Open Access

Identification of nine genes as novel susceptibility loci for early‑onset ischemic stroke, intracerebral hemorrhage, or subarachnoid hemorrhage

  • Authors:
    • Yoshiji Yamada
    • Kimihiko Kato
    • Mitsutoshi Oguri
    • Hideki Horibe
    • Tetsuo Fujimaki
    • Yoshiki Yasukochi
    • Ichiro Takeuchi
    • Jun Sakuma
  • View Affiliations

  • Published online on: May 29, 2018     https://doi.org/10.3892/br.2018.1104
  • Pages: 8-20
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Given that substantial genetic components have been shown in ischemic stroke, intracerebral hemorrhage (ICH), and subarachnoid hemorrhage (SAH), heritability may be higher in early‑onset than late‑onset individuals with these conditions. Although genome‑wide association studies (GWASs) have identified various genes and loci significantly associated with ischemic stroke, ICH, or intracranial aneurysm mainly in European ancestry populations, genetic variants that contribute to susceptibility to these disorders remain to be identified definitively. We performed exome‑wide association studies (EWASs) to identify genetic variants that confer susceptibility to ischemic stroke, ICH, or SAH in early‑onset subjects with these conditions. A total of 6,649 individuals aged ≤65 years were examined. For the EWAS of ischemic or hemorrhagic stroke, 6,224 individuals (450 subjects with ischemic stroke, 5,774 controls) or 6,179 individuals (261 subjects with ICH, 176 subjects with SAH, 5,742 controls), respectively, were examined. EWASs were performed with the use of Illumina Human Exome‑12 v1.2 DNA Analysis BeadChip or Infinium Exome‑24 v1.0 BeadChip. To compensate for multiple comparisons of allele frequencies with ischemic stroke, ICH, or SAH, we applied a false discovery rate (FDR) of <0.05 for statistical significance of association. The association of allele frequencies of 31,245 single nucleotide polymorphisms (SNPs) that passed quality control to ischemic stroke was examined with Fisher's exact test, and 31 SNPs were significantly (FDR <0.05) associated with ischemic stroke. The association of allele frequencies of 31,253 or 30,970 SNPs to ICH or SAH, respectively, was examined with Fisher's exact test, and six or two SNPs were significantly associated with ICH or SAH, respectively. Multivariable logistic regression analysis with adjustment for age, sex, and the prevalence of hypertension and diabetes mellitus revealed that 12 SNPs were significantly [P<0.0004 (0.05/124)] related to ischemic stroke. Similar analysis with adjustment for age, sex, and the prevalence of hypertension revealed that six or two SNPs were significantly [P<0.0016 (0.05/32)] related to ICH or SAH, respectively. After examination of linkage disequilibrium of identified SNPs and results of previous GWASs, we identified HHIPL2, CTNNA3, LOC643770, UTP20, and TRIB3 as susceptibility loci for ischemic stroke, DNTTIP2 and FAM205A as susceptibility loci for ICH, and FAM160A1 and OR52E4 as such loci for SAH. Therefore, to the best of our knowledge, we have newly identified nine genes that confer susceptibility to early‑onset ischemic stroke, ICH, or SAH. Determination of genotypes for the SNPs in these genes may prove informative for assessment of the genetic risk for ischemic stroke, ICH, or SAH in Japanese.

Introduction

Stroke is the leading cause of severe disability and a life-threatening condition (1). In 2015, there were 6.3 million stroke deaths worldwide (11.8% of total deaths), making stroke the second leading cause of death behind ischemic heart disease (2). Of all strokes, 87% are ischemic stroke, 10% are intracerebral hemorrhage (ICH), and 3% are subarachnoid hemorrhage (SAH) in the United States (2). The etiology of common forms of stroke is multifactorial and includes both genetic and environmental factors (2,3). Studies with twins, siblings, and families provided substantial evidence for heritability of stroke (2,4). Given that personalized prevention is important to reduce overall burden of stroke, identification of genetic variants for stroke risk is key both for risk prediction and for potential intervention to avert future cerebrovascular events.

The Trial of ORG 10172 in Acute Stroke Treatment (TOAST) study (5) classified ischemic stroke into five subtypes: i) Large-artery atherosclerosis; ii) cardioembolism; iii) small-vessel occlusion; iv) stroke of other determined etiology; and v) stroke of undetermined etiology. A family history study of 1,000 individuals with ischemic stroke and 800 controls showed that a family history of stroke was a risk factor for both large-vessel atherosclerosis and small-vessel occlusion, especially in cases aged <65 years (6). The heritability of ischemic stroke was estimated to be 40.3% for large-vessel disease, 32.6% for cardioembolic stroke, 16.1% for small-vessel disease, and 37.9% for all ischemic stroke (7). These observations suggest that genetic components play important roles in the pathogenesis of ischemic stroke, in addition to conventional risk factors such as hypertension and diabetes mellitus (2,8,9).

Genome-wide association studies (GWASs) in European ancestry populations identified various genes and loci that confer susceptibility to ischemic stroke (1017). A recent GWAS identified HDAC9 and chromosome 1p13.2 (near TSPAN2) as susceptibility loci for large-vessel disease, PITX2 and ZFHX3 as loci for cardioembolic stroke, and 12q24 (near ALDH2) as a susceptibility locus for small-vessel disease, indicating that ischemic stroke-related loci are subtype specific (18). A more recent multiancestry meta-analysis of GWASs identified 32 loci including 22 new loci that confer susceptibility to stroke (19).

ICH accounts for a large proportion of severe or fatal cases of stroke, with its most important risk factors being hypertension and advanced age (20). Familial aggregation of ICH cases was demonstrated in a prospective study in North Carolina in the United States, showing that 10% of affected individuals had a family history of ICH (21). Genetic factors may influence, not only the development of ICH, but also the prevalence of risk factors for this condition, such as hypertension (22).

ICH has a substantial genetic component with heritability of deep or lobar ICH being estimated at 34 and 73%, respectively (23). Previous genetic association studies suggested several genes and loci are involved in the predisposition to ICH (2427). A meta-analysis of GWASs for ICH in European ancestry populations identified chromosome 12q21.1 (near TRHDE) as a susceptibility locus for lobar ICH and 1q22 (near PMF1-SCL25A44) as a locus for non-lobar ICH (28).

SAH is commonly caused by rupture of an aneurysm in an intracranial artery (29). Given that a family history is an important risk factor for the development of intracranial aneurysm, genetic components may play important roles in the development of SAH (3032). The heritability of SAH was estimated as 41% (33). GWASs have implicated several loci and genes that confer susceptibility to intracranial aneurysm (3438). A meta-analysis of GWASs identified 19 genetic variants associated with intracranial aneurysm (39).

Although several single nucleotide polymorphisms (SNPs) have been found to be significantly associated with ischemic stroke (40,41) or intracranial aneurysm (42) in Japanese subjects, genetic variants that confer susceptibility to ischemic stroke, ICH, or SAH in Japanese remain to be identified definitively.

In a family and twin study of ischemic stroke, heritability was higher in early-onset than late-onset individuals with this condition (6,43,44), suggesting that early-onset ischemic stroke has a strong genetic component. Similar to ischemic stroke, early-onset ICH (45) and SAH (46) were shown to have strong genetic components. Given that a genetic contribution may be greater in early-onset forms than in late-onset forms of ischemic stroke, ICH, and SAH, statistical power of the genetic association study may be increased by focusing on early-onset subjects with diseases (6).

We performed exome-wide association studies (EWASs) with the use of human exome array-based genotyping methods to identify genetic variants that confer susceptibility to ischemic stroke, ICH, or SAH in Japanese individuals. To increase the statistical power of EWASs, we examined early-onset subjects with these conditions.

Materials and methods

Study subjects

In our previous study (47), the median age of subjects with ischemic stroke, ICH, or SAH was 74, 71, or 60 years, respectively. We thus defined patients aged ≤65 years as early-onset cases in the present study. A total of 6,649 individuals aged ≤65 years were examined. For the EWAS of ischemic stroke, 6,224 individuals (450 subjects with ischemic stroke, and 5,774 controls) were examined. For the EWAS of hemorrhagic stroke, 6,179 individuals (261 subjects with ICH, 176 subjects with SAH, and 5,742 controls) were examined. Most control individuals were the same for the studies of ischemic and hemorrhagic stroke. The subjects were recruited from individuals who visited outpatient clinics of or were admitted to participating hospitals in Japan (Gifu Prefectural Tajimi Hospital, Tajimi; Gifu Prefectural General Medical Center, Gifu; Japanese Red Cross Nagoya First Hospital, Nagoya; Northern Mie Medical Center Inabe General Hospital, Inabe; Hirosaki University Hospital and Hirosaki Stroke and Rehabilitation Center, Hirosaki) because of various symptoms or for an annual health checkup between October 2002 and March 2014; or who were community-dwelling individuals recruited to a population-based cohort study in Inabe between March 2010 and September 2014 (48).

The diagnosis of ischemic stroke, ICH, or SAH was based on the occurrence of a new and abrupt focal neurological deficit, with neurological symptoms and signs persisting for >24 h, and it was confirmed by positive findings in computed tomography or magnetic resonance imaging (or both) of the head. The type of stroke was determined according to the Classification of Cerebrovascular Diseases III (49). Given that susceptibility loci for ischemic stroke are subtype-specific (18), we examined subjects with atherothrombotic cerebral infarction (large-vessel disease).

For the study of ischemic stroke, subjects with cardiogenic embolic stroke, lacunar infarction alone, transient ischemic attack, hemorrhagic stroke, cerebrovascular malformations, moyamoya disease, cerebral venous sinus thrombosis, brain tumors, or traumatic cerebrovascular diseases were excluded from enrollment. For the studies of hemorrhagic stroke, individuals with ischemic stroke, lacunar infarction, transient ischemic attack, intracranial hemorrhage resulting from cerebrovascular malformations, moyamoya disease, cerebral venous sinus thrombosis, brain tumors, traumatic cerebrovascular diseases, or subdural hematoma were excluded. The control individuals had no history of ischemic or hemorrhagic stroke; of aortic, coronary, or peripheral artery disease; or of other thrombotic, embolic, or hemorrhagic disorders. Individuals with unruptured intracranial aneurysm were also excluded from controls. The absence of stroke history was evaluated with a detailed questionnaire and was confirmed by the absence of a history of neurological deficits.

EWASs

Venous blood was collected into tubes containing 50 mmol/l ethylenediaminetetraacetic acid (disodium salt), peripheral blood leukocytes were isolated, and genomic DNA was extracted from these cells either with a DNA extraction kit (Genomix, Talent Srl, Trieste, Italy) or SMITEST EX-R&D (Medical & Biological Laboratories, Co., Ltd., Nagoya, Japan). EWASs were performed with the use of a Human Exome-12 v1.2 DNA Analysis BeadChip or Infinium Exome-24 v1.0 BeadChip (Illumina, San Diego, CA, USA), both of which include putative functional exonic variants selected from ~12,000 individual exome and whole-genome sequences. The exonic content of ~244,000 SNPs represents diverse populations including European, African, Chinese, and Hispanic individuals (50). SNPs contained in only one of the exome arrays (~2.6% of all SNPs) were excluded from analysis. We performed quality control (51) as follows: i) Genotyping data with a call rate of <97% were discarded, with the mean call rate for the remaining data being 99.9%. ii) Sex specification was checked for all samples, and those for which sex phenotype in the clinical records was inconsistent with genetic sex were discarded. iii) Duplicated samples and cryptic relatedness were checked by calculation of identity by descent; all pairs of DNA samples showing identity by descent of >0.1875 were inspected, and one sample from each pair was excluded. iv) Heterozygosity of SNPs was calculated for all samples, with those showing extremely low or high heterozygosity (>3 standard deviations from the mean) being discarded. v) SNPs in sex chromosomes or in mitochondrial DNA were excluded from the analysis, as were non-polymorphic SNPs or SNPs with a minor allele frequency of <1.0%. vi) SNPs whose genotype distributions deviated significantly (P<0.01) from Hardy-Weinberg equilibrium in control individuals were discarded. vii) Genotype data were examined for population stratification by principal components analysis (52), and population outliers were excluded from the analysis. Totals of 31,245, 31,253, or 30,970 SNPs that passed quality control for the study of ischemic stroke, ICH, or SAH, respectively, were subjected to analysis.

Statistical analysis

For analysis of characteristics of the study subjects, quantitative data were presented as means ± SD, and were compared between subjects with ischemic stroke, ICH, or SAH and controls with the unpaired Student's t-test. Categorical data were compared between two groups with Pearson's Chi-square test. Allele frequencies were estimated by the gene counting method, and Fisher's exact test was applied to identify departure from Hardy-Weinberg equilibrium. Allele frequencies of SNPs were compared between subjects with ischemic stroke, ICH, or SAH, and corresponding controls with Fisher's exact test. To compensate for multiple comparisons of allele frequencies with ischemic stroke, ICH, or SAH, we applied a false discovery rate (FDR) (53) for statistical significance of association. The significance level was set at a FDR of <0.05 for each EWAS. The inflation factor (λ) was 1.06 for ischemic stroke, 1.10 for ICH, and 1.11 for SAH. Multivariable logistic regression analysis was performed with ischemic stroke as a dependent variable and independent variables including age, sex (0, woman; 1, man), the prevalence of hypertension and diabetes mellitus (0, no history of these conditions; 1, positive history), and genotype of each SNP. A similar analysis was performed with ICH or SAH as a dependent variable and independent variables including age, sex, the prevalence of hypertension, and genotype of each SNP. Genotypes of each SNP were assessed according to dominant [0, AA; 1, AB + BB (A, major allele; B, minor allele)], recessive (0, AA + AB; 1, BB), and additive genetic models, and the P-value, odds ratio, and 95% confidence interval were calculated. Additive models comprised additive 1 (0, AA; 1, AB; 0, BB) and additive 2 (0, AA; 0, AB; 1, BB) scenarios, which were analyzed simultaneously with a single statistical model. The association of genotypes of SNPs to intermediate phenotypes was examined with Pearson's Chi-square test and P-values were shown. Bonferroni's correction was applied to other statistical analyses as indicated. Statistical tests were performed with JMP Genomics version 9.0 software (SAS Institute, Cary, NC, USA).

Results

Characteristics of subjects

The characteristics of the 6,224 subjects enrolled in the ischemic stroke study are shown in Table I. Age, the frequency of men, and the prevalence of hypertension, diabetes mellitus, dyslipidemia, and chronic kidney disease as well as systolic and diastolic blood pressure (BP), fasting plasma glucose (FPG) level, blood glycosylated hemoglobin (hemoglobin A1c) content, and serum concentrations of triglycerides were greater, whereas serum concentration of high density lipoprotein (HDL)-cholesterol and estimated glomerular filtration rate (eGFR) were lower, in subjects with ischemic stroke than in controls.

Table I.

Characteristics of subjects with ischemic stroke and control individuals.

Table I.

Characteristics of subjects with ischemic stroke and control individuals.

CharacteristicControlIschemic strokeP-value
No. of subjects5,774450
Age (years)   50.6±10.256.7±7.1<0.0001
Sex (men/women, %)52.1/47.967.8/32.2<0.0001
Smoking (%)42.535.6   0.0093
Obesity (%)31.033.3   0.3484
Body mass index (kg/m2)23.2±3.523.9±3.8   0.0002
Hypertension (%)31.772.5<0.0001
Systolic BP (mmHg)121±18149±30<0.0001
Diastolic BP (mmHg)   75±13   86±17<0.0001
Diabetes mellitus (%)12.747.5<0.0001
Fasting plasma glucose (mmol/l)   5.66±1.78   7.16±3.00<0.0001
Blood hemoglobin A1c (%)   5.72±0.96   6.52±1.66<0.0001
Dyslipidemia (%)56.966.3   0.0001
Serum triglycerides (mmol/l)   1.32±0.98   1.67±1.03<0.0001
Serum HDL-cholesterol (mmol/l)   1.65±0.45   1.30±0.42<0.0001
Serum LDL-cholesterol (mmol/l)   3.18±0.83   3.13±0.93   0.5012
Chronic kidney disease (%)10.331.2<0.0001
Serum creatinine (µmol/l)   69.8±61.0   88.4±120.2   0.0041
eGFR (ml min−1 1.73 m−2)   78.7±17.1   71.1±23.7<0.0001
Hyperuricemia (%)15.219.1   0.0290
Serum uric acid (µmol/l)321±89337±96   0.0027

[i] Quantitative data are means ± standard deviations and were compared between subjects with ischemic stroke and controls with the unpaired Student's t-test. Categorical data were compared between two groups with Pearson's Chi-square test. Based on Bonferroni's correction, a P-value of <0.0025 (0.05/20) was considered statistically significant. BP, blood pressure; HDL, high density lipoprotein; LDL, low density lipoprotein; eGFR, estimated glomerular filtration rate.

The characteristics of the subjects enrolled in the hemorrhagic stroke study are shown in Table II. Age, the frequency of men, and the prevalence of hypertension, diabetes mellitus, and chronic kidney disease as well as systolic and diastolic BP, FPG level, blood hemoglobin A1c content, and serum concentrations of triglycerides were greater, whereas serum concentrations of HDL-cholesterol and low density lipoprotein (LDL)-cholesterol were lower, in subjects with ICH than in controls. The prevalence of hypertension, diabetes mellitus, and chronic kidney disease as well as systolic and diastolic BP, FPG level, and serum concentrations of triglycerides were greater, whereas the prevalence of dyslipidemia and the serum concentration of HDL-cholesterol were lower, in subjects with SAH than in controls.

Table II.

Characteristics of subjects with ICH or SAH and control individuals.

Table II.

Characteristics of subjects with ICH or SAH and control individuals.

CharacteristicControlICHP-valueSAHP-value
No. of subjects5,742261 176
Age (years)   50.5±10.255.1±7.6<0.000152.2±9.20.0172
Sex (men/women, %)52.1/47.970.9/29.1<0.000142.6/57.40.0134
Smoking (%)42.437.60.191533.00.0434
Obesity (%)30.930.90.995625.40.2094
Body mass index (kg/m2)23.2±3.523.3±3.80.592323.1±3.20.8343
Hypertension (%)31.673.5<0.000159.4<0.0001
Systolic BP (mmHg)121±18150±29<0.0001149±27<0.0001
Diastolic BP (mmHg)   75±13   88±17<0.0001   85±16<0.0001
Diabetes mellitus (%)12.733.1<0.000121.50.0012
Fasting plasma glucose (mmol/l)   5.66±1.83   6.66±2.44<0.0001   6.61±2.440.0002
Blood hemoglobin A1c (%)   5.72±0.97   6.24±1.33<0.0001   5.97±1.280.1712
Dyslipidemia (%)56.754.50.485442.50.0002
Serum triglycerides (mmol/l)   1.31±0.98   1.63±0.91<0.0001   1.89±1.630.0003
Serum HDL-cholesterol (mmol/l)   1.65±0.45   1.27±0.46<0.0001   1.35±0.37<0.0001
Serum LDL-cholesterol (mmol/l)   3.18±0.83   2.90±0.85<0.0001   2.95±0.930.0159
Chronic kidney disease (%)10.318.30.000425.0<0.0001
Serum creatinine (µmol/l)   69.8±61.0   70.7±31.80.7555   68.1±34.50.5044
eGFR (ml min−1 1.73 m−2)   78.7±17.1   79.4±24.10.6912   79.1±27.50.8795
Hyperuricemia (%)15.117.70.261610.30.0817
Serum uric acid (µmol/l)321±89   335±1130.0872   303±1500.2510

[i] Quantitative data are means ± standard deviations and were compared between subjects with ICH or SAH and controls with the unpaired Student's t-test. Categorical data were compared between two groups with Pearson's Chi-square test. Based on Bonferroni's correction, a P-value of <0.0013 (0.05/40) was considered statistically significant. ICH, intracerebral hemorrhage; SAH, subarachnoid hemorrhage; BP, blood pressure; HDL, high density lipoprotein; LDL, low density lipoprotein; eGFR, estimated glomerular filtration rate.

EWAS for ischemic stroke, ICH, or SAH

We examined the association of allele frequencies of 31,245 SNPs that passed quality control to ischemic stroke with the use of Fisher's exact test, and detected that 31 SNPs were significantly (FDR <0.05) associated with ischemic stroke (Table III). The relation of allele frequencies of 31,253 or 30,970 SNPs to ICH or SAH, respectively, was examined with Fisher's exact test. Six or two SNPs were significantly associated with ICH or SAH, respectively (Table IV).

Table III.

The 31 SNPs significantly (FDR <0.05) associated with ischemic stroke in the exome-wide association study.

Table III.

The 31 SNPs significantly (FDR <0.05) associated with ischemic stroke in the exome-wide association study.

GeneSNPNucleotide substitutionaAmino acid substitutionChromosomePositionMAF (%)Allele ORP-value (allele frequency)FDR (allele frequency)
PLCB2rs200787930C/TE1106K15402892981.20.07 3.81×10−9 6.48×10−6
VPS33Brs199921354C/TR80Q15910138411.20.07 5.62×10−9 8.83×10−6
CXCL8rs188378669G/TE31*  4737415681.20.07 5.71×10−9 8.83×10−6
MARCH1rs61734696G/TQ137K  41641973031.20.07 5.89×10−9 8.83×10−6
ADGRL3rs192210727G/TR580I  4619096151.30.07 5.85×10−9 8.83×10−6
TMOD4rs115287176G/AR277W  11511709611.20.08 8.35×10−9 1.22×10−5
COL6A3rs146092501C/TE1386K  22373718611.20.08 1.25×10−8 1.72×10−5
ZNF77rs146879198G/AR340*1929341091.20.08 1.25×10−8 1.72×10−5
NYNRINrs149771079G/AD467N14244091931.23.33 5.03×10−8 6.23×10−5
GOSR2rs1052586T/C 174694109748.70.70 2.01×10−60.0020
rs12662501C/T   6312230737.31.68 1.14×10−50.0104
rs17435433T/C   28821009725.81.39 1.24×10−50.0111
rs7453967T/G   63134646615.31.49 1.85×10−50.0153
HLA-Crs2308557G/AS101N  6312716408.41.62 2.54×10−50.0205
rs3130688T/C   63124243918.61.43 2.77×10−50.0215
HHIPL2rs3748665C/TR394Q  12225402797.40.51 2.88×10−50.0218
MUC22rs11756038A/GT1376A  6310295575.01.73 5.41×10−50.0392
LOC643770rs829881C/A 129848745037.41.33 6.09×10−50.0435
TRIB3rs2295490A/GQ84R2038826123.01.37 6.35×10−50.0449
CDSNrs3130984C/TS143N  63111718713.41.47 6.49×10−50.0449
HLA-DQB1rs1130375C/GA45G  63266504328.30.73 6.49×10−50.0449
CTNNA3rs10997469C/T 106698652726.11.35 7.04×10−50.0477
C6orf15rs2233977T/CV81A  63111211744.00.76 7.44×10−50.0492
rs4713433C/A   63110024944.00.76 7.44×10−50.0492
CDSNrs3130981C/TD527N  63111603613.61.46 7.48×10−50.0492
DDAH1rs12742253T/G   18550529244.90.76 8.45×10−50.0499
HLA-DQB1rs1130370A/CY69D  63266497218.60.69 8.19×10−50.0499
rs3131931A/T   6309774885.61.69 7.72×10−50.0499
CDK18rs77571454G/AG466E  12055313505.11.72 8.4×10−50.0499
CTNNA3rs1925608A/C 106699065432.21.33 8.35×10−50.0499
UTP20rs117417637G/AR1520H121013447041.82.33 8.07×10−50.0499

{ label (or @symbol) needed for fn[@id='tfn3-br-0-0-1104'] } Allele frequencies were analyzed with Fisher's exact test.

a Major allele/minor allele. SNP, single nucleotide polymorphism; FDR, false discovery rate; MAF, minor allele frequency; OR, odds ratio.

Table IV.

The eight SNPs significantly (FDR <0.05) associated with intracerebral hemorrhage or subarachnoid hemorrhage in the exome-wide association study.

Table IV.

The eight SNPs significantly (FDR <0.05) associated with intracerebral hemorrhage or subarachnoid hemorrhage in the exome-wide association study.

GeneSNPNucleotide substitutionaAmino acid substitutionChromosomePositionMAF (%)Allele ORP-value (allele frequency)FDR (allele frequency)
Intracerebral hemorrhage
rs12229654T/G 1211097665722.50.59 7.66×10−60.0093
  FAM205Ars3739881A/CI999S  93472424436.90.66 1.53×10−50.0178
  SVEP1rs7030192G/AA2750V  911040735140.91.47 1.71×10−50.0192
  DNTTIP2rs3747965T/GD309E  19387700842.51.47 2.86×10−50.0316
  ALDH2rs671G/AE504K1211180396227.60.65 4.76×10−50.0483
  ACAD10rs11066015G/A 1211173020527.50.65 4.76×10−50.0483
Subarachnoid hemorrhage
  OR52E4rs11823828T/GF227L11588497336.61.82 2.48×10−60.0063
  FAM160A1rs2709828C/T 415143411633.10.57 5.96×10−60.0145

{ label (or @symbol) needed for fn[@id='tfn5-br-0-0-1104'] } Allele frequencies were analyzed with Fisher's exact test.

a Major allele/minor allele. SNP, single nucleotide polymorphism; FDR, false discovery rate; MAF, minor allele frequency; OR, odds ratio.

Multivariable logistic regression analysis of the association of SNPs to ischemic stroke, ICH, or SAH

The association of the 31 identified SNPs in the EWAS of ischemic stroke was further examined by multivariable logistic regression analysis with adjustment for age, sex, and the prevalence of hypertension and diabetes mellitus (Table V). The 12 SNPs were significantly [P<0.0004 (0.05/124) in at least one genetic model] related to ischemic stroke. The association of the six or two SNPs identified in the EWAS for ICH or SAH, respectively, to these conditions was examined by multivariable logistic regression analysis with adjustment for age, sex, and the prevalence of hypertension (Table VI). The SNPs were significantly [P<0.0016 (0.05/32)] related to ICH or SAH.

Table V.

Association of SNPs to ischemic stroke as determined by multivariable logistic regression analysis.

Table V.

Association of SNPs to ischemic stroke as determined by multivariable logistic regression analysis.

DominantRecessiveAdditive 1Additive 2




GeneSNP P-valueOR95% CIP-valueOR95% CIP-valueOR95% CIP-valueOR95% CI
NYNRINrs149771079G/A<0.00013.212.03–5.07 <0.00013.212.03–5.07
GOSR2rs1052586T/C0.00770.710.55–0.91<0.00010.470.34–0.640.2492 <0.00010.420.29–0.61
HLA-Crs2308557G/A0.00041.611.24–2.090.4809 0.00021.651.27–2.150.5880
rs3130688T/C<0.00011.591.28–1.970.5185 <0.00011.611.28–2.020.2174
HHIPL2rs3748665C/T0.00030.500.34–0.730.4082 0.00050.500.34–0.740.4163
LOC643770rs829881C/A0.00041.511.20–1.90<0.00011.711.31–2.240.01461.361.06–1.73<0.00012.031.50–2.76
TRIB3rs2295490A/G<0.00011.531.24–1.900.00211.881.26–2.810.00151.441.15–1.810.00022.181.44–3.30
CDSNrs3130984C/T<0.00011.621.28–2.040.9814 <0.00011.651.30–2.090.7537
CDSNrs3130981C/T<0.00011.611.28–2.030.9850 <0.00011.641.30–2.080.7588
CDK18rs77571454G/A0.00011.861.36–2.550.2692 <0.00011.901.38–2.610.9969
CTNNA3rs1925608A/C0.00801.341.08–1.670.00181.641.20–2.240.0720 0.00031.831.31–2.56
UTP20rs117417637G/A<0.00012.721.76–4.200.5873 <0.00012.761.79–4.260.9965

[i] Multivariable logistic regression analysis was performed with adjustment for age, sex, and the prevalence of hypertension and diabetes mellitus. Based on Bonferroni's correction; a P-value of <0.0004 (0.05/124) was considered statistically significant. SNP, single nucleotide polymorphism; OR, odds ratio; CI, confidence interval.

Table VI.

Relation of SNPs to intracerebral hemorrhage or subarachnoid hemorrhage as determined by multivariable logistic regression analysis.

Table VI.

Relation of SNPs to intracerebral hemorrhage or subarachnoid hemorrhage as determined by multivariable logistic regression analysis.

DominantRecessiveAdditive 1Additive 2




GeneSNP P-valueOR95% CIP-valueOR95% CIP-valueOR95% CIP-valueOR95% CI
Intracerebral hemorrhage
rs12229654T/G0.00020.580.44–0.770.1592 0.00050.590.44–0.790.0814
  FAM205Ars3739881A/C0.00080.640.50–0.830.00080.420.26–0.700.02140.730.56–0.950.00010.360.22–0.61
  SVEP1rs7030192G/A0.00031.731.28–2.340.00571.551.14–2.110.00271.611.18–2.210.00012.111.45–3.08
  DNTTIP2rs3747965T/G0.00631.521.13–2.050.00021.781.31–2.410.1320 <0.00012.111.45–3.06
  ALDH2rs671G/A0.00040.610.47–0.800.7561 0.00030.580.43–0.780.3117
  ACAD10rs11066015G/A0.00040.610.47–0.810.7534 0.00030.580.44–0.780.3118
Subarachnoid hemorrhage
  OR52E4rs11823828T/G0.03491.481.03–2.14<0.00013.122.14–4.540.9690 <0.00013.112.02–4.78
  FAM160A1rs2709828C/T0.00020.550.41–0.750.00250.310.14–0.660.00470.630.46–0.870.00050.250.12–0.54

[i] Multivariable logistic regression analysis was performed with adjustment for age, sex, and the prevalence of hypertension. Based on Bonferroni's correction, a P-value of <0.0016 (0.05/32) was considered statistically significant. SNP, single nucleotide polymorphism; OR, odds ratio; CI, confidence interval.

Relationship of SNPs associated with ischemic stroke, ICH, or SAH to intermediate phenotypes

We examined the relationship of the 12 SNPs associated with ischemic stroke to intermediate phenotypes of this condition (including hypertension, diabetes mellitus, hypertriglyceridemia, hypo-HDL-cholesterolemia, hyper-LDL-cholesterolemia, chronic kidney disease, obesity, and hyperuricemia) with the use of Pearson's Chi-square test. None of the SNPs was related to intermediate phenotypes (Table VII). The relationship of six or two SNPs associated with ICH or SAH, respectively, to intermediate phenotypes of these conditions was also examined. The rs12229654 at chromosome 12q24.1, rs671 of ALDH2, and rs11066015 of ACAD10 associated with ICH were significantly related to hypertension, hyper-LDL-cholesterolemia, and hyperuricemia, whereas none of SNPs associated with SAH was related to intermediate phenotypes (Table VIII).

Table VII.

Relation of SNPs associated with ischemic stroke to intermediate phenotypes.

Table VII.

Relation of SNPs associated with ischemic stroke to intermediate phenotypes.

GeneSNPHypertensionDMHyper-TGHypo-HDLHyper-LDLCKDObesityHyperuricemia
NYNRINrs149771079G/A0.00540.06860.21170.63400.93850.69420.93540.2993
GOSR2rs1052586T/C0.75870.74830.22890.73870.20920.31070.13090.8628
HLA-Crs2308557G/A0.01880.18810.49740.14090.76320.59040.46660.0329
rs3130688T/C0.39130.47240.07460.43220.01650.53850.44400.7564
HHIPL2rs3748665C/T0.98110.77820.62950.45090.34650.07810.26330.4430
LOC643770rs829881C/A0.48040.18120.28850.52400.77850.12230.12920.5389
TRIB3rs2295490A/G0.64340.69090.50380.11350.16030.07620.28420.2079
CDSNrs3130984C/T0.49460.79360.45410.31640.04510.80390.95440.0920
CDSNrs3130981C/T0.49210.82120.44630.33440.04590.80390.95410.0923
CDK18rs77571454G/A0.51320.79960.74370.71770.92080.56470.26420.3218
CTNNA3rs1925608A/C0.50250.85970.23670.87080.14960.72960.72270.4908
UTP20rs117417637G/A0.20320.91010.58600.19560.03960.00060.19730.4511

[i] Data are P-values. The relationship of genotypes of each SNP to intermediate phenotypes was examined with Pearson's Chi-square test. SNP, single nucleotide polymorphism; DM, diabetes mellitus; hyper-TG, hypertriglyceridemia; hypo-HDL, hypo-HDL-cholesterolemia; hyper-LDL, hyper-LDL-cholesterolemia; CKD, chronic kidney disease. Based on Bonferroni's correction; a P-value of <0.0005 (0.05/96) was considered statistically significant.

Table VIII.

Relationship of SNPs associated with hemorrhagic stroke to intermediate phenotypes.

Table VIII.

Relationship of SNPs associated with hemorrhagic stroke to intermediate phenotypes.

GeneSNPHypertensionDMHyper-TGHypo-HDLHyper-LDLCKDObesityHyperuricemia
Intracerebral hemorrhage
rs12229654T/G <0.00010.04300.15670.47240.00010.52290.0456 <0.0001
  FAM205Ars3739881A/C0.67230.15250.05350.12390.01290.52380.97750.5100
  SVEP1rs7030192G/A0.03170.84340.68590.63140.95870.09070.60290.1534
  DNTTIP2rs3747965T/G0.34060.07960.03160.09210.73140.31820.12420.4615
  ALDH2rs671G/A <0.00010.03730.06770.0286 <0.00010.55590.0086 <0.0001
  ACAD10rs11066015G/A <0.00010.07080.06210.0249 <0.00010.63220.0101 <0.0001
Subarachnoid hemorrhage
  OR52E4rs11823828T/G0.03250.23540.10350.94860.90930.20940.41770.0205
  FAM160A1rs2709828C/T0.51130.94860.81780.31660.16060.14690.98850.7020

[i] Data are P-values. The relation of genotypes of each SNP to intermediate phenotypes was examined with Pearson's Chi-square test. Based on Bonferroni's correction; a P-value of <0.0008 (0.05/64) was considered statistically significant and is shown in bold. SNP, single nucleotide polymorphism; DM, diabetes mellitus; hyper-TG, hypertriglyceridemia; hypo-HDL, hypo-HDL-cholesterolemia; hyper-LDL, hyper-LDL-cholesterolemia; CKD, chronic kidney disease.

Linkage disequilibrium analyses

We examined linkage disequilibrium (LD) among SNPs associated with ischemic stroke or ICH. For the ischemic stroke study, rs3130688 at chromosome 6p21.3 and rs2308557 of HLA-C were in complete LD [square of the correlation coefficient (r2), 1.000], whereas rs3130981 and rs3130984 of CDSN were not in LD. For the ICH study, there was significant LD (r2, 0.650 to 0.995) among rs12229654 at 12q24.1, rs11066015 of ACAD10, and rs671 of ALDH2 (data not shown).

Association of genes, chromosomal loci, and SNPs identified in the present study to phenotypes reported by previously GWASs. In the ischemic stroke study, CDK18 was shown to be related to type 1 diabetes mellitus (T1DM); CDSN to T1DM and serum concentrations of triglycerides; HLA-C to T1DM and serum concentrations of triglycerides and LDL-cholesterol; NYNRIN to serum concentration of LDL-cholesterol; and GOSR2 to systolic BP. The remaining five genes (HHIPL2, CTNNA3, LOC643770, UTP20, TRIB3) were not found to be related to ischemic stroke or other cerebrovascular disease-related phenotypes (Table IX). In the hemorrhagic stroke study, SVEP1 was shown to be related to coronary artery disease (CAD); chromosome 12q24.1 to serum HDL-cholesterol level; ACAD10 to CAD, serum LDL-cholesterol level, T1DM, and diastolic BP; and ALDH2 to CAD, myocardial infarction (MI), serum concentrations of HDL-cholesterol and LDL-cholesterol, T1DM, and systolic and diastolic BP. The remaining four genes (DNTTIP2, FAM205A, FAM160A1, OR52E4) were not related to ICH, SAH, or other cerebrovascular disease-related phenotypes (Table X).

Table IX.

Relationship of genes, chromosomal loci, and SNPs associated with ischemic stroke in the present study to previously reported cerebrovascular disease-related phenotypes.

Table IX.

Relationship of genes, chromosomal loci, and SNPs associated with ischemic stroke in the present study to previously reported cerebrovascular disease-related phenotypes.

Gene/chr. locusSNPChr.PositionPreviously reported phenotypes
CDK18rs775714541205531350Type 1 diabetes (21980299)
HHIPL2rs37486651222540279None
CDSNrs3130981631116036Type 1 diabetes (17554300)
rs3130984631117187triglycerides (20686565)
6p21.3rs3130688631242439None
HLA-Crs2308557631271640Type 1 diabetes (17632545), total cholesterol (20686565), triglycerides (20686565), LDL-cholesterol (20686565)
CTNNA3rs19256081066990654None
LOC643770rs8298811298487450None
UTP20rs11741763712101344704None
NYNRINrs1497710791424409193LDL-cholesterol (20686565), total cholesterol (20686565)
GOSR2rs10525861746941097Systolic blood pressure (21909110, 21909115)
TRIB3rs229549020388261None

[i] Data were obtained from genome-wide repository of associations between SNPs and phenotypes (GRASP) search database (https://grasp.nhlbi.nih.gov/Search.aspx) with a P-value of <1.0×10−6. Numbers in parentheses are PubMed IDs. SNP, single nucleotide polymorphism; Chr., chromosome; LDL, low density lipoprotein.

Table X.

Relationship of genes and SNPs associated with intracerebral hemorrhage or subarachnoid hemorrhage in the present study to previously reported cerebrovascular disease-related phenotypes.

Table X.

Relationship of genes and SNPs associated with intracerebral hemorrhage or subarachnoid hemorrhage in the present study to previously reported cerebrovascular disease-related phenotypes.

Gene/chr. locusSNPChr.PositionPreviously reported phenotypes
Intracerebral hemorrhage
  DNTTIP2rs3747965193877008None
  FAM205Ars3739881934724244None
  SVEP1rs70301929110407351Coronary artery disease (23364394)
  12q24.1rs1222965412110976657HDL-cholesterol (21909109)
  ACAD10rs1106601512111730205Coronary artery disease (23364394, 23202125), LDL-cholesterol (20686565), type 1 diabetes (17554300), diastolic blood pressure (21909115)
  ALDH2rs67112111803962HDL-cholesterol (21572416, 21372407), myocardial infarction (21971053), coronary artery disease (21971053, 21572416, 23202125), diastolic blood pressure (21572416, 21909115), systolic blood pressure (21572416), LDL-cholesterol (21572416, 20686565), type 1 diabetes (17554300)
Subarachnoid hemorrhage
  FAM160A1rs27098284151434116None
  OR52E4rs11823828115884973None

[i] Data were obtained from genome-wide repository of associations between SNPs and phenotypes (GRASP) search database (https://grasp.nhlbi.nih.gov/Search.aspx) with a P-value of <1.0×10−6. Numbers in parentheses are PubMed IDs. SNP, single nucleotide polymorphism; Chr., chromosome; HDL, high density lipoprotein; LDL, low density lipoprotein.

Discussion

Given that stroke is a serious condition and is a global public health problem (1,2,20,29), identification of genetic variants that confer susceptibility to ischemic stroke, ICH, and SAH is clinically important to prevent these conditions. In the present study, we performed EWASs for ischemic stroke, ICH, and SAH in early-onset subjects who may have greater genetic components compared with late-onset individuals.

In the study of ischemic stroke, among 10 genes and one chromosomal locus identified, CDK18, CDSN, HLA-C, NYNRIN and GOSR2 were shown to be related to T1DM (5556), serum concentrations of triglycerides or LDL-cholesterol (57), or systolic BP (58,59) which are risk factors for ischemic stroke. Although rs3130688 at chromosomal 6p21.3 was not related to cerebrovascular phenotype, this region was previously related to CAD (60). We thus identified HHIPL2, CTNNA3, LOC643770, UTP20 and TRIB3 as novel susceptibility loci for ischemic stroke. The five genes associated with ischemic stroke were not related to intermediate phenotypes, although the relation of UTP20 to chronic kidney disease was borderline significance. The underline molecular mechanisms of the association of these genes with ischemic stroke remain unclear.

In the study of hemorrhagic stroke, of the seven genes and one chromosomal locus identified, SVEP1, ACAD10 and ALDH2 were shown to be related to CAD or MI (6062). ACAD10 and ALDH2 were previously related to systolic or diastolic BP (59,63); and these genes as well as chromosome 12q24.1 were previously related to the serum concentrations of HDL-cholesterol or LDL-cholesterol (57,64). These phenotypes are related to cerebrovascular disease. We thus identified DNTTIP2 and FAM205A as new susceptibility loci for ICH, and FAM160A1 and OR52E4 as loci for SAH. Given that the four genes associated with ICH or SAH were not related to intermediate phenotypes, the functional relevance of the association of these genes with ICH or SAH remains to be elucidated.

We previously showed that four, six, or three SNPs were associated with ischemic stroke (P<0.01), ICH (P<0.05), or SAH (P<0.05), respectively, as determined by multivariable logistic regression analysis with adjustment for covariates after the initial EWAS screening among both early- and late-onset subjects with these conditions (47). The relationship of four SNPs to ischemic stroke was not replicated (P<0.05) in the present study. The relation of one of six SNPs [rs138533962 (P=0.0019)] to ICH was replicated in the present study. The association of one of three SNPs [rs117564807 (P=0.0454)] to SAH was replicated in the present study. The results suggest that genetic variants that confer susceptibility to ischemic stroke, ICH, or SAH may differ, in part, between early-onset and late-onset subjects with these conditions.

There are several limitations to our study: i) Given that the results were not replicated, their validation will be necessary in independent study populations or in other ethnic groups. ii) It is possible that SNPs identified in the present study are in LD with other genetic variants in the same gene or in other nearby genes that are actually responsible for the development of ischemic stroke, ICH, or SAH. iii) The functional relevance of identified SNPs to the pathogenesis of ischemic stroke, ICH, or SAH remains to be elucidated.

In conclusion, we have newly identified five (HHIPL2, CTNNA3, LOC643770, UTP20, TRIB3), two (DNTTIP2, FAM205A), or two (FAM160A1, OR52E4) genes as susceptibility loci for early-onset ischemic stroke, ICH, or SAH, respectively. Determination of genotypes for the SNPs in these genes may prove informative for assessment of the genetic risk for ischemic stroke, ICH, or SAH in Japanese subjects.

Acknowledgements

Not applicable.

Funding

This study was supported by CREST (JPMJCR1302), Japan Science and Technology Agency (to YYam, JS and IT).

Availability of data and materials

All data underlying the findings described in the article are available upon request from the corresponding author.

Authors' contributions

YYam contributed to conception and design of the study; to acquisition, analysis, and interpretation of the data; and to drafting of the manuscript. KK, MO, HH and TF each contributed to acquisition of the data and to revision of the manuscript. YYasukochi, IT and JS contributed to analysis and interpretation of the data as well as to revision of the manuscript.

Ethics approval and consent to participate

The study protocol complied with the Declaration of Helsinki and was approved by the Committees on the Ethics of Human Research of Mie University Graduate School of Medicine, Hirosaki University Graduate School of Medicine, and participating hospitals (Gifu Prefectural Tajimi Hospital, Gifu Prefectural General Medical Center, Japanese Red Cross Nagoya First Hospital, Northern Mie Medical Center Inabe General Hospital, and Hirosaki Stroke and Rehabilitation Center). Written informed consent was obtained from all subjects.

Consent for publication

All authors approved submission of the final version of the article for publication.

Competing interests

The authors declare that they have no competing interests.

References

1 

Feigin VL, Forouzanfar MH, Krishnamurthi R, Mensah GA, Connor M, Bennett DA, Moran AE, Sacco RL, Anderson L, Truelsen T, et al: Global Burden of Diseases, Injuries, and Risk Factors Study 2010 (GBD 2010) and the GBD Stroke Experts Group: Global and regional burden of stroke during 1990–2010: Findings from the Global Burden of Disease Study 2010. Lancet. 383:245–254. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Benjamin EJ, Virani SS, Callaway CW, Chang AR, Cheng S, Chiuve SE, Cushman M, Delling FN, Deo R, de Ferranti SD, et al: American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee: Heart disease and stroke statistics-2018 update: A report from the American Heart Association. Circulation. 137:e67–e492. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Tournier-Lasserve E: New players in the genetics of stroke. N Engl J Med. 347:1711–1712. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Bak S, Gaist D, Sindrup SH, Skytthe A and Christensen K: Genetic liability in stroke: A long-term follow-up study of Danish twins. Stroke. 33:769–774. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Adams HP Jr, Bendixen BH, Kappelle LJ, Biller J, Love BB, Gordon DL and Marsh EE III: Classification of subtype of acute ischemic stroke. Definitions for use in a multicenter clinical trial. TOAST. Trial of Org 10172 in Acute Stroke Treatment. Stroke. 24:35–41. 1993. View Article : Google Scholar : PubMed/NCBI

6 

Jerrard-Dunne P, Cloud G, Hassan A and Markus HS: Evaluating the genetic component of ischemic stroke subtypes: A family history study. Stroke. 34:1364–1369. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Bevan S, Traylor M, Adib-Samii P, Malik R, Paul NL, Jackson C, Farrall M, Rothwell PM, Sudlow C, Dichgans M, et al: Genetic heritability of ischemic stroke and the contribution of previously reported candidate gene and genomewide associations. Stroke. 43:3161–3167. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Falcone GJ, Malik R, Dichgans M and Rosand J: Current concepts and clinical applications of stroke genetics. Lancet Neurol. 13:405–418. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Malik R and Dichgans M: Challenges and opportunities in stroke genetics. Cardiovasc Res. 14 Mar. 2018, (Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

10 

Gretarsdottir S, Thorleifsson G, Manolescu A, Styrkarsdottir U, Helgadottir A, Gschwendtner A, Kostulas K, Kuhlenbäumer G, Bevan S, Jonsdottir T, et al: Risk variants for atrial fibrillation on chromosome 4q25 associate with ischemic stroke. Ann Neurol. 64:402–409. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Gudbjartsson DF, Holm H, Gretarsdottir S, Thorleifsson G, Walters GB, Thorgeirsson G, Gulcher J, Mathiesen EB, Njølstad I, Nyrnes A, et al: A sequence variant in ZFHX3 on 16q22 associates with atrial fibrillation and ischemic stroke. Nat Genet. 41:876–878. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Ikram MA, Seshadri S, Bis JC, Fornage M, DeStefano AL, Aulchenko YS, Debette S, Lumley T, Folsom AR, van den Herik EG, et al: Genomewide association studies of stroke. N Engl J Med. 360:1718–1728. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Holliday EG, Maguire JM, Evans TJ, Koblar SA, Jannes J, Sturm JW, Hankey GJ, Baker R, Golledge J, Parsons MW, et al: Australian Stroke Genetics Collaborative; International Stroke Genetics Consortium; Wellcome Trust Case Control Consortium 2: Common variants at 6p21.1 are associated with large artery atherosclerotic stroke. Nat Genet. 44:1147–1151. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Bellenguez C, Bevan S, Gschwendtner A, Spencer CC, Burgess AI, Pirinen M, Jackson CA, Traylor M, Strange A, Su Z, et al: International Stroke Genetics Consortium (ISGC); Wellcome Trust Case Control Consortium 2 (WTCCC2): Genome-wide association study identifies a variant in HDAC9 associated with large vessel ischemic stroke. Nat Genet. 44:328–333. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Traylor M, Farrall M, Holliday EG, Sudlow C, Hopewell JC, Cheng YC, Fornage M, Ikram MA, Malik R, Bevan S, et al: Australian Stroke Genetics Collaborative, Wellcome Trust Case Control Consortium 2 (WTCCC2); International Stroke Genetics Consortium: Genetic risk factors for ischaemic stroke and its subtypes (the METASTROKE collaboration): A meta-analysis of genome-wide association studies. Lancet Neurol. 11:951–962. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Kilarski LL, Achterberg S, Devan WJ, Traylor M, Malik R, Lindgren A, Pare G, Sharma P, Slowik A, Thijs V, et al: GARNET Collaborative Research Group, Wellcome Trust Case Control Consortium 2, Australian Stroke Genetic Collaborative, the METASTROKE Consortium, and the International Stroke Genetics Consortium: Meta-analysis in more than 17,900 cases of ischemic stroke reveals a novel association at 12q24.12. Neurology. 83:678–685. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Chauhan G, Arnold CR, Chu AY, Fornage M, Reyahi A, Bis JC, Havulinna AS, Sargurupremraj M, Smith AV, Adams HHH, et al: Neurology Working Group of the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium, the Stroke Genetics Network (SiGN), and the International Stroke Genetics Consortium (ISGC): Identification of additional risk loci for stroke and small vessel disease: A meta-analysis of genome-wide association studies. Lancet Neurol. 15:695–707. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Pulit SL, McArdle PF, Wong Q, Malik R, Gwinn K, Achterberg S, Algra A, Amouyel P, Anderson CD, Arnett DK, et al: NINDS Stroke Genetics Network (SiGN); International Stroke Genetics Consortium (ISGC): Loci associated with ischaemic stroke and its subtypes (SiGN): A genome-wide association study. Lancet Neurol. 15:174–184. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Malik R, Chauhan G, Traylor M, Sargurupremraj M, Okada Y, Mishra A, Rutten-Jacobs L, Giese AK, van der Laan SW, Gretarsdottir S, et al: AFGen Consortium; Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium; International Genomics of Blood Pressure (iGEN-BP) Consortium; INVENT Consortium; STARNET; BioBank Japan Cooperative Hospital Group; COMPASS Consortium; EPIC-CVD Consortium; EPIC-InterAct Consortium; International Stroke Genetics Consortium (ISGC); METASTROKE Consortium; Neurology Working Group of the CHARGE Consortium; NINDS Stroke Genetics Network (SiGN); UK Young Lacunar DNA Study; MEGASTROKE Consortium; MEGASTROKE Consortium: Multiancestry genome-wide association study of 520,000 subjects identifies 32 loci associated with stroke and stroke subtypes. Nat Genet. 50:524–537. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Qureshi AI, Tuhrim S, Broderick JP, Batjer HH, Hondo H and Hanley DF: Spontaneous intracerebral hemorrhage. N Engl J Med. 344:1450–1460. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Alberts MJ, McCarron MO, Hoffmann KL and Graffagnino C: Familial clustering of intracerebral hemorrhage: A prospective study in North Carolina. Neuroepidemiology. 21:18–21. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Falcone GJ, Biffi A, Devan WJ, Jagiella JM, Schmidt H, Kissela B, Hansen BM, Jimenez-Conde J, Giralt-Steinhauer E, Elosua R, et al: International Stroke Genetics Consortium: Burden of risk alleles for hypertension increases risk of intracerebral hemorrhage. Stroke. 43:2877–2883. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Devan WJ, Falcone GJ, Anderson CD, Jagiella JM, Schmidt H, Hansen BM, Jimenez-Conde J, Giralt-Steinhauer E, Cuadrado-Godia E, Soriano C, et al: International Stroke Genetics Consortium: Heritability estimates identify a substantial genetic contribution to risk and outcome of intracerebral hemorrhage. Stroke. 44:1578–1583. 2013. View Article : Google Scholar : PubMed/NCBI

24 

O'Donnell HC, Rosand J, Knudsen KA, Furie KL, Segal AZ, Chiu RI, Ikeda D and Greenberg SM: Apolipoprotein E genotype and the risk of recurrent lobar intracerebral hemorrhage. N Engl J Med. 342:240–245. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Woo D, Kaushal R, Chakraborty R, Woo J, Haverbusch M, Sekar P, Kissela B, Pancioli A, Jauch E, Kleindorfer D, et al: Association of apolipoprotein E4 and haplotypes of the apolipoprotein E gene with lobar intracerebral hemorrhage. Stroke. 36:1874–1879. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Jeanne M, Labelle-Dumais C, Jorgensen J, Kauffman WB, Mancini GM, Favor J, Valant V, Greenberg SM, Rosand J and Gould DB: COL4A2 mutations impair COL4A1 and COL4A2 secretion and cause hemorrhagic stroke. Am J Hum Genet. 90:91–101. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Rannikmäe K, Davies G, Thomson PA, Bevan S, Devan WJ, Falcone GJ, Traylor M, Anderson CD, Battey TW, Radmanesh F, et al: METASTROKE Consortium; CHARGE WMH Group; ISGC ICH GWAS Study Collaboration; WMH in Ischemic Stroke GWAS Study Collaboration; International Stroke Genetics Consortium: Common variation in COL4A1/COL4A2 is associated with sporadic cerebral small vessel disease. Neurology. 84:918–926. 2015.PubMed/NCBI

28 

Woo D, Falcone GJ, Devan WJ, Brown WM, Biffi A, Howard TD, Anderson CD, Brouwers HB, Valant V, Battey TW, et al: International Stroke Genetics Consortium: Meta-analysis of genome-wide association studies identifies 1q22 as a susceptibility locus for intracerebral hemorrhage. Am J Hum Genet. 94:511–521. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Macdonald RL and Schweizer TA: Spontaneous subarachnoid haemorrhage. Lancet. 389:655–666. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Okamoto K, Horisawa R, Kawamura T, Asai A, Ogino M, Takagi T and Ohno Y: Family history and risk of subarachnoid hemorrhage: A case-control study in Nagoya, Japan. Stroke. 34:422–426. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Bor AS, Rinkel GJ, Adami J, Koffijberg H, Ekbom A, Buskens E, Blomqvist P and Granath F: Risk of subarachnoid haemorrhage according to number of affected relatives: A population based case-control study. Brain. 131:2662–2665. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Tromp G, Weinsheimer S, Ronkainen A and Kuivaniemi H: Molecular basis and genetic predisposition to intracranial aneurysm. Ann Med. 46:597–606. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Korja M, Silventoinen K, McCarron P, Zdravkovic S, Skytthe A, Haapanen A, de Faire U, Pedersen NL, Christensen K, Koskenvuo M, et al: GenomEUtwin Project: Genetic epidemiology of spontaneous subarachnoid hemorrhage: Nordic Twin Study. Stroke. 41:2458–2462. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Bilguvar K, Yasuno K, Niemelä M, Ruigrok YM, von Und Zu Fraunberg M, van Duijn CM, van den Berg LH, Mane S, Mason CE, Choi M, et al: Susceptibility loci for intracranial aneurysm in European and Japanese populations. Nat Genet. 40:1472–1477. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Yasuno K, Bilguvar K, Bijlenga P, Low SK, Krischek B, Auburger G, Simon M, Krex D, Arlier Z, Nayak N, et al: Genome-wide association study of intracranial aneurysm identifies three new risk loci. Nat Genet. 42:420–425. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Foroud T, Koller DL, Lai D, Sauerbeck L, Anderson C, Ko N, Deka R, Mosley TH, Fornage M, Woo D, et al: FIA Study Investigators: Genome-wide association study of intracranial aneurysms confirms role of Anril and SOX17 in disease risk. Stroke. 43:2846–2852. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Foroud T, Lai D, Koller D, Van't Hof F, Kurki MI, Anderson CS, Brown RD Jr, Connolly ES, Eriksson JG, Flaherty M, et al: Familial Intracranial Aneurysm Study Investigators: Genome-wide association study of intracranial aneurysm identifies a new association on chromosome 7. Stroke. 45:3194–3199. 2014. View Article : Google Scholar : PubMed/NCBI

38 

van't Hof FN, Ruigrok YM, Lee CH, Ripke S, Anderson G, de Andrade M, Baas AF, Blankensteijn JD, Böttinger EP, Bown MJ, et al: Aneurysm Consortium; Vascular Research Consortium of New Zealand: Shared genetic risk factors of intracranial, abdominal, and thoracic aneurysms. J Am Heart Assoc. 5:e0026032016. View Article : Google Scholar : PubMed/NCBI

39 

Alg VS, Sofat R, Houlden H and Werring DJ: Genetic risk factors for intracranial aneurysms: A meta-analysis in more than 116,000 individuals. Neurology. 80:2154–2165. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Kubo M, Hata J, Ninomiya T, Matsuda K, Yonemoto K, Nakano T, Matsushita T, Yamazaki K, Ohnishi Y, Saito S, et al: A nonsynonymous SNP in PRKCH (protein kinase C eta) increases the risk of cerebral infarction. Nat Genet. 39:212–217. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Matsushita T, Ashikawa K, Yonemoto K, Hirakawa Y, Hata J, Amitani H, Doi Y, Ninomiya T, Kitazono T, Ibayashi S, et al: Functional SNP of ARHGEF10 confers risk of atherothrombotic stroke. Hum Mol Genet. 19:1137–1146. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Low SK, Takahashi A, Cha PC, Zembutsu H, Kamatani N, Kubo M and Nakamura Y: Genome-wide association study for intracranial aneurysm in the Japanese population identifies three candidate susceptible loci and a functional genetic variant at EDNRA. Hum Mol Genet. 21:2102–2110. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Cheng YC, Cole JW, Kittner SJ and Mitchell BD: Genetics of ischemic stroke in young adults. Circ Cardiovasc Genet. 7:383–392. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Brass LM, Isaacsohn JL, Merikangas KR and Robinette CD: A study of twins and stroke. Stroke. 23:221–223. 1992. View Article : Google Scholar : PubMed/NCBI

45 

Woo D, Sekar P, Chakraborty R, Haverbusch MA, Flaherty ML, Kissela BM, Kleindorfer D, Schneider A, Khoury J, Sauerbeck LR, et al: Genetic epidemiology of intracerebral hemorrhage. J Stroke Cerebrovasc Dis. 14:239–243. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Sundquist J, Li X, Sundquist K and Hemminki K: Risks of subarachnoid hemorrhage in siblings: A nationwide epidemiological study from Sweden. Neuroepidemiology. 29:178–184. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Yamada Y, Sakuma J, Takeuchi I, Yasukochi Y, Kato K, Oguri M, Fujimaki T, Horibe H, Muramatsu M, Sawabe M, et al: Identification of six polymorphisms as novel susceptibility loci for ischemic or hemorrhagic stroke by exome-wide association studies. Int J Mol Med. 39:1477–1491. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Yamada Y, Matsui K, Takeuchi I, Oguri M and Fujimaki T: Association of genetic variants with hypertension in a longitudinal population-based genetic epidemiological study. Int J Mol Med. 35:1189–1198. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Sacco RL, Kasner SE, Broderick JP, Caplan LR, Connors JJ, Culebras A, Elkind MS, George MG, Hamdan AD, Higashida RT, et al: American Heart Association Stroke Council, Council on Cardiovascular Surgery and Anesthesia; Council on Cardiovascular Radiology and Intervention; Council on Cardiovascular and Stroke Nursing; Council on Epidemiology and Prevention; Council on Peripheral Vascular Disease; Council on Nutrition, Physical Activity and Metabolism: An updated definition of stroke for the 21st century: A statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 44:2064–2089. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Grove ML, Yu B, Cochran BJ, Haritunians T, Bis JC, Taylor KD, Hansen M, Borecki IB, Cupples LA, Fornage M, et al: Best practices and joint calling of the HumanExome BeadChip: The CHARGE Consortium. PLoS One. 8:e680952013. View Article : Google Scholar : PubMed/NCBI

51 

Anderson CA, Pettersson FH, Clarke GM, Cardon LR, Morris AP and Zondervan KT: Data quality control in genetic case-control association studies. Nat Protoc. 5:1564–1573. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Price AL, Patterson NJ, Plenge RM, Weinblatt ME, Shadick NA and Reich D: Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet. 38:904–909. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Benjamini Y and Hochberg Y: Controlling the false discovery rate: A practical and powerful approach to multiple testing. J R Stat Soc B. 57:289–300. 1995.

54 

Wellcome Trust Case Control Consortium: Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 447:661–678. 2007. View Article : Google Scholar : PubMed/NCBI

55 

Hakonarson H, Grant SF, Bradfield JP, Marchand L, Kim CE, Glessner JT, Grabs R, Casalunovo T, Taback SP, Frackelton EC, et al: A genome-wide association study identifies KIAA0350 as a type 1 diabetes gene. Nature. 448:591–594. 2007. View Article : Google Scholar : PubMed/NCBI

56 

Bradfield JP, Qu HQ, Wang K, Zhang H, Sleiman PM, Kim CE, Mentch FD, Qiu H, Glessner JT, Thomas KA, et al: A genome-wide meta-analysis of six type 1 diabetes cohorts identifies multiple associated loci. PLoS Genet. 7:e10022932011. View Article : Google Scholar : PubMed/NCBI

57 

Teslovich TM, Musunuru K, Smith AV, Edmondson AC, Stylianou IM, Koseki M, Pirruccello JP, Ripatti S, Chasman DI, Willer CJ, et al: Biological, clinical and population relevance of 95 loci for blood lipids. Nature. 466:707–713. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Wain LV, Verwoert GC, O'Reilly PF, Shi G, Johnson T, Johnson AD, Bochud M, Rice KM, Henneman P, Smith AV, et al: LifeLines Cohort Study; EchoGen consortium; AortaGen Consortium; CHARGE Consortium Heart Failure Working Group; KidneyGen consortium; CKDGen consortium; Cardiogenics consortium; CardioGram: Genome-wide association study identifies six new loci influencing pulse pressure and mean arterial pressure. Nat Genet. 43:1005–1011. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Ehret GB, Munroe PB, Rice KM, Bochud M, Johnson AD, Chasman DI, Smith AV, Tobin MD, Verwoert GC, Hwang SJ, et al: CHARGE-HF consortium: Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature. 478:103–109. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Takeuchi F, Yokota M, Yamamoto K, Nakashima E, Katsuya T, Asano H, Isono M, Nabika T, Sugiyama T, Fujioka A, et al: Genome-wide association study of coronary artery disease in the Japanese. Eur J Hum Genet. 20:333–340. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Lee JY, Lee BS, Shin DJ, Woo Park K, Shin YA, Joong Kim K, Heo L, Young Lee J, Kyoung Kim Y, Jin Kim Y, et al: A genome-wide association study of a coronary artery disease risk variant. J Hum Genet. 58:120–126. 2013. View Article : Google Scholar : PubMed/NCBI

62 

CARDIoGRAMplusC4D Consortium; Deloukas P, Kanoni S, Willenborg C, Farrall M, Assimes TL, Thompson JR, Ingelsson E, Saleheen D, Erdmann J, et al: Large-scale association analysis identifies new risk loci for coronary artery disease. Nat Genet. 45:25–33. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Ehret GB, Munroe PB, Rice KM, Bochud M, Johnson AD, Chasman DI, Smith AV, Tobin MD, Verwoert GC, Hwang SJ, et al: CHARGE-HF consortium: Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature. 478:103–109. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Kato N, Takeuchi F, Tabara Y, Kelly TN, Go MJ, Sim X, Tay WT, Chen CH, Zhang Y, Yamamoto K, et al: Meta-analysis of genome-wide association studies identifies common variants associated with blood pressure variation in east Asians. Nat Genet. 43:531–538. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Kim YJ, Go MJ, Hu C, Hong CB, Kim YK, Lee JY, Hwang JY, Oh JH, Kim DJ, Kim NH, et al: MAGIC consortium: Large-scale genome-wide association studies in East Asians identify new genetic loci influencing metabolic traits. Nat Genet. 43:990–995. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2018
Volume 9 Issue 1

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yamada Y, Kato K, Oguri M, Horibe H, Fujimaki T, Yasukochi Y, Takeuchi I and Sakuma J: Identification of nine genes as novel susceptibility loci for early‑onset ischemic stroke, intracerebral hemorrhage, or subarachnoid hemorrhage. Biomed Rep 9: 8-20, 2018
APA
Yamada, Y., Kato, K., Oguri, M., Horibe, H., Fujimaki, T., Yasukochi, Y. ... Sakuma, J. (2018). Identification of nine genes as novel susceptibility loci for early‑onset ischemic stroke, intracerebral hemorrhage, or subarachnoid hemorrhage. Biomedical Reports, 9, 8-20. https://doi.org/10.3892/br.2018.1104
MLA
Yamada, Y., Kato, K., Oguri, M., Horibe, H., Fujimaki, T., Yasukochi, Y., Takeuchi, I., Sakuma, J."Identification of nine genes as novel susceptibility loci for early‑onset ischemic stroke, intracerebral hemorrhage, or subarachnoid hemorrhage". Biomedical Reports 9.1 (2018): 8-20.
Chicago
Yamada, Y., Kato, K., Oguri, M., Horibe, H., Fujimaki, T., Yasukochi, Y., Takeuchi, I., Sakuma, J."Identification of nine genes as novel susceptibility loci for early‑onset ischemic stroke, intracerebral hemorrhage, or subarachnoid hemorrhage". Biomedical Reports 9, no. 1 (2018): 8-20. https://doi.org/10.3892/br.2018.1104