Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Biochem Biophys Res Commun ; 712-713: 149946, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38643717

RESUMEN

Platelets are small anucleate cells that play a key role in thrombosis and hemostasis. Our group previously identified apolipoprotein A-IV (apoA-IV) as an endogenous inhibitor of thrombosis by competitive blockade of the αIIbß3 integrin on platelets. ApoA-IV inhibition of platelets was dependent on the N-terminal D5/D13 residues, and enhanced with absence of the C-terminus, suggesting it sterically hinders its N-terminal platelet binding site. The C-terminus is also the site of common apoA-IV polymorphisms apoA-IV-1a (T347S) and apoA-IV-2 (Q360H). Interestingly, both are linked with an increased risk of cardiovascular disease, however, the underlying mechanism remains unclear. Here, we generated recombinant apoA-IV and found that the Q360H or T347S polymorphisms dampened its inhibition of platelet aggregation in human platelet-rich plasma and gel-filtered platelets, reduced its inhibition of platelet spreading, and its inhibition of P-selectin on activated platelets. Using an ex vivo thrombosis assay, we found that Q360H and T347S attenuated its inhibition of thrombosis at both high (1800s-1) and low (300s-1) shear rates. We then demonstrate a conserved monomer-dimer distribution among apoA-IV WT, Q360H, and T347S and use protein structure modelling software to show Q360H and T347S enhance C-terminal steric hindrance over the N-terminal platelet-binding site. These data provide critical insight into increased cardiovascular risk for individuals with Q360H or T347S polymorphisms.


Asunto(s)
Apolipoproteínas A , Plaquetas , Agregación Plaquetaria , Trombosis , Humanos , Trombosis/genética , Trombosis/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/genética , Plaquetas/metabolismo , Plaquetas/efectos de los fármacos , Polimorfismo Genético , Apoproteína(a)/genética , Apoproteína(a)/metabolismo , Apoproteína(a)/química , Selectina-P/genética , Selectina-P/metabolismo
2.
Atherosclerosis ; 349: 123-135, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35606072

RESUMEN

Lipoprotein(a) [Lp(a)] became besides LDL cholesterol one of the most attractive targets for intervention in cardiovascular disease. Strong genetic evidence supports the causal association between high Lp(a) concentrations and cardiovascular outcomes. Since specific Lp(a)-lowering therapies are under clinical investigation, the interest in measuring Lp(a) has markedly increased. However, the special structure of the lead protein component of Lp(a), named apolipoprotein(a), creates difficulties for an accurate measurement of Lp(a). A highly homologous repetitive structure, called kringle IV repeat with up to more the 40 repeats, causes a highly polymorphic protein. Antibodies raised against apolipoprotein(a) are mostly directed against the repetitive structure of this protein, which complicates the measurement of Lp(a) in molar terms. Both measurements in mass (mg/dL) and molar terms (nmol/L) are described and a conversion from one into the another unit is only approximately possible. Working groups for standardization of Lp(a) measurements are going to prepare widely available and improved reference materials, which will be a major step for the measurement of Lp(a). This review discusses many aspects of the difficulties in measuring Lp(a). It tries to distinguish between academic and practical concerns and warns to make a mountain out of a molehill, which does no longer allow to see the patient behind that mountain by simply staring at the laboratory issues. On the other hand, the calibration of some assays raises major concerns, which are anything else but a molehill. This should be kept in mind and we should start measuring Lp(a) with the aim of a better risk stratification for the patient and to identify those patients who might be in urgent need for a specific Lp(a)-lowering therapy as soon as it becomes available.


Asunto(s)
Enfermedades Cardiovasculares , Lipoproteína(a) , Apolipoproteínas A , Apoproteína(a)/química , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/genética , LDL-Colesterol/metabolismo , Humanos , Lipoproteína(a)/genética
3.
Cardiovasc Res ; 118(9): 2103-2111, 2022 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34314498

RESUMEN

AIMS: Lipoprotein (a) [Lp(a)] is a lipoprotein species causatively associated with atherosclerosis. Unlike statins, PCSK9 inhibitors (PCSK9i) reduce Lp(a), but this reduction is highly variable. Levels of Lp(a) are chiefly governed by the size of its signature protein, apolipoprotein (a) [apo(a)]. Whether this parameter determines some of the reduction in Lp(a) induced by PCSK9i remains unknown. We aimed to investigate if the Lp(a) lowering efficacy of PCSK9i is modulated by the size of apo(a), which is genetically determined by the variable number of KIV domains present on that protein. METHODS AND RESULTS: The levels of Lp(a) and the size of apo(a) were assessed in plasma samples from 268 patients before and after treatment with PCSK9i. Patients were recruited at the Outpatient Lipid Clinic of the Charité Hospital (Berlin) between 2015 and 2020. They were hypercholesterolaemic at very high cardiovascular disease risk with low-density lipoprotein (LDL)-cholesterol levels above therapeutic targets despite maximally tolerated lipid-lowering therapy. Patients received either Alirocumab (75 or 150 mg) or Evolocumab (140 mg) every 2 weeks. Apo(a), apoB100, and apoE concentrations as well as apoE major isoforms were determined by liquid chromatography high-resolution mass spectrometry. Apo(a) isoforms sizes were determined by western blot. PCSK9i sharply reduced LDL-cholesterol (-57%), apoB100 (-47%), and Lp(a) (-36%). There was a positive correlation between the size of apo(a) and the relative reduction in Lp(a) induced by PCSK9i (r = 0.363, P = 0.0001). The strength of this association remained unaltered after adjustment for baseline Lp(a) levels and all other potential confounding factors. In patients with two detectable apo(a) isoforms, there was also a positive correlation between the size of apo(a) and the reduction in Lp(a), separately for the smaller (r = 0.350, P = 0.0001) and larger (r = 0.324, P = 0.0003) isoforms. The relative contribution of the larger isoform to the total concentration of apo(a) was reduced from 29% to 15% (P < 0.0001). CONCLUSIONS: The size of apo(a) is an independent determinant of the response to PCSK9i. Each additional kringle domain is associated with a 3% additional reduction in Lp(a). This explains in part the variable efficacy of PCSK9i and allows to identify patients who will benefit most from these therapies in terms of Lp(a) lowering.


Asunto(s)
Lipoproteína(a) , Inhibidores de PCSK9 , Apolipoproteínas E , Apoproteína(a)/química , Colesterol , Humanos , Lipoproteína(a)/metabolismo , Proproteína Convertasa 9 , Isoformas de Proteínas
4.
J Lipid Res ; 61(12): 1687-1696, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32907988

RESUMEN

Lipoprotein (a) [Lp(a)] is characterized by an LDL-like composition in terms of lipids and apoB100, and by one copy of a unique glycoprotein, apo(a). The apo(a) structure is mainly based on the repetition of tandem kringle domains with high homology to plasminogen kringles 4 and 5. Among them, kringle IV type 2 (KIV-2) is present in a highly variable number of genetically encoded repeats, whose length is inversely related to Lp(a) plasma concentration and cardiovascular risk. Despite it being the major component of apo(a), the actual function of KIV-2 is still unclear. Here, we describe the first high-resolution crystallographic structure of this domain. It shows a general fold very similar to other KIV domains with high and intermediate affinity for the lysine analog, ε-aminocaproic acid. Interestingly, KIV-2 presents a lysine binding site (LBS) with a unique shape and charge distribution. KIV-2 affinity for predicted small molecule binders was found to be negligible in surface plasmon resonance experiments; and with the LBS being nonfunctional, we propose to rename it "pseudo-LBS". Further investigation of the protein by computational small-molecule docking allowed us to identify a possible heparin-binding site away from the LBS, which was confirmed by specific reverse charge mutations abolishing heparin binding. This study opens new possibilities to define the pathogenesis of Lp(a)-related diseases and to facilitate the design of specific therapeutic drugs.


Asunto(s)
Apoproteína(a)/química , Apoproteína(a)/metabolismo , Kringles , Sitios de Unión , Humanos , Lisina/metabolismo , Modelos Moleculares
5.
Atherosclerosis ; 289: 201-205, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31327478

RESUMEN

BACKGROUND AND AIMS: We aimed to evaluate the effect of statin treatment initiation on lipoprotein(a) [Lp(a)] levels in patients with dyslipidemia, and the interactions with the apolipoprotein(a) [apo(a)] phenotype, LPA single nucleotide polymorphisms (SNPs) and change in LDL cholesterol. METHODS: The study population consisted of patients with dyslipidemia, predominantly familial hypercholesterolemia, who first initiated statin treatment (initiation group; n = 39) or were already on stable statin treatment for at least 4 months (control group; n = 42). Plasma Lp(a) levels were determined with a particle-enhanced immunoturbidimetric assay before and at least 2 months after start of statin treatment in individuals of the initiation group, and at two time points with an interval of at least 2 months in the control group. High and low molecular weight (HMW and LMW, respectively) apo(a) phenotype was determined by immunoblotting, and the common LPA SNPs rs10455872, rs3798220 and rs41272110 by Taqman assay. RESULTS: Plasma Lp(a) levels did not increase significantly in the initiation group (median 20.5 (IQR 10.9-80.7) to 23.3 (10.8-71.8) mg/dL; p = 0.09) nor in the control group (30.9 (IQR 9.2-147.0) to 31.7 (IQR 10.9-164.0) mg/dL; p = 0.61). In patients with the LMW apo(a) phenotype, Lp(a) levels increased significantly from 66.4 (IQR 23.5-148.3) to 97.4 (IQR 24.9-160.4) mg/dL (p = 0.026) in the initiation group, but not in the control group and not in patients characterized by the HMW apo(a) phenotype. Interactions with common LPA SNPs and change in LDL cholesterol were not significant. CONCLUSIONS: Statins affect Lp(a) levels differently in patients with dyslipidemia depending on the apo(a) phenotype. Statins increase Lp(a) levels exclusively in patients with the LMW apo(a) phenotype.


Asunto(s)
Apoproteína(a)/sangre , Enfermedades Cardiovasculares/metabolismo , Dislipidemias/sangre , Dislipidemias/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Adulto , Anciano , Apoproteína(a)/química , Enfermedades Cardiovasculares/prevención & control , LDL-Colesterol/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Peso Molecular , Fenotipo , Polimorfismo de Nucleótido Simple , Riesgo , Adulto Joven
6.
J Am Heart Assoc ; 8(7): e011781, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30897995

RESUMEN

Background Elevated lipoprotein(a) (Lp(a)), a low-density lipoprotein-like particle bound to the polymorphic apolipoprotein(a) (apo(a)), may be causal for cardiovascular disease. However, the metabolism of Lp(a) in humans is poorly understood. Methods and Results We investigated the kinetics of Lp(a)-apo(a) and low-density lipoprotein-apoB-100 in 63 normolipidemic men. The fractional catabolic rate ( FCR ) and production rate PR ) were studied. Plasma apo(a) concentration was significantly and inversely associated with apo(a) isoform size ( r=-0.536, P<0.001) and apo(a) FCR ( r=-0.363, P<0.01), and positively with apo(a) PR ( r=0.877, P<0.001). There were no significant associations between the FCR s of apo(a) and low-density lipoprotein-apoB-100. Subjects with smaller apo(a) isoform sizes (≤22 kringle IV repeats) had significantly higher apo(a) PR ( P<0.05) and lower apo(a) FCR ( P<0.01) than those with larger sizes. Plasma apo(a) concentration was significantly associated with apo(a) PR ( r=0.930, P<0.001), but not with FCR ( r=-0.012, P>0.05) in subjects with smaller apo(a) isoform size. In contrast, both apo(a) PR and FCR were significantly associated with plasma apo(a) concentrations ( r=0.744 and -0.389, respectively, P<0.05) in subjects with larger isoforms. In multiple regression analysis, apo(a) PR and apo(a) isoform size were significant predictors of plasma apo(a) concentration independent of low-density lipoprotein-apoB-100 FCR and background therapy with atorvastatin and evolocumab. Conclusions In normolipidemic men, the plasma Lp(a) concentration is predominantly determined by the rate of production of Lp(a) particles, irrespective of apo(a) isoform size and background therapy with a statin and a proprotein convertase subtilisin-kexin type 9 inhibitor. Our findings underscore the importance of therapeutic targeting of the hepatic synthesis and secretion of Lp(a) particles. Lp(a) particle catabolism may only play a modest role in determining Lp(a) concentration in subjects with larger apo(a) isoform size. Clinical Trial Registration URL : http://www.clinicaltrials.gov . Unique identifier: NCT 02189837.


Asunto(s)
Apoproteína(a)/química , Adolescente , Adulto , Anciano , Anticolesterolemiantes/uso terapéutico , HDL-Colesterol/metabolismo , LDL-Colesterol/metabolismo , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/prevención & control , Lipoproteína(a)/sangre , Lipoproteína(a)/metabolismo , Masculino , Persona de Mediana Edad , Isoformas de Proteínas , Adulto Joven
7.
J Clin Lipidol ; 12(5): 1313-1323, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30100157

RESUMEN

BACKGROUND: Lipoprotein(a) [Lp(a)] is reported as Lp(a) particle mass (mg/dL) or molar concentration of apolipoprotein(a) [apo(a)] (nmol/L), which is considered the gold standard. Values are often converted from one measurement to the other but the validity of this is unknown. OBJECTIVES: To quantify the relationship between Lp(a) molar concentration and Lp(a) mass in the context of various Lp(a) level thresholds and apo(a) isoform size. METHODS: In all samples, Lp(a) levels in molar concentration and apo(a) isoform size were determined at the Northwest Lipid Metabolism and Diabetes Research Laboratories (NLMDRL). Lp(a) mass levels were determined at the University of California, San Diego (UCSD) (1635 samples), by 5 commercially available assays: Denka 1 and Denka 2 (each 80 samples), 2 turbidimetric assays (2545 and 2673 samples, respectively), and an enzyme-linked immunosorbent assay (2605 samples). The ratios between Lp(a) molar concentration and mass (eg, nmol/L/mg/dL) were calculated and related to apo(a) isoform size. RESULTS: The mean (SD) ratios for NLMDRL/UCSD, NLMDRL/Denka1, and NLMDRL/Denka2 were 2.42 (1.25), 1.64 (0.18), and 2.02 (0.22), respectively. The ratios for NLMDRL/UCSD, NLMDRL/Denka1, and NLMDRL/Denka2 increased by Lp(a) cutoffs, with ratios of 1.82, 1.52, and 1.87, respectively, for Lp(a) < 75 nmol/L and 2.80, 1.89, and 2.24, respectively, for Lp(a) > 125 nmol/L. For the commercial turbidimetric assays and enzyme-linked immunosorbent assay, the ratios ranged from <1 to >5. CONCLUSIONS: Lp(a) molar/mass ratios are threshold, method, and isoform dependent. A single conversion factor between assays is not appropriate. These data support the transition of Lp(a) mass assays to molar concentration to improve diagnostic and clinical interpretation of Lp(a)-mediated risk.


Asunto(s)
Apoproteína(a)/química , Lipoproteína(a)/química , Lipoproteína(a)/metabolismo , Apoproteína(a)/metabolismo , Humanos , Lipoproteína(a)/genética , Peso Molecular , Polimorfismo de Nucleótido Simple , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo
8.
J Lipid Res ; 58(10): 2008-2016, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28798072

RESUMEN

An elevated level of lipoprotein (a) [Lp(a)] is a risk factor for CVD. Alirocumab, a monoclonal antibody to proprotein convertase subtilisin/kexin type 9, is reported to reduce Lp(a) levels. The relationship of Lp(a) reduction with apo(a) size polymorphism, phenotype, and dominance pattern and LDL cholesterol (LDL-C) reduction was evaluated in a pooled analysis of 155 hypercholesterolemic patients (75 with heterozygous familial hypercholesterolemia) from two clinical trials. Alirocumab significantly reduced total Lp(a) (pooled median: -21%, P = 0.0001) and allele-specific apo(a), an Lp(a) level carried by the smaller (median: -18%, P = 0.002) or the larger (median: -37%, P = 0.0005) apo(a) isoform, at week 8 versus baseline. The percent reduction in Lp(a) level with alirocumab was similar across apo(a) phenotypes (single vs. double bands) and carriers and noncarriers of a small size apo(a) (≤22 kringles). The percent reduction in LDL-C correlated significantly with the percent reduction in Lp(a) level (r = 0.407, P < 0.0001) and allele-specific apo(a) level associated with the smaller (r = 0.390, P < 0.0001) or larger (r = 0.270, P = 0.0183) apo(a) sizes. In conclusion, alirocumab-induced Lp(a) reduction was independent of apo(a) phenotypes and the presence or absence of a small size apo(a).


Asunto(s)
Anticuerpos Monoclonales/farmacología , Apoproteína(a)/química , Apoproteína(a)/metabolismo , Inhibidores de PCSK9 , Fenotipo , Inhibidores de Proteasas/farmacología , Anticuerpos Monoclonales Humanizados , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo
9.
J Thromb Haemost ; 15(9): 1834-1844, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28632940

RESUMEN

Essentials Elevated lipoproteinp(a) is an independent and causal risk factor for atherothrombotic diseases. rs3798220 (Ile/Met substitution in apo(a) protease-like domain) is associated with disease risk. Recombinant I4399M apo(a) altered clot structure to accelerate coagulation/delay fibrinolysis. Evidence was found for increased solvent exposure and oxidation of Met residue. SUMMARY: Background Lipoprotein(a) (Lp[a]) is a causal risk factor for a variety of cardiovascular diseases. Apolipoprotein(a) (apo[a]), the distinguishing component of Lp(a), is homologous with plasminogen, suggesting that Lp(a) can interfere with the normal fibrinolytic functions of plasminogen. This has implications for the persistence of fibrin clots in the vasculature and hence for atherothrombotic diseases. A single-nucleotide polymorphism (SNP) (rs3798220) in the gene encoding apo(a) has been reported that results in an Ile→Met substitution in the protease-like domain (I4399M variant). In population studies, the I4399M variant has been correlated with elevated plasma Lp(a) levels and higher coronary heart disease risk, and carriers of the SNP had increased cardiovascular benefit from aspirin therapy. In vitro studies suggested an antifibrinolytic role for Lp(a) containing this variant. Objectives We performed a series of experiments to assess the effect of the Ile→Met substitution on fibrin clot formation and lysis, and on the architecture of the clots. Results We found that the Met variant decreased coagulation time and increased fibrin clot lysis time as compared with wild-type apo(a). Furthermore, we observed that the presence of the Met variant significantly increased fibrin fiber width in plasma clots formed ex vivo, while having no effect on fiber density. Mass spectrometry analysis of a recombinant apo(a) species containing the Met variant revealed sulfoxide modification of the Met residue. Conclusions Our data suggest that the I4399M variant differs structurally from wild-type apo(a), which may underlie key differences related to its effects on fibrin clot architecture and fibrinolysis.


Asunto(s)
Apoproteína(a)/sangre , Apoproteína(a)/genética , Coagulación Sanguínea/genética , Fibrinólisis/genética , Lipoproteína(a)/sangre , Lipoproteína(a)/genética , Polimorfismo de Nucleótido Simple , Trombosis/sangre , Trombosis/genética , Adulto , Apoproteína(a)/química , Femenino , Fibrina/química , Fibrina/metabolismo , Predisposición Genética a la Enfermedad , Células HEK293 , Homocigoto , Humanos , Lipoproteína(a)/química , Masculino , Metionina , Persona de Mediana Edad , Simulación de Dinámica Molecular , Oxidación-Reducción , Fenotipo , Conformación Proteica , Proteínas Recombinantes/sangre , Relación Estructura-Actividad , Transfección
10.
Atherosclerosis ; 253: 47-53, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27575936

RESUMEN

BACKGROUND AND AIMS: Circulating lipoprotein (a) [Lp(a)] level relates inversely to apolipoprotein (a) [apo(a)] size. Both smaller apo(a) isoforms and higher Lp(a) levels have been linked to coronary heart disease and stroke, but their independent contributions are less well defined. We examined the role of Lp(a) in younger adults with cryptogenic stroke. METHODS: Lp(a) and apo(a) isoforms were evaluated in a prospectively designed case-control study of patients with unexplained ischemic stroke and stroke-free controls, ages 18 to 64. Serum Lp(a) was measured among 255 cases and 390 controls with both apo(a)-size independent and dependent assays. Apo(a) size was determined by agarose gel electrophoresis. RESULTS: Cases and controls were similar in socio-demographic characteristics, but cases had more hypertension, diabetes, smoking, and migraine with aura. In race-specific analyses, Lp(a) levels showed positive associations with cryptogenic stroke in whites, but not in the smaller subgroups of blacks and Hispanics. After full adjustment, comparison of the highest versus lowest quartile in whites was significant for apo(a)-size-independent (OR = 2.10 [95% CI = 1.04, 4.27], p = 0.040), and near-significant for apo(a)-size-dependent Lp(a) (OR = 1.81 [95% CI = 0.95, 3.47], p = 0.073). Apo(a) size was not associated with cryptogenic stroke in any race-ethnic subgroup. CONCLUSIONS: This study underscores the importance of Lp(a) level, but not apo(a) size, as an independent risk factor for unexplained ischemic stroke in young and middle-aged white adults. Given the emergence of effective Lp(a)-lowering therapies, these findings support routine testing for Lp(a) in this setting, along with further research to assess the extent to which such therapies improve outcomes in this population.


Asunto(s)
Apoproteína(a)/química , Enfermedad Coronaria/sangre , Lipoproteína(a)/sangre , Accidente Cerebrovascular/sangre , Adolescente , Adulto , Índice de Masa Corporal , Estudios de Casos y Controles , Enfermedad Coronaria/epidemiología , Etnicidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Factores de Riesgo , Accidente Cerebrovascular/epidemiología , Resultado del Tratamiento , Adulto Joven
11.
Rapid Commun Mass Spectrom ; 28(10): 1101-6, 2014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24711273

RESUMEN

RATIONALE: Apolipoprotein(a) is a polymorphic glycoprotein covalently bound to apoB100 in Lp(a) particles and has been described to be both atherogenic and prothrombotic, although its exact mechanism of action is not well defined. Apolipoprotein(a) is routinely measured by immunoassays. Unfortunately, the accuracy of the measurement can be affected by the apolipoprotein(a) size (number of kringles) polymorphism in Lp(a) particles. Here we describe an ultra-performance liquid chromatography/mass spectrometry (UPLC/MS) assay that is capable of measuring apolipoprotein(a) concentrations while simultaneously determining the number of kringles present per protein. METHODS: Plasma samples were diluted and proteins de-lipidated with deoxycholate prior to tryptic digestion. Distinct tryptic peptides from different regions of apolipoprotein(a) were measured to determine both concentration and the number of kringles present per protein. Separation and quantitation of tryptic peptides is carried out at 700 µL/min using a 1.7 µm C18 column (2.1 × 100 mm) coupled to a Thermo Vantage triple quadrupole (QQQ) mass spectrometer with a heated electrospray ionization (HESI) source. RESULTS: This method was compared to established methods for measuring concentration (monoclonal antibody based ELISA) and size (gel-electrophoresis) using 80 plasma samples proved by NWLRL. The slope and r(2) value for the correlation of concentrations were determined to be 0.96 and 0.98, demonstrating excellent agreement of absolute values between the UPLC/MS and ELISA methods. As measured by UPLC/MS, the average kringle number or size is smaller than determined by the electrophoretic method. CONCLUSIONS: A single UPLC/MS method was developed capable of measuring apolipoprotein(a) concentration and size (by measuring the number of kringles per protein). This assay passes criteria required for 'fit for purpose' assays including sensitivity, intra and interday reproducibility and freeze/thaw stability. While the agreement between UPLC/MS and ELISA is excellent for concentration and may provide researchers with additional tools for studying apolipoprotein(a), the dissimilarities between UPLC/MS and the electrophoretic method may also be exploited for understanding apolipoprotein(a) structure and function.


Asunto(s)
Apoproteína(a)/análisis , Apoproteína(a)/química , Cromatografía Líquida de Alta Presión/métodos , Espectrometría de Masas/métodos , Secuencia de Aminoácidos , Humanos , Datos de Secuencia Molecular , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/química , Estabilidad Proteica , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
12.
Cardiovasc Res ; 103(1): 28-36, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24760552

RESUMEN

AIMS: The relevance of lipoprotein(a) [Lp(a)] concentrations and low-molecular-weight (LMW) apo(a) phenotypes in peripheral arterial disease (PAD) has only been investigated by few studies. Therefore, we analysed this association in three independent cohorts and performed a Mendelian Randomization approach using instrumental variable regression. METHODS AND RESULTS: Lp(a) concentrations, apo(a) phenotypes, and one SNP in the LPA gene (rs10455872) were measured in the CAVASIC study, including 241 male patients with intermittent claudication and 246 age- and diabetes-matched controls as well as in the two population-based studies KORA F3 (n = 3184) and KORA F4 (n = 3080). In KORA F3/F4, 109/80 persons suffered from intermittent claudication, 200/144 from PAD, and 128/103 showed an ankle-brachial index (ABI) <0.9. In CAVASIC, adjusted logistic regression analyses revealed significant associations between an increase of log-Lp(a) per one standard deviation (SD) (OR = 1.28, P = 0.02) as well as LMW apo(a) phenotypes and symptomatic PAD (OR = 1.65, P = 0.03). Linear regression models with continuous ABI showed a significant association in the combined analyses of KORA F3/F4: an increase in log-Lp(a) per one SD (ß = -0.006, P = 0.005) and the presence of LMW apo(a) phenotypes (ß = -0.011, P = 0.02) or the minor allele of rs10455872 (ß = -0.016, P = 0.03) were associated with a decrease in ABI in the fully adjusted linear and instrumental variable regression models. CONCLUSION: Analyses in three independent populations showed significant associations of Lp(a) concentrations, LMW apo(a) phenotypes, and rs10455872 with PAD. This points to a causal relationship between Lp(a) and PAD since the genetically determined apo(a) phenotypes and SNP alleles are indeed associated with PAD.


Asunto(s)
Apoproteína(a)/sangre , Apoproteína(a)/genética , Lipoproteína(a)/sangre , Lipoproteína(a)/genética , Enfermedad Arterial Periférica/sangre , Enfermedad Arterial Periférica/genética , Adulto , Anciano , Índice Tobillo Braquial , Apoproteína(a)/química , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad , Humanos , Claudicación Intermitente/sangre , Claudicación Intermitente/genética , Claudicación Intermitente/fisiopatología , Modelos Lineales , Masculino , Persona de Mediana Edad , Peso Molecular , Enfermedad Arterial Periférica/fisiopatología , Fenotipo , Polimorfismo de Nucleótido Simple , Factores de Riesgo
13.
Cornea ; 33(4): 405-13, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24452210

RESUMEN

PURPOSE: The aim of this study was to evaluate the antiangiogenic activity of AU6, a novel 6-amino acid peptide derived from Kringle V of human apolipoprotein (a). METHODS: RF/6A rhesus macaque choroid endothelial cells were used for in vitro studies. MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt] assays and modified Boyden chamber and Matrigel assays were used to evaluate the inhibitory effect of AU6 on vascular endothelial growth factor (VEGF)-stimulated endothelial cell functions, including cell proliferation, migration, and tube formation. The chick chorioallantoic membrane model, micropocket corneal neovascularization (CNV) model, and alkali burn CNV model were evaluated in vivo. Bevacizumab (Avastin), the VEGF-neutralizing antibody, and a scrambled peptide (AU6s) were used as positive and negative controls, respectively. RESULTS: AU6 inhibited VEGF-induced RF/6A cell migration, proliferation, and tube formation. It also reduced pathological neovascularization in the chorioallantoic membrane model and in the 2 CNV models, that is, the mouse corneal micropocket model and the rat cornea alkali burn model. CONCLUSIONS: AU6 effectively inhibited pathogenic CNV. This novel peptide shows potential as a new treatment for ocular neovascularization.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Apoproteína(a)/farmacología , Neovascularización de la Córnea/prevención & control , Kringles , Oligopéptidos/farmacología , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Apoproteína(a)/química , Bevacizumab , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Membrana Corioalantoides/efectos de los fármacos , Coroides/irrigación sanguínea , Colágeno , Modelos Animales de Enfermedad , Combinación de Medicamentos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Laminina , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Oligopéptidos/química , Proteoglicanos , Ratas , Ratas Sprague-Dawley , Sales de Tetrazolio , Tiazoles , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/farmacología
14.
J Intern Med ; 276(3): 260-8, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24372611

RESUMEN

OBJECTIVES: Observational and genetic studies have shown that lipoprotein(a) [Lp(a)] levels and apolipoprotein(a) [apo(a)] isoform size are both associated with coronary heart disease (CHD) risk, but the relative independence of these risk factors remains unclear. Clarification of this uncertainty is relevant to the potential of future Lp(a)-lowering therapies for the prevention of CHD. METHODS: Plasma Lp(a) levels and apo(a) isoform size, estimated by the number of kringle IV (KIV) repeats, were measured in 995 patients with CHD and 998 control subjects. The associations between CHD risk and fifths of Lp(a) levels were assessed before and after adjustment for KIV repeats and, conversely, the associations between CHD risk and fifths of KIV repeats were assessed before and after adjustment for Lp(a) levels. RESULTS: Individuals in the top fifth of Lp(a) levels had more than a twofold higher risk of CHD compared with those in the bottom fifth, and this association was materially unaltered after adjustment for KIV repeats [odds ratio (OR) 2.05, 95% confidence interval (CI) 1.38-3.04, P < 0.001]. Furthermore, almost all of the excess risk was restricted to the two-fifths of the population with the highest Lp(a) levels. Individuals in the bottom fifth of KIV repeats had about a twofold higher risk of CHD compared with those in the top fifth, but this association was no longer significant after adjustment for Lp(a) levels (OR 1.13, 95% CI 0.77-1.66, P = 0.94). CONCLUSIONS: The effect of KIV repeats on CHD risk is mediated through their impact on Lp(a) levels, suggesting that absolute levels of Lp(a), rather than apo(a) isoform size, are the main determinant of CHD risk.


Asunto(s)
Enfermedad Coronaria/etiología , Lipoproteína(a)/metabolismo , Apoproteína(a)/química , Apoproteína(a)/metabolismo , Estudios de Casos y Controles , Enfermedad Coronaria/sangre , Femenino , Humanos , Lipoproteína(a)/química , Masculino , Persona de Mediana Edad , Isoformas de Proteínas/metabolismo , Factores de Riesgo
15.
Eur Heart J ; 34(42): 3268-76, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23735860

RESUMEN

Recently published epidemiological and genetic studies strongly suggest a causal relationship of elevated concentrations of lipoprotein (a) [Lp(a)] with cardiovascular disease (CVD), independent of low-density lipoproteins (LDLs), reduced high density lipoproteins (HDL), and other traditional CVD risk factors. The atherogenicity of Lp(a) at a molecular and cellular level is caused by interference with the fibrinolytic system, the affinity to secretory phospholipase A2, the interaction with extracellular matrix glycoproteins, and the binding to scavenger receptors on macrophages. Lipoprotein (a) plasma concentrations correlate significantly with the synthetic rate of apo(a) and recent studies demonstrate that apo(a) expression is inhibited by ligands for farnesoid X receptor. Numerous gaps in our knowledge on Lp(a) function, biosynthesis, and the site of catabolism still exist. Nevertheless, new classes of therapeutic agents that have a significant Lp(a)-lowering effect such as apoB antisense oligonucleotides, microsomal triglyceride transfer protein inhibitors, cholesterol ester transfer protein inhibitors, and PCSK-9 inhibitors are currently in trials. Consensus reports of scientific societies are still prudent in recommending the measurement of Lp(a) routinely for assessing CVD risk. This is mainly caused by the lack of definite intervention studies demonstrating that lowering Lp(a) reduces hard CVD endpoints, a lack of effective medications for lowering Lp(a), the highly variable Lp(a) concentrations among different ethnic groups and the challenges associated with Lp(a) measurement. Here, we present our view on when to measure Lp(a) and how to deal with elevated Lp(a) levels in moderate and high-risk individuals.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Lipoproteína(a)/metabolismo , Apoproteína(a)/química , Recolección de Muestras de Sangre/métodos , Diabetes Mellitus/fisiopatología , Hormonas/fisiología , Humanos , Hipolipemiantes/farmacología , Fallo Renal Crónico/fisiopatología , Lipoproteína(a)/química , Lipoproteína(a)/genética , Hepatopatías/fisiopatología , Guías de Práctica Clínica como Asunto , Valores de Referencia , Medición de Riesgo/métodos
16.
Lipids Health Dis ; 9: 103, 2010 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-20858228

RESUMEN

BACKGROUND: Oxidized lipoproteins play important roles in the atherosclerotic processes. Oxidized lipoprotein(a) (oxLp(a)) may be more potent in atherosclerotic pathophysiology than native Lp(a), a cardiovascular disease-relevant lipoprotein. Increased blood glucose concentrations can induce oxidative modification of lipoproteins. The aim of this study was to investigate the association between circulating oxLp(a) and cardiometabolic variables including blood glucose in healthy volunteers within the normal range of blood glucose. METHODS: Several cardiometabolic variables and serum oxLp(a) (using an ELISA system) were measured among 70 healthy females (mean age, 22 years). RESULTS: Lp(a) and glucose were significantly and positively correlated with oxLp(a) in simple correlation test. Furthermore, a multiple linear regression analysis showed oxLp(a) to have a weakly, but significantly positive and independent correlation with only blood glucose (ß = 0.269, P < 0.05). CONCLUSIONS: These results suggest that increased glucose may enhance the oxidization of Lp(a) even at normal glucose levels.


Asunto(s)
Aterosclerosis/epidemiología , Glucemia/análisis , Lipoproteína(a)/sangre , Lipoproteína(a)/química , Adolescente , Adulto , Apoproteína(a)/sangre , Apoproteína(a)/química , Aterosclerosis/diagnóstico , Presión Sanguínea , Índice de Masa Corporal , Enfermedades Cardiovasculares/epidemiología , Diagnóstico Precoz , Femenino , Humanos , Lípidos/sangre , Lipoproteínas/sangre , Oxidación-Reducción , Factores de Riesgo , Índice de Severidad de la Enfermedad , Estadística como Asunto , Adulto Joven
17.
Biochim Biophys Acta ; 1801(7): 738-45, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20346413

RESUMEN

Human apolipoprotein(a) (apo(a)), synthesized in the liver, contains oxidized phosphatidylcholine (oxPtdPC) adducts probably generated at the hepatic site. Since plasminogen (Plg), also synthesized in the liver, is genetically related and structurally homologous to apo(a), we wanted to determine whether it contains oxPtdPCs and their location. We used Plg isolated from fresh or frozen normal human plasma and several commercial preparations. Some were freed of non-covalently bound lipids by organic solvent extraction. By immunoblot analyses, all products reacted against T15, a natural IgM monoclonal antibody specific for phosphorylcholine -containing oxidized phospholipids (ox-PLs). This immunoreactivity was retained in urokinase type plasminogen activator -generated plasmin and was abrogated in Plg previously digested with lipoprotein-associated phospholipase A(2) (Lp-PLA(2)), a reaction that generated predominantly C16:0 lysophosphatidylcholine species as determined by mass spectrometry. Lyso derivatives were also generated upon the cleavage by Lp-PLA2 of a model ox-PL chemically linked to a lysine-containing pentapeptide. From inorganic phosphorous analyses, we found 2 mol of oxPtdPC/mole of Plg distributed between the kringles 1-4 and mini-Plg domain. OxPtdPCs were also present in the Plg isolated from the serum-free medium of cultured human HepG2 cells. In conclusion, our results provide strong evidence that naturally occurring Plg contains oxPtdPC probably linked by a Schiff base and also suggest that the linkage occurs at the hepatic site. Given the emerging evidence for the cardiovascular pathogenicity of oxPtdPCs, we speculate that they may impart athero-thrombogenic properties to Plg under inflammatory conditions.


Asunto(s)
Apoproteína(a)/química , Fosfatidilcolinas/química , Plasminógeno/química , 1-Alquil-2-acetilglicerofosfocolina Esterasa/química , Animales , Apoproteína(a)/metabolismo , Células Hep G2 , Humanos , Espectrometría de Masas , Ratones , Oxidación-Reducción , Fosfatidilcolinas/metabolismo , Plasminógeno/metabolismo
18.
JAMA ; 301(22): 2331-9, 2009 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-19509380

RESUMEN

CONTEXT: High levels of lipoprotein(a) are associated with increased risk of myocardial infarction (MI). OBJECTIVE: To assess whether genetic data are consistent with this association being causal. DESIGN, SETTING, AND PARTICIPANTS: Three studies of white individuals from Copenhagen, Denmark, were used: the Copenhagen City Heart Study (CCHS), a prospective general population study with 16 years of follow-up (1991-2007, n = 8637, 599 MI events); the Copenhagen General Population Study (CGPS), a cross-sectional general population study (2003-2006, n = 29 388, 994 MI events); and the Copenhagen Ischemic Heart Disease Study (CIHDS), a case-control study (1991-2004, n = 2461, 1231 MI events). MAIN OUTCOME MEASURES: Plasma lipoprotein(a) levels, lipoprotein(a) kringle IV type 2 (KIV-2) size polymorphism genotype, and MIs recorded from 1976 through July 2007 for all participants. RESULTS: In the CCHS, multivariable-adjusted hazard ratios (HRs) for MI for elevated lipoprotein(a) levels were 1.2 (95% confidence interval [CI], 0.9-1.6; events/10,000 person-years, 59) for levels between the 22nd and 66th percentile, 1.6 (95% CI, 1.1-2.2; events/10,000 person-years, 75) for the 67th to 89th percentile, 1.9 (95% CI, 1.2-3.0; events/10,000 person-years, 84) for the 90th to 95th percentile, and 2.6 (95% CI, 1.6-4.1; events/10,000 person-years, 108) for levels greater than the 95th percentile, respectively, vs levels less than the 22nd percentile (events/10,000 person-years, 55) (trend P < .001). Numbers of KIV-2 repeats (sum of repeats on both alleles) ranged from 6 to 99 and on analysis of variance explained 21% and 27% of all variation in plasma lipoprotein(a) levels in the CCHS and CGPS, respectively. Mean lipoprotein(a) levels were 56, 31, 20, and 15 mg/dL for the first, second, third, and fourth quartiles of KIV-2 repeats in the CCHS, respectively (trend P < .001); corresponding values in the CGPS were 60, 34, 22, and 19 mg/dL (trend P < .001). In the CCHS, multivariable-adjusted HRs for MI were 1.5 (95% CI, 1.2-1.9; events/10,000 person-years, 75), 1.3 (95% CI, 1.0-1.6; events/10,000 person-years, 66), and 1.1 (95% CI, 0.9-1.4; events/10,000 person-years, 57) for individuals in the first, second, and third quartiles, respectively, as compared with individuals in the fourth quartile of KIV-2 repeats (events/10,000 person-years, 51) (trend P < .001). Corresponding odds ratios were 1.3 (95% CI, 1.1-1.5), 1.1 (95% CI, 0.9-1.3), and 0.9 (95% CI, 0.8-1.1) in the CGPS (trend P = .005), and 1.4 (95% CI, 1.1-1.7), 1.2 (95% CI, 1.0-1.6), and 1.3 (95% CI, 1.0-1.6) in the CIHDS (trend P = .01). Genetically elevated lipoprotein(a) was associated with an HR of 1.22 (95% CI, 1.09-1.37) per doubling of lipoprotein(a) level on instrumental variable analysis, while the corresponding value for plasma lipoprotein(a) levels on Cox regression was 1.08 (95% CI, 1.03-1.12). CONCLUSION: These data are consistent with a causal association between elevated lipoprotein(a) levels and increased risk of MI.


Asunto(s)
Kringles/genética , Lipoproteína(a)/sangre , Lipoproteína(a)/genética , Infarto del Miocardio/sangre , Infarto del Miocardio/genética , Adulto , Anciano , Apoproteína(a)/sangre , Apoproteína(a)/química , Apoproteína(a)/genética , Dinamarca , Femenino , Genotipo , Humanos , Lipoproteína(a)/química , Masculino , Persona de Mediana Edad , Infarto del Miocardio/epidemiología , Polimorfismo Genético , Riesgo , Población Blanca/genética
19.
Int J Cancer ; 124(4): 843-52, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19035465

RESUMEN

Many angiogenesis inhibitors are derived from large plasma proteins. Previous studies showed that the Kringle5-like domain (termed KV) in human apolipoprotein (a) is a potential antiangiogenic factor. However, its active region and the underling molecular mechanism remain elusive. Here, we identified an 11-amino acid peptide (named KV11) as the key region for the antiangiogenic function of the KV domain of apolipoprotein (a). We demonstrate that KV11 inhibits angiogenesis in vitro by suppressing human umbilical vein endothelial cell migration and microtubule formation. KV11 inhibits angiogenesis in chicken chorioallantoic membrane assays and mouse corneal micropocket angiogenesis assays in vivo. KV11 peptide shows no effect on tumor cell growth or proliferation, but significantly inhibits tumor growth in SCID mouse xenograft tumor model (p < 0.01) by preventing tumor angiogenesis. We elucidate that KV11 peptide suppresses angiogenesis and tumor progression by targeting the c-Src/ERK signaling pathways. Together, these studies provide the first evidence that KV11 from apolipoprotein KV domain has anti-angiogenesis functions and may be an anti-tumor drug candidate.


Asunto(s)
Apolipoproteínas A/química , Apoproteína(a)/fisiología , Células Endoteliales/citología , Neoplasias/patología , Neovascularización Patológica , Fragmentos de Péptidos/química , Péptidos/química , Venas Umbilicales/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Familia-src Quinasas/metabolismo , Animales , Apolipoproteínas A/farmacología , Apoproteína(a)/química , Pollos , Membrana Corioalantoides/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Neoplasias/metabolismo , Fragmentos de Péptidos/farmacología , Fosforilación
20.
FASEB J ; 23(3): 950-6, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19001055

RESUMEN

Lipoprotein (a) [Lp(a)], a cardiovascular risk factor, is a low-density lipoprotein (LDL) variant shown to bind to oxidized phospholipids (oxPLs); however, its binding mode and origin have not been clearly established. We isolated both LDL and Lp(a) from the plasma of a population of high-Lp(a) subjects and in each Lp(a) particle separated apolipoprotein(a) [apo(a)], from the LDL component, Lp(a(-)). These products were assayed by an ELISA using monoclonal antibody T15 with a known specificity for oxPLs. In each subject, the T15 reactivity was confined to apo(a). Moreover, the amount of oxPL bound to apo(a) was unaffected by plasma Lp(a) levels and apo(a) size polymorphism. We have previously shown that kringle V (KV) is the site of oxPL linkage in human apo(a). In this work, we expressed in human embryonic kidney cells a KV-containing recombinant that, when purified from the medium, contained oxPLs. In summary, in human plasma Lp(a), the oxPLs are located in apo(a) and not in the circulating LDLs, suggesting a cellular origin. This latter concept is supported by the studies in which an expressed KV-containing apo(a) microdomain exhibited oxPL reactivity. Thus, apo(a) can undergo potentially pathogenic posttranslational modifications in a cellular environment able to generate oxPL.


Asunto(s)
Apoproteína(a)/metabolismo , Lipoproteínas LDL/metabolismo , Fosfolípidos/metabolismo , Adulto , Anciano , Apoproteína(a)/sangre , Apoproteína(a)/química , Femenino , Humanos , Kringles , Lipoproteínas LDL/química , Masculino , Persona de Mediana Edad , Fosfolípidos/sangre , Fosfolípidos/química , Procesamiento Proteico-Postraduccional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...