Circulation Journal
Online ISSN : 1347-4820
Print ISSN : 1346-9843
ISSN-L : 1346-9843
Reviews
Unraveling New Therapeutic Targets of Coronary Artery Disease by Genetic Approaches
Sang Eun LeeHyo-Soo Kim
著者情報
ジャーナル フリー HTML

2014 年 79 巻 1 号 p. 8-14

詳細
Abstract

Coronary artery disease (CAD) is the most common cause of death and physical disabilities in developed countries, even though efforts to identify and target causal factors such as hypertension and dyslipidemia have brought tremendous improvements in prevention and treatment. A rapid advance in technology has unraveled new genetic variants associated with CAD and also provided great opportunities to identify novel pathogenic mechanisms and to develop new drugs with higher specificity. Whole-genome sequencing and whole-exome sequencing has made it possible to find rare alleles that are responsible for CAD in small, affected families and case-control studies in a very efficient manner. At present, genome-wide association studies have identified more than 50 loci that explain approximately 10% of the heritability of CAD, most of which is unrelated to traditional risk factors. Mendelian randomization studies enable identification of causal factors among numerous biomarkers and to narrow down promising therapeutic targets. This review highlights new genetic approaches and demonstrates the extent to which the outcome contributes to the finding of new therapeutic targets. (Circ J 2015; 79: 8–14)

Coronary artery disease (CAD) is the leading cause of mortality and morbidity worldwide. It is a complex disease resulting from the interplay between multiple genetic variants and environmental factors, and genetic variability explains only a small part of the whole pathogenic mechanism of the disease. Nevertheless, in the goal of detecting new therapeutic targets, reliable identification and thorough understanding of potential causal genetic variants can be crucial. For example, in the case of low-density lipoprotein (LDL) cholesterol and statins, the discovery of mutations affecting the LDL receptor and causing hypercholesterolemia and early-onset myocardial infarction (MI) enabled the use of LDL cholesterol-lowering therapies that remarkably reduce the risk of cardiovascular events.1 In 2003, a finding that gain-of-function mutations in the proprotein, converase stubilisin/kexin9 (PCSK9) is a causal event in 2 families with autosomal dominant hypercholesterolemia turned out to have a positive correlation with the incidence of CAD.2 PCSK9 was then identified as binding the LDL receptor for degradation and reducing the capacity of the liver to bind and remove LDL cholesterol.3,4 Subsequent studies revealed that some patients with PCSK9 loss-of-function mutations had a low LDL cholesterol level and a reduced incidence of CAD.58 These study results raised the possibility that pharmacologic inhibition of PCSK9 might lower the LDL cholesterol level in patients with hypercholesterolemia. In fact, recent clinical trials proved that a human monoclonal antibody to PCSK9 resulted in a significant reduction of LDL cholesterol levels in patients.911

The results from genetic studies can be applied to further research to improve disease risk prediction, pharmacogenomics and identifying new therapeutic targets. This review will mainly deal with the therapeutic aspect: to be more specific, how recent technical advances in the study of genetics help identify novel causative biochemical pathways and therapeutic targets in CAD.

Whole-Genome and Whole-Exome Sequencing in Linkage Analyses

According to the pattern of inheritance, any genetic disease can be divided into 2 classes. A monogenic or Mendelian disease is caused by mutations in 1 gene and follows Mendel’s law of inheritance, whereas a polygenic or complex disease is characterized by complex patterns of inheritance caused by interaction between multiple genetic variants and environmental factors. The latter type of CAD is common; however, there are a few cases of CAD showing a monogenic inheritance pattern and identification of genes associated with this monogenic disease has led to breakthrough understanding of the mechanism of the disease. A representative example is the Nobel Prize-winning discovery that mutations affecting the LDL receptor also cause hypercholesterolemia and early-onset ischemic heart disease (IHD).1

Traditionally, linkage analysis (parametric) was adopted for Mendelian disease and gave researchers a powerful technique for mapping the location of disease-causing loci by identifying genetic markers that are co-inherited with a phenotype of interest (segregation).12 First, a particular region within the genome responsible for a disease of interest must be identified using DNA markers, then a chromosomal region that segregates with the disease can be pinpointed to identify the causal gene and its variant. This type of study also successfully identified the association between ALOX5AP and coronary and cerebrovascular disease,13 between MEF2A and CAD,14 and between genetic variation of PCSK9 and cholesterol metabolism.15

Although previous linkage analyses in affected families followed the sequence of finding the regions first and then identifying the causal gene and the mutation by fine-mapping (positional cloning), so-called next-generation sequencing (NGS) now allows direct screening of the candidate variants and performance of linkage analyses. The NGS technologies are based on parallel sequencing of millions of DNA strands simultaneously. It dramatically decreases the cost and time to sequence the entire human genome. Less than $1,000 and 1 week is all that is needed to sequence a whole genome. Compared with whole-genome sequencing, whole-exome sequencing focuses on an alignment of exons, collectively called ‘the exome’, which constitutes only approximately 1% of the human genome but harbors 85% of occurring mutations that affect disease-related traits.16,17 Detailed technical review of the existing platforms can be found elsewhere.18 A major advantage of these approaches is that less family members are required to have their genome sequenced to identify a particular genetic variation linked to a disease phenotype compared with conventional linkage analysis with positional cloning.1921

Using whole-exome sequencing and linkage analysis, Erdmann et al22 identified 2 heterozygous mutations in 2 functionally related genes, GUCY1A3 and CCT7, which impair nitric oxide signaling in an extended MI family. Only 3 distantly related members of the affected family underwent whole-exome sequencing and it was enough to screen out 4 candidate variants. Another study by Keramati et al also used the same method to identify a founder mutation of DYRK1B in 3 large families with coinheritance of early-onset CAD, central obesity, hypertension, and diabetes.23 These pioneering studies highlight the opportunities of applying NGS to severely affected families to identify new mechanism of the disease and thereby new therapeutic targets.

Genome-Wide Association Studies and Whole-Exome Sequencing in Association Study

Performing a genome-wide association study (GWAS) is a genetic approach to a common disease based on a ‘common disease-common variant hypothesis’ that complex diseases result from cumulative and interactive effects of a large number of loci, each imparting a modest marginal effect to the expression of a phenotype.21 It compares the frequency of repeatedly appearing genetic variants in case-control studies and if significantly more common in cases, it implies that the variant is in a DNA location associated with an increased risk for the disease. The sequencing of the complete human genome and the rise of high-throughput technologies such as DNA microarray of more than 500,000 single-nucleotide polymorphisms (SNPs)24 have triggered an explosion of GWAS aiming to identify disease-associated genetic loci. Furthermore, meta-analyses of multiple GWAS data sets have considerably increased the accuracy of detecting genetic disease associations.25 In this way, many susceptible loci have been proven to be associated with CAD and MI. The advantages of this approach are that it is unbiased and requires no presupposition of which marker or gene is more likely to be a risk candidate.

To date, more than 50 genetic loci associated with CAD have been reported and they only explain approximately 10% of the heritability of CAD.2629 The most interesting finding is that more than two-thirds of the genetic loci are unrelated to traditional risk factors, which highlights that several pathways contributing to the pathogenesis of CAD are yet to be appreciated and there will be more therapeutic targets to unravel26 (Table 1).

Table 1. Genetic Loci Associated With Coronary Artery Disease in Genome-Wide Association Studies
Chromosomal location SNP Related gene(s) Risk allele OR (95% CI) P value
1p13.3 rs599839 SORT1 A 1.29 (1.18–1.40) 4×10−9
1p13.3 rs602633 SORT1 T 1.11 (1.08–1.15) 1×10−8
1p13.3 rs646776 CELSR2, PSRC1, SORT1 T 1.19 (1.13–1.26) 8×10−12
1p32.2 rs17114036 PPAP2B A 1.17 (1.13–1.22) 4×10−19
1p32.3 rs11206510 PCSK9 T 1.15 (1.10–1.21) 1×10−8
1q21.3 rs4845625 IL6R T 1.04 (1.02–1.07) 4×10−8
1q41 rs17465637 MIA3 C 1.14 (1.10–1.19) 1×10−9
2p11.2 rs1561198 VAMP5-VAMP8-GGCX A 1.05 (1.03–1.07) 4×10−9
2p21 rs6544713 ABCG5-ABCG8 T 1.06 (1.04–1.09) 9×10−10
2p24.1 rs2123536 TTC32-WDR35 T 1.12 (1.08–1.16) 7×10−11
2p24.1 rs515135 APOB G 1.08 (1.05–1.11) 5×10−10
2q22.3 rs2252641 ZEB2-AC074093.1 G 1.04 (1.02–1.06) 4×10−8
2q33.2 rs6725887 WDR12 C 1.17 (1.11–1.23) 1×10−8
3q22.3 rs2306374 MRAS C 1.12 (1.07–1.16) 3×10−8
4q31.22 rs1878406 EDNRA T 1.06 (1.02–1.11) 3×10−8
4q32.1 rs7692387 GUCY1A3 G 1.06 (1.03–1.09) 5×10−9
4q32.1 rs1842896 GUCY1A3 T 1.14 (1.10–1.19) 1×10−11
5p15.33 rs11748327 IRX1-LINC01020 N/A 1.25 (1.18–1.33) 5×10−13
5q31.1 rs273909 SLC22A4-SLC22A5 C 1.09 (1.05–1.12) 1×10−8
6p21.2 rs10947789 KCNK5 T 1.06 (1.03–1.08) 2×10−8
6p21.31 rs17609940 ANKS1A G 1.07 (1.05–1.10) 1×10−8
6p21.32 rs9268402 C6orf10-BTNL2 G 1.16 (1.12–1.20) 3×10−15
6p21.33 rs3869109 HLA-C, HLA-B, HCG27 G 1.14 (N/A) 1×10−9
6p24.1 rs12526453 PHACTR1 C 1.12 (1.08–1.17) 1×10−9
6p24.1 rs9369640 PHACTR1 C 1.10 (1.08–1.14) 3×10−11
6p24.1 rs9349379 PHACTR1 G 1.15 (1.10–1.21) 2×10−9
6p24.1 rs6903956 C6orf105 A 1.65 (1.44–1.90) 3×10−13
6q23.2 rs12190287 TCF21 C 1.08 (1.06–1.10) 1×10−12
6q25.3 rs3798220 LPA C 1.51 (1.33–1.70) 3×10−11
6q26 rs4252120 PLG T 1.06 (1.03–1.09) 5×10−9
7p21.1 rs2023938 HDAC9 G 1.07 (1.04–1.11) 5×10−8
7q22.3 rs10953541 BCAP29 C 1.08 (1.05–1.11) 3×10−8
7q32.2 rs11556924 ZC3HC1 C 1.09 (1.07–1.12) 9×10−18
8p21.3 rs264 LPL G 1.05 (1.02–1.08) 5×10−9
8q24.13 rs2954029 TRIB1 A 1.04 (1.02–1.06) 5×10−8
9p21.3 rs1333049 CDKN2A, CDKN2B C 1.37 (1.26–1.48) 2×10−14
9p21.3 rs10757274 CDKN2A, CDKN2B G 1.37 (1.31–1.43) 8×10−45
9q34.2 rs579459 ABO C 1.10 (1.07–1.13) 4×10−14
10p11.23 rs2505083 KIAA1462 C 1.07 (1.04–1.09) 4×10−8
10q11.21 rs1746048 CXCL12 C 1.17 (1.11–1.24) 7×10−9
10q23.31 rs1412444 LIPA T 1.09 (1.07–1.12) 3×10−13
10q24.32 rs12413409 CYP17A1, CNNM2, NT5C2 G 1.12 (1.08–1.16) 1×10−9
11q22.3 rs974819 PDGFD T 1.07 (1.04–1.09) 2×10−9
11q23.3 rs964184 ZNF259, APOA5-A4-C3-A1 G 1.13 (1.10–1.16) 1×10−17
12q21.33 rs7136259 ATP2B1 T 1.11 (1.08–1.15) 6×10−10
12q24.11 rs3782889 MYL2 C 1.26 (1.19–1.34) 4×10−14
12q24.12 rs3184504 SH2B3 T 1.13 (1.08–1.18) 9×10−8
12q24.12 rs11066015 ACAD10, ALDH2, C12orf51,
RPL6-PTPN11
A 1.41 (1.27–1.56) 5×10−11
12q24.13 rs11066280 C12orf51   1.19 (1.13–1.25) 2×10−11
13q12.3 rs9319428 FLT1 A 1.05 (1.03–1.08) 1×10−8
13q34 rs4773144 COL4A1, COL4A2 G 1.07 (1.05–1.09) 4×10−9
14q32.2 rs2895811 HHIPL1 C 1.07 (1.05–1.10) 1×10−10
15q25.1 rs3825807 ADAMTS7 A 1.08 (1.06–1.10) 1×10−12
15q26.1 rs17514846 FURIN-FES A 1.05 (1.03–1.08) 4×10−10
17p11.2 rs12936587 RASD1, SMCR3, PEMT G 1.07 (1.05–1.09) 4×10−10
17p13.3 rs216172 SMG6, SRR C 1.07 (1.05–1.09) 1×10−9
17q21.32 rs46522 UBE2Z, GIP, ATP5G1, SNF8 T 1.06 (1.04–1.08) 2×10−8
19p13.2 rs1122608 LDLR, SMARCA4 G 1.15 (1.10–1.20) 2×10−9
19q13.32 rs2075650 APOE/TOMM40 G 1.14 (1.09–1.19) 3×10−8
21q22.11 rs9982601 KCNE2 T 1.20 (1.14–1.27) 6×10−11

Only P-value <5×10−8. Data from www.genome.gov. CI, confidence interval; OR, odds ratio.

Unfortunately, however, there is a high risk of false-positive results because of multiple comparisons. To alleviate the problem, a stringent statistical significance threshold P-value of 5×10−8 is used and the results must be replicated in an independent population.30 Another drawback is that the identified susceptible loci mostly do not reveal direct information on disease-causing genes or affected pathways, which is essential for a therapeutic drug discovery. Furthermore, it is possible that GWAS just cannot detect rare variants that may explain the missing heritability of CAD. To overcome this limitation, whole-exome sequencing technique is being adopted in a case-control association study of complex diseases. The TG and HDL Working Group of the Exome Sequencing Project identified an aggregate of rare mutations in the gene encoding apolipoprotein C3 (APOC3). They found that the aggregate is associated with a lower plasma triglyceride level and a risk of CAD among the carriers, which was 40% lower than among the noncarriers.31

Mendelian Randomization (MR)

Epidemiologic research has discovered hundreds of biomarkers that associate with CAD.32 Because observational studies cannot draw conclusions about causal relationships, the potential mechanism linking the biomarkers and CAD has been investigated by basic experiments and bench studies.33,34 If the biomarkers are proven to be determinants of the disease in the basic research, the pharmacologists or biotechnologists scrutinize the targets, develop therapeutic agents, and try clinical application. Nevertheless, researchers in field of atherosclerosis biology have rarely succeeded in clinically applying therapeutic agents. Unfortunately, it is only after spending immense time and money that we can judge the validity of the therapeutic agent(s).

A new approach, referred to as MR, can help overcome this limitation and possibly can determine the causal relationship between biomarkers and clinical outcome. It also gives a clue to whether the intervention targeting the biomarker will be effective prior to clinical trials. MR is an observational study that compares the disease state between individuals with and without a certain genotype that predisposes to a phenotype for which we are trying to assess a cause-and-effect relationship. Because genotypes are naturally randomized during sexual reproduction based on Mendel’s second law of independent assortment, the technique is named MR. For example, Timpson et al used SNPs governing the abundance of C-reactive protein (CRP) to assess whether associations between CRP and components of the metabolic syndrome are causal.35

Recent large-scale genetic research including GWAS has created the foundation for MR studies. MR is now rapidly being applied to CAD and many biomarkers have been investigated (Table 2). The results are astonishing enough to challenge previous notions of what is causing CAD. Here are some examples.

Table 2. Mendelian Randomization Studies for CAD
Biomarker SNPs Genes Effect of SNP on CAD Reference
Positive causal relationship
 Blood pressure Multiple (n=30) Multiple + 36
 Fetuin-A Multiple (n=5) AHSG + 37
 IL-6 rs2228145 IL-6R + 38
rs7529229 IL-6R + 39
rs1800795 IL-6 + 40
 LDL rs2228671 LDLR + 41
Multiple (n=3) PCSK9 + 42
 Myeloperoxidase Multiple (n=8) Multiple + 43
 Nonfasting glucose Multiple (n=5) Multiple + 44
 PAI-1 rs1799889 PAI-1 + 45
 Telomere length Multiple (n=7) Multiple + 46
 Triglycerides Multiple (n=44) Multiple + 47
Negative causal relationship
 25-hydroxyvitamin D Multiple (n=6) GC, DHCR7 48
 Bilirubin rs6742078 UGT1A1 49
 CRP Multiple (n=5) Multiple 50
Multiple (n=4) CRP 51
Multiple (n=3) CRP 52
 Fibrinogen Multiple (n=24) Multiple 53
 HDL rs61755018 LIPG 54
Multiple (n=4) ABCA1 55
rs4986970 LCAT 56
 Homocysteine Multiple (n=13) Multiple 57
 Lp-PLA2 rs1051931 PLA2G7 58
 PON1 rs2267829 PON 1 59
 PTX3 Multiple (n=3) Multiple 60
 sPLA2-IIA rs11573156 PLA2G2A 61
 Uric acid rs7442295 SLC2A9 62
rs13129697 SLC2A9 63
Uncertain relationship
 LPA Repeat polymorphism KIV-2 + 64
rs783147/genetic score LPA gene 65
 Adiponectin* Multiple (n=17) Multiple + 66
 Body mass index Multiple (n=14) Multiple 67
Multiple (n=3) Multiple + 68

*Not a true Mendelian randomization trial because pleiotrophic effects of the genetic variants cannot be excluded.

CAD, coronary artery disease; CRP, C-reactive protein; HDL, high-density lipoprotein; IL, interleukin; LDL, low-density lipoprotein; LPA, Lipoprotein (a); SNP, single-nucleotide polymorphism.

High-Density Lipoprotein (HDL) Cholesterol

The most striking example is HDL cholesterol. Although a low serum HDL cholesterol level has been repeatedly observed with the incidence of CAD,6971 the causal relationship has never been established. Fibrates, nicotinic acid, and CETP-inhibitors have been used to increase the HDL cholesterol level in patients in the hope of preventing CAD, yet HDL has not been successfully exploited as therapy.72 The clinical trial and MR study results consistently suggested there is not a causal relationship between the HDL cholesterol level and CAD. Frikke-Schmidt et al used 4 ABCA1 mutations (P1065S, G1216V, N1800H, R2144X) associated with a 17 mg/dl reduction in HDL cholesterol but found no association with IHD.55 The same researchers tested MI using a variant (rs4986970, S208T) in lecithin-cholesterol acyltransferase and had comparable conclusions to their previous study.56 A recent finding is that a SNP in the endothelial lipase gene (LIPG N396S) and a genetic score consisting of 14 common SNPs, which was related to a higher HDL cholesterol level, are not associated with alower risk of MI.54 In contrast to other SNPs specifically related to HDL, cholesterol ester transfer protein (CETP) has reproducibly shown its association with CAD according to the MR study; CETP facilitates the transfer of cholesteryl esters from HDL to LDL cholesterol and dictates the plasma concentration of the 2 lipoproteins.73,74 Therefore, it is unclear at this time whether the effect of lower CETP activity on CAD is related to the change in HDL or LDL cholesterol level.

CRP

Another important biomarker whose causal effect has been thrown into doubt by MR study is CRP, which is an acute-phase protein that increases in response to inflammation. CRP has been shown in several large studies to be strongly associated with IHD and vascular death in a variety of clinical settings.7579 Furthermore, many of the results from basic experiments have suggested a causal role of CRP in the pathogenesis of atherosclerosis and vascular events.8085 Thus, some notable efforts to develop CRP inhibitors to reduce IHD are being made. However, the results from MR studies do not support such efforts. Zacho et al report that genotypic combinations of the 4 CRP polymorphisms (rs3091244, rs1130864, rs1205, and rs3093077) were associated with an increase in CRP levels up to 64%, but that these genotype combinations were not associated with an increased risk of ischemic vascular disease.51 Another study by the C Reactive Protein Coronary Heart Disease Genetics Collaboration (CCGC) used 4 similar CRP genes tagging SNPs (rs3093077, rs1205, rs1130864, rs1800947), each associated with up to 30% per allele difference in the concentration of CRP, and confirmed the genetic variations were not associated with the risk of CAD.52 These findings indicate that CRP alone does not cause cardiovascular disease (CVD) and that lowering CRP will not lessen the occurrence of CVD.

Interleukin-6 (IL-6)

Although CRP is not independently associated with IHD in MR studies, inflammation is considered to play a critical role in atherothrombosis, so several antiinflammatory agents are undergoing evaluation as potential atheroprotective drugs.86 IL-6 is a key target of antiinflammatory treatment for atherosclerosis. Epidemiological data consistently reflect a correlation between a high circulating level of IL-6 and an increased risk of IHD.87,88 IL-6 binds to its receptor (IL-6R) and therefore genetic variation in IL-6R can modulate the inflammatory effect in IHD. IL-6R SNP (rs7529229) marking a non-synonymous IL-6R variant (rs8192284; D358A) was associated with increased circulating levels of IL-6 and also associated with a decreased odds of CAD.39 Another group examined the rs2228145 variant that affects the same amino acid (D358A) and had a consistent result.38 These findings together strongly support the concept that targeting IL-6 signaling may be a novel method of preventing CAD.

Study Limitations

There are key assumptions for MR studies and they must be strictly met for an appropriate interpretation. First, there should be no pleiotropy of a variant. A genetic variant of interest must have no other function than to affect the level of the biomarker of intereset. If it does, the outcome must be independent from the influence of the variant’s other functions. For example, Dastani et al tested whether adiponectin-decreasing alleles influenced the probability of CAD occurrence in the CARDIoGRAM consortium. Among 145 SNPs related to adiponectin level, they found 5 variants that were associated with an increased risk of CAD. However, the study was unable to take into account the difference between SNPs that directly influence CAD through the adiponectin level and those that influence CAD directly but also influence the adiponectin level. Thus, one can say there certainly is evidence of a shared allelic architecture between adiponectin and CAD but never a causal relationship. Second, linkage disequilibrium with other functional variants must be taken into account. Genomic loci that are in close proximity on a given chromosome are more likely to be inherited together, resulting in linkage disequilibrium. If SNP “A” affecting the expression of biomarker A is in linkage disequilibrium with SNP “B” that affects the disease outcome, one may easily conclude that the SNP “A” is responsible for the disease outcome. However, it may or may not be the case. Therefore, when performing a MR study, it is ideal to use specific SNPs that are positioned in genomic regions where the linkage disequilibrium effect can be ruled out.89

Third, there should be no hidden population strata. If a population under investigation is not homogeneous, any disease that runs at a higher prevalence in a subpopulation may display an association with all SNPs that are predominantly found in that subgroup. For example, in a North American cohort a variant may seem to be associated with type 2 diabetes because of Native American descent, which is known to have a greater incidence of the disease.90 Therefore, it is ideal for a MR study to be performed in the same population, although practically it is very hard. Lastly, the sample size must be considered when there is a negative finding. The effect of a SNP on a biomarker can be small because there are multiple genetic variants associated with the biomarker, together with numerous environmental factors influencing the variability of the biomarker. Therefore, a negative result might simply imply that it is hard to say a small difference in the phenotype of a limited sample size is statistically significant.

Conclusions

The traditional approach to developing a new drug mostly used time-consuming epidemiologic studies or laboratory experiments to create a list of potential drug targets. However, those costly methods were fraught with a high risk of failure because the studies can not provide a direct cause of a disease and fail to capture inherent genetic differences between the subjects of interest. However, a ground-breaking advance in technology has brought a new era in genetic studies, providing a reliable list up of potential therapeutic targets of CAD. Specifically, the NGS platform used in linkage analysis or case-control studies and GWAS have offered promising candidate determinants of CAD. Furthermore, MR study has facilitated the unraveling process, with considerable evidence of causality of biomarkers for CAD in humans. These powerful genetic tools are synergistically providing great opportunities to uncover the pathogenic mechanism of CAD and to identify novel therapeutic targets, not only for the satisfaction of geneticists but also for general medical researchers who are sincerely interested in overcoming CAD.

Funding

This research was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HI12C16910000).

References
 
© 2015 THE JAPANESE CIRCULATION SOCIETY
feedback
Top