Next Article in Journal
Homozygous Missense Variant in the Solute Carrier Organic Anion Transporter 2A1 (SLCO2A1) Gene Underlies Isolated Nail Clubbing
Previous Article in Journal
Networks as Biomarkers: Uses and Purposes
Previous Article in Special Issue
Vitamin D Receptor (VDR) Genetic Variants: Relationship of FokI Genotypes with VDR Expression and Clinical Disease Activity in Systemic Lupus Erythematosus Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

MIR149 rs2292832 and MIR499 rs3746444 Genetic Variants Associated with the Risk of Rheumatoid Arthritis

1
Department of Biotechnology, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
2
School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong 999077, China
3
Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
4
King Abdulaziz City for Science and Technology, Riyadh 12354, Saudi Arabia
5
Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
6
Department of Biotechnology, Faculty of Biological Sciences, University of Malakand, Chakdara 18800, Pakistan
7
Peshawar Medical College, Peshawar 25160, Pakistan
8
Department of Medicine, Lady Reading Hospital (LRH), Peshawar 25000, Pakistan
9
Department of Zoology, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
*
Authors to whom correspondence should be addressed.
Genes 2023, 14(2), 431; https://doi.org/10.3390/genes14020431
Submission received: 30 November 2022 / Revised: 1 January 2023 / Accepted: 19 January 2023 / Published: 8 February 2023
(This article belongs to the Special Issue New Insights into the Genetics of Rheumatic Diseases)

Abstract

:
Introduction: MicroRNAs (miRNAs) are small non-coding RNAs that play a key role in post-transcriptional modulation of individual genes’ expression. Several miRNA variants from different populations are known to be associated with an increased risk of rheumatoid arthritis (RA). Aim: This study was undertaken with the aim to investigate the association of single nucleotide variants; namely, rs2292832, rs3746444, rs11614913, rs1044165, and rs767649 of MIR149, MIR499, MIR196, MIR223, and MIR155, respectively, with RA in the Pakistani population. Methods: A case-control study was performed by recruiting and genotyping a total of 600 individuals (300 cases and 300 controls) for these five variants using a TaqMan single-nucleotide polymorphism (SNP) genotyping assay. The resultant genotypic data was statistically analyzed through a chi-squared test for its association with RA under different inheritance models. Results: We found a significant association of rs2292832 with RA at genotypic (co-dominant (p < 0.0001), dominant (CC vs. TT + CT: OR 2.063 (1.437–2.962); p = 0.0001), recessive (TT vs. CT + CC: OR 0.376 (0.259–0.548); p < 0.0001)), and allelic (allele C) levels ((OR 0.506 (0.402–0637); p < 0.0001)). Similarly, the rs3746444 showed a significant association with RA under co-dominant (p = 0.0001), dominant (GG vs. AA + AG: OR 5.246 (3.414–8.061); p < 0.0001), recessive (AA vs. GG + AG: OR 0.653 (0.466–0.916); p = 0.014), and additive models (G vs. A; OR 0.779 (0.620–0.978); p = 0.03). However, we did not observe any significant association of rs11614913, rs1044165, or rs767649 with RA in our subjects. Conclusion: To our knowledge, this was the first study that investigated and found an association between functional polymorphisms in miRNAs and RA in the Pakistani population.

1. Introduction

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease that affects 0.5–1% of the world population with regional variation. The rate of occurrence is three times higher in women than in men [1]. Because it is a chronic immune-mediated inflammatory disease, it results in progressive joint damage, cartilage destruction, and bone and synovial hyperplasia—ultimately leading to extra-articular manifestation and multiple co-morbidities [2,3]. The etiology of RA is complex, and several genetic, environmental, hormonal, infectious, and lifestyle factors that contribute to the pathogenies of RA have been identified [4]. The dynamic interactions between the environment, genotype, and phenotype are likely to be mediated by the epigenetic process [5]. Accumulating manifestations have suggested that aberrant epigenetic alterations play a significant role in the dysregulation of T cells and hence contribute to the emergence of RA [6]. Over the last few decades, more than 150 SNPs present in over 70 genes/loci have been reported for RA through genome-wide association studies (GWAS) and candidate-gene approaches [7,8,9,10,11]. Some of these variants were found in microRNAs (miRNAs), which are an important class of epigenetic factors involved in many autoimmune diseases that include RA [12,13].
MicroRNAs, which are approximately 20–22 nucleotides long, are a class of small non-coding RNAs involved in the post-transcriptional regulation of gene expression [14] in diverse cellular and metabolic pathways; they exert their effects by binding to and silencing their target mRNA [15,16]. The single nucleotide changes in miRNA sequences might change miRNA–mRNA target interaction, thereby deregulating or dysregulating the expression of target genes [17]. Due to their key roles in regulating various biological processes such as cell proliferation, apoptosis, metabolism, stress resistance, the cell cycle, chromosome architecture, stem cell maintenance, etc., their aberrant expression and function have been implicated in a variety of human diseases that include cancer and autoimmune disorders [18,19]. The deregulation of miRNAs results in the abnormal behavior of a variety of cells (T lymphocytes and peripheral blood mononuclear resident cells), which causes inflammation that leads to the degradation of the extracellular matrix (ECM) and damage to synovial fibroblasts and osteoclasts (key effector cells of joint destruction in RA) [12,20]. However, several studies have determined—with conflicting results—the correlation between miRNA variants (rs2292832, rs3746444, rs11614913, and rs767649) and the risk of rheumatoid arthritis [21,22,23,24,25,26,27,28]. Upon successful establishment of their roles, miRNAs can serve as potential therapeutic targets for these conditions [12,13]
For successful treatment, early diagnosis plays an important role, especially in patients with risk of poor prognosis (such as high disease activity, the presence of autoantibodies, and early joint damage) [29]. Although it is now possible to diagnose RA using standard diagnostic criteria, for many the early diagnosis is still undifferentiated arthritis at the time of first presentation in the absence of suitable biomarkers—often, this is the most challenging to find [30]. Accumulating evidence of the role of miRNAs in RA and their aberrant expression in the inflamed synovium and circulation of such patients [31] holds promise for their potential use as novel molecular diagnostic markers for RA [32].
Given the role of miRNAs in autoimmune diseases, particularly RA, we investigated miRNA variants (rs2292832, rs3746444, rs11614913, rs1044165, and rs767649 of MIR149, MIR499, MIR196, MIR223, and MIR155, respectively) for their association with the risk of RA in the Pakistani population.

2. Materials and Methods

2.1. Ethical Approval of the Study

Before enrolling in the study, informed consent was obtained from the patients and controls. Approval for the study was obtained from the ethical committee of Abdul Wali Khan University Mardan (AWKUM) and Lady Reading Hospital (LRH), Peshawar, Pakistan.

2.2. Study Population

The study included 600 individuals: 300 cases (mean age 41.3 ± 13.75) and 300 controls (mean age 41.6 ± 12.3 years). Samples of RA cases were recruited from LRH, Peshawar, Pakistan. RA was diagnosed according to the criteria of the ACR/European League Against Rheumatism (EULAR) 2010 [33]. The control group consisted of healthy, age-matched subjects with no history of autoimmunological diseases. The clinical characteristics of RA and controls are summarized in Table 1.

2.3. Genomic DNA Extraction and Genotyping of Selected SNPs Using TaqMan Assay

Venous blood samples were collected from all of the study subjects via venipuncture. Genomic DNA was isolated from the peripheral blood of controls and patients using an organic phenol–chloroform procedure, and the samples were stored at −80° for future analysis [34]. The rs2292832 in MIR149, rs3746444 in MIR499, rs11614913 in MIR196, rs1044165 in MIR223, and rs767649 in MIR155 were genotyped using a TaqMan SNP genotyping assay. DNA amplification was performed in 384-well plates using an ABI Q6 system (Applied Biosystems, Waltham, MA, USA).

2.4. Statistical Analysis

A chi-squared test was used to determine the Hardy–Weinberg equilibrium (HWE) status of the obtained SNP genotypes. The association between the SNPs and the risk of RA was evaluated using the odds ratio (OR) at a 95% confidence interval (95%CI). Genotype and allele frequencies were determined using direct counting and compared between patients and controls by using chi-squared and Fisher’s exact tests. All co-dominant, homozygous dominant, homozygous recessive, heterozygous, and additive models were used to assess the risk of RA. The genotypes were compared to obtain the p-value for all three SNP genotypes. A p-value of 0.05 was set as the statistical significance.

2.5. In Silico Analysis of miRNAs’ Primary Structures

The following list provides information regarding the screened MIR149 rs2292832, MIR499 rs3746444, and MIR196 rs11614913, as well as their gene names, mature miRNAs, respective chromosome positions, coded alleles, other alleles, and minor allele frequencies, can be found in Table 2. Figure 1 illustrate the positions of the seed region and rs3746444 in the hsa-mir-499-3p. A study was conducted to determine whether the rs2292832, rs3746444, and rs11614913 variants affected the primary structure of MIR149, MIR499, and MIR196, respectively. The RNAeval web server was used to calculate the energies of the RNA sequences. The Vienna RNA website was used to examine RNA secondary and centroid structures.

3. Results

3.1. Genetics Analysis

The genotype distribution of miRNA variants with an RA risk was determined using co-dominant, homozygous dominant, homozygous recessive, heterozygous, and additive models (see Table 3).
The genotype distribution for SNP rs2292832 in MIR149 was significant with RA in the co-dominant (χ2 = 31.23; p < 0.0001), homozygous dominant (CC vs. TT + CT; OR 2.063 (1.437–2.962); p = 0.0001), and homozygous recessive (TT vs. CT + CC; OR 0.376 (0.259–0.548); p < 0.0001) models, as well as in the distribution of allele C in the additive model (C vs. T; OR 0.506 (0.402–0637); p< 0.0001). Similarly, the genotype distribution for SNP rs3746444 in MIR499 showed a significant association with RA in the co-dominant (χ2 = 59.19; p = 0.0001), homozygous dominant (GG vs. AA + AG; OR 5.246 (3.414–8.061); p < 0.0001), homozygous recessive (AA vs. GG + AG; OR 0.653 (0.466–0.916); p = 0.014), and additive models (G vs. A; OR 0.779 (0.620–0.978); p = 0.03). The genotypic distribution of “rs11614913” in MIR196 was more or less random across cases and controls and showed no significant association with RA in the co-dominant (χ2 = 2.35; p = 0.30), homozygous dominant (CC vs. TT + CT; OR 0.930 (0.667–1.298); p = 0.73), homozygous recessive (TT vs. CC + CT; OR 0.826 (0.563–1.213); p = 0.38), and additive models (C vs. T; OR 1.049 (0.834–1.318); p = 0.72). This was also the case for “rs1044165” in MIR223, which showed no meaningful association in the co-dominant model (χ2 = 2.36; p = 0.30), homozygous dominant model (GG vs. AA + AG; OR 1.116 (0656–1.896); p = 0.78), homozygous recessive model (OR 0.368 (0.096–1.404); p = 0.22), or additive model (A vs. T; OR 0.788 (0.488–1.274); p = 0.39). In the same way, no significant differences in the distribution of the genotype for rs767649 in MIR155 across the cases and controls were observed in the co-dominant (χ2 = 4.11; p = 0.12), homozygous dominant (TT vs. CC + CT; OR 1.319 (0.925–1.88); p = 0.14), homozygous recessive (CC vs. TT + CT; OR 2.02 (0.925–8.157); p = 0.50), or additive models (C vs. T; OR 0.833 (0.607–1.144); p = 0.29).

3.2. In Silco Analysis of miRNAs

The rs374644 variant was located in the seed region AC (A/G) UCAC at the position of 20;34,990,448 in the mature sequence of has-mi-499a-3p. The miRNA loop structure increased by one base pair due to this change. The thermodynamic Structure Prediction tool was used to compare the normal and mutant primary miRNA structures, and alterations were observed for the parameters being studied, as shown in Table 4. In the primary structure of miRNA, the rs11614913 variant in MIR196A2 resulted in the substitution of cytosine for uracil. The effect of rs2292832 on the secondary structure of pre-miR-149 was studied using the RNAWeb Server. The precursor miR-149 with and without the rs2292832 variant was modeled to determine the minimum free energy (MFE) and centroid structures. Based on the different alleles of rs2292832, no structural modification was observed for MIR149.

4. Discussion

Rheumatoid arthritis (RA) is characterized as a vital autoimmune disease that hampers the normal physical activities of patients [35,36]. RA causes inflammation in joints, which results in damage to the articular cartilage along with synovial hyperplasia, ultimately leading to consistent pain and permanent disability. The heritability of RA has been reported to be about 60%, whereas the remaining 40% could be due to environmental and other factors [37]. Genome-wide association studies (GWAS) are a common method for elucidating the underlying genetic risk factors. The major histocompatibility complex (MHC) and non-MHC loci have been identified to account for ~23% of the genetic risk factors for RA, which indicates that there are still undiscovered genetic risk factors [7]. Association studies have reported >150 SNPs located at more than 70 gene loci, which provides a glimpse of the underlying genetic factors [7,8,38,39].
The exact etiology and pathogenesis of RA, which are highly complex and involve genetic susceptibility, environmental factors, and epigenetic changes, are still incomplete. miRNAs play a crucial role in maintaining the normal functions of the immune system. In recent decades, research has revealed that dysregulated miRNAs play a role in autoimmune disease pathogenesis because they regulate a wide range of cellular functions at the post-transcriptional level [40]. Recent studies revealed that the occurrence of autoimmune illnesses, particularly the emergence of RA, may be influenced by epigenetic processes such as histone changes, DNA methylation, and microRNAs [41]. Small non-coding miRNAs are the important components of complex genetic regulatory networks. The SNPs or mutations that occur in the miRNA gene region can affect the properties of miRNAs and lead to a hereditary defect in the protein’s translation of a target mRNA [42].
In this study, a population-based association of rs2292832, rs3746444, rs11614913, rs1044165, and rs767649 genotypes with RA was investigated in the Pakistani population. The SNPs rs2292832 in MIR149 and rs3746444 in MIR499 were found to be significantly associated with RA in this population. The gene for MIR149 was located at 2q37.3 and contains one exon which code MIR149 and showed polymorphisms. The pre-miRNA SNP rs2292832 in MIR149 is thought to contribute to RA pathogenesis. The current study found a significant (p < 0.0001) association of rs2292832 in MIR149 with RA in the Pakistani population. Similarly, a case-control study (200 RA patients and 120 controls) that was conducted in the eastern Chinese Han population by Xiao et al. using the RFLP-PCR method reported a significant association with RA at the genotypic and allelic levels [21]. Table 5 shows a list of manuscripts referred in this study.
The rs4746444 in MIR499 is located within the mature miRNA region, thereby altering the pre-miRNA maturation process and binding sites for target mRNA [43]. The MIR499 polymorphism rs3746444 was reported to be involved in RA risk. Several studies have shown that HLADRB1 alleles that encode shared epitope (SE) and STAT4 genes were significantly associated with RA. A target of MIR499 called regulatory factor X4 (RFX4) can influence HLADRB1 expression. Interestingly, we reported in a previous study that HLADRB1 rs6660895 and STAT4 rs7574865 were strongly associated with RA in the Pakistani population (Jalil et al. 2013). Another target was the PADIi4 gene, which encodes the PADI4 enzyme. The synovial tissue and hematological cells of RA patients contained PADI4 mRNA, and it was highly overexpressed in the blood of RA patients. Additionally, the PADI4 enzyme generates autoantigens, which are citrullinated peptides that are specifically recognized by anti-CCP [44]. Inhibition of citrullination occurs when arginine is deaminated to citrulline in the presence of calcium ions [45]. As a result, MIR499 might play a significant role in the pathogenesis and severity of RA.
Previously, the association of rs3746444 in the MIR499 study was conducted in several populations and demonstrated a positive association with RA. For the first time, MIR499 rs3746444 was reported by Yang et al. to be associated with RA in the Chinese Han population [22]. Another case-control study that was conducted by Hashemi et al. found a significant association with RA in the Iranian population [23]. Amal et al. showed a significant association of rs3746444 in MIR499 with RA in Egyptian females [24], and Fatah et al. showed a positive association with RA in the Egyptian population [25]. Ayeldeen et al. also found a significant association with RA in the Egyptian population [46]. Recently, rs3746444 in MIR499 was genotyped in 550 RA patients and 550 controls of Pakistani origin; the study reported a significant association [47]. Rs3746444 in MIR499 was significantly associated with the genotypic (p = 0.0001, p < 0.05) and allelic levels with RA in this study. All these studies were in close agreement with the previous ones that established the association of rs3746444 in MIR499 with RA.
MIR196a2 is located on chromosome 12q13.13 and is expressed from an intergenic region in HOX gene clusters. Rs11614913 is located at the passenger strand near the processing site at nucleotide 78. Only the expression of MIR196a2 has been deregulated in RA synovial fibroblasts (RASFs) with no previous association study of its variants with RA susceptibility [48]. The association of rs11614913 in an MIR196a2 study was conducted for the first time in the Egyptian population by Toraih et al.; the researchers found an insignificant association with RA susceptibility [26]. Another extended study was conducted by Aleman-Avila et al. that found no association of rs11614913 in MIR196a2 in the Mexican population [27]. The association of rs11614913 in MIR-196a2 in the Pakistani population was found to be insignificant, and our results conformed to the previous results of Toraih et al., 2016 and Aleman-Avila et al., 2017.
The rs767649 in the pre-MIR155 promoter region, which affects nuclear factor-kappa B (NF-kB) binding as well as several regulatory factors attached to MIR155, has been shown to be involved in RA pathogenesis [49]. The association of rs7676649 in MIR155 in the Egyptian population was reported by Shaker et al., who established a positive association between RA patients and controls at the genotypic and allelic levels [28]. In this study, we did not find a positive association of rs7676649 in MIR155 between the RA and control samples from in the Pakistani population.
The current study had some limitations. This was a single-cohort-based study; thus, the findings need to be validated in a larger extended cohort with demographic characteristics. Additionally, the current study focused only on seropositive RA, therefore no demographic data were collected. To confirm these findings, the results should be independently replicated in populations with demographic data.
In the nutshell, this study genotyped the miRNA SNPs—including rs2292832, rs3746444, rs11614913, rs1044165, and rs767649—in Pakistani RA patients and in healthy controls to determine their association with RA and to aid in determining the genetic background of RA in the Pakistani population. The study revealed that two SNPs (rs2292832 in MIR149 and rs3746444 in MIR499) are likely to have a role in the pathogenesis of RA in the Pakistani population.

Author Contributions

Conceptualization, Y.A. and F.J.; methodology, Y.A., A.A. (Abdulaziz Alouffi), A.A. (Aisha Aman), M.M.A., A.A. (Abid Ali) and F.J.; validation, Y.A. and A.A. (Aisha Aman); analysis, A.A.S., Y.A., A.A. (Abid Ali), F.J. and Y.C.; resources, I.H., Y.C., F.J., M.M.A., A.A. (Abdulaziz Alouffi), Z.U.R., A.A. (Abid Ali) and F.J.; data curation, Y.A., M.M.A., Z.U.I. and F.J.; writing—original draft preparation, Y.A., Z.U.I., A.A. (Aisha Aman), M.M.A., A.A. (Abdulaziz Alouffi) and A.A. (Abid Ali); writing—review and editing, F.J., A.M., A.A. (Aisha Aman), M.M.A., A.A. (Abdulaziz Alouffi), A.A. (Abid Ali) and A.A.S.; supervision, F.J. and Y.C.; project administration, Y.C. and F.J. All authors have read and agreed to the published version of the manuscript.

Funding

Y.A. was supported by the International Research Support Initiative Program (IRSIP) of the Higher Education Commission of Pakistan. The authors acknowledge the support of the School of Biomedical Sciences of The Chinese University of Hong Kong. The researchers supporting project number (RSP2023R494), King Saud University, Riyadh, Saudi Arabia. The study also was sponsored by the Higher Education Commission, Pakistan (project number NRPU# 5203).

Institutional Review Board Statement

This study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Departmental Ethics Committee of the Department of Biotechnology, Abdul Wali Khan University Mardan (AWKUM\Biotech\2020\45321; Approval date. 5 August 2019).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

All data generated or analyzed during this study were included in this article. Further inquiries can be directed to the corresponding author.

Acknowledgments

The authors acknowledge the support of Higher Education Commission of Pakistan, the School of Biomedical Sciences of The Chinese University of Hong Kong. The researchers supporting project number (RSP2023R494), King Saud University, Riyadh, Saudi Arabia. The authors are thankful to the patients and healthy controls for their participation in this study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Symmons, D.P.M. Epidemiology of rheumatoid arthritis: Determinants of onset, persistence and outcome. Best Pract. Res. Clin. Rheumatol. 2002, 16, 707–722. [Google Scholar] [CrossRef] [PubMed]
  2. Bauer, M.E. Accelerated immunosenescence in rheumatoid arthritis: Impact on clinical progression. Immun. Ageing 2020, 17, 1–14. [Google Scholar] [CrossRef] [PubMed]
  3. Maiuolo, J.; Muscoli, C.; Gliozzi, M.; Musolino, V.; Carresi, C.; Paone, S.; Ilari, S.; Mollace, R.; Palma, E.; Mollace, V. Endothelial dysfunction and extra-articular neurological manifestations in rheumatoid arthritis. Biomolecules 2021, 11, 81. [Google Scholar] [CrossRef] [PubMed]
  4. Deane, K.D.; Demoruelle, M.K.; Kelmenson, L.B.; Kuhn, K.A.; Norris, J.M.; Holers, V.M. Genetic and environmental risk factors for rheumatoid arthritis. Best Pract. Res. Clin. Rheumatol. 2017, 31, 3–18. [Google Scholar] [CrossRef] [PubMed]
  5. Wu, H.; Chen, Y.; Zhu, H.; Zhao, M.; Lu, Q. The pathogenic role of dysregulated epigenetic modifications in autoimmune diseases. Front. Immunol. 2019, 10, 2305. [Google Scholar] [CrossRef]
  6. Patil, K.; Kuttikrishnan, S.; Khan, A.Q.; Ahmad, F.; Alam, M.; Buddenkotte, J.; Ahmad, A.; Steinhoff, M.; Uddin, S. Molecular pathogenesis of Cutaneous T cell Lymphoma: Role of chemokines, cytokines, and dysregulated signaling pathways. Semin. Cancer Biol. 2021, 86, 382–399. [Google Scholar] [CrossRef]
  7. Stahl, E.A.; Raychaudhuri, S.; Remmers, E.F.; Xie, G.; Eyre, S.; Thomson, B.P.; Li, Y.; Kurreeman, F.A.; Zhernakova, A.; Hinks, A. Genome-wide association study meta-analysis identifies seven new rheumatoid arthritis risk loci. Nat. Genet. 2010, 42, 508. [Google Scholar] [CrossRef]
  8. Kim, K.; Bang, S.-Y.; Lee, H.-S.; Bae, S.-C. Update on the genetic architecture of rheumatoid arthritis. Nat. Rev. Rheumatol. 2017, 13, 13–24. [Google Scholar] [CrossRef]
  9. Akhtar, M.; Ali, Y.; Islam, Z.-U.; Arshad, M.; Rauf, M.; Ali, M.; Maodaa, S.N.; Al-Farraj, S.A.; El-Serehy, H.A.; Jalil, F. Characterization of Rheumatoid Arthritis Risk-Associated SNPs and Identification of Novel Therapeutic Sites Using an In-Silico Approach. Biology 2021, 10, 501. [Google Scholar] [CrossRef]
  10. Ali, Y.; Khan, S.; Chen, Y.; Farooqi, N.; Islam, Z.-U.; Akhtar, M.; Aman, A.; Shah, A.A.; Jamal, M.; Jalil, F. Association of AFF3 Gene Polymorphism rs10865035 with Rheumatoid Arthritis: A Population-Based Case-Control Study on a Pakistani Cohort. Genet. Res. 2021, 2021, 5544198. [Google Scholar] [CrossRef]
  11. Akhtar, M.; Khan, S.; Ali, Y.; Haider, S.; ud Din, J.; Islam, Z.-U.; Jalil, F. Association study of CCR6 rs3093024 with Rheumatoid Arthritis in a Pakistani cohort. Saudi J. Biol. Sci. 2020, 27, 3354–3358. [Google Scholar] [CrossRef]
  12. Chen, X.-M.; Huang, Q.-C.; Yang, S.-L.; Chu, Y.-L.; Yan, Y.-H.; Han, L.; Huang, Y.; Huang, R.-Y. Role of micro RNAs in the pathogenesis of rheumatoid arthritis: Novel perspectives based on review of the literature. Medicine 2015, 94, e1326. [Google Scholar] [CrossRef]
  13. Moran-Moguel, M.C.; Petarra-del Rio, S.; Mayorquin-Galvan, E.E.; Zavala-Cerna, M.G. Rheumatoid arthritis and miRNAs: A critical review through a functional view. J. Immunol. Res. 2018, 2018, 2474529. [Google Scholar] [CrossRef]
  14. Manoharan, A.P. The Landscape of Caenorhabditis elegans 3’UTRs. Ph.D. Thesis, University of Michigan, Ann Arbor, MI, USA, 2011. [Google Scholar]
  15. Gurtan, A.M.; Sharp, P.A. The role of miRNAs in regulating gene expression networks. J. Mol. Biol. 2013, 425, 3582–3600. [Google Scholar] [CrossRef]
  16. Imamura, K.; Akimitsu, N. Long non-coding RNAs involved in immune responses. Front. Immunol. 2014, 5, 573. [Google Scholar] [CrossRef]
  17. Gam, R.; Shah, P.; Crossland, R.E.; Norden, J.; Dickinson, A.M.; Dressel, R. Genetic association of hematopoietic stem cell transplantation outcome beyond histocompatibility genes. Front. Immunol. 2017, 8, 380. [Google Scholar] [CrossRef]
  18. Cammaerts, S.; Strazisar, M.; De Rijk, P.; Del Favero, J. Genetic variants in microRNA genes: Impact on microRNA expression, function, and disease. Front. Genet. 2015, 6, 186. [Google Scholar] [CrossRef]
  19. Esteller, M. Non-coding RNAs in human disease. Nat. Rev. Genet. 2011, 12, 861–874. [Google Scholar] [CrossRef]
  20. Li, L.-J.; Gao, L.-B.; Lv, M.-L.; Dong, W.; Su, X.-W.; Liang, W.-B.; Zhang, L. Association between SNPs in pre-miRNA and risk of chronic obstructive pulmonary disease. Clin. Biochem. 2011, 44, 813–816. [Google Scholar] [CrossRef]
  21. Xiao, M.; Ma, Y.; Chen, X.; Kuang, B. Single nucleotide polymorphism of miR-149 and susceptibility of rheumatoid arthritis. Zhong Nan Da Xue Xue Bao. Yi Xue Ban J. Cent. South Univ. Med. Sci. 2015, 40, 495–498. [Google Scholar]
  22. Yang, B.; Chen, J.; Li, Y.; Zhang, J.; Li, D.; Huang, Z.; Cai, B.; Li, L.; Shi, Y.; Ying, B.; et al. Association of polymorphisms in pre-miRNA with inflammatory biomarkers in rheumatoid arthritis in the Chinese Han population. Hum. Immunol. 2012, 73, 101–106. [Google Scholar] [CrossRef]
  23. Hashemi, M.; Eskandari-Nasab, E.; Zakeri, Z.; Atabaki, M.; Bahari, G.; Jahantigh, M.; Taheri, M.; Ghavami, S. Association of pre-miRNA-146a rs2910164 and pre-miRNA-499 rs3746444 polymorphisms and susceptibility to rheumatoid arthritis. Mol. Med. Rep. 2013, 7, 287–291. [Google Scholar] [CrossRef] [PubMed]
  24. Amal, S.; Aly, N.M.; Galil, S.M.A.; Moustafa, M.A.; Kandel, W.A. Association of microRNAs genes polymorphisms with rheumatoid arthritis in Egyptian female patients. Jt. Bone Spine 2013, 80, 626–631. [Google Scholar]
  25. Fattah, S.A.; Ghattas, M.H.; Saleh, S.M.; Abo-Elmatty, D.M. Pre-micro RNA-499 gene polymorphism rs3746444 T/C is associated with susceptibility to rheumatoid arthritis in Egyptian population. Indian J. Clin. Biochem. 2018, 33, 96–101. [Google Scholar] [CrossRef] [PubMed]
  26. Toraih, E.A.; Ismail, N.M.; Toraih, A.A.; Hussein, M.H.; Fawzy, M.S. Precursor miR-499a variant but not miR-196a2 is associated with rheumatoid arthritis susceptibility in an Egyptian population. Mol. Diagn. Ther. 2016, 20, 279–295. [Google Scholar] [CrossRef] [PubMed]
  27. Alemán-Ávila, I.; Jiménez-Morales, M.; Beltrán-Ramírez, O.; Barbosa-Cobos, R.E.; Jiménez-Morales, S.; Sánchez-Muñoz, F.; Valencia-Pacheco, G.; Amezcua-Guerra, L.M.; Juárez-Vicuña, Y.; Hernández, D.M.R.-B.; et al. Functional polymorphisms in pre-miR146a and pre-miR499 are associated with systemic lupus erythematosus but not with rheumatoid arthritis or Graves’ disease in Mexican patients. Oncotarget 2017, 8, 91876. [Google Scholar] [CrossRef]
  28. Shaker, O.G.; Abdelaleem, O.O.; Fouad, N.A.; Ali, A.M.E.A.; Ahmed, T.I.; Ibrahem, E.G.; Abdelghaffar, N.K. Association between miR-155, its polymorphism and ischemia-modified albumin in patients with rheumatoid arthritis. J. Interferon Cytokine Res. 2019, 39, 428–437. [Google Scholar] [CrossRef]
  29. Smolen, J.S.; Landewé, R.; Bijlsma, J.; Burmester, G.; Chatzidionysiou, K.; Dougados, M.; Nam, J.; Ramiro, S.; Voshaar, M.; Van Vollenhoven, R.; et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann. Rheum. Dis. 2017, 76, 960–977. [Google Scholar] [CrossRef]
  30. Smolen, J.S.; Aletaha, D.; Koeller, M.; Weisman, M.H.; Emery, P. New therapies for treatment of rheumatoid arthritis. Lancet 2007, 370, 1861–1874. [Google Scholar] [CrossRef]
  31. Van Rooij, E. The art of microRNA research. Circ. Res. 2011, 108, 219–234. [Google Scholar] [CrossRef]
  32. Filková, M.; Jüngel, A.; Gay, R.E.; Gay, S. MicroRNAs in rheumatoid arthritis. BioDrugs 2012, 26, 131–141. [Google Scholar] [CrossRef]
  33. Aletaha, D.; Neogi, T.; Silman, A.J.; Funovits, J.; Felson, D.T.; Bingham, C.O., III; Birnbaum, N.S.; Burmester, G.R.; Bykerk, V.P.; Cohen, M.D.; et al. 2010 rheumatoid arthritis classification criteria: An American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheum. 2010, 62, 2569–2581. [Google Scholar] [CrossRef]
  34. Sambrook, J.; Fritsch, E.F.; Maniatis, T. Molecular Cloning: A Laboratory Manual; Cold Spring Harbor Laboratory Press: Cold Spring Harbor, NY, USA, 1989. [Google Scholar]
  35. McInnes, I.B.; Schett, G. The pathogenesis of rheumatoid arthritis. N. Engl. J. Med. 2011, 365, 2205–2219. [Google Scholar] [CrossRef]
  36. Shen, L.; Zhang, H.; Yan, T.; Zhou, G.; Liu, R. Association between interleukin 17A polymorphisms and susceptibility to rheumatoid arthritis in a Chinese population. Gene 2015, 566, 18–22. [Google Scholar] [CrossRef]
  37. Karami, J.; Aslani, S.; Jamshidi, A.; Garshasbi, M.; Mahmoudi, M. Genetic implications in the pathogenesis of rheumatoid arthritis; an updated review. Gene 2019, 702, 8–16. [Google Scholar] [CrossRef]
  38. Kochi, Y.; Okada, Y.; Suzuki, A.; Ikari, K.; Terao, C.; Takahashi, A.; Yamazaki, K.; Hosono, N.; Myouzen, K.; Tsunoda, T. A regulatory variant in CCR6 is associated with rheumatoid arthritis susceptibility. Nat. Genet. 2010, 42, 515. [Google Scholar] [CrossRef]
  39. Mohammadi, F.S.; Aslani, S.; Mostafaei, S.; Jamshidi, A.; Riahi, P.; Mahmoudi, M. Are genetic variations in IL-21–IL-23R–IL-17A cytokine axis involved in a pathogenic pathway of rheumatoid arthritis? Bayesian hierarchical meta-analysis. J. Cell. Physiol. 2019, 234, 17159–17171. [Google Scholar] [CrossRef]
  40. Salvi, V.; Gianello, V.; Tiberio, L.; Sozzani, S.; Bosisio, D. Cytokine targeting by miRNAs in autoimmune diseases. Front. Immunol. 2019, 10, 15. [Google Scholar] [CrossRef]
  41. Latini, A.; Ciccacci, C.; Novelli, G.; Borgiani, P. Polymorphisms in miRNA genes and their involvement in autoimmune diseases susceptibility. Immunol. Res. 2017, 65, 811–827. [Google Scholar] [CrossRef]
  42. Li, J.; Wan, Y.; Guo, Q.; Zou, L.; Zhang, J.; Fang, Y.; Zhang, J.; Zhang, J.; Fu, X.; Liu, H. Altered microRNA expression profile with miR-146a upregulation in CD4+ T cells from patients with rheumatoid arthritis. Arthritis Res. Ther. 2010, 12, 1–12. [Google Scholar] [CrossRef]
  43. Song, G.; Bae, S.-C.; Seo, Y.; Kim, J.-H.; Choi, S.; Ji, J.; Lee, Y.H. The association between susceptibility to inflammatory arthritis and miR-146a, miR-499 and IRAK1 polymorphisms. Z. Für Rheumatol. 2015, 74, 637–645. [Google Scholar] [CrossRef] [PubMed]
  44. Schellekens, G.A.; Visser, H.; De Jong, B.A.; Van Den Hoogen, F.H.; Hazes, J.M.; Breedveld, F.C.; Van Venrooij, W.J. The diagnostic properties of rheumatoid arthritis antibodies recognizing a cyclic citrullinated peptide. Arthritis Rheum. 2000, 43, 155–163. [Google Scholar] [CrossRef] [PubMed]
  45. Chavanas, S.; Méchin, M.-C.; Takahara, H.; Kawada, A.; Nachat, R.; Serre, G.; Simon, M. Comparative analysis of the mouse and human peptidylarginine deiminase gene clusters reveals highly conserved non-coding segments and a new human gene, PADI6. Gene 2004, 330, 19–27. [Google Scholar] [CrossRef] [PubMed]
  46. Ayeldeen, G.; Nassar, Y.; Ahmed, H.; Shaker, O.; Gheita, T. Possible use of miRNAs-146a and-499 expression and their polymorphisms as diagnostic markers for rheumatoid arthritis. Mol. Cell. Biochem. 2018, 449, 145–156. [Google Scholar] [CrossRef]
  47. Ul Islam, Z.; Baneen, U.; Khaliq, T.; Nurulain, S.M.; Muneer, Z.; Hussain, S. Association analysis of miRNA-146a and miRNA-499 polymorphisms with rheumatoid arthritis: A case–control and trio-family study. Clin. Exp. Med. 2022, 1–9. [Google Scholar] [CrossRef]
  48. Filkova, M.; Trenkmann, M.; Stanczyk, J.; Frank, M.; Kolling, C.; Huber, L.C.; Michel, B.A.; Gay, R.E.; Senolt, L.; Gay, S. MiR-196a Is An Important Regulator of Synovial Fibroblasts In the Pathogenesis of Rheumatoid Arthritis. In Arthritis and Rheumatism; Wiley: Hoboken, NJ, USA, 2011; p. S986. [Google Scholar]
  49. Mahesh, G.; Biswas, R. MicroRNA-155: A master regulator of inflammation. J. Interferon Cytokine Res. 2019, 39, 321–330. [Google Scholar] [CrossRef]
Figure 1. The structure of MIR499 and the maturity of sequence has-mir-499-3p representing all SNPs.
Figure 1. The structure of MIR499 and the maturity of sequence has-mir-499-3p representing all SNPs.
Genes 14 00431 g001
Table 1. Characteristics of Rheumatoid Arthritis Patients and Controls.
Table 1. Characteristics of Rheumatoid Arthritis Patients and Controls.
VariableCases (n = 300)Controls (n = 300)p-Value
Gender (male/female)113/187119/1810.6121
Mean age in years (±SD)43.5 (±14.5)42 (±12.6)0.217
Mean disease duration in years (±SD)4.1 (±3.7)--
Sero-positive antibody (Me)100% (RF-positive)--
Mean ESR (±SD)40.60 (±15.8)--
Table 2. Detailed information regarding the miRNAs.
Table 2. Detailed information regarding the miRNAs.
SNP IDmiRNANames of Mature miRNA SequencesChromosome No.miRNA
Location (Coordinates)
Coded AlleleOther AlleleMAF
rs2292832 MIR149hsa-miR-149-5p2240456001-240456089TC0.38
hsa-miR-149-3p
rs374644 MIR499Ahsa-miR-499-5p2034990376-34990497 [+]AG0.12
hsa-miR-499-3p
rs11614913MIR196a2hsa-miR-196a-5p1253991738-53991847 [+]CT0.49
hsa-miR-196a-3p
rs1044165MIR223 hsa-miR-223-5pX66018870-66018979GA0.00
hsa-miR-223-3p
rs767649MIR155 hsa-miR-155-5p2125573980-255740444TC0.29
Table 3. Association of RA risk-associated miRNAs in Pakistani case-control study.
Table 3. Association of RA risk-associated miRNAs in Pakistani case-control study.
miRNA SNPs ModelsGenotypesCases
(300)
Controls (300)Odds Ratioχ2-Valuep-Value
MIR149
rs2292832
Co-dominantTT
CT
CC
55 (18.4%)
136 (45.4%)
109 (36.3%)
112 (37.4%)
123 (41.0%)
65 (21.6%)
-31.23<0.0001
DominantCC
TT + CT
109 (36.3%)
291 (63.7%)
65 (21.6%)
235 (78.4%)
2.063
1.437–2.962
-0.0001
RecessiveTT
CC + CT
55 (18.4%)
245 (81.6%)
112 (37.4%)
188 (62.6%)
0.376
0.259–0.548
-<0.0001
HeterozygousCT
CC + TT
136 (45.4%)
164 (54.6%)
123 (41.0%)
177 (59%)
1.267
0.919–1.748
-0.16
AdditiveC
T
264 (44.0%)
354 (56.0%)
347 (57.8%)
253 (42.2%)
0.506
0.402–0.637
-<0.0001
MIR499
rs3746444
Co-dominantAA
AG
GG
72 (24.0%)
110 (36.7%)
118 (39.3%)
129 (43.0%)
138 (46.0%)
33 (11.0%)
-59.190.0001
DominantGG
AA + AG
118 (39.3%)
182 (60.7%)
33 (11.0%)
267 (89.0%)
5.246
3.414–8.061
-<0.0001
RecessiveAA
GG + AG
72 (24.0%)
228 (76.0%)
129 (43.0%)
171 (57.0%)
0.653
0.466–0.916
-0.014
HeterozygousAG
AA + GG
110 (36.7%)
190 (62.3%)
138 (46.0%)
162 (54.0%)
0.679
0.490–0.942
-0.025
AdditiveA
G
254 (42.4%)
346 (57.6%)
291 (58.5%)
309 41.5%)
0.779
0.620–0.978
-0.03
MIR196a2
rs11614913
Co-dominantCC
CT
TT
107 (35.7%)
132 (44.0%)
61 (20.3%)
112 (37.3%)
115 (38.3%)
73 (24.4%)
-2.350.30
DominantCC
TT + CT
107 (35.7%)
193 (64.3%)
112 (37.3%)
188 (42.7%)
0.930
0.667–1.298
-0.73
RecessiveTT
CC + CT
61 (20.3%)
239 (79.6%)
73 (24.4%)
227 (75.6%)
0.826
0.563–1.213
-0.38
HeterozygousCT
CC + TT
132 (44.0%)
168 (56%)
115 (38.3%)
185 (61.7%)
1.264
0.912–1.751
-0.18
AdditiveC
T
346 (57.7%)
254 (42.3%)
339 (56.5%)
261 (43.5%)
1.049
0.834–1.318
-0.72
MIR223
rs1044165
Co-dominantAA
AG
GG
3 (1.0%)
26 (8.7%)
271 (90.3%)
8 (2.7%)
24 (8.0%)
268 (89.3%)
-2.3690.30
DominantGG
AA + AG
271 (90.3%)
29 (9.7%)
268 (89.3%)
32 (10.7%)
1.116
0.656–1.896
-0.78
RecessiveAA
GG + AG
3 (1.0%)
297 (99.0%)
8 (2.7%)
292 (97.3%)
0.368
0.096–1.404
-0.22
HeterozygousAG
AA + GG
26 (8.7%)
274 (91.3%)
24 (8.0%)
276 (92.0%)
1.091
0.611–1.948
-0.88
AdditiveA
G
32 (5.3%)
568 (94.7%)
40 (6.7%)
560 (93.3%)
0.788
0.488–1.274
-0.39
MIR155
rs767649
Co-dominantCC
CT
TT
6 (2.0%)
72 (24.0%)
222 (74.0%)
3 (1.0%)
92(30.7%)
205 (68.3%)
-4.1160.12
DominantTT
CC + CT
222 (74.0%)
78 (26.0%)
205 (68.3%)
95 (31.7%)
1.319
0.925–1.88
0.14
RecessiveCC
TT + CT
6 (2.0%)
294 (98.0%)
3 (1.0%)
297 (99.0%)
2.02
0.50–8.157
-0.50
HeterozygousCT
CC + TT
72 (24.0%)
228 (76%)
92 (30.7%)
208 (69.3%)
0.714
0.497–1.025
-0.08
AdditiveC
T
84 (14.0%)
516 (86.0%)
92 (16.3%)
502 (83.7%)
0.833
0.607–1.144
-0.29
Table 4. Thermodynamic free energy differences between reference mutated sequences.
Table 4. Thermodynamic free energy differences between reference mutated sequences.
ParametersMIR149
rs2292832
MIR499A
rs3746444
MIR196A2
rs11614913
SequencesReference SequenceMutated
Sequence
Reference SequenceMutated
Sequence
Reference SequenceMutated
Sequence
Ensemble
thermodynamic free energy
−54.29
kcal/mol
−56.49
kcal/mol
−64.92
kcal/mol
−59.55
kcal/mol
−52.02
kcal/mol
−42.82
kcal/mol
Ensemble diversity7.217.209.6513.427.184.86
Optimal secondary structure with the lowest free energy−52.70
kcal/mol
−54.90
kcal/mol
−63.20
kcal/mol
−57.70
kcal/mol
−49.90
kcal/mol
−41.60
kcal/mol
Secondary structure of the centroid−52.70
kcal/mol
54.90
kcal/mol
−63.00
kcal/mol
−54.80
kcal/mol
−41.30
kcal/mol
−44.30
kcal/mol
Ensemble MFE structure frequency7.56%7.57%6.14%4.96%6.14%13.85%
Table 5. The basic characteristics of the retrieved studies.
Table 5. The basic characteristics of the retrieved studies.
AuthorYearCountryDiseaseControl SourceGenotype MethodCasesControlsHWE
MIR149 rs22928323
Xiao2015ChinaRAHBPCR-RFLP200120No
MIR499 rs3743444
Yang2012ChinaRAPBPCR-RFLP208240Yes
Hashemi2013IranRAPBT-ARMS-PCR104110Yes
Amal2013EgyptRAPBPCR-RFLP217245Yes
Zhang2013ChinaRAHBMALDI-TOP MS206466Yes
Yang2016ChinaRAPBTaqMan386576Yes
Toraih2016EgyptRAPBReal-time
PCR
95200No
Aleman-avila2017MexicoRAPBTaqMan412486Yes
Ayeldeen2018EgyptRAPBReal-time
PCR
5256Yes
Fataah2018EgyptRAPBPCR-RFLP100100Yes
MIR196a rs 11614913
Toraih2016EgyptRAPBReal-time
PCR
95200Yes
Aleman-avila2017MexicoRAPBTaqMan412486No
MIR155 rs7676649
Shaker2019EgyptRAPBTaqMan7978Yes
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ali, Y.; Chen, Y.; Islam, Z.U.; Aman, A.; Almutairi, M.M.; Alouffi, A.; Mohammed, A.; Shah, A.A.; Rehman, Z.U.; Hussain, I.; et al. MIR149 rs2292832 and MIR499 rs3746444 Genetic Variants Associated with the Risk of Rheumatoid Arthritis. Genes 2023, 14, 431. https://doi.org/10.3390/genes14020431

AMA Style

Ali Y, Chen Y, Islam ZU, Aman A, Almutairi MM, Alouffi A, Mohammed A, Shah AA, Rehman ZU, Hussain I, et al. MIR149 rs2292832 and MIR499 rs3746444 Genetic Variants Associated with the Risk of Rheumatoid Arthritis. Genes. 2023; 14(2):431. https://doi.org/10.3390/genes14020431

Chicago/Turabian Style

Ali, Yasir, Yangchao Chen, Zia Ul Islam, Aisha Aman, Mashal M. Almutairi, Abdulaziz Alouffi, Aymen Mohammed, Aftab Ali Shah, Zia Ur Rehman, Ibrar Hussain, and et al. 2023. "MIR149 rs2292832 and MIR499 rs3746444 Genetic Variants Associated with the Risk of Rheumatoid Arthritis" Genes 14, no. 2: 431. https://doi.org/10.3390/genes14020431

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop