Welcome to the EAS 2023 Interactive Program

The congress will officially run on CET time zone (Central European Time)

225 Presentations

Sort By Relevance

O039 - CERAMIDE-BASED LIPID PROFILES AND THE PREVALENCE OF TYPE 2 DIABETES DIFFER BETWEEN PATIENTS WITH CORONARY ARTERY DISEASE AND THOSE WITH PERIPHERAL ARTERY DISEASE (ID 314)

Session Type
Workshop - Metabolism of Lipids and Lipoproteins
Date
Tue, 23.05.2023
Session Time
11:00 - 12:30
Room
Hall: Rudolf Schönheimer
Lecture Time
12:10 - 12:20

Abstract

Background and Aims

Serum lipids and metabolic diseases, in particular type 2 diabetes (T2D) and non-alcoholic fatty liver disease (NAFLD), predict the atherosclerotic diseases coronary artery disease (CAD) and peripheral arterial disease (PAD). However, it is not known in how far a more detailed characterization including serum lipids improves discrimination of PAD from CAD.

Methods

A cohort of 274 statin-naïve patients with either PAD (n = 89) or stable CAD (n = 185) were referred to metabolic screening and were characterized using nuclear magnetic resonance- and liquid chromatography-tandem mass spectrometry based advanced lipid and lipoprotein analysis. Results were validated in an independent cohort of 1239 patients with PAD or CAD.

Results

referenz - ceramide based lipid profiles.jpgWe found a significant difference in T2D prevalence and in the ceramide-based lipid profile between PAD and CAD patients. However, neither cholesterol-based markers (including LDL-C, HDL-C) and detailed lipoprotein profiles nor the NAFLD status differed significantly between PAD and CAD patients (figure). The difference between ceramide-based lipid profiles of CAD and PAD remained significant also after adjusting for body composition, smoking, inflammatory parameters, and T2D.

Conclusions

We conclude that PAD and CAD differ in ceramide-based lipid profiles and T2D status, but not in other lipid characteristics or metabolic diseases.

Hide

O051 - INTEGRATION OF GENETIC AND CLINICAL DATA, WITH SINGLE-CELL TRANSCRIPTOMICS OF ATHEROSCLEROTIC PLAQUES IDENTIFIES NOVEL SMOOTH MUSCLE CELL GENES (ID 339)

Session Type
Workshop - Pathogenesis of Atherosclerosis
Date
Tue, 23.05.2023
Session Time
15:45 - 17:15
Room
Hall: Heinrich Otto Wieland
Lecture Time
16:35 - 16:45

Abstract

Background and Aims

Smooth muscle cells (SMCs) have been causally implicated with disease processes in atherosclerosis. Single-cell RNA sequencing (scRNAseq) studies of atherosclerotic plaques have identified the presence of transitional mesenchymal cell (MC) populations. Here, we aimed to associate cell fractions from plaques to patient clinical and genetic parameters to identify novel MC-specific and symptom-specific signatures.

Methods

Deconvolution analysis was performed on bulk microarray from carotid plaques in Biobank of Karolinska Endarterectomies (BiKE, n=127) using public scRNAseq data from coronary plaques (n=5). Cell-fraction QTL (cfQTL), and plaque gene expression QTL (plaque eQTL) were calculated for a catalogue of CAD-associated GWAS loci to obtain MC-specific and symptom-specific SNPs. The function of the gene affected by the top SNP was studied in vitro using primary human carotid SMCs.

Results

Deconvolution analyses revealed that SMC cell fractions were significantly reduced in plaques from symptomatic patients. cfQTL and plaque eQTL analyses generated 84 eQTLs highly relevant for MC function. The most significant eQTL was identified as a SNP located at the regulatory region of gene ARNTL. This SNP was associated with expression levels of ARNTL specifically in plaques from symptomatic patients. Silencing ARNTL in SMCs in vitro led to inhibition of proliferation (2-way ANOVA, p < 0.01), increase in contractility (student t-test, p < 0.05), induction of senescence measured by b-galactosidase assay (student t-test, p < 0.05) and senescence markers (student t-test, p < 0.05).

Conclusions

This study identified several novel SNPs that may influence SMC function in plaques and provide insights into novel MC-specific genetic links to symptomatic atherosclerosis.

Hide

O055 - MEDITERRANEAN DIET PRESERVES KIDNEY FUNCTION IN PATIENTS WITH TYPE 2 DIABETES AND OBESITY: FROM THE CORDIOPREV STUDY. (ID 356)

Session Type
Workshop - Prevention and Treatment of CVD
Date
Tue, 23.05.2023
Session Time
15:45 - 17:15
Room
Hall: Rudolf Virchow
Lecture Time
16:25 - 16:35

Abstract

Background and Aims

Type 2 diabetes mellitus (T2DM) is considered an independent risk factor for chronic kidney disease (CKD); however, the distinctive contribution of obesity remains controversial. Our main aim was to establish an accurate contribution of obesity, with T2DM or not, to kidney function in patients with coronary heart disease (CHD), highly predisposed to renal complications, to assign the most appropriate dietary strategy.

Methods

1002 CHD patients from the CORDIOPREV study (Spain) were classified into four groups: Non-Obese/Non-T2DM, Obese/Non-T2DM, Non-Obese/T2DM and Obese/T2DM. Kidney function was assessed by estimated glomerular filtration rate (eGFR) after 5-years of dietary intervention with a low-fat or a Mediterranean diet. Generalized linear model (GLM) analysis were performed to determine the contribution of obesity and T2DM to changes in eGFR.

Results

Obese/T2DM patients showed lower baseline eGFR compared to those without T2DM (p < 0.001). After dietary intervention, the Mediterranean diet produced a lower decline of eGFR only in patients with concomitant T2DM and obesity, compared to low-fat diet (p = 0.014). GLM analysis showed that Obese/T2DM and Non-obese/T2DM patients (95% CI -4.966 to -1.080 and 95% CI -5.386 to -1.117, respectively, compared to Non-obese/Non-T2DM patients) and consumption of a low-fat diet determined more likelihood of declining eGFR.figure 1.jpgfigure 2.jpgtable 1.jpg

Conclusions

The presence of obesity provided an additive effect to T2DM determining a greater impairment of kidney function in CHD patients. Long-term consumption of a Mediterranean diet, compared to low-fat diet, may preserve kidney function, providing a dietary strategy for the reduction of CKD complications in the context of secondary prevention of cardiovascular disease.

Hide

O076 - REVERSING THE FUNCTIONAL- AND REGENERATIVE CAPACITY OF SENESCENT ENDOTHELIAL CELLS BY PHARMACOLOGICAL REPROGRAMMING (ID 366)

Session Type
Workshop - Pathogenesis of Atherosclerosis
Date
Wed, 24.05.2023
Session Time
11:00 - 12:30
Room
Hall: Rudolf Virchow
Lecture Time
12:20 - 12:30

Abstract

Background and Aims

Senescent endothelial cells (EC) are key players in the pathophysiology of cardiovascular diseases contributing to the development of vascular dysfunction with a reduced angiogenic and regenerative potential. Targeting EC senescence and reversing the senescent phenotype might represent a promising therapeutic strategy to improve vascular function. Here we show a reversal of EC senescence following the application of a pharmacological reprogramming strategy by a timely restricted, non-genetic induction of the Yamanaka-factors Oct3/4, Sox2, Klf4 and c-Myc (OSKM).

Methods

Methods to characterize the effects of pharmacological reprogramming included the quantification of gene expression as well as the functional analysis of EC in vitro. In addition, the regenerative capacity of EC was evaluated in an ischemic hind-limb model in vivo.

Results

Application of a pharmacological cocktail of FDA approved substances to replicative senescent EC led to a timely-restricted and robust overexpression of OSKM associated with significantly enhanced functional properties as proliferation, migration and tube formation of the senescent EC compared to untreated control cells (P<0.05). Further, expression of senescent markers such as p16INK4A or p14ARF and expression of cytokines for example TNFa, Il1b and Il-6 was significantly reduced compared to untreated senescent cells. In vivo, a significantly improved blood flow was observed after hind limb ischemia in 22 months old C57BL/6 mice after 7 and 14 days.

Conclusions

In summary, we demonstrate that a short induction of OSKM via a pharmacological approach holds the potential to reverse senescence in EC in vitro and thus to enhance endothelial regenerative capacity in vivo.

Hide

O049 - PEMAFIBRATE INDUCES ATHEROPROTECTION IN LDLR DEFICIENT MICE THROUGH HEPATOCYTE TRANSREPRESSION PPARA ACTIVITY (ID 368)

Session Type
Workshop - Metabolism of Lipids and Lipoproteins
Date
Tue, 23.05.2023
Session Time
15:45 - 17:15
Room
Hall: Anitschkow
Lecture Time
17:05 - 17:15

Abstract

Background and Aims

Beyond lifestyle intervention and statin treatment, residual atherosclerotic cardiovascular disease risk persists in many patients, and hypertriglyceridemia has emerged as a contributing factor in this risk. Therefore, fibrates that target the nuclear receptor peroxisome proliferator-activated receptor α (PPARα), represent interesting therapeutic candidates, given their beneficial effects on lipid metabolism and inflammation. Pemafibrate, a novel highly potent and selective PPARα agonist, has potential in managing cardiovascular risk, but the mechanisms of its atheroprotective effects are not well understood. The aim of our study was to better understand these mechanisms of action, and especially to evaluate the importance of the liver in these effects.

Methods

LDLr-/- PPARα+/+ and LDLr-/- PPARα-/-, as well as LDLr-/- PPARα-/- mice injected either with an AAV8-TBG (Thyroxin Binding Globulin)-GFP (control), an AAV8-TBG-PPARαWT (in order to restore PPARα selectively in the hepatocytes), or an AAV8-TBG-PPARαDISS (mutant of PPARα only owning anti-inflammatory properties), were submitted to a western diet supplemented or not with pemafibrate during 8 weeks.

Results

Pemafibrate induced a strong reduction of atherosclerotic plaque surface (-50%) in LDLr-/- PPARα+/+, but not in LDLr-/- PPARα-/- mice. Under pemafibrate treatment, AAV8-TBG-PPARαWT exhibited a strong reduction of dyslipidemia and atherosclerosis development (-42%), demonstrating a major role of hepatocyte PPARα on pemafibrate-induced atheroprotection. Surprisingly, in AAV8-TBG-PPARαDISS mice, pemafibrate was still able to induce a strong reduction of atherosclerosis lesion surface (-42%), despite no effect on atherogenic dyslipidemia.

Conclusions

Hepatocyte PPARα is the key driver of the beneficial effects of pemafibrate on dyslipidemia and atherosclerosis in LDLr-/- mice. Interestingly, modulation of hepatic inflammation through hepatocyte PPARα is sufficient to mediate pemafibrate-induced atheroprotection.

Hide

O035 - MALONDIALDEHYDE-SPECIFIC IGM ANTIBODIES DEFINE RESIDUAL ATHEROTHROMBOTIC RISK (ID 379)

Session Type
Young Fellows
Date
Tue, 23.05.2023
Session Time
11:00 - 12:30
Room
Hall: Rudolf Virchow
Lecture Time
12:20 - 12:30

Abstract

Background and Aims

Lipid peroxidation products like malondialdehyde(MDA) modify plaque components and are recognized by natural IgM antibodies. Titres of these inversely correlate with cardiovascular events, the mechanism of which is still poorly understood. Amongst others, MDA IgMs inhibit extracellular vesicle(EV)-induced clotting and thrombosis. Besides EVs, other plaque components are prothrombotic and MDA modified. We thus hypothesize that MDA IgMs are protective through inhibition of atherothrombosis.

Methods

We assessed MDA IgMs' anti-thrombotic potential. MDA modified plaque protein or plaque homogenates were used to trigger multiplate platelet aggregation assay, thrombin generation and thromboelastometry in the presence of LR04, an MDA targeting natural IgM, or its IgM isotype control. LR04 or isotype was co-injected with MDA plaque protein to induce pulmonary thrombosis in mice(n=30). Finally, MDA plaque protein IgM levels were measured in 704 cardiovascular patients(427 on statins) and correlated to clinical outcomes.

Results

In vitro, MDA plaque protein or plaque homogenates increased platelet and coagulation cascade activation, which was inhibited by LR04 but not isotype. In vivo, LR04 but not isotype protected from pulmonary thrombosis. Finally, higher levels of IgM targeting MDA modified plaque protein were associated with increased survival particularly in patients on statin therapy(p=0.001, multivariate adjusted, HR=0.431(CI95%:0.284-0.654))

Conclusions

Taken together, we describe a novel protective mechanism of MDA IgMs through atherothrombosis inhibition via reduction of platelet and coagulation cascade activation by MDA modified plaque components. Levels of these antibodies can be used to stratify patients in whom dyslipidaemia is addressed with statin therapy, representing a potential marker of residual risk in patients receiving lipid-lowering therapy.

Hide

O054 - DNMT3A CHIP-DRIVER MUTATIONS IGNITE THE BONE MARROW (ID 406)

Session Type
Workshop - Pathogenesis of Atherosclerosis
Date
Tue, 23.05.2023
Session Time
15:45 - 17:15
Room
Hall: Heinrich Otto Wieland
Lecture Time
17:05 - 17:15

Abstract

Background and Aims

The study aimed to identify phenotypical changes directly attributed to DNMT3A-CHIP driver mutations in blood and bone marrow myeloid cells from patients with coronary artery disease risk.

Methods

We conducted a prospective, observational study to screen CHIP-driver mutations in patients who were admitted for percutaneous coronary intervention. We established a single-cell DNA/RNA parallel sequencing method to acquire whole transcriptome and DNMT3A genotype of the same single blood monocytes from four DNMT3A mutation carriers and three matched non-carriers. We also applied such parallel sequencing method to bone marrow stem and progenitor cells from one DNMT3A hotspot mutation carrier, who underwent sternotomy.

Results

One third of patients in our cohort (53 out of 178) harbored CHIP-driver mutations with DNMT3A and TET2 accounting for 83% of the mutated CHIP genes. Middle-aged DNMT3A or TET2 carriers presented higher severity of coronary artery stenosis than the non-carriers. A total of 1268 blood monocytes were profiled with the parallel sequencing method, whereby we revealed that monocytes from DNMT3A mutation carriers upregulated pro-inflammatory markers, such as the S100A8 and S100A9 alarmins, in comparison with those from the non-carriers. Intra-individual comparison of approximately 100 mutant and 100 non-mutant monocytes of each carrier manifested an identical transcriptome profile, whereas the mutant hematopoietic stem and progenitor cells upregulated secretory cytokines, such as IL-1B and HMGB1, as opposed to their non-mutant counterparts.

Conclusions

Few bone marrow progenitor cells carrying DNMT3A-CHIP driver mutations suffice to induce proatherogenic inflammation in all monocytes –mutant and non-mutant ones alike.

Hide

O015 - ENDOTHELIAL CANNABINOID RECEPTOR 1 UPREGULATION BY PRO-ATHEROGENIC SHEAR STRESS PROMOTES VASCULAR INFLAMMATION (ID 415)

Session Type
Workshop - Pathogenesis of Atherosclerosis
Date
Mon, 22.05.2023
Session Time
15:45 - 17:15
Room
Hall: Heinrich Otto Wieland
Lecture Time
16:25 - 16:35

Abstract

Background and Aims

The cannabinoid receptor CB1 modulates blood pressure and atherogenesis, although the underlying mechanisms regulating CB1 activation are not fully understood. Endothelial cells respond to shear stress, altering gene expression and cell morphology. Here, we addressed whether endothelial CB1 expression is affected by shear stress, thereby modulating key cellular functions in a flow-dependent manner.

Methods

Murine samples were collected from Apoe-/- mice and mice with endothelial CB1 deficiency (Apoe-/-BmxCreCnr1flox/flox). In-situ hybridization for CB1 (Cnr1), immunostainings of ICAM1 and VE-Cadherin were performed with aortic sections and whole mounted arteries. Inner curvature of the aortic arch (OSS) was analysed as atheroprone area, and the descending aorta (LSS) as atheroprotective area. Perfusion experiments were performed to study OSS (4dyn/cm2) and LSS (10dyn/cm2) responses in human umbilical vein endothelial cells (HUVECs) and human aortic endothelial cells (HAECs). Gene expression levels were determined by qPCR.

Results

Endothelial Cnr1 expression was higher in atheroprone compared to atheroprotective areas of Apoe-/- aortas. In HUVECs, CNR1 was upregulated by OSS compared to static/LSS conditions. HAEC treatment with CB1 agonist (ACEA) under LSS, but not static condition, resulted in enhanced expression of markers for lipid transcytosis (CAV1), inflammation (ICAM, VCAM1) and glycolysis (PFKFB3) while decreasing anti-inflammatory gene expression levels. En-face-stained aortas of mice lacking endothelial CB1 revealed decreased ICAM1 in atheroprone areas, suggesting a less inflammatory phenotype.

Conclusions

Our results indicate that CB1 expression is upregulated by atheroprone flow conditions in-vivo and in-vitro. CB1 activation requires mechanical shear force to promote a pro-inflammatory phenotype, which may suggest that CB1 acts as a mechanosensor.

Hide

O056 - METHYLOMIC SIGNATURES MEDIATE THE EFFECT OF PSYCHOSOCIAL STRESS ON CORONARY HEART DISEASE (ID 438)

Session Type
Workshop - Prevention and Treatment of CVD
Date
Tue, 23.05.2023
Session Time
15:45 - 17:15
Room
Hall: Rudolf Virchow
Lecture Time
16:35 - 16:45

Abstract

Background and Aims

Elevated psychosocial stress has been repeatedly associated with altered immune function and increased coronary heart disease (CHD) risk, but the mechanisms underlying these associations are unclear. DNA methylation, a critical epigenetic process in humans, has emerged as a key link between environmental exposures and human health. Our aim was to examine whether methylomic changes mediate the effect of psychosocial stress on incident CHD.

Methods

Analyses included 3584 participants of the Women's Health Initiative (WHI). Stressful life events (SLEs) were measured with a self-administered questionnaire. Incident CHD was ascertained by certified personnel following standardized protocols. Genome-wide DNA (CpG) methylation was measured with the Illumina 450K array. Regression models identified stress-associated CpG sites and tested if their methylation levels statistically mediate the SLEs-CHD relationship. All models controlled for socioeconomic factors, lifestyle parameters, technical batch variables, and methylation-estimated blood cell proportions.

Results

In adjusted analyses, greater stress burden predicted higher incident CHD (HR=1.22, p=0.013) and was associated with methylome-wide differences at 872 CpG sites at false discovery rate (FDR)-adjusted p<0.05, 100 CpGs at FDR p<0.01, and 14 CpGs at the Bonferroni-adjusted p. Methylation risk scores (MRS) derived from the stress-associated CpGs predicted incident CHD (all p<0.008). Moreover, the effect of SLEs on CHD was significantly mediated by MRS100 and MRS14 (p<0.05).

Conclusions

Our findings suggest that methylomic changes mediate the effect of stress burden on incident CHD. These findings are now being replicated in independent human cohorts affiliated with the Trans-Omics for Precision Medicine (TOPMed) program and are further mechanistically dissected in immune cell model of stress.

Hide

O003 - NLRP3 INFLAMMASOME INHIBITION BY THE NOVEL BISPECIFIC ANTIBODY INFLAMAB INHIBITS ATHEROSCLEROSIS IN APOLIPOPROTEIN E-DEFICIENT MICE (ID 467)

Session Type
Workshop - Pathogenesis of Atherosclerosis
Date
Mon, 22.05.2023
Session Time
11:00 - 12:30
Room
Hall: Heinrich Otto Wieland
Lecture Time
12:00 - 12:10

Abstract

Background and Aims

Cardiovascular disease remains the most common cause of mortality worldwide, which is attributable to the underlying chronic inflammatory condition atherosclerosis. The NLRP3 inflammasome is involved in regulating multiple inflammatory diseases, including atherosclerosis. Here, we aimed to determine the efficacy of the novel bispecific antibody InflamAb, designed to target the NLRP3 inflammasome, in inhibiting atherosclerosis.

Methods

We treated western-type diet fed male apoE-/- mice for 3x per week with 100 µg InflamAb or isotype control antibody (n=11-13) during collar-induced atherosclerosis development for 4 weeks, after which atherosclerosis in the carotid artery was analyzed.

Results

Exposure of bone marrow derived macrophages to 25 ng/mL InflamAb prevented the LPA+Alum induced IL-1β release (P<0.05). In vivo, administration of InflamAb significantly reduced circulating IL-1β at 4 hours after LPS injection in western-type diet fed apoE-/- mice (P<0.05). InflamAb treatment significantly inhibited atherosclerotic plaque development from 59±8*103 µm2 in control mice to 36±5*103 µm2 (P<0.05), which was accompanied by a reduction in relative macrophage (control: 36±2% versus InflamAb: 28±3%, P<0.05) and necrotic core content (control: 16±2% versus InflamAb: 8±1%, P<0.05). In apoE-/- mice with established atherosclerosis, InflamAb treatment for 6 weeks did not affect lesion size, but significantly reduced relative macrophage (control: 48±2% versus InflamAb: 42±2%, P<0.05) and necrotic core content (control: 21±1% versus InflamAb: 18±1%, P<0.05) compared to controls, suggesting that InflamAb treatment improved advanced plaque stability.

Conclusions

We here show that inhibition of the NLRP3 inflammasome by the bispecific antibody InflamAb shows promising efficacy in inhibiting atherosclerotic plaque development and destabilization.

Hide

O027 - MYELOID-PCSK9 DEFICIENCY IMPROVES CARDIO-PROTECTION BY REGULATING LYVE1+ MACROPHAGES AGAINST ACUTE MYOCARDIAL INFARCTION (ID 492)

Session Type
Workshop - Metabolism of Lipids and Lipoproteins
Date
Tue, 23.05.2023
Session Time
11:00 - 12:30
Room
Hall: Heinrich Otto Wieland
Lecture Time
11:50 - 12:00

Abstract

Background and Aims

PCSK9 deficiency was reported to suppress inflammatory responses by regulating immune cells in the injury site. However, the effect of PCSK9-deficient macrophages after myocardial infarction (MI) has yet to be investigated. We hypothesized that PCSK9 deletion enabled cardio-protection during MI by regulating myeloid cells in the injury site. To prove this, we tested whether myeloid PCSK9 alters cardiac macrophage heterogeneity to adaptive remodeling after MI.

Methods

C57BL/6J, PCSK9-/-,PCSK9F/F, and Lyz2crePCSK9F/F male mice were subjected to MI using the left anterior descending branch of the coronary artery occlusion or sham operation. We analyzed PCSK9 deficiency functions in cardiac myeloid cells by Echocardiography, Trichrome staining, Flow cytometry, qPCR, and scRNA-seq.

Results

Myeloid-specific PCSK9 deletion alleviated cardiac dysfunction after myocardial injury. As a result of scRNA-seq, PCSK9 mediates macrophage heterogeneity following MI. Thus, the expression of CCR2-Lyve1+ cardiac macrophage subsets increased in PCSK9-/- MI hearts. These CCR2-Lyve1+ macrophages stimulate anti-inflammation against cardiac damage. Deficient PCSK9 in myeloid cells activates the BDNF pathway in the cardiac ischemic state to improve cell survival and wound healing compared to the control. These data elucidated that PCSK9 deletion affects macrophage heterogeneity to activate the BDNF signal, which protects against cardiac dysfunction.

Conclusions

PCSK9 deficiency in the myeloid activates CCR2-Lyve1+ cardiac macrophage to restrain the inflammatory state in the ischemic heart through the BDNF signaling pathway resulting in cardiac protection. Thus, myeloid PCSK9 can be a novel target to prevent adverse remodeling after MI.

Hide

O067 - COMBINING FAMILY HISTORY OF CORONARY HEART DISEASE AND INDIVIDUAL GENETIC PREDISPOSITION TO ACCURATELY PREDICT THE LIFETIME RISK OF MAJOR CORONARY EVENTS (ID 503)

Session Type
Workshop - Prevention and Treatment of CVD
Date
Wed, 24.05.2023
Session Time
11:00 - 12:30
Room
Hall: Heinrich Otto Wieland
Lecture Time
11:40 - 11:50

Abstract

Background and Aims

We aimed at assessing the impact of family history of coronary heart disease (CHD) and genetic predisposition in predicting the individual lifetime risk of major coronary events (MCE).

Methods

Using adjusted Cox proportional hazard models, we estimated the lifetime risk of MCE associated with parental family history of CHD and individual genetic predisposition (estimated through a polygenic score).

Results

A total of 445,744 UK-Biobank participants were selected (mean age 57 years; 54.3% females). Having one parent with a history of CHD increased the lifetime risk of MCE by 75% (HR 1.75, 95%CI 1.70-1.82, p-value<0.0001). Having both parents with a history of CHD further increased the risk (HR 2.78, 95%CI 2.64-2.92, p-value<0.0001) Similarly, a dose-dependent step-wise increase in MCE risk was observed moving from the lowest to the highest decile of the polygenic score. Compared to subjects without family history of CHD and with average level of the polygenic score, having a parental history of CHD determined an increase in lifetime risk of MCE (HR 1.90, 95%CI 1.82-1.98, p-value<0.0001) comparable to belonging to the highest decile of the polygenic score (HR 1.89, 95%CI 1.76-2.02, p-value<0.0001). However, if subjects present both parents with family history of CHD and a very high polygenic predisposition, the risk was even higher (HR 3.54, 95%CI 3.34-3.75, p-value<0.0001), suggesting an additive contribution.

Conclusions

We described the additive impact of family history of CHD and individual polygenic predisposition in predicting lifetime risk of MCE. Therefore, it is essential to retrieve information about both these hereditary components to identify subjects at higher risk.

Hide