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

Interleukin-17A Gene Haplotypes Are Associated with Risk of Premature Coronary Artery Disease in Mexican Patients from the Genetics of Atherosclerotic Disease (GEA) Study

Abstract

Aim

The role of interleukin 17A (IL-17A) in the inflammatory process has caused interest in the potential significance of IL-17A as a biomarker for coronary artery disease (CAD). The aim of the present study was to evaluate the role of IL-17A gene polymorphisms as susceptibility markers for CAD in the Mexican population.

Methods

Four IL-17A gene polymorphisms (rs8193036, rs3819024, rs2275913 and rs8193037) were genotyped by 5’ exonuclease TaqMan assays in a group of 900 patients with premature CAD and 667 healthy controls (with negative calcium score by computed tomography), seeking associations with CAD and other metabolic and cardiovascular risk factors using logistic regression analyses.

Results

No single IL-17A polymorphism was associated with premature CAD, however two haplotypes (CAGG and TAGA) were significantly associated with increased risk of premature CAD (OR = 1.35, 95% CI: 1.00–1.84, P = 0.018 and OR = 2.09, 95% CI: 1.16–3.76, P = 0.003, respectively). Moreover, rs3819024 was associated with increased levels of visceral abdominal fat (P = 0.002) and rs8193036 was significantly associated with risk of central obesity (P = 0.020), hypertriglyceridemia (P = 0.027), and metabolic syndrome (P = 0.027) in the premature CAD group, under dominant models adjusted by age, gender, BMI, smoking history, alcohol consumption, and treatment.

Conclusion

The results suggest that IL-17A haplotypes are involved in the risk of developing premature CAD and some IL-17A polymorphisms are associated with cardiovascular risk factors in Mexican individuals with premature CAD.

Introduction

Coronary artery disease (CAD) is a complex multifactorial and polygenic disorder resulting from an excessive inflammatory response to various forms of injurious stimuli to the arterial wall [13]. Inflammation is recognized as a major contributor to atherogenesis through adverse effects on lipoprotein metabolism and arterial wall biology [4]. Interleukin-17A (IL-17A) is the most widely studied member of the IL-17 cytokine family. It is mainly produced by T-helper (Th)-17 lymphocytes, but is also secreted by natural killer T (NKT) cells, γδ T cells (γδ-17), cytotoxic CD8+ T cells (Tc17), and neutrophils [57]. High circulating IL-17A levels have been observed in patients with chronic inflammatory diseases such as rheumatoid arthritis, multiple sclerosis and inflammatory bowel disease, and IL-17A is considered a proinflammatory cytokine [811].

Several lines of evidence both in animal models and in humans have shown that IL-17A is involved in atherosclerosis. IL-17A plays a proatherogenic inflammatory role during atherogenesis by promoting monocyte/macrophage recruitment into the aortic wall in the mouse model [12], while the administration of IL-17A neutralizing antibodies to apolipoprotein E (ApoE−/−) knockout mice markedly reduced early atherosclerotic lesion area and vulnerability [13]. Moreover IL-17A can increase plaque vulnerability in human specimens [14]. Human coronary artery infiltrating T-cells produce IL-17A and gamma interferon, acting synergistically to induce proinflammatory responses in vascular smooth vessel cells [15]. Other studies have reported the proatherogenic effect of IL-17A [16,17]. These findings have suggested that IL-17A might play a role in the development of cardiovascular diseases.

The IL17A gene is mapped on the chromosome 6 and presents several polymorphisms [18]. Some of these polymorphisms have been associated with risk of developing several disease including cancer, gastro-duodenal disease, chronic periodontal disease, and inflammatory bowel disease [1922]. To our knowledge there is only one report of an association of IL-17A gene variation and coronary artery disease, found in the Han Chinese population [23]. Gene variation association studies may vary among populations due to genetic differences, including differences in allele frequencies and linkage disequilibrium (LD) structures. Therefore, it is important to examine multiple ethnic populations for the identification of ethnicity-specific loci as well as common susceptibility loci. Considering the important role of the IL-17A cytokine in the development of atherosclerosis and the lack of studies of the role of polymorphisms of this gene in coronary artery disease, the aim of the present study was to analyze if IL-17A gene polymorphisms are associated with premature coronary artery disease (CAD) in a case-control association study (GEA or Genetics of Atherosclerotic Disease).

Material and Methods

Subjects

All participants provided written informed consent. The study complies with the Declaration of Helsinki and was approved by the Ethics Committee of the Instituto Nacional de Cardiología Ignacio Chávez (INCICH). The primary aim of the Genetics of Atherosclerotic Disease (GEA) Study is to investigate genetic factors associated with premature CAD, and other coronary risk factors in the Mexican population. All GEA participants are unrelated and of self-reported Mexican-Mestizo ancestry (three generations). A total of 1853 individuals were included in the study, 900 diagnosed with premature CAD and 953 apparently healthy controls. Premature CAD was defined as history of myocardial infarction, angioplasty, revascularization surgery or coronary stenosis >50% on angiography, diagnosed before age 55 in men and before age 65 in women. Controls were apparently healthy asymptomatic individuals without family history of premature CAD, recruited from blood bank donors and through brochures posted in Social Services centers. Exclusion criteria for controls included congestive heart failure, liver, renal, thyroid or oncological disease. The selection of the patients and controls of the GEA study were described in a previous study [24]. Demographic, clinical, anthropometric, biochemical parameter and cardiovascular risk factors were evaluated in both patients and controls. Anthropometric parameters were measured by trained personnel and included waist circumference and body mass index (BMI) calculated as weight in kilograms divided by height in square meters. Blood pressure was measured three different times by sphygmomannometry and the average of the last two measurements was obtained. Obesity was defined as BMI ≥30 kg/m2. Central obesity, hypoalphalipoproteinemia, hypertriglyceridemia, and metabolic syndrome were defined using Adult Treatment Panel III (ATP-III) criteria 2002 [25]. Hypercholesterolemia was defined as total cholesterol (TC) levels ≥200 mg/dL. Hypertension was defined as systolic blood pressure ≥140 mmHg and/or diastolic blood pressure ≥90 mmHg or the use of oral antihypertensive therapy. Type 2 Diabetes mellitus (T2DM) was diagnosed according to World Health Organization criteria.

Computed Tomography of the Chest and Abdomen

Computed tomography of the chest and abdomen were performed using a 64-channel multi-detector helical computed tomography system (Somatom Sensation, Siemens) and interpreted by experienced radiologists. Scans were read to assess and quantify the following: 1) Coronary artery calcification (CAC) score using the Agatston method [26]; 2) total abdominal, subcutaneous and visceral adipose tissue areas as described by Kvist et al. [27] in order to calculate visceral to subcutaneous adipose tissue ratio (VAT/SAT); and 3) hepatic to splenic attenuation ratio (LSAR) as described by Longo et al. [28]. The tomography was performed in all the patients and healthy controls. However, 286 of apparently healthy individuals were positive for CAC (CAC score >0) and were considered as individuals with subclinical atherosclerosis (SA). These individuals were excluded from the case-control analysis, and the control group only included individuals with negative CAC (n = 667).

Genetic analysis

Genomic DNA from whole blood containing EDTA was isolated by standard techniques. The rs8193036, rs3819024, rs2275913 and rs8193037 IL-17A single nucleotide polymorphisms (SNPs) were genotyped using 5’ exonuclease TaqMan genotyping assays on an ABI Prism 7900HT Fast Real-Time PCR system, according to manufacturer’s instructions (Applied Biosystems, Foster City, CA, USA).

Statistical analysis

All calculations were performed using SPSS version 18.0 (SPSS, Chicago, Il) statistical package. Means ± SD and frequencies of baseline characteristics were calculated. Chi-square tests were used to compare frequencies and ANOVA and Students t-test were used to compare means. ANCOVA was used to determine associations between the polymorphisms and metabolic variables, adjusting by age, gender, BMI, smoking history, alcohol consumption, and treatment, as appropriate. Logistic regression analysis was used to test for associations of polymorphisms with premature CAD under different inheritance models. Genotype frequencies did not show deviation from Hardy-Weinberg equilibrium (P > 0.05). Pairwise linkage disequilibrium (LD, D′) estimations between polymorphisms and haplotype reconstruction were performed with Haploview version 4:1 (Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA).

Functional prediction analysis

We predicted the potential effect of the IL-17A SNPs using bioinformatics tools, including FastSNP [29], SNP Function Prediction (http://snpinfo.niehs.nih.gov/snpfunc.htm), Human-transcriptome Database for Alternative Splicing (http://www.h-invitational.jp/h-dbas/), Splice Port: An Interactive Splice Site Analysis Tool (http://www.spliceport.cs.umd.edu/SplicingAnalyser2.html), ESE finder (http://rulai.cshl.edu/cgi-bin/tools/ESE3/esefinger.cgi), HSF (http://www.umd.be/HSF), and SNPs3D (http://www.snps3d.org/).

Results

General characteristics of the population are shown in Tables 1 and 2.

thumbnail
Table 2. Comparison of biochemical parameters in individuals with premature coronary artery disease and controls.

https://doi.org/10.1371/journal.pone.0114943.t002

Association of polymorphisms with premature CAD

The distribution of the studied polymorphisms was similar in premature CAD patients and healthy controls under all inheritance models tested (Table 3), and no associations of single SNPs with CAD were observed.

thumbnail
Table 3. Associations of IL17A polymorphisms with premature CAD.

https://doi.org/10.1371/journal.pone.0114943.t003

Association of the polymorphisms with metabolic cardiovascular risk factors and metabolic parameters

The effect of the polymorphisms on various metabolic parameters and cardiovascular risk factors was analyzed separately in the premature CAD and control (CAC score = 0) groups. No associations were observed in controls; however, associations with some metabolic parameters were observed in the premature CAD group. The rs8193036 polymorphism was associated with increased risk of central obesity (OR = 1.67, 95% CI: 1.14–2.45, Pdom = 0.020), metabolic syndrome (OR = 1.38, 95% CI: 1.05–1.81, Pdom = 0027) and decreased risk of hypertriglyceridemia (OR = 0.75, 95% CI: 0.56–0.97, Pdom = 0.027), all under a dominant model adjusted by age, gender, BMI, smoking history, alcohol consumption, and treatment (Table 4). Moreover, the rs3819024 polymorphism was significantly associated with increased levels of visceral abdominal fat (OR = 1.003, 95% CI: 1.002–1.005, P = 0.002) and the rs8193037 polymorphism was associated with an increased risk of metabolic syndrome (OR = 1.68, 95% CI: 1.16–2.43, Pdom = 0.005) under dominant models adjusted by the same variables (data not shown).

thumbnail
Table 4. Associations of the rs8193036/IL17A variant with metabolic risk factors in premature CAD individuals.

https://doi.org/10.1371/journal.pone.0114943.t004

Haplotype analysis and SNP functional prediction

The IL-17A polymorphisms were in high linkage disequilibrium (D’>0.8 and r2>0.9) and 10 different haplotypes were observed. Two of these haplotypes (H3: CAGG and H6: TAGA) were significantly associated with increased risk of premature CAD (OR = 1.35, 95% CI: 1.00–1.84, P = 0.018 and OR = 2.09, 95% CI: 1.16–3.76, P = 0.003, respectively) (Table 5).

thumbnail
Table 5. IL17A haplotypes frequencies in premature CAD and healthy controls.

https://doi.org/10.1371/journal.pone.0114943.t005

Based on SNP functional prediction software, the rs8193036 polymorphism seems to be functional. The presence of the C allele in this polymorphism produces a DNA binding site for GATA-3 transcription factors with possible consequences on IL-17A expression.

Discussion

IL-17A is a pleiotropic cytokine with effects on multiple cell types to enhance the production of proinflammatory molecules and several lines of evidence have shown it is involved in the pathogenesis of atherosclerosis [12, 1517, 30]. In spite of this known role of the IL-17A in the development of atherosclerosis, only one study has explored whether IL-17A gene variation plays a role in susceptibility to coronary artery disease. Zhang et al. [23] studied 5 IL-17A polymorphisms (rs4711998, rs3819024, rs2275913, rs8193037, and rs3819025) in 1031 CAD patients and 935 healthy controls, reporting a significant association of rs8193037 with CAD in the Chinese Han population. In our study, this polymorphism was not associated with premature CAD, but was associated with metabolic syndrome in the premature CAD group. Moreover, rs3819024 was associated with increased levels of visceral abdominal fat and rs8193036 (not studied by Zhang et al.) was associated with risk of central obesity, metabolic syndrome and hypertriglyceridemia in Mexican CAD patients. It is important to note that Zhang et al. (2011) did not examine the association between these polymorphisms and metabolic parameters or other cardiovascular risk factors, and these observations must be confirmed in other studies.

The functional software used here predicted that rs8193036 as a functional polymorphism. The presence of the rs8193036 C allele produces a DNA binding site for the transcription factors GATA-3 with possible consequences increasing IL-17A expression and its pro-inflammatory effects. The increased inflammation could affect various metabolic parameters and cardiovascular risk factors, explaining the associations observed in the premature CAD group. Although the rs8193036 functional prediction is in agreement with our observations, the results must be interpreted with caution as we did not perform expression analysis and have no evidence that the IL-17A expression is in fact different in premature CAD patients with the risk allele.

In our study, the IL-17A polymorphisms were in strong linkage disequilibrium, and two of the haplotypes (H3: CAGG and H6: TAGA) were associated with premature CAD. To our knowledge, there are no previous studies analyzing associations of these haplotypes with CAD or any other disease.

Study limitations need to be addressed. In this study, only four IL-17A polymorphisms were analyzed. Since this is the first study reporting associations of IL-17A polymorphisms with metabolic parameters and cardiovascular risk factors, replication in other groups of patients is necessary to confirm our results. The predicted functional consequences of the rs8193036 polymorphism using informatics tools need to be corroborated by experimental evidence. A positive point of our work is that the control group only included individuals without subclinical atherosclerosis (individuals without coronary artery calcification).

Conclusions

In summary, our study demonstrates the association of some IL-17A polymorphism with metabolic parameters and cardiovascular risk factor in a group of Mexican CAD patients. The four IL-17A polymorphisms were in high linkage disequilibrium and two haplotypes were significantly associated with premature CAD risk. According to the informatics software, the rs8193036 polymorphism found to be associated with central obesity, metabolic syndrome and hypertriglyceridemia is predicted to have a functional effect. Because the Mexican population has a distinct genetic background [3134], the IL-17A polymorphism associations observed here should be explored in other populations.

Acknowledgments

This work was supported in part by grants from the Consejo Nacional de Ciencia y Tecnología (Project number 156911).

Author Contributions

Conceived and designed the experiments: GVA CPR. Performed the experiments: EAL RPS GCS NPH. Analyzed the data: JAM TVM. Contributed reagents/materials/analysis tools: JMF JMRP JRB JGJR. Wrote the paper: GVA TVM.

References

  1. 1. Ross R (1999) Atherosclerosis an inflammatory disease. N Engl J Med 340:115–126. pmid:9887164
  2. 2. Garcia-Moll X (2005) Inflammatory and Anti-Inflammatory Markers in Acute Coronary Syndromes. Ready for Use in the Clinical Setting?. Rev Esp Cardiol 58:615–617. pmid:15970115
  3. 3. Lusis AJ (2000) Atherosclerosis. Nature 407:233–241. pmid:11001066
  4. 4. Hansson GK, Libby P (2006) The immune response in atherosclerosis: a double-edged sword. Nat Rev Immunol 6:508–519. pmid:16778830
  5. 5. D’Acquisto F, Maione F, Pederzoli-Ribeil M (2010) From IL-15 to IL-33: the never-ending list of new players in inflammation. Is it time to forget the humble aspirin and move ahead? Biochem Pharmacol 79:525–534. pmid:19765545
  6. 6. Onishi RM, Gaffen SL (2010) Interleukin-17 and its target genes: mechanisms of interleukin-17 function in disease. Immunology 129: 311–321. pmid:20409152
  7. 7. Iwakura Y, Ishigame H, Saijo S, Nakae S (2011) Functional specialization of interleukin-17 family members. Immunity 34: 149–162. pmid:21349428
  8. 8. Maione F, Paschalidis N, Mascolo N, Dufton N, Perretti M, et al. (2009) Interleukin 17 sustains rather than induces inflammation. Biochem Pharmacol 77:878–887. pmid:19073154
  9. 9. Cheng X, Yu X, Ding YJ, Fu QQ, Xie JJ, et al. (2008) The Th17/Treg imbalance in patients with acute coronary syndrome. Clin Immunol 127:89–97. pmid:18294918
  10. 10. Leipe J, Grunke M, Dechant C, Reindl C, Kerzendorf U, et al. (2010) Role of Th17 cells in human autoimmune artritis. Arthritis Rheum 62:2876–2885. pmid:20583102
  11. 11. Zhao XF, Pan HF, Yuan H, Zhang WH, Li XP, et al. (2010) Increased serum interleukin 17 in patients with systemic lupus erythematosus. Mol Biol Rep 37:81–85. pmid:19347604
  12. 12. Smith E, Prasad KM, Butcher M, Dobrian A, Kolls JK, et al. (2010) Blockade of interleukin-17A results in reduced atherosclerosis in apolipoprotein E- deficient mice. Circulation 121:1746–1755. pmid:20368519
  13. 13. Erbel C, Chen L, Bea F, Wangler S, Celik S, et al. (2009) Inhibition of IL-17A attenuates atherosclerotic lesion development in ApoE-deficient mice. J Immunol 183:8167–8175. pmid:20007582
  14. 14. de Boer OJ, van der Meer JJ, Teeling P, van der Loos CM, Idu MM, et al. (2010) Differential expression of interleukin-17 family cytokines in intact and complicated human atherosclerotic plaques. J Pathol 220:499–508. pmid:20020510
  15. 15. Eid RE, Rao DA, Zhou J, Lo SF, Ranjbaran H, et al. (2009) Interleukin-17 and interferon-γ are produced concomitantly by human coronary artery-infiltrating T cells and act synergistically on vascular smooth muscle cells. Circulation 119:1424–1432. pmid:19255340
  16. 16. Chen S, Shimada K, Zhang W, Huang G, Crother TR, et al. (2010) IL-17A is proatherogenic in high-fat diet-induced and chlamydia pneumoniae infection- accelerated atherosclerosis in mice. J Immunol 185:5619–5627. pmid:20935201
  17. 17. Xie JJ, Wang J, Tang TT, Chen J, Gao XL, et al. (2010) The Th17/Treg functional imbalance during atherogenesis in ApoE−/− mice. Cytokine 49:185–193. pmid:19836260
  18. 18. Aggarwal S 1, Gurney AL (2002) IL-17: prototype member of an emerging cytokine family. J Leukoc Biol 71:1–8. pmid:11781375
  19. 19. Kadkhodazadeh M 1, Baghani Z, Ebadian AR, Youssefi N, Mehdizadeh AR, et al. (2013) IL-17 gene polymorphism is associated with chronic periodontitis and peri-implantitis in Iranian patients: a cross-sectional study. Immunol Invest 42:156–163. pmid:23323524
  20. 20. Zhang X, Yu P, Wang Y, Jiang W, Shen F, et al. (2013) Genetic polymorphisms of interleukin 17A and interleukin 17F and their association with inflammatory bowel disease in a Chinese Han population. Inflamm Res 62:743–750. pmid:23652560
  21. 21. Hayashi R 1, Tahara T, Shiroeda H, Matsue Y, Minato T, et al. (2012) Association of genetic polymorphisms in IL17A and IL17F with gastroduodenal diseases. J Gastrointestin Liver Dis 21:243–249. pmid:23012664
  22. 22. Qinghai Z, Yanying W, Yunfang C, Xukui Z, Xiaoqiao Z (2014) Effect of interleukin-17A and interleukin-17F gene polymorphisms on the risk of gastric cancer in a Chinese population. Gene 537:328–332. pmid:24315816
  23. 23. Zhang X, Pei F, Zhang M, Yan C, Huang M, et al. (2011) Interleukin-17A gene variants and risk of coronary artery disease: a large angiography-based study. Clin Chim Acta 412:327–331. pmid:21062626
  24. 24. Villarreal-Molina T, Posadas-Romero C, Romero-Hidalgo S, Antúnez-Argüelles E, Bautista-Grande A, et al. (2012) The ABCA1 gene R230C variant is associated with decreased risk of premature coronary artery disease: the genetics of atherosclerotic disease (GEA) study. PLoS One 7:e49285. pmid:23152888
  25. 25. Third report of the National Cholesterol Education Program (NCEP) expert panel on detection, evaluation, and treatment of high blood cholesterol in adults (Adult Treatment Panel III) final report (2002). Circulation 106:3134–3421.
  26. 26. Mautner GC, Mautner SL, Froehlich J, Feuerstein IM, Proschan MA, et al. (1994) Coronary artery calcification: assessment with electron beam CT and histomorphometric correlation. Radiology 192:619–623. pmid:8058924
  27. 27. Kvist H, Chowdhury B, Grangård U, Tylén U, Sjöström L (1988) Total and visceral adipose-tissue volumes derived from measurements with computed tomography in adult men and women: predictive equations. Am J Clin Nutr 48:1351–1361. pmid:3202084
  28. 28. Longo R, Ricci C, Masutti F, Vidimari R, Crocé LS, et al. (1993) Fatty infiltration of the liver. Quantification by 1H localized magnetic resonance spectroscopy and comparison with computed tomography. Invest Radiol 28:297–302. pmid:8478169
  29. 29. Yuan HY, Chiou JJ, Tseng WH, Liu CH, Liu CK, et al. (2006) FASTSNP: an always up-to-date and extendable service for SNP function analysis and prioritization. Nucleic Acids Res 34 (Web Server issue); W635–641 [accessed on 2012]. pmid:16845089
  30. 30. Chang SH, Dong C (2011) Signaling of interleukin-17 family cytokines in immunity and inflammation. Cel Signal 23:1069–1075. pmid:21130872
  31. 31. Lisker R, Perez-Briceño R, Granados J, Babinsky V, de Rubens J, et al. (1986) Gene frequencies and admixture estimates in a Mexico City population. Am J Phys Anthropol 71:203–207. pmid:3099584
  32. 32. Lisker R, Pérez-Briceno R, Granados J, Babinsky V (1988) Gene frequencies and admixture estimates in the state of Puebla, Mexico. Am J Phys Anthropol 76:331–335. pmid:3414796
  33. 33. Lisker R, Ramirez E, Briceño RP, Granados J, Babinsky V (1990) Gene frequencies and admixture estimates in four Mexican urban centers. Hum Biol 62:791–801. pmid:2262203
  34. 34. Juárez-Cedillo T, Zuñiga J, Acuña-Alonzo V, Pérez-Hernández N, Rodríguez-Pérez JM, et al. (2008) Genetic admixture and diversity estimations in the Mexican Mestizo population from Mexico City using 15 STR polymorphic markers. Forensic Sci Int Genet 2:e37–39. pmid:19083813