Open Access

Screening of underlying genetic biomarkers for ankylosing spondylitis

  • Authors:
    • Xutao Fan
    • Bao Qi
    • Longfei Ma
    • Fengyu Ma
  • View Affiliations

  • Published online on: April 24, 2019     https://doi.org/10.3892/mmr.2019.10188
  • Pages: 5263-5274
  • Copyright: © Fan 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

Genetic biomarkers for the diagnosis of ankylosing spondylitis (AS) remain unreported except for human leukocyte antigen B27 (HLA‑B27). Therefore, the aim of the present study was to screen the differentially expressed genes (DEGs), and those that also possess differential single nucleotide polymorphism (SNP) loci in the whole blood of AS patients compared with healthy controls by integrating two mRNA expression profiles (GSE73754 and GSE25101) and SNP microarray data (GSE39428) collected from the Gene Expression Omnibus (GEO). Using the t‑test, 1,056 and 1,073 DEGs were identified in the GSE73754 and GSE25101 datasets, respectively. Among them, 234 DEGs were found to be shared in both datasets, which were subsequently overlapped with 122 differential SNPs of genes in the GSE39428 dataset, resulting in identification of two common genes [eukaryotic translation elongation factor 1 epsilon 1 (EEF1E1) and serpin family A member 1 (SERPINA1)]. Their expression levels were significantly upregulated and the average expression log R ratios of SNP sites in these genes were significantly higher in AS patients than those in controls. Function enrichment analysis revealed that EEF1E1 was involved in AS by influencing the aminoacyl‑tRNA biosynthesis, while SERPINA1 may be associated with AS by participating in platelet degranulation. However, only the genotype and allele frequencies of SNPs (rs7763907 and rs7751386) in EEF1E1 between AS and controls were significantly different between AS and the controls, but not SERPINA1. These findings suggest that EEF1E1 may be an underlying genetic biomarker for the diagnosis of AS.

Introduction

Ankylosing spondylitis (AS) is a common inflammatory rheumatic disease, with an estimated prevalence (per 10,000) of 23.8 in Europe, 16.7 in Asia, 31.9 in North America, 10.2 in Latin America and 7.4 in Africa (1). AS mainly affects the spine and sacroiliac joints in the pelvis to cause low back pain, stiffness and functional disability, which seriously influence the quality of life of patients and impose a heavy economic burden on both family and society (2). Therefore, there is a need for the timely diagnosis and effective treatment of AS.

Although the pathogenesis remains not clearly defined, accumulating evidence has suggested that AS is highly heritable. Human leukocyte antigen (HLA)-B27, a class I surface antigen encoded by B locus in the major histocompatibility complex (MHC) on the short (p) arm of chromosome 6, is one of the convincing genetic factors associated with AS (3). HLA-B27 was reported to be present in 94.3% of patients with AS, but only 9.34% in organ donors (4). The expression of HLA-B27 was found to be significantly higher in patients with AS than that in healthy subjects (5). Meta-analyses indicated that HLA-B27 genetic polymorphism B2704 and B2702 may be risk factors, while B2703, B2706, B2707, B2727, B2729 and B2747 may be protective factors for AS (6,7). HLA-B27-positive patients had a significantly younger age at symptom onset, more uveitis, and a higher frequency of peripheral and hip joint involvement than HLA-B27-negative patients (7,8). Thus, HLA-B27 has been the most commonly used biomarker for the diagnosis of AS (9). However, twin and family studies suggest that HLA-B27 only can explain less than 30% of the overall risk for AS (10,11), meaning there are other genes related with the genetic disorder of AS. Recently, scholars have also aimed to investigate other inflammatory biomarkers for AS, including interleukin (IL)-8 (12), tumor necrosis factor (TNF)-α (13), C-reactive protein (hsCRP) (14) and C-C motif chemokine 11 (CCL11) (15), but studies that have focused on the genetic biomarkers are limited (16,17).

The aim of the present study was to integrate the microarray data of mRNA and the single nucleotide polymorphism (SNP) expression profile in whole blood of AS patients and healthy controls to screen for differentially expressed genes (DEGs), and those that also possess differential SNP loci, which has not been previously performed. These SNP-related DEGs may be crucial genetic biomarkers for AS.

Materials and methods

Microarray data

Three microarray datasets under accession nos. GSE73754 (18), GSE25101 (19) and GSE39428 (20,21) were downloaded from the Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo/). GSE73754 (platform: GPL10558; Illumina HumanHT-12 V4.0 expression BeadChip) detected the gene expression profile in whole blood samples from 52 AS and 20 healthy controls; GSE25101 (platform: GPL6947; Illumina HumanHT-12 V3.0 expression BeadChip) compared the gene expression profile in whole blood samples between 16 AS and 20 healthy controls; and GSE39428 (GPL15779; Illumina custom human SNP VeraCode microarray) analyzed the SNPs in 384 genes of 51 AS and 163 healthy controls.

Data normalization

For the two expression data from the Illumina platform, the TXT. data were downloaded and preprocessed using the Linear Models for Microarray data (LIMMA) method (22) (version 3.34.0; http://www.bioconductor.org/packages/release/bioc/html/limma.html) in the Bioconductor R package (version 3.4.1; http://www.R-project.org/), including base-2 logarithmic (log2) transformation and quantile normalization. The SNP signal spectrum in the GSE39428 dataset was preprocessed using hidden Markov model (HMM)-based program PennCNV (23) (version 1.0.4; http://penncnv.openbioinformatics.org/en/latest/), including the following steps: i) the signal intensity of the A and B alleles in each SNP were extracted and quantile normalized using the quantile method; ii) the normalize_affy_geno_cluster.pl procedure in the PennCNV package was used to calculate the Log R ratio (LRR) and B allele frequency (BAF) in each SNP, resulting in the generation of baf. files; the kcolumn.pl procedure in the PennCNV package was utilized to split the baf. files to signal intensity of single sample; the copy number variation (CNV) was detected using the detect_cnv.pl procedure in the PennCNV package.

Differential analysis of mRNAs and SNPs

The DEGs between control and AS in the GSE73754 and GSE25101 datasets were identified using the LIMMA method (22) based on the t-test where statistical significance was set to |logFC(fold change)| >0.263 and Benjamini and Hochberg adjusted (24) false discovery rate (FDR) <0.05. Hierarchical clustering heatmap illustrating the expression intensity and direction of the common DEGs in two mRNA datasets was constructed using the pheatmap R package (version 1.0.8; http://cran.r-project.org/web/packages/pheatmap) based on Euclidean distance. The differential SNPs were screened by comparing the LRR between AS and controls by using the Student's t-test. The genotype and allele frequencies of SNPs in DEGs between AS and controls were also compared using the Chi-square test (or Fisher's exact test), with P-value <0.05 set as the threshold value.

PPI (protein-protein interaction) network construction

The interaction pairs of the common DEGs were retrieved from the STRING 10.0 (Search Tool for the Retrieval of Interacting Genes; http://string db.org/) database (25) and then the PPI network was visualized using the Cytoscape software (version 3.6.1; www.cytoscape.org/) (26). Four topological characteristics of the genes in the PPI network, including degree [the number of edges (interactions) of a node (protein)], betweenness centrality (BC, the number of shortest paths that run through a node), closeness centrality (CC, the average length of the shortest paths between one node and any other node in the network) and average path length (APL, the average of distances between all pairs of nodes), were calculated using the CytoNCA plugin in Cytoscape software (http://apps.cytoscape.org/apps/cytonca) (27), the overlapped genes of the top 35 in four parameters were suggested as crucial genes.

To identify functionally related and highly interconnected clusters from the PPI network, module analysis was carried out by using the Molecular Complex Detection (MCODE) plugin of Cytoscape software under the followed parameters: Degree cutoff =2, Node score cutoff =0.2 and K-core =2 (ftp://ftp.mshri.on.ca/pub/BIND/Tools/MCODE) (28).

Function enrichment analysis

The underlying functions of common DEGs between two mRNA datasets, genes in the PPI and modules enrichment analyses were predicted using the Database for Annotation, Visualization and Integrated Discovery (DAVID) online tool (version 6.8; http://david.abcc.ncifcrf.gov). P<0.05 was chosen as the threshold to determine the significantly enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Ontology (GO) terms which were visualized using R language.

Results

Identification of DEGs

Based on the threshold (FDR <0.05 and |logFC| >0.263), a total of 1,056 and 1,073 DEGs were identified between AS and controls for GSE73754 and GSE25101 datasets, respectively. After comparison analysis, 105 upregulated and 129 downregulated DEGs were found to be shared in both two datasets. The hierarchical clustering heatmap suggested that these 234 common DEGs could well distinguish AS from control samples (Fig. 1).

Function enrichment analysis for the common DEGs

DAVID database was used to predict the underlying functions of the common DEGs. The results showed that 8 significant KEGG pathways (Fig. 2) were enriched, such as hsa05130:Pathogenic Escherichia coli infection (TLR4, toll like receptor 4) and hsa04145:Phagosome (TLR4) (Table I). In addition, 23 significant GO biological process (BP) terms including GO:0006418~tRNA aminoacylation for protein translation (EEF1E1, eukaryotic translation elongation factor 1 epsilon 1; YARS, tyrosyl-tRNA synthetase), GO:0051092~positive regulation of NF-κB transcription factor activity (TLR4), GO:0050776~regulation of immune response (KLRD1, killer cell lectin like receptor D1), and GO:0032715~negative regulation of interleukin-6 production (TLR4); 6 significant GO molecular function (MF) terms, consisting of GO:0005515~protein binding (SERPINA1, serpin family A member 1; TLR4); and 6 significant GO molecular function (MF), such as GO:0005515~protein binding (SERPINA1, EEF1E1); 26 GO cell component (CC) terms, including GO:0070062~extracellular exosome (SERPINA1, EEF1E1), GO:0005737~cytoplasm (EEF1E1) and GO:0005829~cytosol (EEF1E1); were enriched (Fig. 3 and Table I).

Table I.

Function enrichment for the differentially expressed genes between patients with ankylosing spondylitis and controls.

Table I.

Function enrichment for the differentially expressed genes between patients with ankylosing spondylitis and controls.

CategoryTermP-valueGenes
KEGG_PATHWAYhsa05130:Pathogenic Escherichia coli infection9.97E-03ACTG1, TUBB, EZR, TLR4, TUBA1B
KEGG_PATHWAY hsa04145:Phagosome1.36E-02ACTG1, TUBB, NCF4, TLR4, FCGR2A, M6PR, TUBA1B, HLA-DRA
KEGG_PATHWAYhsa04650:Natural killer cell mediated cytotoxicity1.58E-02IFNAR2, TNFSF10, TNF, CD247, KLRD1, SH2D1B, HCST
KEGG_PATHWAYhsa00061:Fatty acid biosynthesis1.90E-02ACSL1, FASN, ACSL4
KEGG_PATHWAYhsa05164:Influenza A2.56E-02ACTG1, IFNAR2, TNFSF10, TNF, MAP2K4, TLR4, IVNS1ABP, HLA-DRA
KEGG_PATHWAY hsa05140:Leishmaniasis3.01E-02TNF, NCF4, TLR4, FCGR2A, HLA-DRA
KEGG_PATHWAYhsa00071:Fatty acid degradation3.63E-02ACSL1, ECHS1, ACSL4, ALDH9A1
KEGG_PATHWAYhsa01212:Fatty acid metabolism4.52E-02ACSL1, FASN, ECHS1, ACSL4
GOTERM_BP_ DIRECTGO:0007166~cell surface receptor signaling pathway5.44E-05CD8A, CD247, EVL, BIRC2, ADGRG1, IFNAR2, TNFSF10, ADRB2, KLRG1, NUP62, TDP2, CD81, CDA, KLRD1
GOTERM_BP_ DIRECTGO:0006418~tRNA aminoacylation for protein translation1.67E-03YARS, EEF1E1, AARS, EPRS, QARS
GOTERM_BP_ DIRECTGO:0043123~positive regulation of I-kappaB kinase/NF-kappaB signaling4.76E-03CARD11, TNFSF10, TNF, NUP62, PINK1, CXXC5, BIRC2, S100A12
GOTERM_BP_ DIRECTGO:0051092~positive regulation of NF-kappaB transcription factor activity7.34E-03CARD11, IRAK3, NLRC4, TNF, PRKCH, TLR4, S100A12
GOTERM BP_DIRECT_ GO:0050776~regulation of immune response8.11E-03CARD11, CD96, CD8A, CD247, CD81, KLRD1, SH2D1B, HCST
GOTERM_ BP_DIRECTGO:2001240~negative regulation of extrinsic apoptotic signaling pathway in absence of ligand1.17E-02TNF, ZC3HC1, MCL1, CX3CR1
GOTERM_BP_ DIRECTGO:0030890~positive regulation of B cell proliferation1.35E-02CARD11, CD81, TLR4, ADA
GOTERM_BP_ DIRECT GO:2000377~regulation of reactive oxygen species metabolic process2.66E-02TNF, PINK1, BIRC2
GOTERM_BP_ DIRECTGO:0071353~cellular response to interleukin-43.74E-02XBP1, FASN, TUBA1B
GOTERM_BP_ DIRECTGO:0032715~negative regulation of interleukin-6 production4.96E-02IRAK3, TNF, TLR4
GOTERM_MF_ DIRECTGO:0005515~protein binding3.50E-10PDLIM7, PPP2R5A, TLR1, CNOT2, TLR4, RNF216, CCT3, ARID1A, TGFA, SERPINA1
GOTERM_MF_ DIRECTGO:0044822~poly(A) RNA binding1.11E-04ABCF1, CCT3, ZNF207, EXOSC10, HNRNPM, EZR, FASN, APEX1, YARS, MDH2
GOTERM_MF_ DIRECTGO:0005524~ATP binding5.01E-03ABCF1, PINK1, MAP4K1, QARS, CCT3, TRIB1, ACTG1, EPRS, ADK, EIF4A1
GOTERM_MF_ DIRECTGO:0042288~MHC class I protein binding2.42E-02TUBB, CD8A, ATP5A1
GOTERM_MF_ DIRECT GO:0031625~ubiquitin protein ligase binding3.16E-02ACTG1, RPA2, TUBB, XBP1, SLC25A5, RALB, PINK1, TUBA1B, TRIB1
GOTERM_MF_ DIRECTGO:0047485~protein N-terminus binding3.59E-02RPA2, HDAC1, BIRC2, GLRX, FEZ1
GOTERM_CC_ DIRECT GO:0070062~extracellular exosome3.20E-06HIST2H2AA3, CAPZA2, PTGS1, CCT3, PDHB, RTN3, ACTG1, N4BP2L2, CCNY, LILRA5
GOTERM_CC_ DIRECT GO:0005737~cytoplasm1.87E-05ABCF1, C9ORF72, E2F3, PDLIM7, AGTPBP1, PPP2R5A, PTGS1, CNOT2, PINK1, SHOC2
GOTERM_CC_ DIRECT GO:0005829~cytosol1.39E-04ABCF1, AGTPBP1, CAPZA2, CNOT2, PINK1, DPH2, RNF216, QARS, ARHGAP17, CCT3
GOTERM_CC_ DIRECT GO:0030529~intracellular ribonucleoprotein complex2.73E-04ZFP36L2, HNRNPM, NUP62, CSNK1E, RPL22, SNRPB, EPRS, DYRK2, HNRNPR
GOTERM_CC_DIRECT GO:0016020~membrane1.212E-03ABCF1, KCNJ15, GNAI3, TNF, MCL1, PPP2R5A, CAPZA2, TLR1, CD247, CNOT2

[i] KEGG, Kyoto encyclopedia of Genes and Genomes; GO, Gene Ontology; BP, biological process; MF, molecular function; CC, cell component.

PPI network

After mapping the DEGs to the STRING database, 356 interaction pairs were obtained which were used for constructing the PPI network where 154 nodes (64 upregulated and 88 downregulated) were included (Fig. 4). By calculating degree, BC, CC and APL, and comparing genes ranked as the top 30, HDAC1 (histone deacetylase 1), YARS, EPRS (glutamyl-prolyl-tRNA synthetase), APEX1 (apurinic/apyrimidinic endodeoxyribonuclease 1), ACTG1 (actin γ 1), MDH2 (malate dehydrogenase 2), TNF (tumor necrosis factor), CCT3 (chaperonin containing TCP1 subunit 3), TLR4 (Toll-like receptor 4), TUBB (tubulin β class I), FCGR2A (Fc fragment of IgG receptor IIa), KLRD1 (killer cell lectin-like receptor D1) and FASN (fatty acid synthase) were found to be shared by these 4 topological characteristics, suggesting they were hub genes for AS (Tables II and III).

Table II.

Topological characteristics.

Table II.

Topological characteristics.

A, Degree

GenesValue
TNF24
EPRS19
ACTG117
YARS16
TLR414
HDAC114
CCT714
NOP5613
MDH213
IMP312
CCT312
EIF4A112
ATP5A111
POLR1C11
GNAI310
CS10
ATIC10
APEX19
NOP29
SNRPB9
CD2479
KLRD19
DDX478
AARS8
MCL18
SRSF58
TUBB8
FASN8
FCGR2A7

B, Closeness centrality

GenesValue

RNF1261.0000
KLHL21.0000
FBXO211.0000
GPBP11.0000
PLEKHF11.0000
RTN31.0000
RRAGD1.0000
TNF0.4000
HDAC10.3946
ACTG10.3852
CCT30.3605
YARS0.3596
EPRS0.3570
ALDH9A10.3510
FCGR2A0.3427
MDH20.3387
APEX10.3349
ADA0.3333
FASN0.3318
CS0.3288
ATIC0.3281
TLR40.3274
EZR0.3244
DDB10.3237
CCT70.3230
AARS0.3223
KLRD10.3216
TUBB0.3216
CYB5R40.3216

C, Betweenness centrality

GenesValue

TNF0.2996
ACTG10.2278
HDAC10.1883
YARS0.0824
EPRS0.0783
TLR40.0753
GNAI30.0745
CD2470.0743
APEX10.0688
RALB0.0622
ALDH9A10.0559
EIF4A10.0558
ADA0.0508
FASN0.0507
KLRD10.0499
TUBB0.0491
FCGR2A0.0430
MDH20.0382
EZR0.0360
CYB5R40.0359
CCT30.0337
PRKCH0.0316
MCL10.0315
NUP2140.0289
HIST2H2AA30.0288
SERPINA10.0286
TDP20.0282
SHOC20.0274
MAP4K10.0273

D, Average path length

GenesValue

RNF1261.0000
KLHL21.0000
FBXO211.0000
GPBP11.0000
PLEKHF11.0000
RTN31.0000
RRAGD1.0000
TNF2.5000
HDAC12.5342
ACTG12.5959
CCT32.7740
YARS2.7808
EPRS2.8014
ALDH9A12.8493
FCGR2A2.9178
MDH22.9521
APEX12.9863
ADA3.0000
FASN3.0137
CS3.0411
ATIC3.0479
TLR43.0548
EZR3.0822
DDB13.0890
CCT73.0959
AARS3.1027
KLRD13.1096
TUBB3.1096
CYB5R43.1096

Table III.

Overlapping DEGs according to topological features (degree, closeness centrality, betweenness centrality and average path length).

Table III.

Overlapping DEGs according to topological features (degree, closeness centrality, betweenness centrality and average path length).

Common genesExpression
HDAC1Down
YARSDown
EPRSDown
APEX1Down
ACTG1Down
MDH2Down
TNFDown
CCT3Down
TLR4Up
TUBBDown
FCGR2AUp
KLRD1Down
FASNDown

Subsequently, four functionally related and highly interconnected modules were screened (Fig. 5). The genes in module 1 were associated with aminoacyl-tRNA biosynthesis (YARS) (Fig. 5A); the genes in module 2 were related with natural killer cell mediated cytotoxicity (KLRD1) and immune response (KLRD1) (Fig. 5B); the genes in module 3 were relevant with metabolic pathways (EPRS) (Fig. 5C); and the genes in module 4 were enriched in GO terms of platelet degranulation (SERPINA1) (Fig. 5D) (Table IV).

Table IV.

Function enrichment for genes in modules.

Table IV.

Function enrichment for genes in modules.

CategoryTermP-valueGenes
1KEGG_PATHWAY hsa00970:Aminoacyl-tRNA biosynthesis8.99E-05YARS, AARS, QARS
GOTERM_BP_DIRECTGO:0006418~tRNA aminoacylation for protein translation3.31E-05YARS, AARS, QARS
GOTERM_BP_DIRECTGO:0006457~protein folding6.76E-04CCT7, AARS, CCT3
GOTERM_BP_DIRECTGO:1904871~positive regulation of protein localization to Cajal body1.90E-03CCT7, CCT3
GOTERM_BP_DIRECTGO:1904874~positive regulation of telomerase RNA localization to Cajal body3.57E-03CCT7, CCT3
GOTERM_BP_DIRECTGO:0032212~positive regulation of telomere maintenance via telomerase7.60E-03CCT7, CCT3
GOTERM_BP_DIRECTGO:0007339~binding of sperm to zona pellucida8.31E-03CCT7, CCT3
GOTERM_BP_DIRECTGO:1901998~toxin transport8.55E-03CCT7, CCT3
GOTERM_BP_DIRECTGO:0050821~protein stabilization3.20E-02CCT7, CCT3
2KEGG_PATHWAYhsa04650:Natural killer cell mediated cytotoxicity4.23E-03TNFSF10, CD247, KLRD1, HCST
KEGG_PATHWAYhsa03013:RNA transport1.10E-02NUP214, NUP62, EIF4A1, GEMIN4
GOTERM_BP_DIRECTGO:0007166~cell surface receptor signaling pathway3.33E-04TNFSF10, NUP62, CD247, BIRC2, KLRD1
GOTERM_BP_DIRECTGO:0016032~viral process4.64E-04NUP214, NUP62, HDAC1, CD247, EIF4A1
GOTERM_BP_DIRECTGO:0043066~negative regulation of apoptotic process2.21E-03MCL1, NUP62, HDAC1, BCL2A1, BIRC2
GOTERM_BP_DIRECTGO:0043123~positive regulation of I-kappaB kinase/NF-kappaB signaling1.70E-02TNFSF10, NUP62, BIRC2
GOTERM_BP_DIRECT GO:0050776~regulation of immune response2.06E-02CD247, KLRD1, HCST
GOTERM_BP_DIRECT GO:0043044~ATP-dependent chromatin remodeling2.84E-02HDAC1, SMARCA5
GOTERM_BP_DIRECTGO:0006364~rRNA processing2.90E-02EXOSC10, NOP56, GEMIN4
GOTERM_BP_DIRECTGO:0006409~tRNA export from nucleus3.93E-02NUP214, NUP62
GOTERM_BP_DIRECT GO:0010827~regulation of glucose transport4.05E-02NUP214, NUP62
GOTERM_BP_DIRECT GO:0097192~extrinsic apoptotic signaling pathway in absence of ligand4.17E-02MCL1, BCL2A1
3KEGG_PATHWAYhsa01100:Metabolic pathways1.94E-02ATIC, EPRS, ATP5A1, MDH2
GOTERM_BP_DIRECTGO:0006888~ER to Golgi vesicle-mediated transport1.32E-03TGFA, SERPINA1, PROS1
GOTERM_BP_DIRECT GO:0048566~embryonic digestive tract development5.70E-03TNF, ADA
GOTERM_BP_DIRECTGO:0048208~COPII vesicle coating2.16E-02TGFA, SERPINA1
4 GOTERM_BP_DIRECTGO:0002576~platelet degranulation3.62E-02SERPINA1, PROS1
GOTERM_BP_DIRECT GO:0000187~activation of MAPK activity3.76E-02TNF, TGFA
GOTERM_BP_DIRECTGO:0010951~negative regulation of endopeptidase activity4.25E-02SERPINA1, PROS1

[i] KEGG, Kyoto encyclopedia of Genes and Genomes; GO, Gene Ontology; BP, biological process.

Integration of SNP microarray and expression profile data

The LRR of each SNP for 384 genes in AS and control samples was computed. The LRR in most samples were lower than 1, indicating the presence of copy number deletions. Subsequently, the statistical difference in LRR of each SNP between AS and control samples were determined by Student's t-test, with 122 differential SNP identified. After overlapping the genes having differential SNP with the DEGs, two common genes (EEF1E1 and SERPINA1) were obtained. SERPINA1 was upregulated in AS (Fig. 6A) and the average expression LRR of the rs6575424 polymorphism in AS samples was significantly higher than that in the controls (0.05 vs. −0.14, P=6.57E-07) (Fig. 6B); EEF1E1 was also upregulated in AS (Fig. 6A) and the average expression LRRs of rs7763907 (−4.88 vs. −5.91, P=0.048), rs9328453 (0.07 vs. −0.12, P=3.69E-05) (Fig. 6B), rs7751386 (−0.85 vs. −1.49, P=2.52E-04), and rs12660697 (0.08 vs. −0.02, P=0.02) polymorphisms in AS samples were significantly higher than that in controls.

Furthermore, the genotype and allele frequencies of SNPs in EEF1E1 and SERPINA1 between AS and controls were compared using the Chi-square (or Fisher's exact) test. The results showed there were significant differences in the genotype and allele frequencies of rs7763907 between AS and control samples. The genotype frequency of rs7751386 between AS and control samples was also significantly differential. These findings suggest that these two polymorphic sites of the EEF1E1 gene may be associated with the susceptibility to acquire AS (Table V).

Table V.

Genotype and allele frequency of SNP loci for SERPINA1 and EEF1E1.

Table V.

Genotype and allele frequency of SNP loci for SERPINA1 and EEF1E1.

Genotype Allele


GenesSNP ASControlP-value ASControlP-value
SERPINA1rs6575424AA9120.077A25790.665
AB1667 B42151
BB2684
EEF1E1rs7763907AB10<0.001A440.047
BB135 B145
NC37154
AA04
rs9328453AB031.000A031.000
BB51163 B51166
rs7751386AA72<0.001A12411.000
AB539 B2580
BB2041
NC1981
rs12660697AA010.631A4100.749
AB49 B51162
BB47153

[i] SNP, single nucleotide polymorphism; AS, ankylosing spondylitis.

Discussion

In the present study, two crucial genes (EEF1E1 and SERPINA1) were identified for the diagnosis of ankylosing spondylitis (AS) by analyzing two mRNA expression profile datasets and one single nucleotide polymorphism (SNP) dataset. Their expression levels were significantly upregulated and the average expression LRRs of SNP sites in these genes were significantly higher in AS patients that those in the controls. EEF1E1 was involved in AS by influencing aminoacyl-tRNA biosynthesis, while SERPINA1 may be associated with AS by participating in platelet degranulation.

EEF1E1, also known as aminoacyl-tRNA synthetase-interacting multifunctional protein 3 (AIMP3/p18), was initially found to encode an auxiliary component of the macromolecular aminoacyl-tRNA synthase complex that catalyzes the ligation of a specific amino acid to its compatible cognate tRNA to form an aminoacyl-tRNA to initiate protein translation (29,30). Thus, EEF1E1 may be upregulated to promote the development of various types of cancer (31). However, recent studies indicate that EEF1E1 may also function as a tumor-suppressor (32,33) by upregulating the growth factor- or Ras-dependent induction of p53 (34,35). Cells with loss of EEF1E1 were found to exhibit impaired p53 transactivity and genomic instability and thus were found to became susceptible to cell malignant transformation (34,36), while overexpression of EEF1E1 induced cellular senescence phenotypes (37). It was also demonstrated that the p53 level was significantly higher in the peripheral blood supernatant of a rheumatoid arthritis (RA) group than the level in control groups and there was a positive correlation between p53 levels and the disease activity score in the RA group (38). In addition, in RA synovial tissues, 80% of p53-positive cells were found to be TUNEL-positive (39). These results indicate that upregulation of the p53 gene may result in chronic inflammation and apoptosis in RA patients. In addition, other members of the AIMP families, such as AIMP1, were also found to promote the expression of pro-inflammatory genes in monocytes/macrophages and dendritic cells (40) and induce cytokine (i.e. TNF-α)-dependent apoptosis (41). The antibody atliximab was reported to neutralize the expression of AIMP1 and then block the AIMP1-mediated production of inflammatory cytokines, ultimately attenuating collagen-induced arthritis (42). Accordingly, we speculate that EEF1E1 may also be involved in inflammation of AS by upregulating p53 and pro-inflammatory cytokines. In line with this hypothesis, our results showed that EEF1E1 was upregulated in the whole blood of AS patients compared with the control. Upregulation of EEF1E1 may be attributed to genetic mutations (rs7763907 and rs7751386) since the LRR of AS was significantly higher than that of controls and the genotype and allele frequencies were significantly different. However, further experimental validation is needed as studies investigating the SNPs of EEF1E1 are limited apart from the study of Liu et al that showed the number of risk alleles of rs12199241 in AIMP3 to be significantly associated with high DNA damage level (43).

SERPINA1 is a gene that encodes alpha-1-antitrypsin (AAT). It was found that the AAT concentration was higher in AS patients under active phase than the patients with remission/partial remission (44). In addition, the carboxyl terminal fragment of AAT was demonstrated to significantly induce the production of pro-inflammatory molecules (gelatinase B, monocyte chemoattractant protein-1 and IL-6) in human monocytes by interactions with the CD36 scavenger receptor and low density lipoprotein (LDL) receptor (45). These findings suggest that SERPINA1 may be a potential biomarker for the diagnosis of AS and evaluation of the efficacy of treatment by influencing inflammation. In line with these studies, we also found that SERPINA1 was upregulated in AS patients and it participated in GO terms of platelet degranulation. Platelet-specific degranulation gene Munc13-4 knockout mice were shown to display a reduction in airway hyper-responsiveness and eosinophilic inflammation, indirectly confirming the pro-inflammatory roles of SERPINA1 in AS (46). Importantly, a study was conducted to use TaqMan method to genotype tag SNPs (rs2753934, rs2749531 and rs6575424) in SERPINA1 of 56 AS cases and 160 healthy controls. The results revealed an increased expression of AAT in synovial membranes of AS compared with control samples, but no significant association was observed between the AAT polymorphism and AS (47). This also seems to be in accordance with our results and indicates that SERPINA1 may not be a genetically related biomarker for AS.

However, there were some limitations to the present study. First, this study was only performed to preliminarily screen the potential genetic biomarkers for AS. Further experiments are necessary, including clinical confirmation of the association between the polymorphism of EEF1E1 and SERPINA1 and the risk of AS and patient prognosis; clinical validation of the expression of EEF1E1 and SERPINA1; clinical (correlation analysis), in vitro (site-directed mutagenesis to construct the expression vector with different alleles, transfection of monocytes or osteoblasts followed by detection of cell proliferation, inflammatory factor release or mineralization) and in vivo (mutation knockout in animal models followed by assessment of histology and bone joint) verification of the association between gene polymorphisms and their expressions as well as corresponding phenotypic changes. Second, the SNP microarray used in this study only analyzed the SNPs in specific 384 genes, but not all the genes. Additional SNP discovery by deep sequencing with a larger sample size is essential to obtain more genetic biomarkers.

In conclusion, our findings preliminarily suggest that EEF1E1 may be an underlying novel, important genetic biomarker for the diagnosis of AS. Its rs7763907 and rs7751386 polymorphisms may lead to its upregulated expression and then promote the transcription of p53 and pro-inflammatory cytokines, leading to the development of AS.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The microarray data GSE73754 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE73754), GSE25101 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE25101) and GSE39428 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE39428) were downloaded from the GEO database in NCBI.

Authors' contributions

XF was involved in the conception and design, analysis and interpretation of data and drafted the initial manuscript. BQ collected the data. LM and FM contributed to the interpretation of the data. BQ, LM and FM revised the manuscript critically for important intellectual content. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Dean LE, Jones GT, MacDonald AG, Downham C, Sturrock RD and Macfarlane GJ: Global prevalence of ankylosing spondylitis. Rheumatology (Oxford). 53:650–657. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Healey EL, Haywood KL, Jordan KP, Garratt AM and Packham JC: Patients with well-established ankylosing spondylitis show limited deterioration in a ten-year prospective cohort study. Clin Rheumatol. 32:67–72. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Chen B, Li J, He C, Li D, Tong W, Zou Y and Xu W: Role of HLA-B27 in the pathogenesis of ankylosing spondylitis. Mol Med Rep. 15:1943–1951. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Fernández-Sueiro JL, Alonso C, Blanco FJ, Rodríguez-Gómez M, Galdo F and González-Gay MA: Prevalence of HLA-B27 and subtypes of HLA-B27 associated with ankylosing spondylitis in Galicia, Spain. Clin Exp Rheumatol. 22:465–468. 2004.PubMed/NCBI

5 

Cauli A, Dessole G, Fiorillo MT, Vacca A, Mameli A, Bitti P, Passiu G, Sorrentino R and Mathieu A: Increased level of HLA-B27 expression in ankylosing spondylitis patients compared with healthy HLA-B27-positive subjects: A possible further susceptibility factor for the development of disease. Rheumatology (Oxford) 41(12). 1375–1379. 2002. View Article : Google Scholar

6 

Yang T, Duan Z, Wu S, Liu S, Zeng Z, Li G, Wang S, Fan D, Ye D, Xu S, et al: Association of HLA-B27 genetic polymorphisms with ankylosing spondylitis susceptibility worldwide: A meta-analysis. Mod Rheumatol. 24:150–161. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Lin H and Gong YZ: Association of HLA-B27 with ankylosing spondylitis and clinical features of the HLA-B27-associated ankylosing spondylitis: A meta-analysis. Rheumatol Int. 37:1267–1280. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Kim TJ, Na KS, Lee HJ, Lee B and Kim TH: HLA-B27 homozygosity has no influence on clinical manifestations and functional disability in ankylosing spondylitis. Clin Exp Rheumatol. 27:574–579. 2009.PubMed/NCBI

9 

Zhang L, Zhang YJ, Chen J, Huang XL, Fang GS, Yang LJ, Duan Y and Wang J: The association of HLA-B27 and Klebsiella pneumoniae in ankylosing spondylitis: A systematic review. Microb Pathog. 117:49–54. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Brown MA, Kennedy LG, Macgregor AJ, Darke C, Duncan E, Shatford JL, Taylor A, Calin A and Wordsworth P: Susceptibility to ankylosing spondylitis in twins: The role of genes, HLA, and the environment. Arthritis Rheum. 40:1823–1828. 1997. View Article : Google Scholar : PubMed/NCBI

11 

Brophy S, Hickey S, Menon A, Taylor G, Bradbury L, Hamersma J and Calin A: Concordance of disease severity among family members with ankylosing spondylitis? J Rheumatol. 31:1775–1778. 2004.PubMed/NCBI

12 

Azevedo VF, Faria-Neto JR, Stinghen A, Lorencetti PG, Miller WP, Gonçalves BP, Szyhta CC and Pecoits-Filho R: IL-8 but not other biomarkers of endothelial damage is associated with disease activity in patients with ankylosing spondylitis without treatment with anti-TNF agents. Rheumatol Int. 33:1779–1783. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Gonza-Lezlopez L, Fajardo-Robledo NS, Saldaña-Cruz AM, Moreno-Sandoval IV, Bonilla-Lara D, Zavaleta-Muñiz S, Nava-Zavala AH, Hernandez-Cuervo P, Rocha-Muñoz A, Rodriguez-Jimenez NA, et al: Association of adipokines, interleukin-6, and tumor necrosis factor-α concentrations with clinical characteristics and presence of spinal syndesmophytes in patients with ankylosing spondylitis: A cross-sectional study. J Int Med Res. 45:1024–1035. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Sundström B, Ljung L and Wållberg-Jonsson S: Exercise habits and C-reactive protein may predict development of spinal immobility in patients with ankylosing spondylitis. Clin Rheumatol. 37:2881–2885. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Sohn DH, Jeong H, Roh JS, Lee HN, Kim E, Koh JH and Lee SG: Serum CCL11 level is associated with radiographic spinal damage in patients with ankylosing spondylitis. Rheumatol Int. 38:1455–1464. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Ruan WF, Xie JT, Jin Q, Wang WD and Ping AS: The diagnostic and prognostic role of interleukin 12B and interleukin 6R gene polymorphism in patients with ankylosing spondylitis. J Clin Rheumatol. 24:18–24. 2018.PubMed/NCBI

17 

Ma HJ, Yin QF, Wu Y and Guo MH: TNF-α-308 polymorphism determines clinical manifestations and therapeutic response of ankylosing spondylitis in Han chinese. Med Clin (Barc). 149:517–522. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Gracey E, Yao Y, Green B, Qaiyum Z, Baglaenko Y, Lin A, Anton A, Ayearst R, Yip P and Inman RD: Sexual dimorphism in the Th17 signature of ankylosing spondylitis. Arthritis Rheumatol. 68:679–689. 2015. View Article : Google Scholar

19 

Pimentel-Santos FM, Ligeiro D, Matos M, Mourão AF, Costa J, Santos H, Barcelos A, Godinho F, Pinto P, Cruz M, et al: Whole blood transcriptional profiling in ankylosing spondylitis identifies novel candidate genes that might contribute to the inflammatory and tissue-destructive disease aspects. Arthritis Res Ther. 13:R572011. View Article : Google Scholar : PubMed/NCBI

20 

Chang X, Xu B, Wang L, Wang Y, Wang Y and Yan S: Investigating a pathogenic role for TXNDC5 in tumors. Int J Oncol. 43:1871–1884. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Chang X, Zheng Y, Yang Q, Wang L, Pan J, Xia Y, Yan X and Han J: Carbonic anhydrase I (CA1) is involved in the process of bone formation and is susceptible to ankylosing spondylitis. Arthritis Res Ther. 14:R1762012. View Article : Google Scholar : PubMed/NCBI

22 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

23 

Wang K, Li M, Hadley D, Liu R, Glessner J, Grant SFA, Hakonarson H and Bucan M: PennCNV: An integrated hidden Markov model designed for high-resolution copy number variation detection in whole-genome SNP genotyping data. Genome Res. 17:1665–1674. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Benjamini Y, Drai D, Elmer G, Kafkafi N and Golani I: Controlling the false discovery rate in behavior genetics research. Behav Brain Res. 125:279–284. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, et al: STRING v10: Protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res 43(Database Issue). D447–D452. 2015. View Article : Google Scholar

26 

Kohl M, Wiese S and Warscheid B: Cytoscape: Software for visualization and analysis of biological networks. Methods Mol Biol. 696:291–303. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Tang Y, Li M, Wang J, Pan Y and Wu FX: CytoNCA: A cytoscape plugin for centrality analysis and evaluation of protein interaction networks. Biosystems. 127:67–72. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Bader GD and Hogue CW: An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics. 4:22003. View Article : Google Scholar : PubMed/NCBI

29 

Ku MJ and Lee SY: Contributions of aminoacyl-tRNA synthetase-interacting multifunctional protein-3 to mammalian translation initiation. Amino Acids. 44:1241–1245. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Kang T, Kwon NH, Lee JY, Park MC, Kang E, Kim HH, Kang TJ and Kim S: AIMP3/p18 controls translational initiation by mediating the delivery of charged initiator tRNA to initiation complex. J Mol Biol. 423:475–481. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Hassan MK, Kumar D, Naik M and Dixit M: The expression profile and prognostic significance of eukaryotic translation elongation factors in different cancers. PLoS One. 13:e01913772018. View Article : Google Scholar : PubMed/NCBI

32 

Yu X, Zheng H, Chan MT and Wu WK: HULC: An oncogenic long non-coding RNA in human cancer. J Cell Mol Med. 21:410–417. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Kim SS, Hur SY, Kim YR, Yoo NJ and Lee SH: Expression of AIMP1, 2 and 3, the scaffolds for the multi-tRNA synthetase complex, is downregulated in gastric and colorectal cancer. Tumori. 97:380–385. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Park BJ, Oh YS, Park SY, Choi SJ, Rudolph C, Schlegelberger B and Kim S: AIMP3 haploinsufficiency disrupts oncogene-induced p53 activation and genomic stability. Cancer Res. 66:6913–6918. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Han JM, Park BJ, Sang GP, Oh YS, Choi SJ, Sang WL, Hwang SK, Chang SH, Cho MH and Kim S: AIMP2/p38, the scaffold for the multi-tRNA synthetase complex, responds to genotoxic stresses via p53. Proc Natl Acad Sci USA. 105:11206–11211. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Gurung PM, Veerakumarasivam A, Williamson M, Counsell N, Douglas J, Tan WS, Feber A, Crabb SJ, Short SC, Freeman A, et al: Loss of expression of the tumour suppressor gene AIMP3 predicts survival following radiotherapy in muscle-invasive bladder cancer. Int J Cancer. 136:709–720. 2015.PubMed/NCBI

37 

Lee S, Yu KR, Ryu YS, Oh YS, Hong IS, Kim HS, Lee JY, Kim S, Seo KW and Kang KS: miR-543 and miR-590-3p regulate human mesenchymal stem cell aging via direct targeting of AIMP3/p18. Age (Dordr). 36:97242014. View Article : Google Scholar : PubMed/NCBI

38 

Abou-Shousha SA, Salah E and Wagdy E: Study of P53 in peripheral blood and synovial mononuclear cells of rheumatoid arthritis and osteoarthritis patients and its relation to the degree of disease activity. Egypt J Immunol. 12:61–70. 2005.PubMed/NCBI

39 

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. 2015. View Article : Google Scholar

40 

Liang D, Halpert MM, Konduri V and Decker WK: Stepping out of the cytosol: AIMp1/p43 potentiates the link between innate and adaptive immunity. Int Rev Immunol. 34:367–381. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Choi JW, Kim DG, Park MC, Um JY, Han JM, Park SG, Choi EC and Kim S: AIMP2 promotes TNFalpha-dependent apoptosis via ubiquitin-mediated degradation of TRAF2. J Cell Sci. 122:2710–2715. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Hong SH, Cho JG, Yoon KJ, Lim DS, Kim CH, Lee SW and Park SG: The antibody atliximab attenuates collagen-induced arthritis by neutralizing AIMP1, an inflammatory cytokine that enhances osteoclastogenesis. Biomaterials. 44:45–54. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Liu J, Zhu M, Chen W, Xie K, Shen W, Yuan J, Cheng Y, Geng L, Wang Y, Jin G, et al: Genetic variants in multisynthetase complex genes are associated with DNA damage levels in chinese populations. Mutat Res. 786:8–13. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Ozgocmen S, Godekmerdan A and Ozkurt-Zengin F: Acute-phase response, clinical measures and disease activity in ankylosing spondylitis. Joint Bone Spine. 74:249–253. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Janciauskiene S, Moraga F and Lindgren S: C-terminal fragment of alpha1-antitrypsin activates human monocytes to a pro-inflammatory state through interactions with the CD36 scavenger receptor and LDL receptor. Atherosclerosis. 158:41–51. 2001. View Article : Google Scholar : PubMed/NCBI

46 

Cardenas EI, Breaux K, Da Q, Flores JR, Ramos MA, Tuvim MJ, Burns AR, Rumbaut RE and Adachi R: Platelet Munc13-4 regulates hemostasis, thrombosis and airway inflammation. Haematologica. 103:1235–1244. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Sun S, Fang K, Zhao Y, Yan X and Chang X: Increased expression of alpha 1-anti-trypsin in the synovial tissues of patients with ankylosing spondylitis. Clin Exp Rheumatol. 30:39–44. 2012.PubMed/NCBI

Related Articles

Journal Cover

June-2019
Volume 19 Issue 6

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
Fan X, Qi B, Ma L and Ma F: Screening of underlying genetic biomarkers for ankylosing spondylitis. Mol Med Rep 19: 5263-5274, 2019
APA
Fan, X., Qi, B., Ma, L., & Ma, F. (2019). Screening of underlying genetic biomarkers for ankylosing spondylitis. Molecular Medicine Reports, 19, 5263-5274. https://doi.org/10.3892/mmr.2019.10188
MLA
Fan, X., Qi, B., Ma, L., Ma, F."Screening of underlying genetic biomarkers for ankylosing spondylitis". Molecular Medicine Reports 19.6 (2019): 5263-5274.
Chicago
Fan, X., Qi, B., Ma, L., Ma, F."Screening of underlying genetic biomarkers for ankylosing spondylitis". Molecular Medicine Reports 19, no. 6 (2019): 5263-5274. https://doi.org/10.3892/mmr.2019.10188