). Although synoviocytes are present in all joints, only synoviocytes from RA patients can produce IL-26 (44).

MafB was another gene found as differentially expressed in our study. The MafB protein is a putative tumor suppressor in the myeloid lineage, with a key role in monocytopoiesis (45), as well as in monocyte-dendritic cell differentiation (46).

The GO functional annotation analysis showed that DEGs are enriched for a total of 309 GO terms. The top 10 GO terms mainly referred to anatomical structure development, membrane formation and stability, and biological adhesion. It is notable that alterations in biological adhesion are involved in RA. For example, COL5A1 (collagen, type V, α1) was expressed at significantly higher levels in chondrocytes from the damaged region of osteoarthritic cartilage than in those from the intact region (47).

KEGG signaling pathway analysis revealed that the DEGs are predicted to be involved in a total of 131 pathways, including Wnt and calcium signaling, as well as CAM-related pathways. The Wnt signaling pathway plays a key role in cell renewal. A few studies showed that it is also involved in RA pathogenesis (48,49). In the synovial membrane of patients with RA, the Wnt and Fz genes are expressed at higher levels compared to those observed in patients without RA (50). The Wnt proteins are glycoproteins that bind to the Fz receptors on the cell surface, thereby affecting a number of important biological processes, such as cell differentiation, embryonic development, limb development, and joint formation (51). Enhanced knowledge of the role(s) of the Wnt signaling pathway in RA is expected to improve our understanding of the different RA clinical features and improve prognosis. Both calcium signaling and CAM-related pathways, upregulated in RA patients showing a traditional Chinese medicine heat pattern, have been suggested to be important for T-lymphocyte interactions, and thus, constitute candidate targets for RA therapy (52).

Cross-referencing with disease databases revealed that 16 of the identified DEGs have been previously associated with RA; these include genes encoding MMP-1, interleukin-1 receptor type 1 (IL1R1) and PADI2. The aggressive phenotype of synovial fibroblasts in RA is characterised by the increased expression of MMP-1 (53). Collagenases MMP-1 and -13 play an important role in collagen degradation in RA and osteoarthritis, while gelatinases MMP-2 and -9 may be involved in arthritis by degrading non-collagen matrix components in the joints (54). The IL1R1 receptor, once activated upon IL-1 binding, activates NF-κB, which is a modulator of expression of inflammatory and immune genes (55). Nevertheless, further experiments are needed to study the roles that these genes may play in the occurrence of RA.

Overall, our study provided a list of candidate molecular targets for RA treatment and further research. In recent years, gene-targeted therapy of RA has become a popular approach, and related ongoing research appears promising. Despite significant therapeutic advances, RA treatment remains an unsolved medical issue. Therefore, the molecular mechanism(s) underlying this disease need to be further investigated.

Acknowledgements

This study was supported by the National Natural Science Foundation of China of the year 2011 (81171734) and the Applied Basic Research Project of Yunnan Province (2011FZ313).

References

1 

Marrelli A, Cipriani P, Liakouli V, et al: Angiogenesis in rheumatoid arthritis: a disease specific process or a common response to chronic inflammation? Autoimmun Rev. 10:595–598. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Lajas C, Abasolo L, Bellajdel B, et al: Costs and predictors of costs in rheumatoid arthritis: a prevalence-based study. Arthritis Rheum. 49:64–70. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Smolen JS, Aletaha D, Koeller M, Weisman MH and Emery P: New therapies for treatment of rheumatoid arthritis. Lancet. 370:1861–1874. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Aletaha D, Neogi T, Silman AJ, et al: 2010 Rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheum. 62:2569–2581. 2010. View Article : Google Scholar

5 

Bartok B and Firestein GS: Fibroblast-like synoviocytes: key effector cells in rheumatoid arthritis. Immunol Rev. 233:233–255. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Tobon GJ, Youinou P and Saraux A: The environment, geo-epidemiology, and autoimmune disease: Rheumatoid arthritis. Autoimmun Rev. 9:A288–A292. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Smolen JS, Aletaha D, Bijlsma JW, et al: Treating rheumatoid arthritis to target: recommendations of an international task force. Ann Rheum Dis. 69:631–637. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Guma M, Hammaker D, Topolewski K, et al: Antiinflammatory functions of p38 in mouse models of rheumatoid arthritis: advantages of targeting upstream kinases MKK-3 or MKK-6. Arthritis Rheum. 64:2887–2895. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Le Goff B, Blanchard F, Berthelot JM, Heymann D and Maugars Y: Role for interleukin-6 in structural joint damage and systemic bone loss in rheumatoid arthritis. Joint Bone Spine. 77:201–205. 2010.PubMed/NCBI

10 

Damjanov N, Kauffman RS and Spencer-Green GT: Efficacy, pharmacodynamics, and safety of VX-702, a novel p38 MAPK inhibitor, in rheumatoid arthritis: results of two randomized, double-blind, placebo-controlled clinical studies. Arthritis Rheum. 60:1232–1241. 2009. View Article : Google Scholar

11 

Buch MH and Emery P: New therapies in the management of rheumatoid arthritis. Curr Opin Rheumatol. 23:245–251. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Isomaki HA, Hakulinen T and Joutsenlahti U: Excess risk of lymphomas, leukemia and myeloma in patients with rheumatoid arthritis. J Chronic Dis. 31:691–696. 1978. View Article : Google Scholar : PubMed/NCBI

13 

Smitten AL, Simon TA, Hochberg MC and Suissa S: A meta-analysis of the incidence of malignancy in adult patients with rheumatoid arthritis. Arthritis Res Ther. 10:R452008. View Article : Google Scholar : PubMed/NCBI

14 

Olson AL, Swigris JJ, Sprunger DB, et al: Rheumatoid arthritis-interstitial lung disease-associated mortality. Am J Respir Crit Care Med. 183:372–378. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Cloonan N, Forrest AR, Kolle G, et al: Stem cell transcriptome profiling via massive-scale mRNA sequencing. Nat Methods. 5:613–619. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Mortazavi A, Williams BA, McCue K, Schaeffer L and Wold B: Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods. 5:621–628. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Wang Z, Gerstein M and Snyder M: RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet. 10:57–63. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Ozsolak F and Milos PM: RNA sequencing: advances, challenges and opportunities. Nat Rev Genet. 12:87–98. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Heruth DP, Gibson M, Grigoryev DN, Zhang LQ and Ye SQ: RNA-seq analysis of synovial fibroblasts brings new insights into rheumatoid arthritis. Cell Biosci. 2:432012. View Article : Google Scholar : PubMed/NCBI

20 

Zhang LQ, Cheranova D, Gibson M, et al: RNA-seq reveals novel transcriptome of genes and their isoforms in human pulmonary microvascular endothelial cells treated with thrombin. PLoS One. 7:e312292012. View Article : Google Scholar

21 

Cheranova D, Gibson M, Chaudhary S, et al: RNA-seq analysis of transcriptomes in thrombin-treated and control human pulmonary microvascular endothelial cells. J Vis Exp. e43932013. View Article : Google Scholar

22 

Cox MP, Peterson DA and Biggs PJ: SolexaQA: At-a-glance quality assessment of Illumina second-generation sequencing data. BMC Bioinformatics. 11:4852010. View Article : Google Scholar : PubMed/NCBI

23 

Trapnell C, Pachter L and Salzberg SL: TopHat: discovering splice junctions with RNA-Seq. Bioinformatics. 25:1105–1111. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Trapnell C, Williams BA, Pertea G, et al: Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 28:511–515. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Trapnell C, Roberts A, Goff L, et al: Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc. 7:562–578. 2012. View Article : Google Scholar : PubMed/NCBI

26 

R Core Team. R: A language and environment for statistical computing. R Foundation for Statistical Computing; Vienna, Austria: 2013

27 

Beissbarth T and Speed TP: GOstat: find statistically overrepresented Gene Ontologies within a group of genes. Bioinformatics. 20:1464–1465. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Moriya Y, Itoh M, Okuda S, Yoshizawa A and Kanehisa M: KAAS: an automatic genome annotation and pathway reconstruction server. Nucleic Acids Res. 35:W182–W185. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Xie C, Mao X, Huang J, et al: KOBAS 2.0: a web server for annotation and identification of enriched pathways and diseases. Nucleic Acids Res. 39:W316–W322. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Brooker DS: Rheumatoid arthritis: otorhinolaryngological manifestations. Clin Otolaryngol Allied Sci. 13:239–246. 1988. View Article : Google Scholar : PubMed/NCBI

31 

Gierut A, Perlman H and Pope RM: Innate immunity and rheumatoid arthritis. Rheum Dis Clin North Am. 36:271–296. 2010. View Article : Google Scholar

32 

Scott DL, Wolfe F and Huizinga TW: Rheumatoid arthritis. Lancet. 376:1094–1108. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Felson DT, Smolen JS, Wells G, et al: American College of Rheumatology/European League against Rheumatism provisional definition of remission in rheumatoid arthritis for clinical trials. Ann Rheum Dis. 70:404–413. 2011. View Article : Google Scholar

34 

Maeda K, Kobayashi Y, Udagawa N, et al: Wnt5a-Ror2 signaling between osteoblast-lineage cells and osteoclast precursors enhances osteoclastogenesis. Nat Med. 18:405–412. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Sonomoto K, Yamaoka K, Oshita K, et al: Interleukin-1β induces differentiation of human mesenchymal stem cells into osteoblasts via the Wnt-5a/receptor tyrosine kinase-like orphan receptor 2 pathway. Arthritis Rheum. 64:3355–3363. 2012.

36 

Srivastava SK, Antal P, Gal J, et al: Lack of evidence for association of two functional SNPs of CHI3L1 gene (HC-gp39) with rheumatoid arthritis. Rheumatol Int. 31:1003–1007. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Kazakova M, Batalov A, Deneva T, Mateva N, Kolarov Z and Sarafian V: Relationship between sonographic parameters and YKL-40 levels in rheumatoid arthritis. Rheumatol Int. 33:341–346. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Chen CC, Llado V, Eurich K, Tran HT and Mizoguchi E: Carbohydrate-binding motif in chitinase 3-like 1 (CHI3L1/YKL-40) specifically activates Akt signaling pathway in colonic epithelial cells. Clin Immunol. 140:268–275. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Johansen JS, Stoltenberg M, Hansen M, et al: Serum YKL-40 concentrations in patients with rheumatoid arthritis: relation to disease activity. Rheumatology (Oxford). 38:618–626. 1999. View Article : Google Scholar : PubMed/NCBI

40 

Malemud CJ: Matrix metalloproteinases (MMPs) in health and disease: an overview. Front Biosci. 11:1696–1701. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Tchetverikov I, Ronday HK, Van El B, et al: MMP profile in paired serum and synovial fluid samples of patients with rheumatoid arthritis. Ann Rheum Dis. 63:881–883. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Jungel A, Ospelt C, Lesch M, et al: Effect of the oral application of a highly selective MMP-13 inhibitor in three different animal models of rheumatoid arthritis. Ann Rheum Dis. 69:898–902. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Corvaisier M, Delneste Y, Jeanvoine H, et al: IL-26 is overexpressed in rheumatoid arthritis and induces proinflammatory cytokine production and Th17 cell generation. PLoS Biol. 10:e10013952012. View Article : Google Scholar : PubMed/NCBI

44 

Sedwick C: IL-26 kick-starts rheumatoid arthritis. PLoS Biol. 10:e10013982012. View Article : Google Scholar : PubMed/NCBI

45 

Gemelli C, Montanari M, Tenedini E, et al: Virally mediated MafB transduction induces the monocyte commitment of human CD34+ hematopoietic stem/progenitor cells. Cell Death Differ. 13:1686–1696. 2006.PubMed/NCBI

46 

Bakri Y, Sarrazin S, Mayer UP, et al: Balance of MafB and PU.1 specifies alternative macrophage or dendritic cell fate. Blood. 105:2707–2716. 2005. View Article : Google Scholar : PubMed/NCBI

47 

Sato T, Konomi K, Yamasaki S, et al: Comparative analysis of gene expression profiles in intact and damaged regions of human osteoarthritic cartilage. Arthritis Rheum. 54:808–817. 2006. View Article : Google Scholar : PubMed/NCBI

48 

Trenkmann M, Brock M, Gay RE, et al: Expression and function of EZH2 in synovial fibroblasts: epigenetic repression of the Wnt inhibitor SFRP1 in rheumatoid arthritis. Ann Rheum Dis. 70:1482–1488. 2011. View Article : Google Scholar : PubMed/NCBI

49 

de Rooy DP, Yeremenko NG, Wilson AG, et al: Genetic studies on components of the Wnt signalling pathway and the severity of joint destruction in rheumatoid arthritis. Ann Rheum Dis. 72:769–775. 2013.PubMed/NCBI

50 

Sen M, Reifert J, Lauterbach K, et al: Regulation of fibronectin and metalloproteinase expression by Wnt signaling in rheumatoid arthritis synoviocytes. Arthritis Rheum. 46:2867–2877. 2002. View Article : Google Scholar : PubMed/NCBI

51 

Sen M: Wnt signalling in rheumatoid arthritis. Rheumatology (Oxford). 44:708–713. 2005. View Article : Google Scholar : PubMed/NCBI

52 

Lu C, Xiao C, Chen G, et al: Cold and heat pattern of rheumatoid arthritis in traditional Chinese medicine: distinct molecular signatures indentified by microarray expression profiles in CD4-positive T cell. Rheumatol Int. 32:61–68. 2012. View Article : Google Scholar

53 

Maciejewska-Rodrigues H, Karouzakis E, Strietholt S, et al: Epigenetics and rheumatoid arthritis: the role of SENP1 in the regulation of MMP-1 expression. J Autoimmun. 35:15–22. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Kim KS, Choi HM, Lee YA, et al: Expression levels and association of gelatinases MMP-2 and MMP-9 and collagenases MMP-1 and MMP-13 with VEGF in synovial fluid of patients with arthritis. Rheumatol Int. 31:543–547. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Nakki A, Kouhia ST, Saarela J, et al: Allelic variants of IL1R1 gene associate with severe hand osteoarthritis. BMC Med Genet. 11:502010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2014
Volume 10 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang X, Xia S and Fu B: RNA‑seq analysis of synovial fibroblasts in human rheumatoid arthritis. Mol Med Rep 10: 241-247, 2014
APA
Wang, X., Xia, S., & Fu, B. (2014). RNA‑seq analysis of synovial fibroblasts in human rheumatoid arthritis. Molecular Medicine Reports, 10, 241-247. https://doi.org/10.3892/mmr.2014.2182
MLA
Wang, X., Xia, S., Fu, B."RNA‑seq analysis of synovial fibroblasts in human rheumatoid arthritis". Molecular Medicine Reports 10.1 (2014): 241-247.
Chicago
Wang, X., Xia, S., Fu, B."RNA‑seq analysis of synovial fibroblasts in human rheumatoid arthritis". Molecular Medicine Reports 10, no. 1 (2014): 241-247. https://doi.org/10.3892/mmr.2014.2182