Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 612
Filtrar
1.
Biochem J ; 481(19): 1277-1296, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39302109

RESUMEN

Elevated plasma levels of lipoprotein(a) (Lp(a)) are a prevalent, independent, and causal risk factor for atherosclerotic cardiovascular disease and calcific aortic valve disease. Lp(a) consists of a lipoprotein particle resembling low density lipoprotein and the covalently-attached glycoprotein apolipoprotein(a) (apo(a)). Novel therapeutics that specifically and potently lower Lp(a) levels are currently in advanced stages of clinical development, including in large, phase 3 cardiovascular outcomes trials. However, fundamental unanswered questions remain concerning some key aspects of Lp(a) biosynthesis and catabolism as well as the true pathogenic mechanisms of the particle. In this review, we describe the salient biochemical features of Lp(a) and apo(a) and how they underlie the disease-causing potential of Lp(a), the factors that determine plasma Lp(a) concentrations, and the mechanism of action of Lp(a)-lowering drugs.


Asunto(s)
Enfermedades Cardiovasculares , Lipoproteína(a) , Humanos , Lipoproteína(a)/sangre , Lipoproteína(a)/metabolismo , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/sangre , Animales , Aterosclerosis/metabolismo , Aterosclerosis/sangre
2.
Curr Opin Lipidol ; 35(5): 234-240, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39145610

RESUMEN

PURPOSE OF REVIEW: There is growing literature that supports the testing of Lp(a). However, few patients are tested, including those with a personal or family history of cardiovascular disease (CVD). One often noted barrier to more widespread testing is uncertainty regarding what to do with an elevated Lp(a) level. Although guidelines vary, there is agreement on the use of Lp(a) as a risk enhancer to guide medical care and shared decision-making. This review will discuss a clinical approach with supporting evidence for management of patients with elevated Lp(a). RECENT FINDINGS: At the minimum, elevated Lp(a) increases cardiovascular risk and can be incorporated into existing risk stratification paradigms. The cornerstone of management is aggressive management of traditional cardiovascular risk factors, including LDL-cholesterol (LDL-C). More recent studies have highlighted the potential role for proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), aspirin in primary prevention, and prolonged dual antiplatelet therapy in secondary prevention. SUMMARY: Although there is optimism for Lp(a)-targeted therapies in the near future, an elevated Lp(a) level is actionable today, and uncertainty regarding the management of patients with elevated Lp(a) should not be a barrier to more widespread testing.


Asunto(s)
Lipoproteína(a) , Humanos , Lipoproteína(a)/sangre , Lipoproteína(a)/metabolismo , Lipoproteína(a)/genética , Enfermedades Cardiovasculares/prevención & control , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/metabolismo
3.
Int J Mol Sci ; 25(16)2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39201505

RESUMEN

The evaluation of serum Lp(a) values in childhood and adolescence has been widely debated, and in the last few years, many authors have tried to better define Lp(a) role in atherosclerosis pathogenesis, starting from childhood. In our narrative review, we have evaluated the main historical stages of Lp(a) studies in childhood, trying to focus on pathogenic mechanisms linked to elevated serum Lp(a) values, starting from ischemic stroke and vascular damage, and to its possible direct involvement in premature atherosclerosis from childhood onwards. Historic manuscripts on Lp(a) in pediatric patients have mainly focused on serum Lp(a) values and increased stroke risk. More recently, many studies have evaluated Lp(a) as a coronary vascular disease (CVD) risk factor starting from childhood, especially related to a positive family history of premature CVD. Finally, only a few studies evaluated the role of Lp(a) in premature atherosclerotic processes and endothelial and vascular damage in pediatric patients. Lastly, we have hypothesized a future perspective, with the hope that plasma Lp(a) levels will be treated with a tailored pharmacologic approach, and Lp(a) will become a precocious therapeutic target to control the atherosclerotic pathways from the first years of life.


Asunto(s)
Enfermedades Cardiovasculares , Lipoproteína(a) , Humanos , Lipoproteína(a)/sangre , Lipoproteína(a)/metabolismo , Niño , Adolescente , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/metabolismo , Factores de Riesgo , Aterosclerosis/sangre
5.
Curr Atheroscler Rep ; 26(8): 383-394, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38878139

RESUMEN

PURPOSE OF REVIEW: The primary objective of this review is to explore the pathophysiological roles and clinical implications of lipoprotein(a) [Lp(a)] in the context of atherosclerotic cardiovascular disease (ASCVD). We seek to understand how Lp(a) contributes to inflammation and arteriosclerosis, aiming to provide new insights into the mechanisms of ASCVD progression. RECENT FINDINGS: Recent research highlights Lp(a) as an independent risk factor for ASCVD. Studies show that Lp(a) not only promotes the inflammatory processes but also interacts with various cellular components, leading to endothelial dysfunction and smooth muscle cell proliferation. The dual role of Lp(a) in both instigating and, under certain conditions, mitigating inflammation is particularly noteworthy. This review finds that Lp(a) plays a complex role in the development of ASCVD through its involvement in inflammatory pathways. The interplay between Lp(a) levels and inflammatory responses highlights its potential as a target for therapeutic intervention. These insights could pave the way for novel approaches in managing and preventing ASCVD, urging further investigation into Lp(a) as a therapeutic target.


Asunto(s)
Aterosclerosis , Inflamación , Lipoproteína(a) , Humanos , Lipoproteína(a)/metabolismo , Lipoproteína(a)/sangre , Aterosclerosis/metabolismo , Aterosclerosis/inmunología , Inflamación/metabolismo , Animales , Factores de Riesgo
6.
Curr Atheroscler Rep ; 26(7): 317-329, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38753254

RESUMEN

PURPOSE OF REVIEW: Low-density lipoprotein (LDL) poses a risk for atherosclerotic cardiovascular disease (ASCVD). As LDL comprises various subtypes differing in charge, density, and size, understanding their specific impact on ASCVD is crucial. Two highly atherogenic LDL subtypes-electronegative LDL (L5) and Lp(a)-induce vascular cell apoptosis and atherosclerotic changes independent of plasma cholesterol levels, and their mechanisms warrant further investigation. Here, we have compared the roles of L5 and Lp(a) in the development of ASCVD. RECENT FINDINGS: Lp(a) tends to accumulate in artery walls, promoting plaque formation and potentially triggering atherosclerosis progression through prothrombotic or antifibrinolytic effects. High Lp(a) levels correlate with calcific aortic stenosis and atherothrombosis risk. L5 can induce endothelial cell apoptosis and increase vascular permeability, inflammation, and atherogenesis, playing a key role in initiating atherosclerosis. Elevated L5 levels in certain high-risk populations may serve as a distinctive predictor of ASCVD. L5 and Lp(a) are both atherogenic lipoproteins contributing to ASCVD through distinct mechanisms. Lp(a) has garnered attention, but equal consideration should be given to L5.


Asunto(s)
Aterosclerosis , Lipoproteína(a) , Humanos , Lipoproteína(a)/sangre , Lipoproteína(a)/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/sangre , Lipoproteínas LDL/sangre , Lipoproteínas LDL/metabolismo , Apoptosis , Animales
7.
Nature ; 629(8013): 945-950, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38720069

RESUMEN

Lipoprotein(a) (Lp(a)), an independent, causal cardiovascular risk factor, is a lipoprotein particle that is formed by the interaction of a low-density lipoprotein (LDL) particle and apolipoprotein(a) (apo(a))1,2. Apo(a) first binds to lysine residues of apolipoprotein B-100 (apoB-100) on LDL through the Kringle IV (KIV) 7 and 8 domains, before a disulfide bond forms between apo(a) and apoB-100 to create Lp(a) (refs. 3-7). Here we show that the first step of Lp(a) formation can be inhibited through small-molecule interactions with apo(a) KIV7-8. We identify compounds that bind to apo(a) KIV7-8, and, through chemical optimization and further application of multivalency, we create compounds with subnanomolar potency that inhibit the formation of Lp(a). Oral doses of prototype compounds and a potent, multivalent disruptor, LY3473329 (muvalaplin), reduced the levels of Lp(a) in transgenic mice and in cynomolgus monkeys. Although multivalent molecules bind to the Kringle domains of rat plasminogen and reduce plasmin activity, species-selective differences in plasminogen sequences suggest that inhibitor molecules will reduce the levels of Lp(a), but not those of plasminogen, in humans. These data support the clinical development of LY3473329-which is already in phase 2 studies-as a potent and specific orally administered agent for reducing the levels of Lp(a).


Asunto(s)
Descubrimiento de Drogas , Lipoproteína(a) , Macaca fascicularis , Animales , Femenino , Humanos , Masculino , Ratones , Administración Oral , Kringles , Lipoproteína(a)/antagonistas & inhibidores , Lipoproteína(a)/sangre , Lipoproteína(a)/química , Lipoproteína(a)/metabolismo , Ratones Transgénicos , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/química , Plasminógeno/química , Plasminógeno/metabolismo , Especificidad de la Especie , Ensayos Clínicos Fase II como Asunto , Apolipoproteínas A/química , Apolipoproteínas A/metabolismo
8.
Curr Probl Cardiol ; 49(8): 102586, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38653440

RESUMEN

Cardiovascular disease (CVD) remains a significant global health challenge despite advancements in prevention and treatment. Elevated Lipoprotein(a) [Lp(a)] levels have emerged as a crucial risk factor for CVD and aortic stenosis, affecting approximately 20 of the global population. Research over the last decade has established Lp(a) as an independent genetic contributor to CVD and aortic stenosis, beginning with Kare Berg's discovery in 1963. This has led to extensive exploration of its molecular structure and pathogenic roles. Despite the unknown physiological function of Lp(a), studies have shed light on its metabolism, genetics, and involvement in atherosclerosis, inflammation, and thrombosis. Epidemiological evidence highlights the link between high Lp(a) levels and increased cardiovascular morbidity and mortality. Newly emerging therapies, including pelacarsen, zerlasiran, olpasiran, muvalaplin, and lepodisiran, show promise in significantly lowering Lp(a) levels, potentially transforming the management of cardiovascular disease. However, further research is essential to assess these novel therapies' long-term efficacy and safety, heralding a new era in cardiovascular disease prevention and treatment and providing hope for at-risk patients.


Asunto(s)
Enfermedades Cardiovasculares , Lipoproteína(a) , Humanos , Lipoproteína(a)/metabolismo , Lipoproteína(a)/sangre , Enfermedades Cardiovasculares/prevención & control , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Biomarcadores/sangre , Biomarcadores/metabolismo , Factores de Riesgo de Enfermedad Cardiaca , Hipolipemiantes/uso terapéutico , Factores de Riesgo
9.
Harefuah ; 163(3): 185-190, 2024 Mar.
Artículo en Hebreo | MEDLINE | ID: mdl-38506362

RESUMEN

INTRODUCTION: Lipoprotein(a) [Lp(a)] is composed of 2 major protein components, a low-density lipoprotein (LDL) cholesterol-like particle containing apolipoprotein B (apo B) that is covalently bound to apolipoprotein(a). Its level is predominantly genetically determined, and it is estimated that 20% to 25% of the population have Lp(a) levels that are associated with increased cardiovascular risk. Elevated Lp(a) is related to increased vascular inflammation, calcification, atherogenesis and thrombosis, and is considered an independent and potentially causal risk factor for atherosclerotic cardiovascular diseases and calcified aortic valve stenosis. Recent data demonstrate that Lp(a) testing has the potential to reclassify patients' risk and improve cardiovascular risk prediction, and therefore could inform clinical decision-making regarding risk management. Statins and ezetimibe are ineffective in lowering Lp(a) levels, whereas proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors have a modest effect on Lp(a) reduction. Nevertheless, RNA interference-based therapies with potent Lp(a)-lowering effects are in advanced stages of development, and clinical trials are underway to confirm their benefit in reducing cardiovascular events. This scientific consensus document was developed by a committee that consisted of representatives from the Israeli Society for the Research, Prevention and Treatment of Atherosclerosis, and the Israeli Society for Clinical Laboratory Sciences, in order to create uniformity in Lp(a) measurement methods, indications for testing and reporting of the results, aiming to improve the diagnosis and management of elevated Lp(a) in clinical practice.


Asunto(s)
Estenosis de la Válvula Aórtica , Válvula Aórtica/patología , Aterosclerosis , Calcinosis , Proproteína Convertasa 9 , Humanos , Israel , Ciencia del Laboratorio Clínico , Aterosclerosis/diagnóstico , Aterosclerosis/prevención & control , Lipoproteína(a)/metabolismo , Factores de Riesgo
10.
J Bioenerg Biomembr ; 56(3): 247-259, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38483739

RESUMEN

Cardiovascular diseases (CVDs) are the leading cause of death globally, attributed to a complex etiology involving metabolic, genetic, and protein-related factors. Lipoprotein(a) (Lp(a)), identified as a genetic risk factor, exhibits elevated levels linked to an increased risk of cardiovascular diseases. The lipoprotein(a) kringle domains have recently been identified as a potential target for the treatment of CVDs, in this study we utilized a fragment-based drug design approach to design a novel, potent, and safe inhibitor for lipoprotein(a) kringle domain. With the use of fragment library (61,600 fragments) screening, combined with analyses such as MM/GBSA, molecular dynamics simulation (MD), and principal component analysis, we successfully identified molecules effective against the kringle domains of Lipoprotein(a). The hybridization process (Breed) of the best fragments generated a novel 249 hybrid molecules, among them 77 exhibiting superior binding affinity (≤ -7 kcal/mol) compared to control AZ-02 (-6.9 kcal/mol), Importantly, the top ten molecules displayed high similarity to the control AZ-02. Among the top ten molecules, BR1 exhibited the best docking energy (-11.85 kcal/mol ), and higher stability within the protein LBS site, demonstrating the capability to counteract the pathophysiological effects of lipoprotein(a) [Lp(a)]. Additionally, principal component analysis (PCA) highlighted a similar trend of motion during the binding of BR1 and the control compound (AZ-02), limiting protein mobility and reducing conformational space. Moreover, ADMET analysis indicated favorable drug-like properties, with BR1 showing minimal violations of Lipinski's rules. Overall, the identified compounds hold promise as potential therapeutics, addressing a critical need in cardiovascular medicine. Further preclinical and clinical evaluations are needed to validate their efficacy and safety, potentially ushering in a new era of targeted therapies for CVDs.


Asunto(s)
Enfermedades Cardiovasculares , Diseño de Fármacos , Kringles , Lipoproteína(a) , Lipoproteína(a)/metabolismo , Lipoproteína(a)/química , Enfermedades Cardiovasculares/tratamiento farmacológico , Humanos , Simulación de Dinámica Molecular
11.
Kardiol Pol ; 82(4): 363-374, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38493469

RESUMEN

Patients with transient ST-segment elevation myocardial infarction or spontaneous reperfusion, which occurs in approximately 20% of patients with ST-segment elevation myocardial infarction (STEMI), have smaller infarcts and more favorable clinical outcomes than patients without spontaneous reperfusion. Understanding the mechanisms underlying spontaneous reperfusion is therefore important since this may identify possible novel therapeutic targets to improve outcomes in patients with STEMI. In this review, we discuss some of the possible determinants of spontaneous reperfusion including pro-thrombotic profile, endogenous fibrinolytic status, lipoprotein(a) (Lp[a]), inflammatory markers, and neutrophil extracellular traps (NETs). Effective (rapid) endogenous fibrinolysis, as assessed in whole blood in vitro, using a point-of-care technique assessment of global thrombotic status, has been strongly linked to spontaneous reperfusion. Lp(a), which has a high degree of homology to plasminogen, may impair fibrinolysis through competitive inhibition of tissue plasminogen activator-mediated plasminogen activation as well as tissue plasminogen activator-mediated clot lysis and contribute to pathogenic clot properties by decreasing fibrin clot permeation. NETs appear to negatively modulate clot lysis by increasing thrombin fiber diameter and inhibiting plasmin-driven lysis of plasma clots. There are limited data that oral anticoagulation may modulate endogenous fibrinolysis but antiplatelet agents currently appear to have no impact. Phase III trials involving subcutaneous P2Y12 or glycoprotein IIb/IIIa inhibitors, oral factor XIa inhibitors, interleukin-6 inhibitors, and apolipoprotein(a) antisense oligonucleotides in patients with cardiovascular disease are ongoing. Future studies will be needed to determine the impact of these novel antithrombotic, anti-inflammatory, and lipid-lowering therapies on endogenous fibrinolysis and spontaneous reperfusion.


Asunto(s)
Infarto del Miocardio con Elevación del ST , Humanos , Infarto del Miocardio con Elevación del ST/terapia , Infarto del Miocardio con Elevación del ST/fisiopatología , Fibrinólisis , Trampas Extracelulares/metabolismo , Reperfusión Miocárdica , Lipoproteína(a)/metabolismo , Lipoproteína(a)/sangre
12.
Int J Biol Macromol ; 264(Pt 1): 130388, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38417756

RESUMEN

Among the major Surface Exposed Colonization Proteins (SECPs) of Campylobacter jejuni (C. jejuni), Jejuni lipoprotein A (JlpA) plays a crucial role in host cell adhesion specifically by binding to the N-terminal domain of the human heat shock protein 90α (Hsp90α-NTD). Although the JlpA binding to Hsp90α activates NF-κB and p38 MAP kinase pathways, the underlying mechanism of JlpA association with the cellular receptor remains unclear. To this end, we predicted two potential receptor binding sites within the C-terminal domain of JlpA: one spanning from amino acid residues Q332-A354 and the other from S258-T295; however, the latter exhibited weaker binding. To assess the functional attributes of these predicted sequences, we generated two JlpA mutants (JlpAΔ1: S258-T295; JlpAΔ2: Q332-A354) and assessed the Hsp90α-binding affinity-kinetics by in vitro and ex vivo experiments. Our findings confirmed that the residues Q332-A354 are of greater importance in host cell adhesion with a measurable impact on cellular responses. Further, thermal denaturation by circular dichroism (CD) confirmed that the reduced binding affinity of the JlpAΔ2 to Hsp90α is not associated with protein folding or stability. Together, this study provides a possible framework for determining the molecular function of designing rational inhibitors efficiently targeting JlpA.


Asunto(s)
Campylobacter jejuni , Lipoproteína(a) , Humanos , Lipoproteína(a)/metabolismo , Campylobacter jejuni/genética , Campylobacter jejuni/metabolismo , Ligandos , Proteínas de Choque Térmico/metabolismo , FN-kappa B/metabolismo
13.
Curr Atheroscler Rep ; 26(4): 111-118, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38311667

RESUMEN

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.


Asunto(s)
Enfermedades Cardiovasculares , Lipoproteína(a) , Humanos , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/etiología , Factores de Riesgo de Enfermedad Cardiaca , Lipoproteína(a)/efectos de los fármacos , Lipoproteína(a)/genética , Lipoproteína(a)/metabolismo , Oligonucleótidos Antisentido/uso terapéutico , Factores de Riesgo
14.
Eur J Clin Invest ; 54(6): e14179, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38363025

RESUMEN

BACKGROUND: Emerging data suggested that lipoprotein(a) [Lp(a)] is an independent risk factor for atherosclerotic cardiovascular disease. Previous studies indicated fibrinogen (Fib) had synergetic effect on Lp(a)-induced events. However, combined impact of Fib and Lp(a) on ischemic stroke has not been elucidated. METHODS: In this prospective study, we consecutively enrolled 8263 patients with stable coronary artery diseases (CAD) from 2011 to 2017. Patients were categorized into three groups according to tertiles of Lp(a) levels [Lp(a)-low, Lp(a)-medium, and Lp(a)-high] and further divided into nine groups by Lp(a) and Fib levels. All subjects were followed up for the occurrence of ischemic stroke. RESULTS: During a median follow-up of 37.7 months, 157 (1.9%) ischemic strokes occurred. Stroke incidence increased by Lp(a) (1.1 vs. 2.1 vs. 2.5%, Cochran-Armitage p < .001) and Fib (1.1 vs. 2.0 vs. 2.6%, Cochran-Armitage p < .001) categories. When further classified into nine groups by Lp(a) and Fib levels, the incidence of ischemic stroke in group 9 [Lp(a)-high and Fib-high] was significantly higher than that in group 1 [Lp(a)-low and Fib-low] (3.1 vs. 6%, p < .001). The group 9 was associated with a highest risk for ischemic stroke (adjusted HR 4.907, 95% CI: 2.154-11.18, p < .001), compared with individuals in the Lp(a)-high (adjusted HR 2.290, 95% CI: 1.483-3.537, p < .001) or Fib-high (adjusted HR 1.184, 95% CI: 1.399-3.410, p = .001). Furthermore, combining Lp(a) with Fib increased C-statistics by .045 (p = .004). CONCLUSIONS: Current study first demonstrated that elevated Lp(a) combining with Fib evaluation enhanced the risk of ischemic stroke in patients with CAD beyond Lp(a) or Fib alone.


Asunto(s)
Enfermedad de la Arteria Coronaria , Fibrinógeno , Accidente Cerebrovascular Isquémico , Lipoproteína(a) , Humanos , Lipoproteína(a)/sangre , Lipoproteína(a)/metabolismo , Fibrinógeno/metabolismo , Masculino , Femenino , Enfermedad de la Arteria Coronaria/epidemiología , Persona de Mediana Edad , Anciano , Estudios Prospectivos , Accidente Cerebrovascular Isquémico/epidemiología , Accidente Cerebrovascular/epidemiología , Incidencia , Factores de Riesgo
15.
Arterioscler Thromb Vasc Biol ; 44(3): 720-740, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38269588

RESUMEN

BACKGROUND: Oxidized phospholipids play a key role in the atherogenic potential of lipoprotein(a) (Lp[a]); however, Lp(a) is a complex particle that warrants research into additional proinflammatory mediators. We hypothesized that additional Lp(a)-associated lipids contribute to the atherogenicity of Lp(a). METHODS: Untargeted lipidomics was performed on plasma and isolated lipoprotein fractions. The atherogenicity of the observed Lp(a)-associated lipids was tested ex vivo in primary human monocytes by RNA sequencing, ELISA, Western blot, and transendothelial migratory assays. Using immunofluorescence staining and single-cell RNA sequencing, the phenotype of macrophages was investigated in human atherosclerotic lesions. RESULTS: Compared with healthy individuals with low/normal Lp(a) levels (median, 7 mg/dL [18 nmol/L]; n=13), individuals with elevated Lp(a) levels (median, 87 mg/dL [218 nmol/L]; n=12) demonstrated an increase in lipid species, particularly diacylglycerols (DGs) and lysophosphatidic acid (LPA). DG and the LPA precursor lysophosphatidylcholine were enriched in the Lp(a) fraction. Ex vivo stimulation with DG(40:6) demonstrated a significant upregulation in proinflammatory pathways related to leukocyte migration, chemotaxis, NF-κB (nuclear factor kappa B) signaling, and cytokine production. Functional assessment showed a dose-dependent increase in the secretion of IL (interleukin)-6, IL-8, and IL-1ß after DG(40:6) and DG(38:4) stimulation, which was, in part, mediated via the NLRP3 (NOD [nucleotide-binding oligomerization domain]-like receptor family pyrin domain containing 3) inflammasome. Conversely, LPA-stimulated monocytes did not exhibit an inflammatory phenotype. Furthermore, activation of monocytes by DGs and LPA increased their transendothelial migratory capacity. Human atherosclerotic plaques from patients with high Lp(a) levels demonstrated colocalization of Lp(a) with M1 macrophages, and an enrichment of CD68+IL-18+TLR4+ (toll-like receptor) TREM2+ (triggering receptor expressed on myeloid cells) resident macrophages and CD68+CASP1+ (caspase) IL-1B+SELL+ (selectin L) inflammatory macrophages compared with patients with low Lp(a). Finally, potent Lp(a)-lowering treatment (pelacarsen) resulted in a reduction in specific circulating DG lipid subspecies in patients with cardiovascular disease with elevated Lp(a) levels (median, 82 mg/dL [205 nmol/L]). CONCLUSIONS: Lp(a)-associated DGs and LPA have a potential role in Lp(a)-induced monocyte inflammation by increasing cytokine secretion and monocyte transendothelial migration. This DG-induced inflammation is, in part, NLRP3 inflammasome dependent.


Asunto(s)
Lisofosfolípidos , Monocitos , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Diglicéridos/metabolismo , Inflamasomas/metabolismo , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lipoproteína(a)/metabolismo , Monocitos/metabolismo , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
16.
Annu Rev Pharmacol Toxicol ; 64: 135-157, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-37506332

RESUMEN

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.


Asunto(s)
Enfermedades Cardiovasculares , Humanos , LDL-Colesterol/metabolismo , LDL-Colesterol/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/genética , Factores de Riesgo , Lipoproteína(a)/genética , Lipoproteína(a)/metabolismo , Lipoproteína(a)/uso terapéutico , Factores de Riesgo de Enfermedad Cardiaca
17.
Curr Opin Clin Nutr Metab Care ; 27(2): 136-143, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-37997792

RESUMEN

PURPOSE OF REVIEW: Lp(a) is one of the most atherogenic lipoproteins, and significant progress has been made to understand its pathophysiology over the last 20 years. There are now selective therapies in late-stage clinical trials to lower Lp(a). Yet there are many outstanding questions about Lp(a). This review outlines 10 of the most burning questions and tries to answer some of them. RECENT FINDINGS: Antisense oligonucleotide (ASO) treatment is currently the most advanced therapy to lower plasma Lp(a) by 60-80%. There are, however, also two small molecule medications in early stage of development with similar efficacy. SUMMARY: This review aims to answer important preclinical and clinical questions about the metabolism and physiological role of Lp(a) and also outlines possible therapeutic approaches with nutraceuticals, currently available lipid-lowering therapies and new medications. In addition, ways are illustrated to use Lp(a) as a marker to better predict cardiovascular risk.


Asunto(s)
Aterosclerosis , Enfermedades Cardiovasculares , Lipoproteína(a) , Humanos , Aterosclerosis/tratamiento farmacológico , Lipoproteína(a)/antagonistas & inhibidores , Lipoproteína(a)/metabolismo , Oligonucleótidos Antisentido/uso terapéutico , Factores de Riesgo , Animales
18.
Curr Opin Clin Nutr Metab Care ; 27(1): 77-86, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37650693

RESUMEN

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.


Asunto(s)
Estenosis de la Válvula Aórtica , Calcinosis , Humanos , Válvula Aórtica/cirugía , Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/epidemiología , Estenosis de la Válvula Aórtica/etiología , Estenosis de la Válvula Aórtica/cirugía , Calcinosis/genética , Calcinosis/metabolismo , Lipoproteína(a)/genética , Lipoproteína(a)/metabolismo
19.
Protein Sci ; 32(10): e4781, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37703013

RESUMEN

The 11 lytic transglycosylases of Pseudomonas aeruginosa have overlapping activities in the turnover of the cell-wall peptidoglycan. Rare lipoprotein A (RlpA) is distinct among the 11 by its use of only peptidoglycan lacking peptide stems. The spatial localization of RlpA and its interactome within P. aeruginosa are unknown. We employed suppression of introduced amber codons at sites in the rlpA gene for the introduction of the unnatural-amino-acids Νζ -[(2-azidoethoxy)carbonyl]-l-lysine (compound 1) and Nζ -[[[3-(3-methyl-3H-diazirin-3-yl)propyl]amino]carbonyl]-l-lysine (compound 2). In live P. aeruginosa, full-length RlpA incorporating compound 1 into its sequence was fluorescently tagged using strained-promoted alkyne-azide cycloaddition and examined by fluorescence microscopy. RlpA is present at low levels along the sidewall length of the bacterium, and at higher levels at the nascent septa of replicating bacteria. In intact P. aeruginosa, UV photolysis of full-length RlpA having compound 2 within its sequence generated a transient reactive carbene, which engaged in photoaffinity capture of neighboring proteins. Thirteen proteins were identified. Three of these proteins-PBP1a, PBP5, and MreB-are members of the bacterial divisome. The use of the complementary methodologies of non-canonical amino-acid incorporation, photoaffinity proximity analysis, and fluorescent microscopy confirm a dominant septal location for the RlpA enzyme of P. aeruginosa, as a divisome-associated activity. This accomplishment adds to the emerging recognition of the value of these methodologies for identification of the intracellular localization of bacterial proteins.


Asunto(s)
Lipoproteína(a) , Pseudomonas aeruginosa , Lipoproteína(a)/metabolismo , Codón de Terminación/metabolismo , Peptidoglicano/metabolismo , Lisina/metabolismo
20.
Stem Cell Res ; 72: 103205, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37734317

RESUMEN

Elevated circulating lipoprotein(a) (Lp(a)) is a genetically determined risk factor for coronary artery disease and aortic valve stenosis (Tsimikas, 2017). Importantly, the LPA gene, which encodes the apolipoprotein(a) (protein-component of Lp(a)), is missing in most species, and human liver cell-lines do not secrete Lp(a). There is a need for the development of human in vitro models suitable for investigating biological mechanisms involved in Lp(a) metabolism. We here generated and characterized iPSCs from a patient with extremely high Lp(a) plasma levels genetically determined (Coassin et al., 2022). This unique cellular model offers great opportunities and new perspectives for investigations on biological mechanisms involved in Lp(a) metabolism.


Asunto(s)
Estenosis de la Válvula Aórtica , Enfermedad de la Arteria Coronaria , Células Madre Pluripotentes Inducidas , Humanos , Lipoproteína(a)/genética , Lipoproteína(a)/metabolismo , Válvula Aórtica/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Estenosis de la Válvula Aórtica/etiología , Estenosis de la Válvula Aórtica/genética , Enfermedad de la Arteria Coronaria/etiología , Enfermedad de la Arteria Coronaria/genética , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA