Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 678
Filtrer
1.
J Am Heart Assoc ; 13(12): e033654, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38879448

RÉSUMÉ

Elevated lipoprotein(a) is a genetically transmitted codominant trait that is an independent risk driver for cardiovascular disease. Lipoprotein(a) concentration is heavily influenced by genetic factors, including LPA kringle IV-2 domain size, single-nucleotide polymorphisms, and interleukin-1 genotypes. Apolipoprotein(a) is encoded by the LPA gene and contains 10 subtypes with a variable number of copies of kringle -2, resulting in >40 different apolipoprotein(a) isoform sizes. Genetic loci beyond LPA, such as APOE and APOH, have been shown to impact lipoprotein(a) levels. Lipoprotein(a) concentrations are generally 5% to 10% higher in women than men, and there is up to a 3-fold difference in median lipoprotein(a) concentrations between racial and ethnic populations. Nongenetic factors, including menopause, diet, and renal function, may also impact lipoprotein(a) concentration. Lipoprotein(a) levels are also influenced by inflammation since the LPA promoter contains an interleukin-6 response element; interleukin-6 released during the inflammatory response results in transient increases in plasma lipoprotein(a) levels. Screening can identify elevated lipoprotein(a) levels and facilitate intensive risk factor management. Several investigational, RNA-targeted agents have shown promising lipoprotein(a)-lowering effects in clinical studies, and large-scale lipoprotein(a) testing will be fundamental to identifying eligible patients should these agents become available. Lipoprotein(a) testing requires routine, nonfasting blood draws, making it convenient for patients. Herein, we discuss the genetic determinants of lipoprotein(a) levels, explore the pathophysiological mechanisms underlying the association between lipoprotein(a) and cardiovascular disease, and provide practical guidance for lipoprotein(a) testing.


Sujet(s)
Maladies cardiovasculaires , Lipoprotéine (a) , Humains , Lipoprotéine (a)/sang , Lipoprotéine (a)/génétique , Maladies cardiovasculaires/génétique , Maladies cardiovasculaires/sang , Maladies cardiovasculaires/épidémiologie , Facteurs de risque de maladie cardiaque , Prédisposition génétique à une maladie , Appréciation des risques , Phénotype
2.
Genome Biol ; 25(1): 167, 2024 06 26.
Article de Anglais | MEDLINE | ID: mdl-38926899

RÉSUMÉ

BACKGROUND: Variable number tandem repeats (VNTRs) are highly polymorphic DNA regions harboring many potentially disease-causing variants. However, VNTRs often appear unresolved ("dark") in variation databases due to their repetitive nature. One particularly complex and medically relevant VNTR is the KIV-2 VNTR located in the cardiovascular disease gene LPA which encompasses up to 70% of the coding sequence. RESULTS: Using the highly complex LPA gene as a model, we develop a computational approach to resolve intra-repeat variation in VNTRs from largely available short-read sequencing data. We apply the approach to six protein-coding VNTRs in 2504 samples from the 1000 Genomes Project and developed an optimized method for the LPA KIV-2 VNTR that discriminates the confounding KIV-2 subtypes upfront. This results in an F1-score improvement of up to 2.1-fold compared to previously published strategies. Finally, we analyze the LPA VNTR in > 199,000 UK Biobank samples, detecting > 700 KIV-2 mutations. This approach successfully reveals new strong Lp(a)-lowering effects for KIV-2 variants, with protective effect against coronary artery disease, and also validated previous findings based on tagging SNPs. CONCLUSIONS: Our approach paves the way for reliable variant detection in VNTRs at scale and we show that it is transferable to other dark regions, which will help unlock medical information hidden in VNTRs.


Sujet(s)
Maladies cardiovasculaires , Répétitions minisatellites , Humains , Maladies cardiovasculaires/génétique , Variation génétique , Analyse de séquence d'ADN/méthodes , Lipoprotéine (a)/génétique , Prédisposition génétique à une maladie
3.
Lipids Health Dis ; 23(1): 175, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38851763

RÉSUMÉ

BACKGROUND: The causal associations of lipids and the drug target genes with atrial fibrillation (AF) risk remain obscure. We aimed to investigate the causal associations using genetic evidence. METHODS: Mendelian randomization (MR) analyses were conducted using summary-level genome-wide association studies (GWASs) in European and East Asian populations. Lipid profiles (low-density lipoprotein cholesterol, triglyceride, and lipoprotein[a]) and lipid-modifying drug target genes (3-hydroxy-3-methylglutaryl-CoA reductase, proprotein convertase subtilisin/kexin type 9, NPC1-like intracellular cholesterol transporter 1, apolipoprotein C3, angiopoietin-like 3, and lipoprotein[a]) were used as exposures. AF was used as an outcome. The inverse variance weighted method was applied as the primary method. Summary-data-based Mendelian randomization analyses were performed for further validation using expression quantitative trait loci data. Mediation analyses were conducted to explore the indirect effect of coronary heart disease. RESULTS: In the European population, MR analyses demonstrated that elevated levels of lipoprotein(a) increased AF risk. Moreover, analyses focusing on drug targets revealed that the genetically proxied target gene LPA, which simulates the effects of drug intervention by reducing lipoprotein(a), exhibited an association with AF risk. This association was validated in independent datasets. There were no consistent and significant associations observed for other traits when analyzed in different datasets. This finding was also corroborated by Summary-data-based Mendelian randomization analyses between LPA and AF. Mediation analyses revealed that coronary heart disease plays a mediating role in this association. However, in the East Asian population, no statistically significant evidence was observed to support these associations. CONCLUSIONS: This study provided genetic evidence that Lp(a) may be a causal factor for AF and that LPA may represent a promising pharmacological target for preventing AF in the European population.


Sujet(s)
Fibrillation auriculaire , Étude d'association pangénomique , Hydroxymethylglutaryl-CoA reductases , Lipoprotéine (a) , Analyse de randomisation mendélienne , Proprotéine convertase 9 , Humains , Protéine-3 de type angiopoïétine , Fibrillation auriculaire/génétique , Fibrillation auriculaire/traitement médicamenteux , Cholestérol LDL/sang , Prédisposition génétique à une maladie , Génomique/méthodes , Hydroxymethylglutaryl-CoA reductases/génétique , Lipoprotéine (a)/génétique , Lipoprotéine (a)/sang , Protéines de transport membranaire , Polymorphisme de nucléotide simple , Proprotéine convertase 9/génétique , Locus de caractère quantitatif , Facteurs de risque , Triglycéride/sang , /génétique , Peuples d'Asie de l'Est/génétique
4.
Arterioscler Thromb Vasc Biol ; 44(7): 1683-1693, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38779854

RÉSUMÉ

BACKGROUND: Heterozygous familial hypercholesterolemia (FH) is among the most common genetic conditions worldwide that affects ≈ 1 in 300 individuals. FH is characterized by increased levels of low-density lipoprotein cholesterol (LDL-C) and increased risk of coronary artery disease (CAD), but there is a wide spectrum of severity within the FH population. This variability in expression is incompletely explained by known risk factors. We hypothesized that genome-wide genetic influences, as represented by polygenic risk scores (PRSs) for cardiometabolic traits, would influence the phenotypic severity of FH. METHODS: We studied individuals with clinically diagnosed FH (n=1123) from the FH Canada National Registry, as well as individuals with genetically identified FH from the UK Biobank (n=723). For all individuals, we used genome-wide gene array data to calculate PRSs for CAD, LDL-C, lipoprotein(a), and other cardiometabolic traits. We compared the distribution of PRSs in individuals with clinically diagnosed FH, genetically diagnosed FH, and non-FH controls and examined the association of the PRSs with the risk of atherosclerotic cardiovascular disease. RESULTS: Individuals with clinically diagnosed FH had higher levels of LDL-C, and the incidence of atherosclerotic cardiovascular disease was higher in individuals with clinically diagnosed compared with genetically identified FH. Individuals with clinically diagnosed FH displayed enrichment for higher PRSs for CAD, LDL-C, and lipoprotein(a) but not for other cardiometabolic risk factors. The CAD PRS was associated with a risk of atherosclerotic cardiovascular disease among individuals with an FH-causing genetic variant. CONCLUSIONS: Genetic background, as expressed by genome-wide PRSs for CAD, LDL-C, and lipoprotein(a), influences the phenotypic severity of FH, expanding our understanding of the determinants that contribute to the variable expressivity of FH. A PRS for CAD may aid in risk prediction among individuals with FH.


Sujet(s)
Cholestérol LDL , Maladie des artères coronaires , Prédisposition génétique à une maladie , Étude d'association pangénomique , Hyperlipoprotéinémie de type II , Lipoprotéine (a) , Hérédité multifactorielle , Phénotype , Enregistrements , Humains , Hyperlipoprotéinémie de type II/génétique , Hyperlipoprotéinémie de type II/sang , Hyperlipoprotéinémie de type II/diagnostic , Hyperlipoprotéinémie de type II/épidémiologie , Femelle , Mâle , Adulte d'âge moyen , Cholestérol LDL/sang , Maladie des artères coronaires/génétique , Maladie des artères coronaires/épidémiologie , Maladie des artères coronaires/sang , Maladie des artères coronaires/diagnostic , Appréciation des risques , Lipoprotéine (a)/sang , Lipoprotéine (a)/génétique , Adulte , Sujet âgé , Canada/épidémiologie , Royaume-Uni/épidémiologie , Indice de gravité de la maladie , Facteurs de risque , Études cas-témoins , Marqueurs biologiques/sang , Incidence
5.
J Lipid Res ; 65(4): 100528, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38458338

RÉSUMÉ

Dyslipidemia has long been implicated in elevating mortality risk; yet, the precise associations between lipid traits and mortality remained undisclosed. Our study aimed to explore the causal effects of lipid traits on both all-cause and cause-specific mortality. One-sample Mendelian randomization (MR) with linear and nonlinear assumptions was conducted in a cohort of 407,951 European participants from the UK Biobank. Six lipid traits, consisting of low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), triglycerides, apolipoprotein A1 (ApoA1), apolipoprotein B (ApoB), and lipoprotein(a), were included to investigate the causal associations with mortality. Two-sample MR was performed to replicate the association between each lipid trait and all-cause mortality. Univariable MR results showed that genetically predicted higher ApoA1 was significantly associated with a decreased all-cause mortality risk (HR[95% CI]:0.93 [0.89-0.97], P value = 0.001), which was validated by the two-sample MR analysis. Higher lipoprotein(a) was associated with an increased risk of all-cause mortality (1.03 [1.01-1.04], P value = 0.002). Multivariable MR confirmed the direct causal effects of ApoA1 and lipoprotein(a) on all-cause mortality. Meanwhile, nonlinear MR found no evidence for nonlinearity between lipids and all-cause mortality. Our examination into cause-specific mortality revealed a suggestive inverse association between ApoA1 and cancer mortality, a significant positive association between lipoprotein(a) and cardiovascular disease mortality, and a suggestive positive association between lipoprotein(a) and digestive disease mortality. High LDL-C was associated with an increased risk of cardiovascular disease mortality but a decreased risk of neurodegenerative disease mortality. The findings suggest that implementing interventions to raise ApoA1 and decrease lipoprotein(a) levels may improve overall health outcomes and mitigate cancer and digestive disease mortality.


Sujet(s)
Lipides , Analyse de randomisation mendélienne , Humains , Mâle , Femelle , Lipides/sang , Adulte d'âge moyen , Facteurs de risque , Apolipoprotéine A-I/sang , Apolipoprotéine A-I/génétique , Lipoprotéine (a)/sang , Lipoprotéine (a)/génétique , Cause de décès , Sujet âgé
6.
J Clin Lab Anal ; 38(5): e24998, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38444303

RÉSUMÉ

BACKGROUND: Lipoprotein(a) [Lp(a)] level variability, related to atherothrombotic risk increase, is mainly attributed to LPA gene, encoding apolipoprotein(a), with kringle IV type 2 (KIV2) copy number variation (CNV) acting as the primary genetic determinant. Genetic characterization of Lp(a) is in continuous growth; nevertheless, the peculiar structural characteristics of this variant constitute a significant challenge to the development of effective detection methods. The aim of the study was to compare quantitative real-time PCR (qPCR) and digital droplet PCR (ddPCR) in the evaluation of KIV2 repeat polymorphism. METHODS: We analysed 100 subjects tested for cardiovascular risk in which Lp(a) plasma levels were assessed. RESULTS: Correlation analysis between CNV values obtained with the two methods was slightly significant (R = 0.413, p = 0.00002), because of the wider data dispersion in qPCR compared with ddPCR. Internal controls C1, C2 and C3 measurements throughout different experimental sessions revealed the superior stability of ddPCR, which was supported by a reduced intra/inter-assay coefficient of variation determined in this method compared to qPCR. A significant inverse correlation between Lp(a) levels and CNV values was confirmed for both techniques, but it was higher when evaluated by ddPCR than qPCR (R = -0.393, p = 0.000053 vs R = -0.220, p = 0.028, respectively). When dividing subjects into two groups according to 500 mg/L Lp(a) cut-off value, a significantly lower number of KIV2 repeats emerged among subjects with greater Lp(a) levels, with stronger evidence in ddPCR than in qPCR (p = 0.000013 and p = 0.001, respectively). CONCLUSIONS: Data obtained support a better performance of ddPCR in the evaluation of KIV2 repeat polymorphism.


Sujet(s)
Variations de nombre de copies de segment d'ADN , Kringles , Humains , Kringles/génétique , Variations de nombre de copies de segment d'ADN/génétique , Lipoprotéine (a)/génétique , Polymorphisme génétique , Réaction de polymérisation en chaine en temps réel/méthodes
7.
Int J Mol Sci ; 25(6)2024 Mar 21.
Article de Anglais | MEDLINE | ID: mdl-38542510

RÉSUMÉ

Lipoprotein(a) [Lp(a)] consists of a low-density lipoprotein-like molecule and an apolipoprotein(a) [apo(a)] particle. Lp(a) has been suggested to be an independent risk factor of atherosclerotic cardiovascular disease (ASCVD). Lp(a) plasma levels are considered to be 70-90% genetically determined through the codominant expression of the LPA gene. Therefore, Lp(a) levels are almost stable during an individual's lifetime. This lifelong stability, together with the difficulties in measuring Lp(a) levels in a standardized manner, may account for the scarcity of available drugs targeting Lp(a). In this review, we synopsize the latest data regarding the structure, metabolism, and factors affecting circulating levels of Lp(a), as well as the laboratory determination measurement of Lp(a), its role in the pathogenesis of ASCVD and thrombosis, and the potential use of various therapeutic agents targeting Lp(a). In particular, we discuss novel agents, such as antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs) that are currently being developed and target Lp(a). The promising role of muvalaplin, an oral inhibitor of Lp(a) formation, is then further analyzed.


Sujet(s)
Athérosclérose , Maladies cardiovasculaires , Humains , Lipoprotéine (a)/génétique , Athérosclérose/traitement médicamenteux , Facteurs de risque , Apoprotéine A , Apolipoprotéines A
8.
Curr Probl Cardiol ; 49(4): 102439, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38301917

RÉSUMÉ

BACKGROUND: Rising incidence of heart failure (HF) in the Western world despite advanced clinical care necessitate exploration of further preventive tools and strategies. Lipoprotein(a) [Lp(a)], recognized as one of the major cardiovascular risk factors has also been implicated as a risk factor for HF. However, existing evidence remains inconclusive and that has led us to perform this meta-analysis. METHODS: PubMed/Medline, EMBASE and Scopus were systematically searched for studies evaluating an association of Lp(a) with occurrence of HF from inception-till November 2023. Random effects models and I2 statistics were used for pooled odds ratio (OR) and heterogeneity assessment. We performed leave one out sensitivity analyses by sequentially removing one study at a time and recalculating the pooled effect size. RESULT: Our search rendered in total 360 studies and after final screening this resulted in 7 Mendelian randomization (MR) design. According to the MR analysis, increasing Lp(a) level were significantly associated with increased risk of HF (OR 1.064, 95 % CI: 1.043-1.086, I2= 97.59 %, P < 0.001). In addition, Leave-one-out sensitivity analysis showed that the effect size did not change substantially by removal of any particular study in MR studies and ORs ranged from 1.051 (when excluding Levin) to a maximum of 1.111 (when excluding Wang or Jiang), hereby confirming the association. CONCLUSION: We were able to show that by meta-analysis of MR data, increasing lipoprotein (a) levels are associated with an increased risk of HF. Whether this is due to a direct effect on heart muscle contraction or whether this is due to an increased risk of ischemic cardiac disease remains to be elucidated.


Sujet(s)
Maladie des artères coronaires , Défaillance cardiaque , Humains , Facteurs de risque de maladie cardiaque , Défaillance cardiaque/épidémiologie , Défaillance cardiaque/génétique , Lipoprotéine (a)/génétique , Analyse de randomisation mendélienne
9.
Curr Atheroscler Rep ; 26(4): 111-118, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38311667

RÉSUMÉ

PURPOSE OF REVIEW: Lipoprotein(a) is an important causal risk factor for cardiovascular disease but currently no available medication effectively reduces lipoprotein(a). This review discusses recent findings regarding lipoprotein(a) as a causal risk factor and therapeutic target in cardiovascular disease, it reviews current clinical recommendations, and summarizes new lipoprotein(a) lowering drugs. RECENT FINDINGS: Epidemiological and genetic studies have established lipoprotein(a) as a causal risk factor for cardiovascular disease and mortality. Guidelines worldwide now recommend lipoprotein(a) to be measured once in a lifetime, to offer patients with high lipoprotein(a) lifestyle advise and initiate other cardiovascular medications. Clinical trials including antisense oligonucleotides, small interfering RNAs, and an oral lipoprotein(a) inhibitor have shown great effect on lowering lipoprotein(a) with reductions up to 106%, without any major adverse effects. Recent clinical phase 1 and 2 trials show encouraging results and ongoing phase 3 trials will hopefully result in the introduction of specific lipoprotein(a) lowering drugs to lower the risk of cardiovascular disease.


Sujet(s)
Maladies cardiovasculaires , Lipoprotéine (a) , Humains , Maladies cardiovasculaires/traitement médicamenteux , Maladies cardiovasculaires/étiologie , Facteurs de risque de maladie cardiaque , Lipoprotéine (a)/effets des médicaments et des substances chimiques , Lipoprotéine (a)/génétique , Lipoprotéine (a)/métabolisme , Oligonucléotides antisens/usage thérapeutique , Facteurs de risque
10.
Int J Mol Sci ; 25(2)2024 Jan 06.
Article de Anglais | MEDLINE | ID: mdl-38255810

RÉSUMÉ

Lipoprotein(a) (Lp(a)) is an independent risk factor for future coronary events. Variants rs10455872 and rs3798220 in the gene encoding Lp(a) are associated with an increased Lp(a) concentration and risk of coronary artery disease. We aimed to determine whether in high-risk coronary artery disease patients these two genetic variants and the kringle IV type 2 (KIV-2) repeats are associated with impairment of inflammatory and hemostatic parameters. Patients after myocardial infarction with elevated Lp(a) levels were included. Blood samples underwent biochemical and genetic analyses. In carriers of the AC haplotype, the concentrations of tumor necrosis factor (TNF)-α (4.46 vs. 3.91 ng/L, p = 0.046) and plasminogen activator inhibitor-1 (PAI-1) (p = 0.026) were significantly higher compared to non-carriers. The number of KIV-2 repeats was significantly associated with the concentration of high-sensitivity C-reactive protein (ρ = 0.251, p = 0.038) and overall fibrinolytic potential (r = -0.253, p = 0.038). In our patients, a direct association between the AC haplotype and both TNF-α and PAI-1 levels was observed. Our study shows that the number of KIV-2 repeats not only affects proatherosclerotic and proinflammatory effects of Lp(a) but is also associated with its antifibrinolytic properties.


Sujet(s)
Maladie des artères coronaires , Infarctus du myocarde , Humains , Fibrinolyse/génétique , Inhibiteur-1 d'activateur du plasminogène/génétique , Maladie des artères coronaires/génétique , Haplotypes , Infarctus du myocarde/génétique , Inflammation/génétique , Lipoprotéine (a)/génétique , Facteur de nécrose tumorale alpha
11.
Sci Rep ; 14(1): 2564, 2024 01 31.
Article de Anglais | MEDLINE | ID: mdl-38297066

RÉSUMÉ

Heterozygous familial hypercholesterolemia (HeFH) is a common autosomal-dominant inherited disorder associated with atherosclerotic cardiovascular disease (ASCVD). HeFH subjects have a higher lipoprotein(a), i.e. Lp(a), concentration than the general population. Patients with FH are exposed to elevated levels of LDL from birth and ox-LDL may induce other oxidation pathways. The aim of the study was to determine the levels of markers of oxidative stress and DNA damage in patients with HeFH and describe the effect of Lp(a) on the resulting damage. Higher DNA damage was identified in patients with HeFH compared to the normolipidemic ones, and ASCVD was associated with greater damage. Oxidative stress markers were elevated in HeFH patients; however, only ox-LDL was higher in the ASCVD group and its level correlated with DNA damage. A positive correlation was found between DNA damage and Lp(a) concentration in the HeFH patients. Higher levels of Lp(a) were associated with greater DNA damage, especially in patients with HeFH and ASCVD. In HeFH patients, the optimal Lp(a) cut-off point associated with ASCVD is > 23.45 nmol/L, i.e. much lower than for the general population; however this cut-off point needs validation in a larger group of HeFH patients.


Sujet(s)
Athérosclérose , Hypercholestérolémie , Hyperlipoprotéinémie de type II , Humains , Lipoprotéine (a)/génétique , Hyperlipoprotéinémie de type II/métabolisme , Hypercholestérolémie/complications , Athérosclérose/complications , Altération de l'ADN
12.
J Am Coll Cardiol ; 83(3): 385-395, 2024 01 23.
Article de Anglais | MEDLINE | ID: mdl-38233012

RÉSUMÉ

BACKGROUND: Lipoprotein(a) (Lp(a)) is recognized as a causal factor for coronary heart disease (CHD) but its atherogenicity relative to that of low-density lipoprotein (LDL) on a per-particle basis is indeterminate. OBJECTIVES: The authors addressed this issue in a genetic analysis based on the fact that Lp(a) and LDL both contain 1 apolipoprotein B (apoB) per particle. METHODS: Genome-wide association studies using the UK Biobank population identified 2 clusters of single nucleotide polymorphisms: one comprising 107 variants linked to Lp(a) mass concentration, the other with 143 variants linked to LDL concentration. In these Lp(a) and LDL clusters, the relationship of genetically predicted variation in apoB with CHD risk was assessed. RESULTS: The Mendelian randomization-derived OR for CHD for a 50 nmol/L higher Lp(a)-apoB was 1.28 (95% CI: 1.24-1.33) compared with 1.04 (95% CI: 1.03-1.05) for the same increment in LDL-apoB. Likewise, use of polygenic scores to rank subjects according to difference in Lp(a)-apoB vs difference in LDL-apoB revealed a greater HR for CHD per 50 nmol/L apoB for the Lp(a) cluster (1.47; 95% CI: 1.36-1.58) compared with the LDL cluster (1.04; 95% CI: 1.02-1.05). From these data, we estimate that the atherogenicity of Lp(a) is approximately 6-fold (point estimate of 6.6; 95% CI: 5.1-8.8) greater than that of LDL on a per-particle basis. CONCLUSIONS: We conclude that the atherogenicity of Lp(a) (CHD risk quotient per unit increase in particle number) is substantially greater than that of LDL. Therefore, Lp(a) represents a key target for drug-based intervention in a significant proportion of the at-risk population.


Sujet(s)
Maladie coronarienne , Lipoprotéine (a) , Humains , Lipoprotéine (a)/génétique , Étude d'association pangénomique , Cholestérol LDL , Apolipoprotéines B/génétique , Maladie coronarienne/épidémiologie , Maladie coronarienne/génétique , Facteurs de risque
13.
Eur J Clin Invest ; 54(5): e14168, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38239089

RÉSUMÉ

BACKGROUND: In this study we used Mendelian randomization (MR) to investigate the potential causal association of lipoprotein (a) [Lp(a)] levels with pulse wave velocity (PWV). METHODS: Genetic variants associated with Lp(a) were retrieved from the UK Biobank GWAS (N = 290,497). A non- overlapping GWAS based on a European cohort (N = 7,000) was used to obtain genetic associations with PWV (outcome) and utilized two different measures for the same trait, brachial-ankle (baPWV) and carotid-femoral (cfPWV) PWV. We applied a two-sample MR using the inverse variance weighting method (IVW) and a series of sensitivity analyses for 170 SNPs that were selected as instrumental variables (IVs). RESULTS: Our analyses do not support a causal association between Lp(a) and PWV for neither measurement [ßiwv(baPWV) = -.0005, p = .8 and ßiwv(cfPWV) = -.006, p = .16]. The above findings were consistent across sensitivity analyses including weighted median, mode-based estimation, MR-Egger regression and MR-PRESSO. CONCLUSION: We did not find evidence indicating that Lp(a) is causally associated with PWV, the gold standard marker of arterial stiffness.


Sujet(s)
Lipoprotéine (a) , Rigidité vasculaire , Humains , Lipoprotéine (a)/génétique , Rigidité vasculaire/génétique , Analyse de l'onde de pouls , Analyse de randomisation mendélienne , Causalité
14.
Curr Atheroscler Rep ; 26(3): 75-82, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38252372

RÉSUMÉ

PURPOSE OF REVIEW: This paper reviews the evidence why lipoprotein(a) (Lp(a)) is a causal risk factor for cardiovascular disease and how high Lp(a) concentrations should be managed now and with an outlook to the future. REVIEW FINDINGS: No optimal and widely available animal models exist to study the causality of the association between Lp(a) and cardiovascular disease. This has been a major handicap for the entire field. However, genetic studies turned the page. Already in the early 1990s, the principle of Mendelian randomization studies was applied for the first time ever (even if they were not named so at that time). Genetic variants of the LPA gene such as the apolipoprotein(a) isoform size, the number and sum of kringle IV repeats and later single nucleotide polymorphisms are strongly associated with life-long exposure to high Lp(a) concentrations as well as cardiovascular outcomes. This evidence provided a basis for the development of specific Lp(a)-lowering drugs that are currently in clinical testing phase. Lp(a) is one of the most important genetically determined risk factors for cardiovascular disease. With the specific Lp(a)-lowering therapies, we might get tools to fight this common risk factor in case the outcome trials will be positive.


Sujet(s)
Maladies cardiovasculaires , Lipoprotéine (a) , Animaux , Humains , Lipoprotéine (a)/génétique , Maladies cardiovasculaires/génétique , Facteurs de risque , Polymorphisme de nucléotide simple
15.
Annu Rev Pharmacol Toxicol ; 64: 135-157, 2024 Jan 23.
Article de Anglais | MEDLINE | ID: mdl-37506332

RÉSUMÉ

Lipoprotein(a) [Lp(a)] is a molecule bound to apolipoprotein(a) with some similarity to low-density lipoprotein cholesterol (LDL-C), which has been found to be a risk factor for cardiovascular disease (CVD). Lp(a) appears to induce inflammation, atherogenesis, and thrombosis. Approximately 20% of the world's population has increased Lp(a) levels, determined predominantly by genetics. Current clinical practices for the management of dyslipidemia are ineffective in lowering Lp(a) levels. Evolving RNA-based therapeutics, such as the antisense oligonucleotide pelacarsen and small interfering RNA olpasiran, have shown promising results in reducing Lp(a) levels. Phase III pivotal cardiovascular outcome trials [Lp(a)HORIZON and OCEAN(a)] are ongoing to evaluate their efficacy in secondary prevention of major cardiovascular events in patients with elevated Lp(a). The future of cardiovascular residual risk reduction may transition to a personalized approach where further lowering of either LDL-C, triglycerides, or Lp(a) is selected after high-intensity statin therapy based on the individual risk profile and preferences of each patient.


Sujet(s)
Maladies cardiovasculaires , Humains , Cholestérol LDL/métabolisme , Cholestérol LDL/usage thérapeutique , Maladies cardiovasculaires/traitement médicamenteux , Maladies cardiovasculaires/génétique , Facteurs de risque , Lipoprotéine (a)/génétique , Lipoprotéine (a)/métabolisme , Lipoprotéine (a)/usage thérapeutique , Facteurs de risque de maladie cardiaque
18.
Curr Opin Clin Nutr Metab Care ; 27(1): 77-86, 2024 01 01.
Article de Anglais | MEDLINE | ID: mdl-37650693

RÉSUMÉ

PURPOSE OF REVIEW: Calcific aortic valve disease (CAVD), the most common cause of aortic stenosis (AS), is characterized by slowly progressive fibrocalcific remodelling of the valve cusps. Once symptomatic, severe AS is associated with poor survival unless surgical or transcatheter valve replacement is performed. Unfortunately, no pharmacological interventions have been demonstrated to alter the natural history of CAVD. Lipoprotein(a) [Lp(a)], a low-density lipoprotein-like particle, has been implicated in the pathophysiology of CAVD. RECENT FINDINGS: The mechanisms by which Lp(a) results in CAVD are not well understood. However, the oxidized phospholipids carried by Lp(a) are considered a crucial mediator of the disease process. An increasing number of studies demonstrate a causal association between plasma Lp(a) levels and frequency of AS and need for aortic valve replacement, which is independent of inflammation, as measured by plasma C-reactive protein levels. However, not all studies show an association between Lp(a) and increased progression of calcification in individuals with established CAVD. SUMMARY: Epidemiologic, genetic, and Mendelian randomization studies have collectively suggested that Lp(a) is a causal risk factor for CAVD. Whether Lp(a)-lowering can prevent initiation or slow progression of CAVD remains to be demonstrated.


Sujet(s)
Sténose aortique , Calcinose , Humains , Valve aortique/chirurgie , Valve aortique/métabolisme , Sténose aortique/épidémiologie , Sténose aortique/étiologie , Sténose aortique/chirurgie , Calcinose/génétique , Calcinose/métabolisme , Lipoprotéine (a)/génétique , Lipoprotéine (a)/métabolisme
19.
Curr Probl Cardiol ; 49(1 Pt A): 102024, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37553064

RÉSUMÉ

Lipoprotein (a) (Lp[a]) is an established risk factor for atherosclerotic cardiovascular disease (ASCVD). However, data on association of Lp(a) with risk of atrial fibrillation (AF) is still limited. We searched PubMed/Medline, Scopus, and EMBASE for studies evaluating the association of Lp(a) with the occurrence of AF until July 2023. Random effects models and I2 statistics were used for pooled odds ratios (OR), and heterogeneity assessments. A subgroup analysis was performed based on the cohort population, and a one-out sensitivity analysis was performed. This meta-analysis comprised 275,647 AF cases and 2,100,172 Lp(a) participants. An increase in Lp(a) was associated with an increased risk of AF in mendelian randomization (MR) studies (OR 1.024, 95% CI: 1.007-1.042, I2 = 87.72%, P < 0.001). Leave-one-out sensitivity analysis confirmed equivalent results in MR studies. Subgroup analysis of MR studies revealed a higher risk of AF in the European cohort (OR 1.023, 95% CI: 1.007-1.040, I2 = 89.05%, P < 0.001) and a low risk (OR 0.940, 95% CI: 0.893-0.990) in the Chinese population. Meta-analysis of the MR data suggested higher levels of Lp(a) were associated with increased risk of AF. Future robust prospective studies are warranted to validate these findings.


Sujet(s)
Fibrillation auriculaire , Humains , Fibrillation auriculaire/épidémiologie , Fibrillation auriculaire/génétique , Lipoprotéine (a)/génétique , Analyse de randomisation mendélienne , Études prospectives , Facteurs de risque
20.
Indian Heart J ; 76 Suppl 1: S117-S120, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38160790

RÉSUMÉ

Lp(a) is a genetically determined, heritable, independent and causal risk factor for ASCVD. About 1 in 5 people worldwide have elevated Lp(a) (>50 mg/dL or >125 nmol/L) whereas in Indians it is 25 %. Epidemiological, genome-wide association and mendelian randomization studies have demonstrated an association between elevated Lp(a) levels and increased incidence of myocardial infarction, aortic valve stenosis, ischemic stroke, heart failure, CV and all-cause mortality. The increased Lp(a)-mediated CV risk is mediated by pro-inflammatory, pro-thrombotic and pro-atherogenic processes, leading to progression of atherosclerosis and increased risk of thrombosis. Lp(a) level reaches peak by 5 years of age and remains stable over time. Levels are not much influenced by dietary and environmental factors but it can vary in certain clinical situations like thyroid diseases, chronic kidney disease, inflammation and sepsis. It should be measured at least once in life time. Cascade testing for high Lp(a) is recommended in the settings of FH, family history of (very) high Lp(a), and personal or family history of ASCVD. In the absence of specific Lp(a)-lowering therapies, comprehensive risk factor management is recommended as per guidelines for individuals with elevated Lp(a). PCSK9 inhibitors and Inclisiran reduce Lp(a) by 25%. Pelacarsen is an antisense oligonucleotide and is found to reduce Lp(a) by 80%. In a recent Indian study of 1,021 CAD patients, presence of elevated Lp(a) (>50 mg/dL) correlated with severe angiographic disease. 37% of ACS patients exhibited elevated Lp(a) and it was higher in young CAD patients with FH (43%).


Sujet(s)
Athérosclérose , Hyperlipidémies , Humains , Proprotéine convertase 9 , Lipoprotéine (a)/génétique , Étude d'association pangénomique , Facteurs de risque , Athérosclérose/diagnostic , Athérosclérose/épidémiologie , Athérosclérose/étiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...