Skip to main content
Erschienen in: BMC Medicine 1/2023

Open Access 01.12.2023 | Research article

Genome-wide association analyses identified novel susceptibility loci for pulmonary embolism among Han Chinese population

verfasst von: Zhu Zhang, Haobo Li, Haoyi Weng, Geyu Zhou, Hong Chen, Guoru Yang, Ping Zhang, Xiangyan Zhang, Yingqun Ji, Kejing Ying, Bo Liu, Qixia Xu, Yongjun Tang, Guangfa Zhu, Zhihong Liu, Shuyue Xia, Xiaohong Yang, Lixia Dong, Ling Zhu, Mian Zeng, Yadong Yuan, Yuanhua Yang, Nuofu Zhang, Xiaomao Xu, Wenyi Pang, Meng Zhang, Yu Zhang, Kaiyuan Zhen, Dingyi Wang, Jieping Lei, Sinan Wu, Shi Shu, Yunxia Zhang, Shuai Zhang, Qian Gao, Qiang Huang, Chao Deng, Xi Fu, Gang Chen, Wenxin Duan, Jun Wan, Wanmu Xie, Peng Zhang, Shengfeng Wang, Peiran Yang, Xianbo Zuo, Zhenguo Zhai, Chen Wang, on behalf of the China pUlmonary Thromboembolism REgistry Study (CURES) investigators

Erschienen in: BMC Medicine | Ausgabe 1/2023

Abstract

Background

A large proportion of pulmonary embolism (PE) heritability remains unexplained, particularly among the East Asian (EAS) population. Our study aims to expand the genetic architecture of PE and reveal more genetic determinants in Han Chinese.

Methods

We conducted the first genome-wide association study (GWAS) of PE in Han Chinese, then performed the GWAS meta-analysis based on the discovery and replication stages. To validate the effect of the risk allele, qPCR and Western blotting experiments were used to investigate possible changes in gene expression. Mendelian randomization (MR) analysis was employed to implicate pathogenic mechanisms, and a polygenic risk score (PRS) for PE risk prediction was generated.

Results

After meta-analysis of the discovery dataset (622 cases, 8853 controls) and replication dataset (646 cases, 8810 controls), GWAS identified 3 independent loci associated with PE, including the reported loci FGG rs2066865 (p-value = 3.81 × 10−14), ABO rs582094 (p-value = 1.16 × 10−10) and newly reported locus FABP2 rs1799883 (p-value = 7.59 × 10−17). Previously reported 10 variants were successfully replicated in our cohort. Functional experiments confirmed that FABP2-A163G(rs1799883) promoted the transcription and protein expression of FABP2. Meanwhile, MR analysis revealed that high LDL-C and TC levels were associated with an increased risk of PE. Individuals with the top 10% of PRS had over a fivefold increased risk for PE compared to the general population.

Conclusions

We identified FABP2, related to the transport of long-chain fatty acids, contributing to the risk of PE and provided more evidence for the essential role of metabolic pathways in PE development.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12916-023-02844-4.
Zhu Zhang, Haobo Li, Haoyi Weng, Geyu Zhou, and Hong Chen contribute equally as the first authors.
Xianbo Zuo, Zhenguo Zhai, and Chen Wang contributed equally as the corresponding authors.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AA
African American
AUC
Area under the curve
DVT
Deep vein thrombosis
EAS
East Asian
EUR
European
GWAS
Genome-wide association study
LD
Linkage disequilibrium
LDL
Low-density lipoprotein
MAF
Minor allele frequency
MR
Mendelian randomization
PCs
Principal components
PE
Pulmonary embolism
PRS
Polygenic risk score
ROC
Receiver operating characteristic curve
SNP
Single nucleotide polymorphism
VTE
Venous thromboembolism

Background

Pulmonary embolism (PE) is a complex and multifactorial disease, together with deep vein thrombosis (DVT), commonly referred as venous thromboembolism (VTE). Twin studies have estimated the heritability of VTE to be approximately 50%, indicating that genetic factors may play a significant role in the pathogenesis of the disease [1]. Over the past decades, family and population studies have discovered dozens of variants across the genome that contribute to the genetic risk of PE or VTE [2, 3]. The largest meta-analysis of genome-wide association study (GWAS) for VTE has identified 34 independent genetic signals [4]. Most of the reported associated loci regulate the coagulation and anticoagulation functions [4, 5], which are vital disease-causing mechanisms in PE. Additionally, platelet, inflammation and erythrocytes have also been associated with the risk [4]. However, we still have limited knowledge of the genetic architecture of PE, leaving a large proportion of heritability unexplained [6, 7].
The global disease burden of PE has been steadily increasing in the past decade, affecting 100–200 per 100,000 individuals in western countries. Nevertheless, the prevalence of PE in EAS is 1/3–1/5 of that in EUR [8]. Little is known about the genetic and other factors accounting for PE prevalence between East Asians (EAS) and European ancestry (EUR). Studies have suggested that ancestry-specific allele frequencies may explain the differences [9]. Genetic studies with diverse populations are valuable for identifying more genetic risk factors of PE and maximizing the relevance of findings across populations [10]. However, there have been few genetic studies for PE among EAS.
Genetic studies of VTE have been performed in subjects with EUR or African American (AA) ancestry. However, due to varying minor allele frequencies (MAF) across different populations, some of the associated variants identified in one population may not be replicated in another population. For example, rs6025 in F5 (Factor V Leiden, FVL), the well-known leading single nucleotide polymorphism (SNP) in EUR VTE patients, has been rarely reported among EAS, with a MAF reported to be 0.024 in EUR and 0 in EAS [1113]. Similarly, variants in THBD have been reported to be associated with VTE in AA but not in EUR [1416]. Such inconsistency makes it unreliable to generalize the genetic findings from EUR to other populations. Direct application of the PE risk assessment models with genetic factors discovered in the EUR may lead to inaccurate estimates of the actual PE risk among EAS, exacerbating health disparities in diverse populations [17, 18].
Till now, there is no solid evidence of the PE risk assessment base on genetic study in EAS. To accelerate our understanding of the genetic basis for PE in EAS, we performed a large-scale genome-wide association study in the Han Chinese population and developed a population-specific polygenic risk score (PRS) to identify sub-populations at higher risk of PE.

Methods

Study design and participants

DNA samples and phenotype data were collected from a Han descent population (Additional File 1: Fig. S1). All samples were collected from the China Pulmonary Thromboembolism Registry Study (CURES), recruited from 2016 to 2020 [19]. We identified patients with our inclusion criteria, while ancestry-matched controls were obtained in collaboration with WeGene [20].
This study was conducted under the human and ethical research principles of The Ministry of Science and Technology, People’s Republic of China (Regulation of the Administration of Human Genetic Resources, July 1, 2019). All the participants provided informed consent and agreed to participate in this research under a protocol approved by the Ethical Committee of China-Japan Friendship Hospital (Cases) and WeGene (Controls). The study’s ethical approval was obtained from the Ethics Committee in China-Japan Friendship Hospital (2016-SSW-7).

Genotyping and genome-wide quality-control procedures

DNA of cases and controls were extracted from whole blood or saliva samples. All participants were genotyped at WeGene Lab using a customized Illumina WeGene V2 Array by Illumina iScan System, which contained roughly 700,000 markers covering the whole genome. The customized array was originally designed from the Infinium Global Screening Array BeadChip, which specifically included around 560,000 genome-wide backbone markers for optimized genome-wide association studies in the Chinese population and other markers for clinical research and quality control. Genotype imputation was conducted using ChinaMAP (http://​www.​mbiobank.​com/​login/​?​next=​%2Fimputation%2F), an online imputation server for East Asian population genotype imputation [21, 22]. Genotype calling was performed using Illumina GenomeStudio software. Quality control was performed before further analyses. Individuals were excluded based on gender mismatches, disproportionate levels of individual missingness (> 0.05), evidence of relatedness (removing one from each pair within 2nd-degree identified by KING [23], inbreeding coefficient above 0.2 or below − 0.2), and being of non-Han Chinese. The patients of discovery dataset were recruited from 2016 to 2018, while the patients of replication dataset were recruited from 2018 to 2020. The top ten principal components (PCs) were calculated using GCTA [24].

Genome-wide association testing, and meta-analysis

For each phase, all genotyped variants passing quality control on autosomal chromosomes were tested for association with PE through logistic regression adjusting for age, sex, and top ten principal components (PCs) using PLINK [25]. The genome-wide significance threshold was set at p-value < 5 × 10−8. Association summary statistics were combined for variants common to discovery stage and replication stage, and then for variants common to all two phases, in fixed effects models using METAL [26]. Cochran’s Q statistic was used to test for heterogeneity and the I2 statistic was used to quantify variation due to heterogeneity. To visualize the results, a Manhattan plot and a quantile–quantile (Q-Q) plot were generated using the R package “qqman.” A regional association plot for the genomic region within 500 Kb of the top hit was generated using LocusZoom software [27], and a forest plot for the most significant SNP association was generated using revman.

Gene-based testing analysis

Gene-based testing was performed using FUMA [28, 29] software. Input SNPs were mapped to 15,756 protein-coding genes, so we set 3.17 × 10−6 as the Bonferroni-corrected significance threshold (0.05/15756 = 3.173 × 10−6). The python package assocplots was used to produce Manhattan plots and QQ-plots [30].

Cellular experiments

Cells and reagents

The HEK293T cells (ATCC, CRL-3216, LabWecom, China) were maintained in DMEM (11,965,092, Gibco, America) supplemented with 10% FBS (10099141C, Gibco, America) and Penicillin–Streptomycin (15,140,163, Gibco, America) with indicated proportion.

Plasmids and antibodies

The GFP-Tagged full-length target gene (Wild Type, WT) and its mutant type (MT) were constructed into eukaryotic expression PEGFP-N1, respectively. Recombinant PEGFP-N1s encoding the target gene and its MT were constructed by PCR-based amplification of cDNA from the sequence. The WT and MT cDNA of the target gene were subcloned into the eukaryotic expression PEGFP-N1, respectively. The antibody to GFP (AE012, human-specific) was purchased from Abclonal.

Plasmid transfection and immunofluorescence assay

HEK293T cells were cultured and then seeded in 6-well plates and the cell density was 350,000 per well. After overnight incubation, cells were transfected with PEGFP-N1, target gene-WT and target gene-MT, respectively. Plasmids were pre-incubated with Opti-MEM™ (31,985,070, GIbco, America) and jetPRIME® Transfection Reagent (114–15, Polyplus, France) as instructed on the manual. After 72 h, fluorescence microscopy were used to assess the transfection efficiency.

Western blot analysis

Western blotting was performed as described previously [31]. Briefly, cells were lysed with lysis buffer (1% TritonX 100, 20 mM Tris–HCl pH 8.0, 250 mM NaCl, 3 mM EDTA pH 8.0), 3 mM EGTA (pH 8.0) with the pH adjusted to 7.6, and complete protease inhibitor cocktail (CW2200, CWBIO, China) on ice for 30 min. Lysates were eluted by boiling 10 min with 2 Χ sample buffer (100 mM Tris–HCl, pH 6.8, 2% SDS, 10% glycerol, 0.1% bromophenol blue, 1% β-mercaptoethanol) and were separated by 10% SDS/PAGE, followed by examination of expression levels of the indicated proteins. β-Tubulin served as an internal control.

Quantitative PCR analysis

Gene expression was analyzed by three-step q-RT–PCR (qPCR). Total RNA were extracted from HEK293T cells transfected plasmids using TRIzol reagent (T9424, Sigma, Germany). Following the manufacturer’s instructions, RNA were reverse-transcribed in a 20 μl reaction volume (42 °C, 30 min; 95 °C, 5 min) using a QuantiTect Reverse Transcription Kit (KR118, TIANGEN, China). cDNA was then amplified using a SYBR Green I Master mix (FP205, TIANGEN, China) and the ABI 7500 Fast Real Time PCR system (ABI, America). All tests were carried out on duplicate reaction mixtures in 96-well plates. The relative expression of the gene of interest was determined using the 2–ΔΔCt method, with GAPDH as the internal control.

Mendelian randomization(MR) analysis

In order to detect the cause relationship between PE and lipoprotein associated triat, we performed two-sample Mendelian randomization (MR) analysis. Summary statistics for exposure were obtained from Biobank Japan Project (http://​jenger.​riken.​jp/​en/​), which contained individuals of similar EAS ancestry as Chinese population to avoid population stratification. We estimated the causal effect of exposure on outcome using two‐sample MR method. The inverse variance weighted (IVW) method [32] was used in the main MR analyses, and the maximum likelihood weighted median and penalized weighted median [33] approaches were employed as sensitivity analysis. MR-Egger [34] method was used to detect the directional pleiotropy according to the intercept of weighted linear regression of the SNP‐outcome coefficients on SNP‐exposure coefficients. Results were considered statistically significant at p-value < 0.017 (0.05/3). The MR analysis was performed using the TwoSampleMR [35] package and MR-PRESSO [33] package in R software (version 3.4).

Heritability and LD-score regression

LDSC v.1.0.1 was used to calculate the heritability on the liability scale [36].

Polygenic risk score analysis

We derived the PRS from PE associated variants in the Han Chinese population. The PRS training cohort including cases and controls of discovery stage, and the PRS testing cohort including cases and controls of replication stage. PRSs of the test cohort were calculated using the Polygenic Risk Score software (PRSice-2) [37], based on the summary statistics of the training group. The performance of a series of cutoff of PE association p-values for selection of SNP markers was assessed by the Area Under the Curve (AUC) for ROC. The p-value cutoff with the largest AUC was adopted. According to the PRS, individuals were divided into seven intervals from low to high (< 10th, 10–20th, 20–30th, 30–70th, 70–80th, 80–90th, < 90th), and the odds ratio of each interval relative to the baseline data (30–70th) is calculated. Finally, we used PLINK to calculate the performance of other PRS available in the literature in the testing cohort and compared them with our PRS.

Results

GWAS and replication analysis

In the discovery stage, we consecutively recruited 624 cases (Fig. 1). 622 PE cases passed QC and were analyzed, of which the mean (+ / − SD) age was 62 (+ / − 19) years old, and 323 (52%) were male. A total number of 8853 controls were collected. Although there are statistically significant differences (p-value < 0.05) in age and sex between cases and controls, the association results are unlikely to be biased as the susceptible variants are generally independent regardless of age and sex.
In replication stage, 647 acute PE cases and 8820 controls were included as additional independent samples. Six hundred forty-six PE cases and 8810 controls passed QC and were analyzed. The mean (+ / − SD) age was 62 (+ / − 15) years old, and 341 (53%) were male in the case group. The characteristics of the participants were presented in Table 1. Only FABP2 rs1799883 and FGG rs2066865 reached genome-wide significance in the discovery stage. We then performed the GWAS meta-analysis based on the discovery and replication stage. Finally, we identified 16 genome-wide significant variants in the meta-analysis, where FGG rs2066865 (p-value = 3.81 × 10−14) and ABO rs582094 (p-value = 1.16 × 10−10) were known loci in the coagulation pathway, and FABP2 rs1799883 (p-value = 7.59 × 10−17) was newly reported (Figs. 2 and 3). The three variants were independent of each other and the leading SNPs are in strong linkage disequilibrium with considerable imputed variants of similar statistical associations (Table 2, Additional File 1: Fig. S1), the remaining 13 loci were neither genome-wide significant in the discovery stage nor previously known to be associated with VTE (Additional File 2: Table S1, Additional File 2: Table S2). PCA (Additional File 1: Fig. S2) and QQ plot (Fig. 2) showed no population stratification and inflation of test statistics (λGC = 1.053).
Table 1
Demographic, clinical characteristics, and risk factors of the patients included
Characteristics
Discovery stage
Replication stage
 
Case
Control
Case
Control
 
N = 622
N = 8853
N = 646
N = 8810
Demographic characteristics
 Age, year, mean (SD)
62 (19)
59 (9)
62 (15)
59 (8)
 Male, n (%)
323 (52)
4041 (46)
341 (53)
4041 (46)
 BMI, kg/m2, mean (SD)
24.4 (3.9)
-
24.5 (3.6)
-
 PE alone, n (%)
331 (53)
-
340 (53)
-
 PE combined DVT, n (%)
291 (47)
-
306 (47)
-
Complications and risk factors
 
-
 
-
 Cardiovascular disease, n (%)
311 (50)
-
311 (48)
-
 Chronic pulmonary disease, n (%)
105 (17)
-
118 (18)
-
 Cancer, n (%)
60 (10)
-
62 (10)
-
 Metabolic and endocrine diseases, n (%)
108 (17)
-
101 (16)
-
 Smoking history, n (%)
198 (32)
-
182 (28)
-
 
N = 218
 
N = 220
 
Thrombophilia test
 
-
 
-
 Plasma antithrombin activity < 70%, n/N (%)
32 (15)
-
32 (15)
-
 Plasma protein S activity < 60%, n/N (%)
36 (17)
-
42 (19)
-
 Plasma protein C activity < 70%, n/N, n/N (%)
42 (19)
-
49 (22)
-
BMI Body mass index, PE Pulmonary embolism, SD Standard deviation
Table 2
Independent genome-wide significant lead SNPs verified to be associated with PE
Chromosome position
rsID
EA/OA
Nearest gene
Discovery stage
Replication stage
Meta
    
EAF (case/control)
OR (95% CI)
P-value
EAF (case/control)
OR (95% CI)
P-value
OR (95% CI)
P-value
I2
4:120,241,902
rs1799883
T/C
FABP2
0.31 (0.39/0.31)
1.41 (1.25–1.59)
1.05 × 10−8
0.31 (0.39/0.30)
1.43 (1.28–1.61)
1.25 × 10−9
1.42 (1.31–1.54)
7.59 × 10−17
0
4:155,525,276
rs2066865
A/G
FGG
0.51 (0.57/0.52)
1.41 (1.25–1.58)
9.85 × 10−9
0.52 (0.55/0.52)
1.34 (1.20–1.50)
6.19 × 10−7
1.37 (1.26–1.49)
3.81 × 10−14
0
9:136,145,484
rs582094
T/A
ABO
0.43 (0.50/0.43)
1.29 (1.15–1.45)
2.25 × 10−5
0.44 (0.49/0.43)
1.33 (1.19–1.49)
1.18 × 10−6
1.31 (1.21–1.42)
1.16 × 10−10
0
Chromosome position: based on hg19/GRCh37 build. rsID rs number, EA Effect allele, OA Other allele, EAF Effect allele frequency, OR Odds ratio, CI confidence interval

Replication of associations peviously identified loci in EAS population

We compiled VTE susceptibility SNPs from the pevious GWAS literature and assessed the associations of 22 previously reported VTE associated variants in our Han Chinese samples [3, 16, 3846]. For 10 out of 22 variants, the association signals were successfully replicated at the Bonferroni-corrected significance level of 2.27 × 10−3 (0.05/22). The most significant SNP was rs505922 near ABO, with p-value of 3.81 × 10−14 and odds ratio (OR, 95% confidence interval [CI]) of 1.31 (1.20–1.42). Nine additional SNPs, at five gene, also reached the threshold for significance. These were rs657152, rs630014 and rs687289 near ABO, rs6825454 and rs2070011 near FGA, rs2066865 and rs6536024 near FGG, rs4253399 near F11, and rs13084580 near CSRNP1. Other previously reported candidate genes (F2, F5, PROS1, PROCR, NME7, SLC44A2, THBD, SMAP1/B3GAT2, PEPD, GP6) for recurrent VTE showed no or very weak association in our cohort (Additional File 2: Table S3).

Gene-based analysis

We further performed a gene-based analysis using functional mapping and annotation (FUMA) MAGMA to prioritize candidate genes associated with PE. After aggregating the association signal of all SNPs in each gene, we identified two genes (FABP2 and FGG) reaching the Bonferroni-corrected threshold (p-value < 3.17 × 10−6), and FGG was previously known to be associated with PE (p-value = 1.45 × 10−6) (Additional File 1: Fig. S3, Additional File 2: Table S4). However, we noted that ABO is annotated as a “processed transcript” in the database and, therefore, it was not incorporated into this analysis.

FABP2-A163G promotes gene transcription and protein expression of FABP2

To explore the influence of the variant on FABP2 expression, PEGFP-N1, FABP2-WT, FABP2-A163G plasmids were transfected into HEK293T cells, and the qPCR results suggested that FABP2-A163G promoted FABP2 transcription compared to FABP2-WT (Fig. 4A, Additional File 1: Fig. S4).
To further examine the effect on FABP2 protein expression, the whole cell lysate from HEK293T cells transfected with EGFP-N1, FABP2-WT, and FABP2-A163G plasmids were harvested. Western blotting results suggested that FABP2-A163G markedly promoted FABP2 protein expression compared to FABP2-WT (Fig. 4B, C).

Mendelian randomization (MR) of metabolism phenotypes and PE

Our GWAS analysis has implicated a strong relationship between FABP2 polymorphism and PE. FABP2 polymorphism is known to be significantly associated with serum total cholesterol and LDL-C [47], which is also proven by our data (Additional File 1: Fig. S5). Previous studies [4, 38] have implied that lipid metabolic traits may be involved in PE pathogenesis. To further investigate the potential causal relationship, we conducted MR analysis of PE and 3 metabolic related phenotypes: total cholesterol (TC), triglycerides (TG), and low-density lipoprotein-cholesterol (LDL-C). The results indicated that high TC, and LDL-C levels were associated with increased risk of PE while TG was not causally related with the risk of PE (TC: OR = 1.42; 95% CI = 1.24–1.61; p-value < 0. 001; LDL-C: OR = 1.21; 95% CI = 1.13–1.29; p-value < 0. 001; TG: p-value = 0.357). Results from maximal likelihood and MR-PRESSO analysis were consistent with IVW (Additional File 1: Fig. S6).

Heritability and LD-score regression

Using LDSC and common variants outside of the HLA region, we found that the estimated heritability of PE was comparable between EAS and EUR (0.16 ± 0.03). The LD score regression generated an intercept of 1.01 ± 0.01 with a p-value = 0.152.

PRS analysis

We generated a 288-variant (Additional File 2: Table S5) PRS under a penalized regression framework using discoverage stage as the training set and replication stage as the testing set. The receiver operating characteristic curve (ROC) in the testing set achieved an area under the curve (AUC) of 0.765 (Additional File 1: Fig. S7). The distributions of standardized PRS for cases and controls in the testing data were illustrated in Fig. 5A. Individuals in the top 10% group of PRS had a 5.08-fold of PE risk relative to the general population (30th–70th quantile) (Fig. 5B, Additional File 2: Table S6). Our 288-variant PRS had a better performance than three publicly available genome-wide PRSs in the testing set (Additional File 1: Fig. S8).

Discussion

As most large-scale genetic studies of VTE have been conducted in EUR ancestry [4, 45, 48, 49], we performed the first GWAS in the Han Chinese population to expand the genetic landscape of PE. We identified three genome-wide significant loci, of which two were known to be associated with PE (FGG, ABO). Another locus at FABP2 reached the significance threshold both in the discovery stage and meta-analysis. The risk allele at the FABP2 locus (rs1799883) is reported as a functional variant to increase the gene expression by qPCR and Western-blot expreriments. The results showed that the carriers of that mutation have higher blood TC and LDL-C [50]. We further performed MR analysis and found that increased levels of LDC-C and TC were associated with a higher risk of PE, which implied the inhibition of LDL-C and TC to be a potential measure of PE prevention.
Some significant loci for the risk of VTE have been reported in European ancestry in the past but have not been validated in the Asian population, we attempted to replicate the previously known loci identified among the EUR population in the current study. In our study, 10 of the 22 leading variants were replicated, located on FGG, F11, ABO, CSRNP1, and FGA. The lack of replication in the current study might be partially attributed to insufficient power and diverse LD patterns across ancestries rather than different biological effects. These findings further emphasize the importance of including diverse ancestral groups in genomic studies to maximize the power for detecting disease associations.
Fatty acid binding proteins (FABPs) are key proteins in lipid transport. FABP2 can traffic lipids from the intestinal lumen to enterocytes and bind superfluous fatty acids to maintain a steady pool of fatty acids in the epithelium. FABP2 polymorphism is known to be significantly associated with serum total cholesterol and LDL-C [47]. Based on the above evidence, we speculate that variants in FABP2 may contribute to PE through lipid metabolism function. Our findings were consistent with previous studies on the role of metabolic traits in PE [51], which is the first time to be verified among the EAS population. Lipid-lowering drugs for prevention or even adjunctive therapy of PE have been proposed in many clinical trials [52]. For example, statins contribute to PE prevention through anti-inflammatory and LDL-lowering effects [53]. Proprotein convertase subtilisin/kexin type (PCSK9) inhibitor has also been identified to lower the risk of VTE by LDL reduction [54]. Our study provided a shred of robust evidence that lipid-lowering therapy may also be considered to prevent PE occurrence in the EAS population.
In addition to FABP2, FGG, and ABO, there were also 13 loci reaching genome-wide significance in the meta-analysis. However, these loci did not achieve genome-wide significance in the discovery stage, nor had they been previously identified as PE-associated variants. There was currently insufficient evidence to support the reliability of those association results so we put emphasis on the three loci (FABP2, FGG, and ABO). More East Asian cohorts are needed to verify the associated loci in the future.
PRS has been widely used in the prediction of common diseases, and the PRS model for VTE had been validated in European ancestry. The early genetic risk model of VTE mainly focused on two loci, rs6025 and rs1799963.The Caprini risk assessment model, primarily relying on these two loci, is extensively utilized to predict VTE risk [17]. However, these two variants are almost absent in the EAS population. With the entry into the GWAS era, more loci were used for risk stratification, Crous-Bou et al. established a new risk model based on the 16 SNPs and found that the risk of VTE in patients with high PRS score was 2.02 times that of patients with low PRS score, and achieved better results [55]. Klarin generated a 297 variant polygenic risk score to predict VTE events among patients [45]. Previous research has indicated a reduced accuracy of PRS models when transferred across ancestries [18]. We, therefore, constructed a 288-variant PRS obtained from the EAS population for PE risk prediction. The PRS incorporated population-specific variants and outperformed in EAS population with an AUC of 0.765. Individuals in the top 10% group of PRS had a 5.08-fold of PE risk relative to the general population (30th–70th quantile). However, the model needs further validation in independent datasets with larger sample sizes.
As GWAS have uncovered hundreds of common genetic variants involved in PE susceptibility, our study shed new light on the genetic architecture of PE among Han Chinese population. Nevertheless, like most complex diseases, the common variants discovered in GWAS only explain a fraction of total disease heritability. The rare variants across the whole genome could also play an important role in disease development[56]. Therefore, large-scale sequencing studies of PE in the EAS population are needed to measure the relationship between rare variants and PE risk.
There are several limitations in the current study. Since associations do not imply causation, further research is required to clarify the functional consequences of these novel signals in PE development. We acknowledge the imbalance of sample size and age and sex differences between cases and controls. However, we employed PLINK firth logistic regression with age and sex as covariates in GWAS analysis to control for type-I error issues. Considering the potential risk of inducing biased and spurious associations, we opted not to perform whole genome imputation. Instead, we restricted our imputation to the genomic regions within + / − 500kbp of the FABP2, FGG, and ABO loci, which constituted the main findings of our study. While we acknowledge that this approach may have led to the neglection of genetic signals, we held that these loci exhibited sound reliability. We look forward to expanding our analysis by incorporating more extensive genetic data in future studies.
Nevertheless, our study represents the first multicenter PE genetic study in diverse areas across China, which is a good representative of the Han Chinese population. We revealed through extensive genetic analyses that FABP2 polymorphism is associated with PE risk and the lipid-metabolic pathways are crucial in the PE development. Although more studies are needed to confirm the value of FABP2, the inhibition of FABP2 is promising to benefit from early intervention in reducing the risk of thrombosis. Our study also demonstrated the utility of applying a population-specific PRS model for PE risk prediction. The clinical use of PRS has the potential to recognize high-risk patients and improve health outcomes through eventual routine implementation as clinical biomarkers.

Conclusions

In conclusion, this is the first large-scale genetic PE study in EAS. We identified novel risk loci of FABP2 to expand the global genetic architecture of PE. MR analysis highlighted the importance of lipid-metabolic pathways in PE development. Pharmacological agents modulating blood lipids could be considered in the future for Chinese people to prevent PE. Moreover, we established a population-specific PRS model with improved performance in the EAS population compared to models trained from EUR data. Our study emphasizes the value of investigating diverse ancestral populations in genomic studies, especially for those ethnic groups that are less studied in the global population.

Acknowledgements

We appreciate the continuous support and contributions from Hongyu Zhao (Yale University), Xiaohui Wang (The First Affiliated Hospital of Chongqing Medical University), Guohua Yu (Weifang No.2 People's Hospital), Ruhong Xu (Dongguan People’s Hospital), Weijia Liu (Guizhou Provincial People’s Hospital), Zhonghe Zhang, Jun An (The First Affiliated Hospital of Dalian Medical University), Guofeng Ma, Chao Yan (Sir Run Run Shaw Hospital, Zhejiang University School of Medicine), Lijun Suo (Linzi District People’s Hospital), Xiaoqing Li, Yingying Pang, Beilei Gong (The First Affiliated Hospital of Bengbu Medical College), Wei Yang (Xiangya Hospital Central South University), Wenmei Zhang (Beijing Anzhen Hospital, Capital Medical University), Qin Luo (Fuwai Hospital, Chinese Academy of Medical Science; National Center for Cardiovascular Diseases), Hui Jia, Yunxia Liu (Central Hospital Affiliated to Shenyang Medical College), Ying Chen (People’s Hospital of Xinjiang Uygur Autonomous Region), Wei Zhou (Tianjin Medical University General Hospital), Ling Zhu, Yi Liu (Shandong Provincial Hospital), Xia Li (The First Affiliated Hospital, Sun Yat-Sen University), Xiaowei Gong (The Second Hospital of Hebei Medical University), Jifeng Li (Beijing Chao-Yang Hospital, Capital Medical University), Linli Duan, Simin Qing, Chunli Liu (The First Affiliated Hospital of Guangzhou Medical University), Baomin Fang, He Yang (Beijing Hospital), Chaosheng Deng, Minxia Yang, Dawen Wu (The First Affiliated Hospital of Fujian Medical University), Songping Huang, Qinghua Xu (Quanzhou First Hospital), Faguang Jin, Ning Wang, Yanli Chen, Yanyan Li (Tangdu Hospital), Jingping Yang, Xiyuan Xu, Baoying Bu (The Third Affiliated Hospital of Inner Mongolia Medical University), Chunxiao Yu, Zhenfang Lu, Jing Hua (Beijing Jingmei Group General Hospital), Chaobo Cui, Ning Wang (Harrison International Peace Hospital), Zhenyang Xu, Hongxia Zhang, Jinxiang Wang (Beijing Luhe Hospital, Capital Medical University), Shudong Zhang, Lijun Kang (Yantaishan Hospital), Lu Guo, Jing Zhang (Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital), Gang Chen, Yunxia Zhao, Zixiao Liu (The Third Hospital of Hebei Medical University), Jinming Liu, Qinhua Zhao (Shanghai Pulmonary Hospital), Xiaoyun Hu, Fangfang Fan (The First Hospital of Shanxi Medical University). Also, we would like to thank the research participants of WeGene and the members of WeGene Research Team for making this work possible. We would like to thank the patient participants of CURES and centers.

Declarations

This study has been approved by the Ethics Committee in China-Japan Friendship Hospital (2016-SSW-7). All patients signed a written informed consent to participate in the study.
N/A.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Gerhardt A, Scharf RE, Greer IA, Zotz RB. Hereditary risk factors for thrombophilia and probability of venous thromboembolism during pregnancy and the puerperium. Blood. 2016;128(19):2343–9.PubMedPubMedCentralCrossRef Gerhardt A, Scharf RE, Greer IA, Zotz RB. Hereditary risk factors for thrombophilia and probability of venous thromboembolism during pregnancy and the puerperium. Blood. 2016;128(19):2343–9.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Hotoleanu C. Genetic Risk Factors in Venous Thromboembolism. Adv Exp Med Biol. 2017;906:253–72.PubMedCrossRef Hotoleanu C. Genetic Risk Factors in Venous Thromboembolism. Adv Exp Med Biol. 2017;906:253–72.PubMedCrossRef
3.
Zurück zum Zitat Germain M, Chasman DI, de Haan H, Tang W, Lindstrom S, Weng LC, de Andrade M, de Visser MC, Wiggins KL, Suchon P, et al. Meta-analysis of 65,734 individuals identifies TSPAN15 and SLC44A2 as two susceptibility loci for venous thromboembolism. Am J Hum Genet. 2015;96(4):532–42.PubMedPubMedCentralCrossRef Germain M, Chasman DI, de Haan H, Tang W, Lindstrom S, Weng LC, de Andrade M, de Visser MC, Wiggins KL, Suchon P, et al. Meta-analysis of 65,734 individuals identifies TSPAN15 and SLC44A2 as two susceptibility loci for venous thromboembolism. Am J Hum Genet. 2015;96(4):532–42.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Lindstrom S, Wang L, Smith EN, Gordon W, van Hylckama VA, de Andrade M, Brody JA, Pattee JW, Haessler J, Brumpton BM, et al. Genomic and transcriptomic association studies identify 16 novel susceptibility loci for venous thromboembolism. Blood. 2019;134(19):1645–57.PubMedPubMedCentralCrossRef Lindstrom S, Wang L, Smith EN, Gordon W, van Hylckama VA, de Andrade M, Brody JA, Pattee JW, Haessler J, Brumpton BM, et al. Genomic and transcriptomic association studies identify 16 novel susceptibility loci for venous thromboembolism. Blood. 2019;134(19):1645–57.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Tang W, Stimson MR, Basu S, Heckbert SR, Cushman M, Pankow JS, Folsom AR, Pankratz N. Burden of rare exome sequence variants in PROC gene is associated with venous thromboembolism: a population-based study. J Thromb Haemost. 2020;18(2):445–53.PubMedCrossRef Tang W, Stimson MR, Basu S, Heckbert SR, Cushman M, Pankow JS, Folsom AR, Pankratz N. Burden of rare exome sequence variants in PROC gene is associated with venous thromboembolism: a population-based study. J Thromb Haemost. 2020;18(2):445–53.PubMedCrossRef
7.
Zurück zum Zitat Zöller B, Li X, Ohlsson H, Ji J, Sundquist J, Sundquist K. Family history of venous thromboembolism as a risk factor and genetic research tool. Thromb Haemost. 2015;114(5):890–900.PubMed Zöller B, Li X, Ohlsson H, Ji J, Sundquist J, Sundquist K. Family history of venous thromboembolism as a risk factor and genetic research tool. Thromb Haemost. 2015;114(5):890–900.PubMed
8.
Zurück zum Zitat Zhang Z, Lei J, Shao X, Dong F, Wang J, Wang D, Wu S, Xie W, Wan J, Chen H, et al. Trends in Hospitalization and In-Hospital Mortality From VTE, 2007 to 2016. China Chest. 2019;155(2):342–53.PubMedCrossRef Zhang Z, Lei J, Shao X, Dong F, Wang J, Wang D, Wu S, Xie W, Wan J, Chen H, et al. Trends in Hospitalization and In-Hospital Mortality From VTE, 2007 to 2016. China Chest. 2019;155(2):342–53.PubMedCrossRef
9.
Zurück zum Zitat Park L. Population-specific long-range linkage disequilibrium in the human genome and its influence on identifying common disease variants. Sci Rep. 2019;9(1):11380.PubMedPubMedCentralCrossRef Park L. Population-specific long-range linkage disequilibrium in the human genome and its influence on identifying common disease variants. Sci Rep. 2019;9(1):11380.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Lam M, Chen CY, Li Z, Martin AR, Bryois J, Ma X, Gaspar H, Ikeda M, Benyamin B, Brown BC, et al. Comparative genetic architectures of schizophrenia in East Asian and European populations. Nat Genet. 2019;51(12):1670–8.PubMedPubMedCentralCrossRef Lam M, Chen CY, Li Z, Martin AR, Bryois J, Ma X, Gaspar H, Ikeda M, Benyamin B, Brown BC, et al. Comparative genetic architectures of schizophrenia in East Asian and European populations. Nat Genet. 2019;51(12):1670–8.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Ribeiro DD, Lijfering WM, Rosendaal FR, Cannegieter SC. Risk of venous thrombosis in persons with increased body mass index and interactions with other genetic and acquired risk factors. J Thromb Haemost. 2016;14(8):1572–8.PubMedCrossRef Ribeiro DD, Lijfering WM, Rosendaal FR, Cannegieter SC. Risk of venous thrombosis in persons with increased body mass index and interactions with other genetic and acquired risk factors. J Thromb Haemost. 2016;14(8):1572–8.PubMedCrossRef
12.
Zurück zum Zitat Choe HJ, Suh KJ, Lee JY, Kim M, Kim MJ, Park SS, Kim JW, Kim SH, Kim JW, Lee JO, et al. Acute pulmonary thromboembolism caused by factor V Leiden mutation in South Korea: A case report. Medicine (Baltimore). 2019;98(28):e16318.PubMedCrossRef Choe HJ, Suh KJ, Lee JY, Kim M, Kim MJ, Park SS, Kim JW, Kim SH, Kim JW, Lee JO, et al. Acute pulmonary thromboembolism caused by factor V Leiden mutation in South Korea: A case report. Medicine (Baltimore). 2019;98(28):e16318.PubMedCrossRef
13.
Zurück zum Zitat Huang SS, Liu Y, Jing ZC, Wang XJ, Mao YM. Common genetic risk factors of venous thromboembolism in Western and Asian populations. Genet Mol Res. 2016;15(1):15017644.PubMedCrossRef Huang SS, Liu Y, Jing ZC, Wang XJ, Mao YM. Common genetic risk factors of venous thromboembolism in Western and Asian populations. Genet Mol Res. 2016;15(1):15017644.PubMedCrossRef
14.
Zurück zum Zitat Ahmad A, Sundquist K, Zoller B, Svensson PJ, Sundquist J, Memon AA. Thrombomodulin gene c.1418C>T polymorphism and risk of recurrent venous thromboembolism. J Thromb Thrombolysis. 2016;42(1):135–41.PubMedCrossRef Ahmad A, Sundquist K, Zoller B, Svensson PJ, Sundquist J, Memon AA. Thrombomodulin gene c.1418C>T polymorphism and risk of recurrent venous thromboembolism. J Thromb Thrombolysis. 2016;42(1):135–41.PubMedCrossRef
15.
Zurück zum Zitat Hinds DA, Buil A, Ziemek D, Martinez-Perez A, Malik R, Folkersen L, Germain M, Malarstig A, Brown A, Soria JM, et al. Genome-wide association analysis of self-reported events in 6135 individuals and 252 827 controls identifies 8 loci associated with thrombosis. Hum Mol Genet. 2016;25(9):1867–74.PubMedPubMedCentralCrossRef Hinds DA, Buil A, Ziemek D, Martinez-Perez A, Malik R, Folkersen L, Germain M, Malarstig A, Brown A, Soria JM, et al. Genome-wide association analysis of self-reported events in 6135 individuals and 252 827 controls identifies 8 loci associated with thrombosis. Hum Mol Genet. 2016;25(9):1867–74.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Hernandez W, Gamazon ER, Smithberger E, O’Brien TJ, Harralson AF, Tuck M, Barbour A, Kittles RA, Cavallari LH, Perera MA. Novel genetic predictors of venous thromboembolism risk in African Americans. Blood. 2016;127(15):1923–9.PubMedPubMedCentralCrossRef Hernandez W, Gamazon ER, Smithberger E, O’Brien TJ, Harralson AF, Tuck M, Barbour A, Kittles RA, Cavallari LH, Perera MA. Novel genetic predictors of venous thromboembolism risk in African Americans. Blood. 2016;127(15):1923–9.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Cronin M, Dengler N, Krauss ES, Segal A, Wei N, Daly M, Mota F, Caprini JA. Completion of the Updated Caprini Risk Assessment Model (2013 Version). Clin Appl Thromb Hemost. 2019;25:1076029619838052.PubMedPubMedCentralCrossRef Cronin M, Dengler N, Krauss ES, Segal A, Wei N, Daly M, Mota F, Caprini JA. Completion of the Updated Caprini Risk Assessment Model (2013 Version). Clin Appl Thromb Hemost. 2019;25:1076029619838052.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Martin AR, Kanai M, Kamatani Y, Okada Y, Neale BM, Daly MJ. Clinical use of current polygenic risk scores may exacerbate health disparities. Nat Genet. 2019;51(4):584–91.PubMedPubMedCentralCrossRef Martin AR, Kanai M, Kamatani Y, Okada Y, Neale BM, Daly MJ. Clinical use of current polygenic risk scores may exacerbate health disparities. Nat Genet. 2019;51(4):584–91.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Zhai Z, Wang D, Lei J, Yang Y, Xu X, Ji Y, Yi Q, Chen H, Hu X, Liu Z, et al. Trends in risk stratification, in-hospital management and mortality of patients with acute pulmonary embolism: an analysis from China pUlmonary thromboembolism REgistry Study (CURES). Eur Respir J. 2021;58(4):2002963. Zhai Z, Wang D, Lei J, Yang Y, Xu X, Ji Y, Yi Q, Chen H, Hu X, Liu Z, et al. Trends in risk stratification, in-hospital management and mortality of patients with acute pulmonary embolism: an analysis from China pUlmonary thromboembolism REgistry Study (CURES). Eur Respir J. 2021;58(4):2002963.
20.
Zurück zum Zitat Kang K, Sun X, Wang L, Yao X, Tang S, Deng J, Wu X, Yang C, Chen G, WeGene Research T. Direct-to-consumer genetic testing in China and its role in GWAS discovery and replication. Quant Biol. 2021;9(2):201–15. Kang K, Sun X, Wang L, Yao X, Tang S, Deng J, Wu X, Yang C, Chen G, WeGene Research T. Direct-to-consumer genetic testing in China and its role in GWAS discovery and replication. Quant Biol. 2021;9(2):201–15.
22.
Zurück zum Zitat Li L, Huang P, Sun X, Wang S, Xu M, Liu S, Feng Z, Zhang Q, Wang X, Zheng X, et al. The ChinaMAP reference panel for the accurate genotype imputation in Chinese populations. Cell Res. 2021;31(12):1308–10.PubMedPubMedCentralCrossRef Li L, Huang P, Sun X, Wang S, Xu M, Liu S, Feng Z, Zhang Q, Wang X, Zheng X, et al. The ChinaMAP reference panel for the accurate genotype imputation in Chinese populations. Cell Res. 2021;31(12):1308–10.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Manichaikul A, Mychaleckyj JC, Rich SS, Daly K, Sale M, Chen WM. Robust relationship inference in genome-wide association studies. Bioinformatics. 2010;26(22):2867–73.PubMedPubMedCentralCrossRef Manichaikul A, Mychaleckyj JC, Rich SS, Daly K, Sale M, Chen WM. Robust relationship inference in genome-wide association studies. Bioinformatics. 2010;26(22):2867–73.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Chang CC, Chow CC, Tellier LC, Vattikuti S, Purcell SM, Lee JJ. Second-generation PLINK: rising to the challenge of larger and richer datasets. Gigascience. 2015;4:7.PubMedPubMedCentralCrossRef Chang CC, Chow CC, Tellier LC, Vattikuti S, Purcell SM, Lee JJ. Second-generation PLINK: rising to the challenge of larger and richer datasets. Gigascience. 2015;4:7.PubMedPubMedCentralCrossRef
26.
27.
Zurück zum Zitat Pruim RJ, Welch RP, Sanna S, Teslovich TM, Chines PS, Gliedt TP, Boehnke M, Abecasis GR, Willer CJ. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics. 2010;26(18):2336–7.PubMedPubMedCentralCrossRef Pruim RJ, Welch RP, Sanna S, Teslovich TM, Chines PS, Gliedt TP, Boehnke M, Abecasis GR, Willer CJ. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics. 2010;26(18):2336–7.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Watanabe K, Taskesen E, van Bochoven A, Posthuma D. Functional mapping and annotation of genetic associations with FUMA. Nat Commun. 2017;8(1):1826.PubMedPubMedCentralCrossRef Watanabe K, Taskesen E, van Bochoven A, Posthuma D. Functional mapping and annotation of genetic associations with FUMA. Nat Commun. 2017;8(1):1826.PubMedPubMedCentralCrossRef
29.
30.
Zurück zum Zitat Khramtsova EA, Stranger BE. Assocplots: a Python package for static and interactive visualization of multiple-group GWAS results. Bioinformatics. 2017;33(3):432–4.PubMedCrossRef Khramtsova EA, Stranger BE. Assocplots: a Python package for static and interactive visualization of multiple-group GWAS results. Bioinformatics. 2017;33(3):432–4.PubMedCrossRef
31.
Zurück zum Zitat Yang X, Jiang X, Chen G, Xiao Y, Geng S, Kang C, Zhou T, Li Y, Guo X, Xiao H, et al. T cell Ig mucin-3 promotes homeostasis of sepsis by negatively regulating the TLR response. J Immunol. 2013;190(5):2068–79.PubMedCrossRef Yang X, Jiang X, Chen G, Xiao Y, Geng S, Kang C, Zhou T, Li Y, Guo X, Xiao H, et al. T cell Ig mucin-3 promotes homeostasis of sepsis by negatively regulating the TLR response. J Immunol. 2013;190(5):2068–79.PubMedCrossRef
32.
Zurück zum Zitat Burgess S, Bowden J, Fall T, Ingelsson E, Thompson SG. Sensitivity Analyses for Robust Causal Inference from Mendelian Randomization Analyses with Multiple Genetic Variants. Epidemiology. 2017;28(1):30–42.PubMedCrossRef Burgess S, Bowden J, Fall T, Ingelsson E, Thompson SG. Sensitivity Analyses for Robust Causal Inference from Mendelian Randomization Analyses with Multiple Genetic Variants. Epidemiology. 2017;28(1):30–42.PubMedCrossRef
33.
Zurück zum Zitat Verbanck M, Chen CY, Neale B, Do R. Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nat Genet. 2018;50(5):693–8.PubMedPubMedCentralCrossRef Verbanck M, Chen CY, Neale B, Do R. Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nat Genet. 2018;50(5):693–8.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Bowden J, Davey Smith G, Burgess S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int J Epidemiol. 2015;44(2):512–25.PubMedPubMedCentralCrossRef Bowden J, Davey Smith G, Burgess S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int J Epidemiol. 2015;44(2):512–25.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Hemani G, Zheng J, Elsworth B, Wade KH, Haberland V, Baird D, Laurin C, Burgess S, Bowden J, Langdon R, et al. The MR-Base platform supports systematic causal inference across the human phenome. Elife. 2018;7:e34408. Hemani G, Zheng J, Elsworth B, Wade KH, Haberland V, Baird D, Laurin C, Burgess S, Bowden J, Langdon R, et al. The MR-Base platform supports systematic causal inference across the human phenome. Elife. 2018;7:e34408.
36.
Zurück zum Zitat Bulik-Sullivan BK, Loh PR, Finucane HK, Ripke S, Yang J. Schizophrenia Working Group of the Psychiatric Genomics C, Patterson N, Daly MJ, Price AL, Neale BM: LD Score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat Genet. 2015;47(3):291–5.PubMedPubMedCentralCrossRef Bulik-Sullivan BK, Loh PR, Finucane HK, Ripke S, Yang J. Schizophrenia Working Group of the Psychiatric Genomics C, Patterson N, Daly MJ, Price AL, Neale BM: LD Score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat Genet. 2015;47(3):291–5.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Choi SW, O'Reilly PF. PRSice-2: Polygenic Risk Score software for biobank-scale data. Gigascience. 2019;8(7):giz082. Choi SW, O'Reilly PF. PRSice-2: Polygenic Risk Score software for biobank-scale data. Gigascience. 2019;8(7):giz082.
38.
Zurück zum Zitat Zöller B, Svensson PJ, Dahlbäck B, Lind-Hallden C, Hallden C, Elf J. Genetic risk factors for venous thromboembolism. Expert Rev Hematol. 2020;13(9):971–81.PubMedCrossRef Zöller B, Svensson PJ, Dahlbäck B, Lind-Hallden C, Hallden C, Elf J. Genetic risk factors for venous thromboembolism. Expert Rev Hematol. 2020;13(9):971–81.PubMedCrossRef
39.
Zurück zum Zitat Tregouet DA, Heath S, Saut N, Biron-Andreani C, Schved JF, Pernod G, Galan P, Drouet L, Zelenika D, Juhan-Vague I, et al. Common susceptibility alleles are unlikely to contribute as strongly as the FV and ABO loci to VTE risk: results from a GWAS approach. Blood. 2009;113(21):5298–303.PubMedCrossRef Tregouet DA, Heath S, Saut N, Biron-Andreani C, Schved JF, Pernod G, Galan P, Drouet L, Zelenika D, Juhan-Vague I, et al. Common susceptibility alleles are unlikely to contribute as strongly as the FV and ABO loci to VTE risk: results from a GWAS approach. Blood. 2009;113(21):5298–303.PubMedCrossRef
40.
Zurück zum Zitat Germain M, Saut N, Greliche N, Dina C, Lambert JC, Perret C, Cohen W, Oudot-Mellakh T, Antoni G, Alessi MC, et al. Genetics of venous thrombosis: insights from a new genome wide association study. PLoS ONE. 2011;6(9):e25581.PubMedPubMedCentralCrossRef Germain M, Saut N, Greliche N, Dina C, Lambert JC, Perret C, Cohen W, Oudot-Mellakh T, Antoni G, Alessi MC, et al. Genetics of venous thrombosis: insights from a new genome wide association study. PLoS ONE. 2011;6(9):e25581.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Tang W, Teichert M, Chasman DI, Heit JA, Morange PE, Li G, Pankratz N, Leebeek FW, Pare G, de Andrade M, et al. A genome-wide association study for venous thromboembolism: the extended cohorts for heart and aging research in genomic epidemiology (CHARGE) consortium. Genet Epidemiol. 2013;37(5):512–21.PubMedPubMedCentralCrossRef Tang W, Teichert M, Chasman DI, Heit JA, Morange PE, Li G, Pankratz N, Leebeek FW, Pare G, de Andrade M, et al. A genome-wide association study for venous thromboembolism: the extended cohorts for heart and aging research in genomic epidemiology (CHARGE) consortium. Genet Epidemiol. 2013;37(5):512–21.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Herrera-Rivero M, Stoll M, Hegenbarth JC, Ruhle F, Limperger V, Junker R, Franke A, Hoffmann P, Shneyder M, Stach M, et al. Single- and Multimarker Genome-Wide Scans Evidence Novel Genetic Risk Modifiers for Venous Thromboembolism. Thromb Haemost. 2021;121(9):1169–80.PubMedCrossRef Herrera-Rivero M, Stoll M, Hegenbarth JC, Ruhle F, Limperger V, Junker R, Franke A, Hoffmann P, Shneyder M, Stach M, et al. Single- and Multimarker Genome-Wide Scans Evidence Novel Genetic Risk Modifiers for Venous Thromboembolism. Thromb Haemost. 2021;121(9):1169–80.PubMedCrossRef
43.
Zurück zum Zitat Klarin D, Emdin CA, Natarajan P, Conrad MF, Consortium I, Kathiresan S. Genetic Analysis of Venous Thromboembolism in UK Biobank Identifies the ZFPM2 Locus and Implicates Obesity as a Causal Risk Factor. Circ Cardiovasc Genet. 2017;10(2):e001643. Klarin D, Emdin CA, Natarajan P, Conrad MF, Consortium I, Kathiresan S. Genetic Analysis of Venous Thromboembolism in UK Biobank Identifies the ZFPM2 Locus and Implicates Obesity as a Causal Risk Factor. Circ Cardiovasc Genet. 2017;10(2):e001643.
44.
Zurück zum Zitat Lindstrom S, Loomis S, Turman C, Huang H, Huang J, Aschard H, Chan AT, Choi H, Cornelis M, Curhan G, et al. A comprehensive survey of genetic variation in 20,691 subjects from four large cohorts. PLoS ONE. 2017;12(3):e0173997.PubMedPubMedCentralCrossRef Lindstrom S, Loomis S, Turman C, Huang H, Huang J, Aschard H, Chan AT, Choi H, Cornelis M, Curhan G, et al. A comprehensive survey of genetic variation in 20,691 subjects from four large cohorts. PLoS ONE. 2017;12(3):e0173997.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Klarin D, Busenkell E, Judy R, Lynch J, Levin M, Haessler J, Aragam K, Chaffin M, Haas M, Lindstrom S, et al. Genome-wide association analysis of venous thromboembolism identifies new risk loci and genetic overlap with arterial vascular disease. Nat Genet. 2019;51(11):1574–9.PubMedPubMedCentralCrossRef Klarin D, Busenkell E, Judy R, Lynch J, Levin M, Haessler J, Aragam K, Chaffin M, Haas M, Lindstrom S, et al. Genome-wide association analysis of venous thromboembolism identifies new risk loci and genetic overlap with arterial vascular disease. Nat Genet. 2019;51(11):1574–9.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Ruhle F, Witten A, Barysenka A, Huge A, Arning A, Heller C, Krumpel A, Mesters R, Franke A, Lieb W, et al. Rare genetic variants in SMAP1, B3GAT2, and RIMS1 contribute to pediatric venous thromboembolism. Blood. 2017;129(6):783–90.PubMedCrossRef Ruhle F, Witten A, Barysenka A, Huge A, Arning A, Heller C, Krumpel A, Mesters R, Franke A, Lieb W, et al. Rare genetic variants in SMAP1, B3GAT2, and RIMS1 contribute to pediatric venous thromboembolism. Blood. 2017;129(6):783–90.PubMedCrossRef
47.
Zurück zum Zitat Shabana, Hasnain S. The fatty acid binding protein 2 (FABP2) polymorphism Ala54Thr and obesity in Pakistan: A population based study and a systematic meta-analysis. Gene. 2015;574(1):106–11.PubMedCrossRef Shabana, Hasnain S. The fatty acid binding protein 2 (FABP2) polymorphism Ala54Thr and obesity in Pakistan: A population based study and a systematic meta-analysis. Gene. 2015;574(1):106–11.PubMedCrossRef
48.
Zurück zum Zitat Thibord F, Klarin D, Brody JA, Chen MH, Levin MG, Chasman DI, Goode EL, Hveem K, Teder-Laving M, Martinez-Perez A, et al. Cross-Ancestry Investigation of Venous Thromboembolism Genomic Predictors. Circulation. 2022;146(16):1225–42.PubMedCrossRef Thibord F, Klarin D, Brody JA, Chen MH, Levin MG, Chasman DI, Goode EL, Hveem K, Teder-Laving M, Martinez-Perez A, et al. Cross-Ancestry Investigation of Venous Thromboembolism Genomic Predictors. Circulation. 2022;146(16):1225–42.PubMedCrossRef
49.
Zurück zum Zitat Ghouse J, Tragante V, Ahlberg G, Rand SA, Jespersen JB, Leinøe EB, Vissing CR, Trudsø L, Jonsdottir I, Banasik K, et al. Genome-wide meta-analysis identifies 93 risk loci and enables risk prediction equivalent to monogenic forms of venous thromboembolism. Nat Genet. 2023;55(3):399-409. Ghouse J, Tragante V, Ahlberg G, Rand SA, Jespersen JB, Leinøe EB, Vissing CR, Trudsø L, Jonsdottir I, Banasik K, et al. Genome-wide meta-analysis identifies 93 risk loci and enables risk prediction equivalent to monogenic forms of venous thromboembolism. Nat Genet. 2023;55(3):399-409.
50.
Zurück zum Zitat Zhao T, Nzekebaloudou M. lv J: Ala54Thr polymorphism of fatty acid-binding protein 2 gene and fasting blood lipids: a meta-analysis. Atherosclerosis. 2010;210(2):461–7.PubMedCrossRef Zhao T, Nzekebaloudou M. lv J: Ala54Thr polymorphism of fatty acid-binding protein 2 gene and fasting blood lipids: a meta-analysis. Atherosclerosis. 2010;210(2):461–7.PubMedCrossRef
51.
Zurück zum Zitat Ay C, Tengler T, Vormittag R, Simanek R, Dorda W, Vukovich T, Pabinger I. Venous thromboembolism–a manifestation of the metabolic syndrome. Haematologica. 2007;92(3):374–80.PubMedCrossRef Ay C, Tengler T, Vormittag R, Simanek R, Dorda W, Vukovich T, Pabinger I. Venous thromboembolism–a manifestation of the metabolic syndrome. Haematologica. 2007;92(3):374–80.PubMedCrossRef
52.
Zurück zum Zitat Zaccardi F, Kunutsor SK, Seidu S, Davies MJ, Khunti K. Is the lower risk of venous thromboembolism with statins related to low-density-lipoprotein reduction? A network meta-analysis and meta-regression of randomised controlled trials. Atherosclerosis. 2018;271:223–31.PubMedCrossRef Zaccardi F, Kunutsor SK, Seidu S, Davies MJ, Khunti K. Is the lower risk of venous thromboembolism with statins related to low-density-lipoprotein reduction? A network meta-analysis and meta-regression of randomised controlled trials. Atherosclerosis. 2018;271:223–31.PubMedCrossRef
53.
Zurück zum Zitat Bordbar M, de Mutsert R, Cevval M, Rosendaal FR, Jukema JW, Lijfering WM. Differential effect of statin use on coagulation markers: an active comparative analysis in the NEO study. Thromb J. 2021;19(1):45.PubMedPubMedCentralCrossRef Bordbar M, de Mutsert R, Cevval M, Rosendaal FR, Jukema JW, Lijfering WM. Differential effect of statin use on coagulation markers: an active comparative analysis in the NEO study. Thromb J. 2021;19(1):45.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Marston NA, Gurmu Y, Melloni GEM, Bonaca M, Gencer B, Sever PS, Pedersen TR, Keech AC, Roselli C, Lubitz SA, et al. The Effect of PCSK9 (Proprotein Convertase Subtilisin/Kexin Type 9) Inhibition on the Risk of Venous Thromboembolism. Circulation. 2020;141(20):1600–7.PubMedPubMedCentralCrossRef Marston NA, Gurmu Y, Melloni GEM, Bonaca M, Gencer B, Sever PS, Pedersen TR, Keech AC, Roselli C, Lubitz SA, et al. The Effect of PCSK9 (Proprotein Convertase Subtilisin/Kexin Type 9) Inhibition on the Risk of Venous Thromboembolism. Circulation. 2020;141(20):1600–7.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Crous-Bou M, De Vivo I, Camargo CA Jr, Varraso R, Grodstein F, Jensen MK, Kraft P, Goldhaber SZ, Lindström S, Kabrhel C. Interactions of established risk factors and a GWAS-based genetic risk score on the risk of venous thromboembolism. Thromb Haemost. 2016;116(4):705–13.PubMedPubMedCentralCrossRef Crous-Bou M, De Vivo I, Camargo CA Jr, Varraso R, Grodstein F, Jensen MK, Kraft P, Goldhaber SZ, Lindström S, Kabrhel C. Interactions of established risk factors and a GWAS-based genetic risk score on the risk of venous thromboembolism. Thromb Haemost. 2016;116(4):705–13.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, McCarthy MI, Ramos EM, Cardon LR, Chakravarti A, et al. Finding the missing heritability of complex diseases. Nature. 2009;461(7265):747–53.PubMedPubMedCentralCrossRef Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, McCarthy MI, Ramos EM, Cardon LR, Chakravarti A, et al. Finding the missing heritability of complex diseases. Nature. 2009;461(7265):747–53.PubMedPubMedCentralCrossRef
Metadaten
Titel
Genome-wide association analyses identified novel susceptibility loci for pulmonary embolism among Han Chinese population
verfasst von
Zhu Zhang
Haobo Li
Haoyi Weng
Geyu Zhou
Hong Chen
Guoru Yang
Ping Zhang
Xiangyan Zhang
Yingqun Ji
Kejing Ying
Bo Liu
Qixia Xu
Yongjun Tang
Guangfa Zhu
Zhihong Liu
Shuyue Xia
Xiaohong Yang
Lixia Dong
Ling Zhu
Mian Zeng
Yadong Yuan
Yuanhua Yang
Nuofu Zhang
Xiaomao Xu
Wenyi Pang
Meng Zhang
Yu Zhang
Kaiyuan Zhen
Dingyi Wang
Jieping Lei
Sinan Wu
Shi Shu
Yunxia Zhang
Shuai Zhang
Qian Gao
Qiang Huang
Chao Deng
Xi Fu
Gang Chen
Wenxin Duan
Jun Wan
Wanmu Xie
Peng Zhang
Shengfeng Wang
Peiran Yang
Xianbo Zuo
Zhenguo Zhai
Chen Wang
on behalf of the China pUlmonary Thromboembolism REgistry Study (CURES) investigators
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Medicine / Ausgabe 1/2023
Elektronische ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-023-02844-4

Weitere Artikel der Ausgabe 1/2023

BMC Medicine 1/2023 Zur Ausgabe

Leitlinien kompakt für die Allgemeinmedizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Facharzt-Training Allgemeinmedizin

Die ideale Vorbereitung zur anstehenden Prüfung mit den ersten 49 von 100 klinischen Fallbeispielen verschiedener Themenfelder

Mehr erfahren

Bei Herzinsuffizienz muss „Eisenmangel“ neu definiert werden

16.05.2024 Herzinsuffizienz Nachrichten

Bei chronischer Herzinsuffizienz macht es einem internationalen Expertenteam zufolge wenig Sinn, die Diagnose „Eisenmangel“ am Serumferritin festzumachen. Das Team schlägt vor, sich lieber an die Transferrinsättigung zu halten.

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.

Betalaktam-Allergie: praxisnahes Vorgehen beim Delabeling

16.05.2024 Pädiatrische Allergologie Nachrichten

Die große Mehrheit der vermeintlichen Penicillinallergien sind keine. Da das „Etikett“ Betalaktam-Allergie oft schon in der Kindheit erworben wird, kann ein frühzeitiges Delabeling lebenslange Vorteile bringen. Ein Team von Pädiaterinnen und Pädiatern aus Kanada stellt vor, wie sie dabei vorgehen.

Diabetestechnologie für alle?

15.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Eine verbesserte Stoffwechseleinstellung und höhere Lebensqualität – Diabetestechnologien sollen den Alltag der Patienten erleichtern. Dass CGM, AID & Co. bei Typ-1-Diabetes helfen, ist belegt. Bei Typ-2 gestaltet sich die Sache komplizierter.

Update Allgemeinmedizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.