Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

The Clinical Significance of Interleukin–1 Receptor Antagonist +2018 Polymorphism in Rheumatoid Arthritis

  • Endom Ismail,

    Affiliation School of Bioscience & Biotechnology, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia

  • Omimah Khaled Jaber Nofal,

    Affiliation School of Bioscience & Biotechnology, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia

  • Rajalingham Sakthiswary ,

    sakthis5@hotmail.com

    Affiliation Faculty of Medicine, Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Cheras, Kuala Lumpur, Malaysia

  • Syahrul Sazliyana Shaharir,

    Affiliation Faculty of Medicine, Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Cheras, Kuala Lumpur, Malaysia

  • Radhika Sridharan

    Affiliation Faculty of Medicine, Department of Radiology, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Cheras, Kuala Lumpur, Malaysia

Abstract

Objective

Interleukin-1 receptor antagonist (IL-1Ra) acts as an inhibitor of IL-1; which is one of the culprit cytokines in rheumatoid arthritis (RA). Although +2018 polymorphism of IL-1Ra has been implicated in the pathogenesis of RA, its importance remains poorly understood. Hence, the purpose of this study was to determine the clinical significance of interleukin-1 receptor antagonist (IL-1Ra) +2018 polymorphism in RA.

Methods

Polymerase chain reaction (PCR) and sequencing were used to determine the genotypes of the IL-1Ra +2018 for 77 RA patients and 18 healthy controls. All RA patients were assessed for the disease activity score that includes 28 joints (DAS28) and radiographic disease damage based on Modified Sharp Score (MSS).

Results

The frequency of the T/T and C/T genotypes did not differ significantly (p = 0.893) between the RA patients and the controls. The C/T genotype had significantly higher mean disease activity (DAS 28) and disease damage (MSS) scores with p values of 0.017 and 0.004, respectively. Additionally, the ESR (erythrocyte sedimentation rate), CRP (C-reactive protein), the number of swollen and tender joints were higher for the C/T individuals. On multivariate analysis the CRP, swollen joint count and MSS remained significant with the following p values i.e. 0.045, 0.046 and less than 0.05.

Conclusions

C/T genotype of IL-1Ra +2018 prognosticates more aggressive disease in RA.

Introduction

Rheumatoid arthritis (RA) is a chronic systemic and inflammatory joint disease that leads to bone and cartilage destruction [1]. It is an autoimmune disease with a complex pathogenesis, with strong contribution by environmental and genetic factors [2].

Cytokines with polymorphic gene sequences have received a great deal of research interest in the recent years. One of the candidate cytokine genes is interleukin 1 (IL-1), which is composed of IL-1α, IL-1β and IL-1 receptor antagonist (IL-1Ra). Genes encoding this family are mapped on chromosome 2q14 [3]. Both IL-1α and IL-1β are potent pro-inflammatory mediators, implicated in human joint destruction [4]. In contrast, IL-1Ra is a naturally occurring anti-inflammatory molecule that inhibits the action of IL-1-induced pro-inflammatory activity [5] and therefore has been shown to prevent joint erosion and damage in RA [2].

Variable number of tandem repeats (VNTR) and +2018 polymorphisms of IL-1Ra have been investigated in several inflammatory diseases including rheumatoid arthritis, systemic lupus erythematosus, alopecia areata and ulcerative cholitis [68]. +2018 is a single nucleotide polymorphism (SNP), located in exon 2 of IL-1Ra gene at 2018 nucleotide position. The T allele was much more common in healthy human populations, compared to its other form i.e. the C allele [9, 10]. The C allele has been associated with many human diseases [1012], albeit data from Asian populations in this regard, remains scarce [13, 14]. The exact roles of the T and C alleles of the IL-1Ra gene in RA have not been fully elucidated. This has prompted us to embark on a study to determine the clinical significance of IL-1Ra +2018 polymorphism in Malaysian RA patients.

Methods

Study Design

This was a cross-sectional, case-control study involving RA patients who were under follow-up at the Universiti Kebangsaan Malaysia Medical Centre between January 2014 and May 2015. This study was approved by the Ethics Committee of the institution. All subjects gave written consent to participate in this study. We freshly recruited subjects for this study without using the same subjects of our previous study on interleukin 23 in RA [11]. The subjects were assessed for DAS28 (disease activity score based on 28 joints) and Stanford Health Assessment Questionnaire (HAQ) 8-item Disability Index (HAQ-DI). Seventy seven patients and 18 healthy controls voluntarily consented and were tested for +2018 genetic analysis. The hand radiographs of the subjects were scored using the Modified Sharp Score (MSS) by a radiologist who was blinded to the subjects.

DNA Extraction and Polymerase Chain Reaction

Genomic DNA was isolated from 200μl whole blood samples using Qiagen QIAamp DNA kit, following the manufacturer’s instructions. Genomic DNA was amplified by PCR using the respective forward 5'-GCCTCTTAACCATTGTCAGCC-3' and reverse 5'-GCCTTCTGGTATTTGCCCTT-3 primer following these conditions: initial pre-denaturation 95°C 5 minutes; 34 cycle of denaturation 95°C 1 minute, annealing 66°C 1 minute, extension 72°C 1 minute; and final extension 72°C 10 minutes.

DNA Purification and Sequencing

The PCR product was purified using the MEGAquick-spin Purification kit. Purified PCR products were sequenced by First BASE Laboratories. Genotype distribution was determined by repeating unit size of PCR products.

Statistical Analysis

The statistical analyses were performed using the Statistical Package for Social Sciences (SPSS) package for Windows version 21. Continuous variables were expressed as mean ± SD. Differences between genotype groups were analysed using the Fisher’s exact test for the categorical data whereas the independent t-test was used for continuous variables. The odds ratio (OR) was determined using binary logistic regression analysis for variables with significant p values (p < 0.05) on univariate analysis.

Results

Demographic Data of the Subjects

A total of 77 RA patients and 18 healthy controls were enrolled in this study. Female RA patients outnumbered the males; 69 (89.6%) versus 8 (10.4%). The mean age of the subjects with RA was 58.6 ± 10.3 years whereas for the controls was 36.4 ± 7.3 years. The demographic characteristics were matched between the RA patients and the controls (Table 1). Our study subjects were mainly of Malay, Chinese and Indian ethnicities reflecting the multiracial Malaysian population. There was a single RA patient who belonged to the Kadazan ethnic group which was categorised as ‘others’. Apart from age and gender, the distribution of the ethnic groups was matched between the RA patients and controls (p = 0.237) as ethnic variation can affect the genetic makeup. The frequency of the T/T and the C/T genotypes did not differ significantly (p = 0.893) between the RA patients and the controls. There was likewise no appreciable difference in the distribution of the allele frequencies.

thumbnail
Table 1. Sociodemographic data of the RA patients and the Controls.

https://doi.org/10.1371/journal.pone.0153752.t001

Comparison between the T/T and C/T Genotypes in RA

The C/T genotype had significantly higher mean disease activity (DAS 28) and disease damage (MSS) scores with p values of 0.017 and 0.004, respectively. Besides, the ESR(erythrocyte sedimentation rate), CRP (C-reactive protein), the number of swollen and tender joints were higher in the C/T genotype arm. The differences in the CRP and swollen joint count was statistically significant (p<0.050 and 0.033, respectively) (Table 2). The above differences were observed despite the matched frequency of seropositivity, disease duration and usage of medications between both the genotypes. On multivariate analysis (Table 3), the CRP, swollen joint count and MSS remained significant with the following p values: 0.045,0.046 and <0.05.

Discussion

Advances in genetic studies have lead to the discovery of a wide variety of genetic loci which may contribute to the development and course of RA [12]. HLA alleles, particularly HLA-DRB1 shared epitope alleles, have been shown to dominate the genetic risk in RA, while other less predominant loci includes the non-HLA variants [13] and multiple single nucleotide polymorphisms (SNPs) such as VEGFA [14], RANKL[15], MMP1-3 [14] and PTPN22 [16].

IL-1 signalling pathway is a critical cytokine pathway in RA which is involved in cell migration and inflammation [17]. The regulation and secretion of the cytokine has been demonstrated to be under genetic control through genetic polymorphism in their coding and promoter gene sequences [18]. Therefore, IL-1Ra polymorphism, has been postulated to have a pivotal role in RA [1922]. However, the data is still inconclusive owing to the conflicting findings of many of the studies in this regard [23].

We found no significant difference in the frequency of the IL-Ra C/T and T/T genotype expression between the RA patients and the controls. In keeping with our findings, Arman et al [24] revealed that their 94 RA patients and the 104 healthy controls had similar +2018 genotype distribution. The same finding was reported by another Asian study by Lee et al [10].

The novel findings of our study include the associations of the C/T genotype of IL-1Ra+2018 with significantly higher disease activity based on DAS28 scores and disease damage based on MSS scores. A few studies have consistently pointed out the role of IL-Ra gene polymorphism in the disease activity of RA. For instance, RA patients with persistent active disease, had significantly increased expression of IL-1RN*1 and IL-1RN*2 [25]. Apart from that, another study which involved Spanish RA patients also found a significant association between homozygosity for IL1RN*2 with increased number of affected joints [20]. In parallel with our findings, an Italian study demonstrated that their RA patients with C/T genotype of IL-1Ra had more aggressive disease with poor response to methotrexate therapy [22]. Polymorphisms in the regions of cytokine genes that affect transcription may influence cytokine production [26]. It is tempting to speculate that the higher disease activity among RA patients with C/T genotype of +2018 was due to increased production of IL-1 by monocytes. Unfortunately, data on the production of cytokines in relation to single chromosomal markers in the IL-1 locus are prone to experimental errors and hence, remain controversial.

Unlike disease activity, not many studies in the past have disclosed a link between IL-1Ra genotypes and the disease damage in RA. Recent studies by Gaafar et al [27], Cantagrel et al [28] and Buchs et al [29], for instance, failed to demonstrate that IL-1Ra polymorphism was predictive of more severe joint destruction. Kazkaz et al [30] found that tumour necrosis factor alpha promoter -238 but not IL-Ra +2018; was associated with joint destruction based on Larsen scores of the right wrist. However, Lubbe et al [31] disclosed that the +2018 “C”allele predicted higher Larsen scores which was consistent with our findings of higher disease damage with C/T genotype. Cox et al.[32] found 4 IL-1 gene cluster markers which were associated with joint erosions. It is plausible that the more severe joint destruction is due to the stimulation of bone resorption by IL-1 due to genetic linkage disequilibrium [33]

In conclusion, C/T genotype of +2018 prognosticates more aggressive disease in RA. The above genotype is associated with higher disease activity and more extensive radiographic joint damage. These findings, albeit interesting, have to be considered preliminary and will need to be confirmed by future genetic studies. However, owing to the polygenic aetiology, ethnic variations and complexity of the disease itself, there may be lack of reproducibility of genetic associations in RA.

Author Contributions

Conceived and designed the experiments: EI RS. Performed the experiments: ON R. Sakthiswary SSS R. Sridharan. Analyzed the data: ON R. Sridharan. Contributed reagents/materials/analysis tools: ON EI. Wrote the paper: R. Sakthiswary SSS. Assessment of the disease activity of the subjects: R. Sakthiswary.

References

  1. 1. Choy EH, Panayi GS. Cytokine pathways and joint inflammation in rheumatoid arthritis. N Engl J Med. 2001 Mar 22;344(12):907–16. pmid:11259725
  2. 2. John S, Worthington J. Genetic epidemiology. Approaches to the genetic analysis of rheumatoid arthritis. Arthritis Res. 2001;3(4):216–20. pmid:11438039
  3. 3. Cantagrel A, Navaux F, Loubet-Lescoulié P, Nourhashemi F, Enault G, Abbal M, et al. Interleukin-1β, interleukin-1 receptor antagonist, interleukin-4, and interleukin-10 gene polymorphisms: Relationship to occurrence and severity of rheumatoid arthritis. Arthritis & Rheumatism. 1999;42(6):1093–100.
  4. 4. Arend WP, Dayer JM. Inhibition of the production and effects of interleukin-1 and tumor necrosis factor alpha in rheumatoid arthritis. Arthritis Rheum. 1995 Feb;38(2):151–60. pmid:7848304
  5. 5. Steinkasserer A, Spurr NK, Cox S, Jeggo P, Sim RB. The human IL-1 receptor antagonist gene (IL1RN) maps to chromosome 2q14–q21, in the region of the IL-1α and IL-1β loci. Genomics. 1992;13(3):654–7. pmid:1386337
  6. 6. Blakemore AI, Tarlow JK, Cork MJ, Gordon C, Emery P, Duff GW. Interleukin-1 receptor antagonist gene polymorphism as a disease severity factor in systemic lupus erythematosus. Arthritis and rheumatism. 1994 Sep;37(9):1380–5. pmid:7945503
  7. 7. Carter MJ, Jones S, Camp NJ, Cox A, Mee J, Warren B, et al. Functional correlates of the interleukin-1 receptor antagonist gene polymorphism in the colonic mucosa in ulcerative colitis. Genes Immun. 2004 Jan;5(1):8–15. pmid:14735144
  8. 8. Huang CM, Wu MC, Wu JY, Tsai FJ. Interleukin-1 receptor antagonist gene polymorphism in chinese patients with systemic lupus erythematosus. Clinical rheumatology. 2002 Jun;21(3):255–7. pmid:12111633
  9. 9. Harrison P, Pointon JJ, Chapman K, Roddam A, Wordsworth BP. Interleukin-1 promoter region polymorphism role in rheumatoid arthritis: a meta-analysis of IL-1B-511A/G variant reveals association with rheumatoid arthritis. Rheumatology (Oxford). 2008 Dec;47(12):1768–70.
  10. 10. Lee YH, Kim HJ, Rho YH, Choi SJ, Ji JD, Song GG. Interleukin-1 receptor antagonist gene polymorphism and rheumatoid arthritis. Rheumatology international. 2004 May;24(3):133–6. pmid:14872281
  11. 11. Dalila AS, Mohd Said MS, Shaharir SS, Asrul AW, Low SF, Shamsul AS, et al. Interleukin-23 and its correlation with disease activity, joint damage, and functional disability in rheumatoid arthritis. Kaohsiung J Med Sci. 2014 Jul;30(7):337–42. pmid:24924839
  12. 12. Raychaudhuri S. Recent advances in the genetics of rheumatoid arthritis. Curr Opin Rheumatol. 2010 Mar;22(2):109–18. pmid:20075733
  13. 13. Eyre S, Bowes J, Diogo D, Lee A, Barton A, Martin P, et al. High-density genetic mapping identifies new susceptibility loci for rheumatoid arthritis. Nat Genet. 2012 Dec;44(12):1336–40. pmid:23143596
  14. 14. Chen Y, Mattey DL. Age at onset of rheumatoid arthritis: association with polymorphisms in the vascular endothelial growth factor A(VEGFA) gene and an intergenic locus between matrix metalloproteinase (MMP) 1 and 3 genes. Clin Exp Rheumatol. 2012 Nov-Dec;30(6):894–8. pmid:22776467
  15. 15. Tan W, Wu H, Zhao J, Derber LA, Lee DM, Shadick NA, et al. A functional RANKL polymorphism associated with younger age at onset of rheumatoid arthritis. Arthritis Rheum. 2010 Oct;62(10):2864–75. pmid:20533289
  16. 16. Karlson EW, Chibnik LB, Cui J, Plenge RM, Glass RJ, Maher NE, et al. Associations between human leukocyte antigen, PTPN22, CTLA4 genotypes and rheumatoid arthritis phenotypes of autoantibody status, age at diagnosis and erosions in a large cohort study. Ann Rheum Dis. 2008 Mar;67(3):358–63. pmid:17666451
  17. 17. Choy E. Understanding the dynamics: pathways involved in the pathogenesis of rheumatoid arthritis. Rheumatology (Oxford). 2012 July 1, 2012;51(suppl 5):v3–v11.
  18. 18. Bidwell J, Keen L, Gallagher G, Kimberly R, Huizinga T, McDermott MF, et al. Cytokine gene polymorphism in human disease: on-line databases. Genes Immun. 1999 Sep;1(1):3–19. pmid:11197303
  19. 19. Kapetanovic MC, Lindqvist E, Sturfelt G, Saxne T, Truedsson L, Eberhardt K. The impact of IL-1Ra and MBL gene polymorphisms on joint damage after 5 and 10 years in patients with early rheumatoid arthritis. J Rheumatol. 2007 Apr;34(4):894–5. pmid:17407253
  20. 20. Carreira PE, Gonzalez-Crespo MR, Ciruelo E, Pablos JL, Santiago B, Gomez-Camara A, et al. Polymorphism of the interleukin-1 receptor antagonist gene: a factor in susceptibility to rheumatoid arthritis in a Spanish population. Arthritis Rheum. 2005 Oct;52(10):3015–9. pmid:16200608
  21. 21. Huang CM, Tsai FJ, Wu JY, Wu MC. Interleukin-1beta and interleukin-1 receptor antagonist gene polymorphisms in rheumatoid arthritis. Scand J Rheumatol. 2001;30(4):225–8. pmid:11578018
  22. 22. Tolusso B, Pietrapertosa D, Morelli A, De Santis M, Gremese E, Farina G, et al. IL-1B and IL-1RN gene polymorphisms in rheumatoid arthritis: relationship with protein plasma levels and response to therapy. Pharmacogenomics. 2006 Jul;7(5):683–95. pmid:16886894
  23. 23. Xue J ML, Lu X, Wu H. No association of interleukin-1 receptor antagonist VNTR polymorphism and rheumatoid arthritis susceptibility: a meta analysis. Clin Exp Rheumatol 2010;28(5):654–60. pmid:20863445
  24. 24. Arman A, Yilmaz B, Coker A, Inanc N, Direskeneli H. Interleukin-1 receptor antagonist (IL-1RN) and interleukin-1B gene polymorphisms in Turkish patients with rheumatoid arthritis. Clin Exp Rheumatol. 2006 Nov-Dec;24(6):643–8. pmid:17207379
  25. 25. Swellam M, Gabal KMA, Youssef SS. Interleukin-1 receptor antagonist gene polymorphism and hepcidin in rheumatoid arthritis: Correlations with clinical and laboratory indices of disease activity. IUBMB Life. 2013;65(10):883–8. pmid:24014495
  26. 26. Danis VA, Millington M, Hyland VJ, Grennan D. Cytokine production by normal human monocytes: inter-subject variation and relationship to an IL-1 receptor antagonist (IL-1Ra) gene polymorphism. Clin Exp Immunol. 1995 Feb;99(2):303–10. pmid:7851026
  27. 27. Gaafar T, Bayoumi F, El-Senousy W, Raafat H, Sherif M, Morcos B. Influence of Interleukine-1 Beta and Interleukine-1 Receptor Antagonist Genes Polymorphism on Rheumatoid Arthritis. World Applied Sci J. 2013;27(5):574–84.
  28. 28. Cantagrel A, Navaux F, Loubet-Lescoulie P, Nourhashemi F, Enault G, Abbal M, et al. Interleukin-1beta, interleukin-1 receptor antagonist, interleukin-4, and interleukin-10 gene polymorphisms: relationship to occurrence and severity of rheumatoid arthritis. Arthritis Rheum. 1999 Jun;42(6):1093–100. pmid:10366101
  29. 29. Buchs N, di Giovine FS, Silvestri T, Vannier E, Duff GW, Miossec P. IL-1B and IL-1Ra gene polymorphisms and disease severity in rheumatoid arthritis: interaction with their plasma levels. Genes Immun. 2001 Jun;2(4):222–8. pmid:11477478
  30. 30. Kazkaz L, Marotte H, Hamwi M, Angelique Cazalis M, Roy P, Mougin B, et al. Rheumatoid arthritis and genetic markers in Syrian and French populations: different effect of the shared epitope. Ann Rheum Dis. 2007 Feb;66(2):195–201. pmid:17068065
  31. 31. Lubbe S, Tikly M, van der Merwe L, Hodkinson B, Ramsay M. Interleukin-1 receptor antagonist gene polymorphisms are associated with disease severity in Black South Africans with rheumatoid arthritis. Joint Bone Spine. 2008 Jul;75(4):422–5. pmid:18468936
  32. 32. Cox A, Camp NJ, Cannings C, di Giovine FS, Dale M, Worthington J, et al. Combined sib-TDT and TDT provide evidence for linkage of the interleukin-1 gene cluster to erosive rheumatoid arthritis. Hum Mol Genet. 1999 Sep;8(9):1707–13. pmid:10441334
  33. 33. Mino T, Sugiyama E, Taki H, Kuroda A, Yamashita N, Maruyama M, et al. Interleukin-1alpha and tumor necrosis factor alpha synergistically stimulate prostaglandin E2-dependent production of interleukin-11 in rheumatoid synovial fibroblasts. Arthritis and rheum. 1998 Nov;41(11):2004–13.