Next Article in Journal
A High-Copy Suppressor Screen Reveals a Broad Role of Prefoldin-like Bud27 in the TOR Signaling Pathway in Saccharomyces cerevisiae
Next Article in Special Issue
Identification and Validation of an m7G-Related lncRNAs Signature for Prognostic Prediction and Immune Function Analysis in Endometrial Cancer
Previous Article in Journal
Spatiotemporal Regulation of Circular RNA Expression during Liver Development of Chinese Indigenous Ningxiang Pigs
Previous Article in Special Issue
Differential Expression Profiles and Bioinformatics Analysis of tRNA-Derived Small RNAs in Muscle-Invasive Bladder Cancer in a Chinese Population
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genetic Variants of MIR27A, MIR196A2 May Impact the Risk for the Onset of Coronary Artery Disease in the Pakistani Population

1
Department of Biotechnology, University of Malakand, Chakdara 18000, Pakistan
2
Department of Biotechnology, Abdul Wali Khan University Mardan (AWKUM), Mardan 23200, Pakistan
3
School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Genes 2022, 13(5), 747; https://doi.org/10.3390/genes13050747
Submission received: 19 December 2021 / Revised: 6 January 2022 / Accepted: 7 January 2022 / Published: 24 April 2022
(This article belongs to the Special Issue Non-coding RNAs in Human Health and Diseases)

Abstract

:
Genetic variants in microRNA genes have a detrimental effect on miRNA-mediated regulation of gene expression and may contribute to coronary artery disease (CAD). CAD is the primary cause of mortality worldwide. Several environmental, genetic, and epigenetic factors are responsible for CAD susceptibility. The contribution of protein-coding genes is extensively studied. However, the role of microRNA genes in CAD is at infancy. The study is aimed to investigate the impact of rs895819, rs11614913, and rs2168518 variants in MIR27A, MIR196A2, and MIR4513, respectively, in CAD using allele-specific PCR. Results: For variant rs11614913, significant distribution of the genotypes among the cases and controls was determined by co-dominant [χ2 = 54.4; p value ≤ 0.0001], dominant (C/C vs. C/T + T/T) [OR = 0.257 (0.133–0.496); p value ≤ 0.0001], recessive (T/T vs. C/T + C/C) [OR = 1.56 (0.677–0.632); p value = 0.398], and additive models [OR = 0.421 (0.262–0.675); p value = 0.0004]. Similarly, a significant association of rs895819 was determined by co-dominant [χ2 = 9.669; p value ≤ 0.008], dominant (A/A vs. A/G + G/G) [OR = 0.285 (0.1242–0.6575); p value ≤ 0.0034], recessive (G/G vs. A/G + A/A) [OR = 0.900 (0.3202–3.519); p value = 1.000], and additive models [OR = 0.604 (0.3640–1.002); p value = 0.05] while no significant association of rs2168518 with CAD was found. Conclusion: The variants rs895819 and rs11614913 are the susceptibility factors for CAD.

1. Introduction

MicroRNAs (miRNAs) are a group of small non-coding RNAs molecules (20–24 nucleotides) that repress mRNAs post-transcriptionally [1,2,3,4]. MicroRNA regulates the biological phenomenon at the post-transcriptional level [5]. MiRNAs play a pivotal role in the post-transcriptional regulation of protein-coding genes [6]. Dysregulation of miRNA expression is connected with several human diseases including coronary artery disease (CAD) [7]. Since the first discovery of miRNA, there are 38,589 entries representing hairpin precursor microRNAs, from 271 organisms. The human genome contains 1917 annotated hairpin precursors and 2654 mature sequences [8]. The seed region comprises 2–8 nucleotides of the miRNA and the recognition of the target genes depends mainly on their pairing [9]. Target miRNA cleavage or translational inhibition are two independent ways of miRNA-mediated translational control [10]. About half of miRNAs are transcribed from introns (intragenic) and relatively few are from exons of protein-coding genes. Similarly, few are intergenic and transcribed independently [11,12,13] while some miRNAs are located as clusters and their transcription has resulted in one long transcript [14]. However, the interaction of miRNAs with other regions in the target mRNA including the 5′-untranslated regions, coding sequence, and gene promoters, have also been reported [15]. The miRNAs lead the argonaut proteins to specific target messenger RNAs to suppress their stability and translation.
Aberrant miRNA expression is mostly due to single nucleotide polymorphism (SNP) in miRNA genes [16]. SNP is the most common form of single base-pair changes in a genome. The previous study has shown that SNPs have a profound influence on miRNA function, stability, and targeting [17]. SNPs are commonly identified in the miRNA genes or the binding site of mRNA of miRNAs. SNPs in the miRNA binding site can modify miRNA by generating or removing a miRNA binding site in the target mRNA [18,19]. Several studies showed that SNPs in the target site of miRNA genes are involved in a wide range of diseases [7,20,21]. It was investigated that the SNP (rs531564) is associated with an increased risk of cervical cancer, diabetic retinopathy, and CAD [22,23,24]. Coronary artery disease (CAD) is a pathological condition linked with atherosclerotic plaque aggregation in the epicardial arteries, whether obstructive or non-obstructive [25]. The association of mir-499-SNP (rs3746444) with ischemic stroke in the Asian population was reported [26]. It was found that miRNAs play a role in the pathophysiology of Cardiomyopathy [27]. The core heptameric sequence of a mature miRNA term as “seed region,” includes 2–8 nucleotides and plays a crucial role in target gene recognition and interaction [4,28]. Genetic variations in the seed regions have crucial impacts on gene expression and disease susceptibility in humans [1]. The distribution pattern of genetic variation in miRNA seed regions might be related to miRNA function [2,3,29].
A previous study indicates that CAD leads to one-third of mortality in women regardless of their ethnicity [30]. An earlier study showed that more than six percent of the adult population is suffering from CAD [31]. Atherosclerosis is a condition in which plaque builds up inside the arteries that provide oxygenated blood to the heart and are leading to CAD [32,33]. The plaque is made over the years that lead to narrowing of the coronary artery lumen. Consequently, limits the flow of blood to the artery [34]. Several studies have shown the contribution of alteration in protein-coding genes with CAD [35,36]. However, the association of genetic alteration in non-coding genomes especially microRNA genes, and pathophysiology of CAD is investigated very rarely [37,38,39]. Mature microRNA plays a pivotal role in the endothelial function in cardiometabolic disorders. In CAD patients, miR-206 has been demonstrated to reduce the viability and invasion of endothelial progenitor cells while increasing their death [25,40,41,42,43]. A previous study has shown that miR-92a-3p is up-regulated in CAD [44]. It was also noted high expression of miR-330 blocked the formation of plaques in atherosclerosis [45].
It was also noted that MiR-1 plays a pivotal role in the progression of CAD, as well as in cardiogenesis and cardiac hypertrophy. It was investigated that miR-1 is upregulated in CAD patients through regulation of cardiac arrhythmogenic potential by targeting several ion channel genes [46,47]. Interestingly it was noted that miR-322 was upregulated in unstable plaques compared to stable plaques that are formed during CAD progression [48]. The current study is focused on the genetic alteration in microRNA (miRNA) genes and their association with CAD in the Pakistani population. Therefore, SNPs in the miRNA seed region are likely to change the target genes expression and influence the corresponding phenotypes.

2. Materials and Methods

2.1. Study Population

This current case-control study was designed to investigate the genetic-based risk factors for CAD. In this study, we examined 223 coronary artery disease individuals (CAD), as well as 150 healthy controls as shown in Table 1. The patients were clinically diagnosed by a certified cardiologist. A properly designed questionnaire was used to record the demographic and clinical data of each participant and informed consent was obtained from all participants or their guardians.

2.2. Inclusion and Exclusion Criteria for CAD Patients and Healthy Controls

This study was conducted on clinically confirmed cases of CAD patients and healthy controls with no history of CAD. Those CAD patients were selected who had visited a hospital for the evaluation of stable chest pain by elective angiography, electrocardiogram (ECG or EKG), and/or echocardiogram (echo). Patients with a previous history of any chronic disease or those who had performed coronary bypass surgery were excluded from the current study.
Relevant biochemical tests were also performed to confirm the previous history of CAD. Those healthy controls who have previous cardiac/angina and/or myocardial infarction history were excluded.

2.3. Blood Samples Collection and Genomic DNA Extraction

About 3–5 mL of whole blood was collected from February 2019 till January 2020 in EDTA tubes from all patients and healthy controls from District Head Quarter (DHQ) Hospitals of Dir lower and Malakand, Khyber Pakhtunkhwa, Pakistan. DNA was isolated by Phenol/Chloroform method, dissolved in distilled water, and stored at +4 °C until further processing. The quality of extracted DNA was confirmed by a spectrophotometer (Evolution 300 BB, Ser No. EV3 131505, Thermo ELECTRON CORPORATION, Waltham, MA, USA).

2.4. Primers for Allele-Specific PCR and Genotyping of Rs895819, Rs11614913, and Rs2168518

The previously published allele-specific PCR primers were used for genotyping of rs895819, rs11614913, and rs2168518 [49,50,51]. PCR products were run on 2% agarose gel and the nature of each genotype (homozygous/heterozygous) was recorded using the visual inspection method of the gel.

2.5. Statistical Analysis

Chi-square analysis and Fisher exact test were used to calculate allelic and genotypic frequencies of rs895819, rs11614913, and rs2168518 variants in MIR27A, MIR196A2, and MIR4513, respectively, for both CAD patients and healthy controls. The associations between the studied SNPs and the risk of CAD patients were estimated by calculating the odds ratios (ORs), and with 95% confidence intervals (CIs). GraphPad Prism 6 was used for data analysis.

2.6. In-Silico Analyses of the Primary Structure of miRNAs

Detailed information about the screened rs895819, rs11614913, and rs2168518 in MIR27A, MIR196A2, and MIR4513, their approved gene names, mature miRNAs, location (coordinates) on their respective chromosomes, coded alleles, another allele, and minor allele frequencies are shown in Table 2. Figure 1 shows the locations of the seed region and rs895819 in hsa-mir-27a-3p as an example. To find out whether the rs895819, rs11614913, and rs2168518 variations have some impact on the primary structures of miR-27a, miR-196a2, and miR-4513. The energies of the RNA sequences were determined using the RNAeval web server. Analysis of RNA secondary and centroid structures was carried out with the help of the Vienna RNA website [52].

3. Results

3.1. Association of Rs2168518, Rs895819, and Rs11614913 with CAD

The current study showed that rs11614913 and rs895819 is linked with the risk of CAD by using different inheritance models as shown in Table 3. The rs11614913 and rs895819 were assessed through different statistical models such as co-dominant, homozygous dominant, homozygous recessive, and additive models. The rs895819 genotypes showed significant association in co-dominant model [χ2 = 54.4; p < 0.0001], homozygous dominant model [OR = 0.257 (0.133–1.496); p < 0.0001], and additive model [OR = 0.421 (0.262–0.675); p < 0.0004], but the association of its genotypes was insignificant at homozygous recessive model [OR = 0.156 (0.677–0.632); p = 0.398]. Likewise, significant association of rs11614913 with CAD was noted using co-dominant model [χ2 = 9.669; p < 0.008], homozygous dominant model [OR = 0.285 (0.1242–0.6575); p < 0.0034] and additive model [OR = 0.604 (0.364–1.002); p = 0.05].

3.2. Consequences of Variant Rs2168518, Rs895819, Rs11614913 on miRNA Structure and Properties

Since the variant rs2168518 is in the seed region of MIR4513, therefore, there is the substitution of Cytosine with Uracil at position 18 in the mature sequence of hsa-mir-4513-3p. This change leads to an increase in the primary miRNA loop structure by one base pair. By comparing, the normal and mutant primary miRNA structure using the thermodynamic Structure Prediction tool, alterations were also observed in the studied parameters as shown in Table 4. It was observed that the variant rs11614913 in MIR196A2 resulted in the substitution of Cytosine base by Uracil in the miRNA primary structure. Similarly, due to rs895819, there are 13 bases in the terminal loop structure of wild-type miR-27a centroid structure while 15 bases in the corresponding terminal loop structure of the mutant miR-27a.

4. Discussion

It is already known that several environmental, genetic, as well as epigenetic factors, are responsible for the pathophysiology of CAD. Arterial thrombosis is one of the main causes of CAD. Arterial thrombosis is characterized by the development of a blood clot in an artery. Blood vessel occlusion usually occurs when the erosion of associated arterial sclerosis plaque causes wounds particularly in tissues with a terminal tube bed [53]. Arterial sclerosis plaque severely influences the local hemodynamics in coronary arteries [54,55]. The previous study has indicated that the geometry of a plaque is associated with the possibility of clinical incidents, including myocardial infarction in patients with CAD [56]. Coronary plaque geometry is vital for the pathophysiology and enhancing the diagnosis and cure of CAD [57]. There are also serval genetic factors that contribute to arterial thrombosis. Several studies have shown the contribution of SNPs in the protein-coding part of the genome [58,59,60]. Although the role of SNPs in the non-coding part of the genome especially miRNA genes are not fully elucidated. The previous study has shown that SNPs in miRNAs are related to the development of CAD [61]. SNPs located in miRNAs also known as mirSNPs play a pivotal role in the development of various types of diseases. The previous study has explored the role of mirSNPs in CAD in different populations worldwide, however, the role of rs895819, rs11614913, and rs2168518 in CAD in the Pakistani population is in infancy. This study was designed to evaluate the influence of rs895819, rs11614913, and rs2168518 variants located in MIR27A and MIR196A2 and MIR4513, respectively, on the risk of CAD in the selected population. The previous study has shown that rs2168518, is significantly linked with blood pressure, LDL, total cholesterol and fasting glucose [49]. It was shown that the expression of miR-4513 is significantly reduced by RNA induced silencing complex (RISC) loading and RNA degradation mechanisms due to the rs2168518 variant in the seed region of MIR4513 [62,63,64]. Furthermore, it was also investigated that there is a strong association of rs2168518 with the high mortality rate in CAD patients [65]. These data may have significant clinical implications on evaluating the risk of cardiovascular events or the possibility of intensive treatment interventions in CAD patients. The variant rs11614913 is intensively studied in different types of cancers and their association was established in different populations worldwide [66,67,68]. It was investigated that the T allele of the variant rs11614913 C/T in MIR196A2 was strongly linked with CAD [69]. The current study also confirmed that the T allele of rs11614913 is the risk factor for CAD in the Pakistani Population. The variant rs895819 is one of the broadly studied SNP in different diseases like Type 2 Diabetes Mellitus [70], colorectal cancer [71], and breast cancer [51]. It was also explored that the SNP rs895819 in the primary structure of MIR27A (pre-miR-27a) is associated with susceptibility to myocardial infarction (MI) in the Chinese Hans population. This is following the present study. It is, therefore, recommended that this study should be repeated on a larger cohort. Functional studies of these SNPs should be conducted to investigate their exact role in the pathophysiology of CAD. Furthermore, an expression study should be conducted to provide the basis for the development of blood-borne miRNA-based novel biomarkers for CAD in the future. It is, thus, concluded that these three variants rs895819 and rs11614913 are associated with CAD in the Pakistani population. In the current study, a total of 223 CAD and 150 healthy individuals were genotyped. One of the major limitations of the study is the small sample size. Secondly, the samples may be further validated through the DNA sequencing approach. Moreover, confirmation of these SNPs in CAD patients may be screened on a larger sample size.

Author Contributions

A.A.S. and F.J. designed the study, analyzed the data. T.U.H. and A.Z. collected blood samples along with clinical data and performed laboratory experiments. The co-authors participated in manuscript writing. Y.A. and Y.C. edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work is not funded by any funding agency. All the experiments were performed in the Department of Biotechnology, University of Malakand, Khyber Pakhtunkhwa, Pakistan.

Institutional Review Board Statement

The present study was conducted following the ethical criteria of the 1964 Helsinki declaration. This study was approved by the Advanced Study and Research Board (ASRB) in its 53rd meeting, (Agenda item No. 04), University of Malakand, Khyber Pakhtunkhwa, Pakistan.

Informed Consent Statement

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

Acknowledgments

The authors are highly thankful to the patients and healthy controls who participated in this study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hill, C.G.; Jabbari, N.; Matyunina, L.V.; McDonald, J.F. Functional and evolutionary significance of human microRNA seed region mutations. PLoS ONE 2014, 9, e115241. [Google Scholar] [CrossRef]
  2. Klum, S.M.; Chandradoss, S.D.; Schirle, N.T.; Joo, C.; MacRae, I.J. Helix-7 in Argonaute2 shapes the microRNA seed region for rapid target recognition. EMBO J. 2018, 37, 75–88. [Google Scholar] [CrossRef] [PubMed]
  3. Nigita, G.; Acunzo, M.; Romano, G.; Veneziano, D.; Lagana, A.; Vitiello, M.; Wernicke, D.; Ferro, A.; Croce, C.M. microRNA editing in seed region aligns with cellular changes in hypoxic conditions. Nucleic Acids Res. 2016, 44, 6298–6308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Chai, J.; Chen, L.; Luo, Z.; Zhang, T.; Chen, L.; Lou, P.; Sun, W.; Long, X.; Lan, J.; Wang, J.; et al. Spontaneous single nucleotide polymorphism in porcine microRNA-378 seed region leads to functional alteration. Biosci. Biotechnol. Biochem. 2018, 82, 1081–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Bronze-da-Rocha, E. MicroRNAs expression profiles in cardiovascular diseases. BioMed Res. Int. 2014, 2014, 985408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. He, L.; Hannon, G.J. MicroRNAs: Small RNAs with a big role in gene regulation. Nat. Rev. Genet. 2004, 5, 522–531. [Google Scholar] [CrossRef] [PubMed]
  7. Ullah, S.; John, P.; Bhatti, A. MicroRNAs with a role in gene regulation and in human diseases. Mol. Biol. Rep. 2014, 41, 225–232. [Google Scholar] [CrossRef] [PubMed]
  8. Kozomara, A.; Birgaoanu, M.; Griffiths-Jones, S. miRBase: From microRNA sequences to function. Nucleic Acids Res. 2019, 47, D155–D162. [Google Scholar] [CrossRef] [PubMed]
  9. Ludwig, N.; Becker, M.; Schumann, T.; Speer, T.; Fehlmann, T.; Keller, A.; Meese, E. Bias in recent miRBase annotations potentially associated with RNA quality issues. Sci. Rep. 2017, 7, 5162. [Google Scholar] [CrossRef]
  10. van den Berg, A.; Mols, J.; Han, J. RISC-target interaction: Cleavage and translational suppression. Biochim. Biophys. Acta 2008, 1779, 668–677. [Google Scholar] [CrossRef] [Green Version]
  11. Chen, J.F.; Mandel, E.M.; Thomson, J.M.; Wu, Q.; Callis, T.E.; Hammond, S.M.; Conlon, F.L.; Wang, D.Z. The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nat. Genet. 2006, 38, 228–233. [Google Scholar] [CrossRef] [PubMed]
  12. Agarwal, V.; Bell, G.W.; Nam, J.W.; Bartel, D.P. Predicting effective microRNA target sites in mammalian mRNAs. eLife 2015, 4, e05005. [Google Scholar] [CrossRef] [PubMed]
  13. Kim, Y.K.; Kim, V.N. Processing of intronic microRNAs. EMBO J. 2007, 26, 775–783. [Google Scholar] [CrossRef] [Green Version]
  14. Tanzer, A.; Stadler, P.F. Molecular evolution of a microRNA cluster. J. Mol. Biol. 2004, 339, 327–335. [Google Scholar] [CrossRef]
  15. Paraskevopoulou, M.D.; Georgakilas, G.; Kostoulas, N.; Vlachos, I.S.; Vergoulis, T.; Reczko, M.; Filippidis, C.; Dalamagas, T.; Hatzigeorgiou, A.G. DIANA-microT web server v5.0: Service integration into miRNA functional analysis workflows. Nucleic Acids Res. 2013, 41, W169–W173. [Google Scholar] [CrossRef] [Green Version]
  16. Wilk, G.; Braun, R. regQTLs: Single nucleotide polymorphisms that modulate microRNA regulation of gene expression in tumors. PLoS Genet. 2018, 14, e1007837. [Google Scholar] [CrossRef] [PubMed]
  17. Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef] [PubMed]
  18. Broughton, J.P.; Lovci, M.T.; Huang, J.L.; Yeo, G.W.; Pasquinelli, A.E. Pairing beyond the Seed Supports MicroRNA Targeting Specificity. Mol. Cell. 2016, 64, 320–333. [Google Scholar] [CrossRef] [Green Version]
  19. Vasudevan, S. Posttranscriptional upregulation by microRNAs. Wiley Interdiscip. Rev. RNA 2012, 3, 311–330. [Google Scholar] [CrossRef]
  20. Hayes, J.; Peruzzi, P.P.; Lawler, S. MicroRNAs in cancer: Biomarkers, functions and therapy. Trends Mol. Med. 2014, 20, 460–469. [Google Scholar] [CrossRef]
  21. Huang, W. MicroRNAs: Biomarkers, Diagnostics, and Therapeutics. Methods Mol. Biol. 2017, 1617, 57–67. [Google Scholar] [PubMed]
  22. Moazeni-Roodi, A.; Aftabi, S.; Sarabandi, S.; Karami, S.; Hashemi, M.; Ghavami, S.; Taheri, M. Association Between miR-146a rs2910164 Polymorphism and Breast Cancer Susceptibility: An Updated Meta-analysis of 9545 Cases and 10030 Controls. Microrna 2021, 10, 191–199. [Google Scholar]
  23. Massignam, E.T.; Dieter, C.; Pellenz, F.M.; Assmann, T.S.; Crispim, D. Involvement of miR-126 rs4636297 and miR-146a rs2910164 polymorphisms in the susceptibility for diabetic retinopathy: A case-control study in a type 1 diabetes population. Acta Ophthalmol. 2021, 99, e461–e469. [Google Scholar] [CrossRef] [PubMed]
  24. Mir, R.; Elfaki, I.; Jha, C.; Javid, J.; Rehman, S.; Banu, S.; Mir, M.M.; Babakr, A.T.; Chahal, S.M.S. Molecular Evaluation of MicroRNA-146 Gene Variability (rs2910164 C > G) and its Association with Increased Susceptibility to Coronary Artery Disease. Microrna 2020, 9, 363–372. [Google Scholar] [CrossRef] [PubMed]
  25. Knuuti, J.; Wijns, W.; Saraste, A.; Capodanno, D.; Barbato, E.; Funck-Brentano, C.; Prescott, E.; Storey, R.F.; Deaton, C.; Cuisset, T. 2019 ESC Guidelines for the diagnosis and management of chronic coronary syndromes: The Task Force for the diagnosis and management of chronic coronary syndromes of the European Society of Cardiology (ESC). Eur. Heart J. 2020, 41, 407–477. [Google Scholar] [CrossRef] [PubMed]
  26. Zhu, R.; Wang, Q.W.; Zhao, J.; Liu, X.; He, Z. miR-149 and miR-499 gene polymorphism and the incident of ischemic stroke in the Asian population: From a case-control study to meta-analysis. Clin. Neurol. Neurosurg. 2020, 193, 105789. [Google Scholar] [CrossRef]
  27. Ahmed, U.; Khaliq, S.; Ahmad, H.U.; Ahmad, I.; Ashfaq, U.A.; Qasim, M.; Masoud, M.S. Pathogenesis of Diabetic Cardiomyopathy and Role of miRNA. Crit. Rev. Eukaryot. Gene Expr. 2021, 31, 79–92. [Google Scholar] [CrossRef] [PubMed]
  28. Fawzy, M.S.; Toraih, E.A.; Hamed, E.O.; Hussein, M.H.; Ismail, H.M. Association of MIR-499a expression and seed region variant (rs3746444) with cardiovascular disease in Egyptian patients. Acta Cardiol. 2018, 73, 131–140. [Google Scholar]
  29. Mullany, L.E.; Herrick, J.S.; Wolff, R.K.; Slattery, M.L. MicroRNA Seed Region Length Impact on Target Messenger RNA Expression and Survival in Colorectal Cancer. PLoS ONE 2016, 11, e0154177. [Google Scholar] [CrossRef] [PubMed]
  30. Thom, T.; Haase, N.; Rosamond, W.; Howard, V.J.; Rumsfeld, J.; Manolio, T.; Zheng, Z.J.; Flegal, K.; O’Donnell, C.; Kittner, S.; et al. Heart disease and stroke statistics—2006 update: A report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 2006, 113, e85–e151. [Google Scholar]
  31. Mozaffarian, D.; Benjamin, E.J.; Go, A.S.; Arnett, D.K.; Blaha, M.J.; Cushman, M.; de Ferranti, S.; Despres, J.P.; Fullerton, H.J.; Howard, V.J.; et al. Heart disease and stroke statistics—2015 update: A report from the American Heart Association. Circulation 2015, 131, e29–e322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kumari, A.; Kristensen, K.K.; Ploug, M.; Winther, A.L. The Importance of Lipoprotein Lipase Regulation in Atherosclerosis. Biomedicines 2021, 9, 782. [Google Scholar] [CrossRef]
  33. Pella, Z.; Pella, D.; Paralic, J.; Vanko, J.I.; Fedacko, J. Analysis of Risk Factors in Patients with Subclinical Atherosclerosis and Increased Cardiovascular Risk Using Factor Analysis. Diagnostics 2021, 11, 1284. [Google Scholar] [CrossRef]
  34. Cagle, S.D., Jr.; Cooperstein, N. Coronary Artery Disease: Diagnosis and Management. Prim. Care 2018, 45, 45–61. [Google Scholar] [CrossRef]
  35. Hughes, M.F.; Lenighan, Y.M.; Godson, C.; Roche, H.M. Exploring Coronary Artery Disease GWAs Targets With Functional Links to Immunometabolism. Front. Cardiovasc. Med. 2018, 5, 148. [Google Scholar] [CrossRef] [PubMed]
  36. Nikpay, M.; Goel, A.; Won, H.H.; Hall, L.M.; Willenborg, C.; Kanoni, S.; Saleheen, D.; Kyriakou, T.; Nelson, C.P.; Hopewell, J.C.; et al. A comprehensive 1000 Genomes-based genome-wide association meta-analysis of coronary artery disease. Nat. Genet. 2015, 47, 1121–1130. [Google Scholar]
  37. Cavarretta, E.; Frati, G. MicroRNAs in Coronary Heart Disease: Ready to Enter the Clinical Arena? BioMed Res. Int. 2016, 2016, 2150763. [Google Scholar] [CrossRef] [Green Version]
  38. Schulte, C.; Zeller, T. microRNA-based diagnostics and therapy in cardiovascular disease-Summing up the facts. Cardiovasc. Diagn. Ther. 2015, 5, 17–36. [Google Scholar]
  39. Thanikachalam, P.V.; Ramamurthy, S.; Wong, Z.W.; Koo, B.J.; Wong, J.Y.; Abdullah, M.F.; Chin, Y.H.; Chia, C.H.; Tan, J.Y.; Neo, W.T.; et al. Current attempts to implement microRNA-based diagnostics and therapy in cardiovascular and metabolic disease: A promising future. Drug Discov. Today 2018, 23, 460–480. [Google Scholar] [CrossRef]
  40. Churov, A.; Summerhill, V.; Grechko, A.; Orekhova, V.; Orekhov, A. MicroRNAs as potential biomarkers in atherosclerosis. Int. J. Mol. Sci. 2019, 20, 5547. [Google Scholar] [CrossRef] [Green Version]
  41. Ghafouri-Fard, S.; Gholipour, M.; Taheri, M. Role of microRNAs in the pathogenesis of coronary artery disease. Front. Cardiovasc. Med. 2021, 8, 632392. [Google Scholar] [CrossRef]
  42. Wang, M.; Ji, Y.; Cai, S.; Ding, W. MiR-206 Suppresses the Progression of Coronary Artery Disease by Modulating Vascular Endothelial Growth Factor (VEGF) Expression. Med. Sci. Monit. 2016, 22, 5011–5020. [Google Scholar] [CrossRef] [Green Version]
  43. Kaur, A.; Mackin, S.T.; Schlosser, K.; Wong, F.L.; Elharram, M.; Delles, C.; Stewart, D.J.; Dayan, N.; Landry, T.; Pilote, L. Systematic review of microRNA biomarkers in acute coronary syndrome and stable coronary artery disease. Cardiovasc. Res. 2020, 116, 1113–1124. [Google Scholar] [CrossRef]
  44. Liu, Y.; Li, Q.; Hosen, M.R.; Zietzer, A.; Flender, A.; Levermann, P.; Schmitz, T.; Frühwald, D.; Goody, P.; Nickenig, G. Atherosclerotic conditions promote the packaging of functional microRNA-92a-3p into endothelial microvesicles. Circ. Res. 2019, 124, 575–587. [Google Scholar] [CrossRef]
  45. Ren, J.; Ma, R.; Zhang, Z.B.; Li, Y.; Lei, P.; Men, J.L. Effects of microRNA-330 on vulnerable atherosclerotic plaques formation and vascular endothelial cell proliferation through the WNT signaling pathway in acute coronary syndrome. J. Cell. Biochem. 2018, 119, 4514–4527. [Google Scholar] [CrossRef]
  46. Yang, B.; Lin, H.; Xiao, J.; Lu, Y.; Luo, X.; Li, B.; Zhang, Y.; Xu, C.; Bai, Y.; Wang, H. The muscle-specific microRNA miR-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2. Nat. Med. 2007, 13, 486–491. [Google Scholar] [CrossRef]
  47. Sayed, D.; Hong, C.; Chen, I.-Y.; Lypowy, J.; Abdellatif, M. MicroRNAs play an essential role in the development of cardiac hypertrophy. Circ. Res. 2007, 100, 416–424. [Google Scholar] [CrossRef] [Green Version]
  48. Chen, Y.-C.; Bui, A.V.; Diesch, J.; Manasseh, R.; Hausding, C.; Rivera, J.; Haviv, I.; Agrotis, A.; Htun, N.M.; Jowett, J. A novel mouse model of atherosclerotic plaque instability for drug testing and mechanistic/therapeutic discoveries using gene and microRNA expression profiling. Circ. Res. 2013, 113, 252–265. [Google Scholar] [CrossRef] [Green Version]
  49. Ghanbari, M.; de Vries, P.S.; de Looper, H.; Peters, M.J.; Schurmann, C.; Yaghootkar, H.; Dorr, M.; Frayling, T.M.; Uitterlinden, A.G.; Hofman, A.; et al. A genetic variant in the seed region of miR-4513 shows pleiotropic effects on lipid and glucose homeostasis, blood pressure, and coronary artery disease. Hum. Mutat. 2014, 35, 1524–1531. [Google Scholar] [CrossRef]
  50. Mir, R.; Jha, C.K.; Elfaki, I.; Javid, J.; Rehman, S.; Khullar, N.; Banu, S.; Chahal, S.M.S. Incidence of MicroR-4513C/T Gene Variability in Coronary Artery Disease—A Case-Control Study. Endocr. Metab. Immune Disord.-Drug Targets 2019, 19, 1216–1223. [Google Scholar] [CrossRef]
  51. Mashayekhi, S.; Saeidi Saedi, H.; Salehi, Z.; Soltanipour, S.; Mirzajani, E. Effects of miR-27a, miR-196a2 and miR-146a polymorphisms on the risk of breast cancer. Br. J. Biomed. Sci. 2018, 75, 76–81. [Google Scholar] [CrossRef]
  52. Ding, Y.; Chan, C.Y.; Lawrence, C.E. RNA secondary structure prediction by centroids in a Boltzmann weighted ensemble. RNA 2005, 11, 1157–1166. [Google Scholar] [CrossRef] [Green Version]
  53. Previtali, E.; Bucciarelli, P.; Passamonti, S.M.; Martinelli, I. Risk factors for venous and arterial thrombosis. Blood Transfus. 2011, 9, 120–138. [Google Scholar] [PubMed]
  54. Libby, P.; Buring, J.E.; Badimon, L.; Hansson, G.K.; Deanfield, J.; Bittencourt, M.S.; Tokgözoğlu, L.; Lewis, E.F. Atherosclerosis. Nat. Rev. Dis. Primers 2019, 5, 56. [Google Scholar] [CrossRef] [PubMed]
  55. Haverich, A.; Boyle, E.C. Atherosclerosis Pathogenesis and Microvascular Dysfunction; Springer: Berlin/Heidelberg, Germany, 2019. [Google Scholar]
  56. Choi, G.; Lee, J.M.; Kim, H.-J.; Park, J.-B.; Sankaran, S.; Otake, H.; Doh, J.-H.; Nam, C.-W.; Shin, E.-S.; Taylor, C.A. Coronary artery axial plaque stress and its relationship with lesion geometry: Application of computational fluid dynamics to coronary CT angiography. Cardiovasc. Imaging 2015, 8, 1156–1166. [Google Scholar]
  57. Ratiu, M.; Chitu, M.; Benedek, I.; Benedek, T.; Kovacs, I.; Rat, N.; Rezus, C. Impact of coronary plaque geometry on plaque vulnerability and its association with the risk of future cardiovascular events in patients with chest pain undergoing coronary computed tomographic angiography—The GEOMETRY study: Protocol for a prospective clinical trial. Medicine 2018, 97, e13498. [Google Scholar]
  58. Sachidanandam, R.; Weissman, D.; Schmidt, S.C.; Kakol, J.M.; Stein, L.D.; Marth, G.; Sherry, S.; Mullikin, J.C.; Mortimore, B.J.; Willey, D.L.; et al. A map of human genome sequence variation containing 1.42 million single nucleotide polymorphisms. Nature 2001, 409, 928–933. [Google Scholar]
  59. Hu, G.; Modrek, B.; Riise Stensland, H.M.; Saarela, J.; Pajukanta, P.; Kustanovich, V.; Peltonen, L.; Nelson, S.F.; Lee, C. Efficient discovery of single-nucleotide polymorphisms in coding regions of human genes. Pharm. J. 2002, 2, 236–242. [Google Scholar] [CrossRef] [Green Version]
  60. Robert, F.; Pelletier, J. Exploring the Impact of Single-Nucleotide Polymorphisms on Translation. Front. Genet. 2018, 9, 507. [Google Scholar] [CrossRef] [Green Version]
  61. Condorelli, G.; Latronico, M.V.; Cavarretta, E. microRNAs in cardiovascular diseases: Current knowledge and the road ahead. J. Am. Coll. Cardiol. 2014, 63, 2177–2187. [Google Scholar] [CrossRef] [Green Version]
  62. Khvorova, A.; Reynolds, A.; Jayasena, S.D. Functional siRNAs and miRNAs exhibit strand bias. Cell 2003, 115, 209–216. [Google Scholar] [CrossRef] [Green Version]
  63. Krol, J.; Sobczak, K.; Wilczynska, U.; Drath, M.; Jasinska, A.; Kaczynska, D.; Krzyzosiak, W.J. Structural features of microRNA (miRNA) precursors and their relevance to miRNA biogenesis and small interfering RNA/short hairpin RNA design. J. Biol. Chem. 2004, 279, 42230–42239. [Google Scholar] [CrossRef] [Green Version]
  64. Kawamata, T.; Seitz, H.; Tomari, Y. Structural determinants of miRNAs for RISC loading and slicer-independent unwinding. Nat. Struct. Mol. Biol. 2009, 16, 953–960. [Google Scholar] [CrossRef]
  65. Li, Q.; Chen, L.; Chen, D.; Wu, X.; Chen, M. Influence of microRNA-related polymorphisms on clinical outcomes in coronary artery disease. Am. J. Transl. Res. 2015, 7, 393–400. [Google Scholar]
  66. Yang, S.; Zheng, Y.; Zhou, L.; Jin, J.; Deng, Y.; Yao, J.; Yang, P.; Yao, L.; Wu, Y.; Zhai, Z.; et al. miR-499 rs3746444 and miR-196a-2 rs11614913 Are Associated with the Risk of Glioma, but Not the Prognosis. Mol. Ther. Nucleic Acids 2020, 22, 340–351. [Google Scholar] [CrossRef]
  67. Ahmad, M.; Shah, A.A. Predictive role of single nucleotide polymorphism (rs11614913) in the development of breast cancer in Pakistani population. Pers. Med. 2020, 17, 213–227. [Google Scholar] [CrossRef]
  68. Liu, Y.; He, A.; Liu, B.; Zhong, Y.; Liao, X.; Yang, J.; Chen, J.; Wu, J.; Mei, H. rs11614913 polymorphism in miRNA-196a2 and cancer risk: An updated meta-analysis. OncoTargets Ther. 2018, 11, 1121–1139. [Google Scholar] [CrossRef] [Green Version]
  69. Soltanian, A.R.; Hosseini, B.; Mahjub, H.; Bahreini, F.; Nazemalhosseini Mojarad, E.; Ghaffari, M.E. Association between rs11614913 Polymorphism of The MiR-196-a2 Gene and Colorectal Cancer in The Presence of Departure from Hardy-Weinberg Equilibrium. Cell J. 2021, 23, 313–318. [Google Scholar]
  70. Zhu, Z.; Zhang, Y.; Bai, R.; Yang, R.; Shan, Z.; Ma, C.; Yang, J.; Sun, D. Association of Genetic Polymorphisms in MicroRNAs With Type 2 Diabetes Mellitus in a Chinese Population. Front. Endocrinol. 2020, 11, 587561. [Google Scholar] [CrossRef]
  71. Zhang, S.; Han, Q.; Zhu, K.; Wang, Q. The association of miR-27a rs895819 polymorphism with colorectal cancer risk in Chinese population. J. Clin. Lab. Anal. 2020, 34, e23497. [Google Scholar] [CrossRef]
Figure 1. The up-ward arrows show the mature sequence of hsa-mir-27a-3p while the red arrows indicate the seed region (6 nts). Stare (*) Indicates the location of all SNPs in hsa-mir-27a-5p.
Figure 1. The up-ward arrows show the mature sequence of hsa-mir-27a-3p while the red arrows indicate the seed region (6 nts). Stare (*) Indicates the location of all SNPs in hsa-mir-27a-5p.
Genes 13 00747 g001
Table 1. Pre-clinical data about age, gender, BMI, RBS, TC, TG, HDL, and LDL of the CAD patients, as well as healthy controls.
Table 1. Pre-clinical data about age, gender, BMI, RBS, TC, TG, HDL, and LDL of the CAD patients, as well as healthy controls.
CategoriesAge (Year)GenderBMI (kg/m2)RBS (mg/dL)TC (mg/dL)TG (mg/dL)HDL (mg/dL)LDL (mg/dL)
CAD55.2 (27–91)Male = 18323.3 (12.2–38.2)245.8 (114–415)222.7 (156–262)199.3 (110–395)38.2 (21–58)126.8 (24–271)
Female = 40
Controls±45Male = 138±22.3±112.4±200±162.5±35±100
Female = 12
BMI: Body mass index, RBS: Random blood sugar, TC: Total cholesterol, TG: Triglyceride, HDL: High-density lipoprotein, LDL: Low-density lipoprotein.
Table 2. Shows the list of studied SNPs, their official name, mature miRNA sequences, chromosomal location, and MAF.
Table 2. Shows the list of studied SNPs, their official name, mature miRNA sequences, chromosomal location, and MAF.
SNP IDmiRNA Gene NameName of Mature miRNA SequencesChromosome No.miRNA Location (Coordinates)Coded AlleleOther AllelesMAF
rs895819MIR27Ahsa-miR-27a-5p1913836440-13836517 [−]TA/C/G0.50
hsa-miR-27a-3p
rs11614913MIR196A2hsa-miR-196a-5p1253991738-53991847 [+]CT0.49
hsa-miR-196a-3p
rs2168518MIR4513hsa-miR-45131574788672-74788757 [−]GA0.47
Table 3. Inheritance models for investigating the association of rs895819, rs11614913, and rs2168518 with CAD.
Table 3. Inheritance models for investigating the association of rs895819, rs11614913, and rs2168518 with CAD.
Gene (Accession Number)Statistical ModelsGenotypesCasesControlOdds Ratiο (95% CI)χ2-Value, dfp-Value
MIR196A2 (rs11614913)Co-dominantCC CT TT24 40 1650 19 1154.4, 2<0.0001
DominantCC CT + TT24 5650 300.257 (0.133–0.496)<0.0001
RecessiveTT CT + CC16 6411 691.56 (0.677–0.632)0.398
AdditiveC T88 72119 410.421 (0.262–0.675)0.0004
MIR27A (rs895819)Co-dominantAA AG GG10 46 428 35 59.669, 2<0.008
DominantAA AG + GG10 5028 400.285 (0.1242–0.6575)<0.0034
RecessiveGG AG + AA4 565 630.900 (0.3202–3.519)1.000
AdditiveA G66 5491 450.604 (0.3640–1.002)0.05
MIR4513 (rs2168518)Co-dominantGG GA AA14 105 244 47 193.682, 20.1586
DominantGG GA + AA14 1294 661.791 (0.5668–5.658)0.4340
RecessiveAA GA + GG24 11919 510.5414 (0.2727–1.075)0.1012
AdditiveG A133 15355 851.343 (0.8905–2.027)0.1773
Table 4. Information about the free energy of the thermodynamic ensemble, the frequency of the MFE structure in the ensemble, the ensemble diversity, the optimal secondary structure with minimum free energy, and the centroid secondary structure of studied reference SNPs and their mutated variants.
Table 4. Information about the free energy of the thermodynamic ensemble, the frequency of the MFE structure in the ensemble, the ensemble diversity, the optimal secondary structure with minimum free energy, and the centroid secondary structure of studied reference SNPs and their mutated variants.
ParametersReferenceMutatedReferenceMutatedReferenceMutated
MIR4513 rs2168518MIR4513 rs2168518MIR27A rs895819MIR27A rs895819MIR196A2 rs11614913MIR196A2 rs11614913
Free energy of the thermodynamic ensemble−41.83 kcal/mol−42.34 kcal/mol−38.24 kcal/mol−38.28 kcal/mol.−52.02 kcal/mol−46.52 kcal/mol
The frequency of the MFE structure in the ensemble26.16%18.55%15.62%.14.84%.6.14%5.18%
The ensemble diversity3.583.504.414.557.187.49
The optimal secondary structure with a minimum free energy−41.00 kcal/mol −41.30 kcal/mol−34.40 kcal/mol−34.40 kcal/mol−50.30 kcal/mol−44.70 kcal/mol
The centroid secondary structure−41.00 kcal/mol−41.30 kcal/mol−37.10 kcal/mol−37.10 kcal/mol−49.90 kcal/mol−44.30 kcal/mol
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Haq, T.U.; Zahoor, A.; Ali, Y.; Chen, Y.; Jalil, F.; Shah, A.A. Genetic Variants of MIR27A, MIR196A2 May Impact the Risk for the Onset of Coronary Artery Disease in the Pakistani Population. Genes 2022, 13, 747. https://doi.org/10.3390/genes13050747

AMA Style

Haq TU, Zahoor A, Ali Y, Chen Y, Jalil F, Shah AA. Genetic Variants of MIR27A, MIR196A2 May Impact the Risk for the Onset of Coronary Artery Disease in the Pakistani Population. Genes. 2022; 13(5):747. https://doi.org/10.3390/genes13050747

Chicago/Turabian Style

Haq, Taqweem Ul, Abdul Zahoor, Yasir Ali, Yangchao Chen, Fazal Jalil, and Aftab Ali Shah. 2022. "Genetic Variants of MIR27A, MIR196A2 May Impact the Risk for the Onset of Coronary Artery Disease in the Pakistani Population" Genes 13, no. 5: 747. https://doi.org/10.3390/genes13050747

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