Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 156
Filtrar
1.
J Clin Lab Anal ; 38(5): e24998, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38444303

RESUMEN

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.


Asunto(s)
Variaciones en el Número de Copia de ADN , Kringles , Humanos , Kringles/genética , Variaciones en el Número de Copia de ADN/genética , Lipoproteína(a)/genética , Polimorfismo Genético , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos
2.
Arterioscler Thromb Vasc Biol ; 42(3): 289-304, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35045727

RESUMEN

BACKGROUND: Elevated plasma Lp(a) (lipoprotein(a)) levels are associated with increased risk for atherosclerotic cardiovascular disease and aortic valve stenosis. However, the cell biology of Lp(a) biosynthesis remains poorly understood, with the locations of the noncovalent and covalent steps of Lp(a) assembly unclear and the nature of the apoB-containing particle destined for Lp(a) unknown. We, therefore, asked if apo(a) and apoB interact noncovalently within hepatocytes and if this impacts Lp(a) biosynthesis. METHODS: Using human hepatocellular carcinoma cells expressing 17K (17 kringle) apo(a), or a 17KΔLBS7,8 variant with a reduced ability to bind noncovalently to apoB, we performed coimmunoprecipitation, coimmunofluorescence, and proximity ligation assays to document intracellular apo(a):apoB interactions. We used a pulse-chase metabolic labeling approach to measure apo(a) and apoB secretion rates. RESULTS: Noncovalent complexes containing apo(a)/apoB are present in lysates from cells expressing 17K but not 17KΔLBS7,8, whereas covalent apo(a)/apoB complexes are absent from lysates. 17K and apoB colocalized intracellularly, overlapping with staining for markers of endoplasmic reticulum trans-Golgi, and early endosomes, and less so with lysosomes. The 17KΔLBS7,8 had lower colocalization with apoB. Proximity ligation assays directly documented intracellular 17K/apoB interactions, which were dramatically reduced for 17KΔLBS7,8. Treatment of cells with PCSK9 (proprotein convertase subtilisin/kexin type 9) enhanced, and lomitapide reduced, apo(a) secretion in a manner dependent on the noncovalent interaction between apo(a) and apoB. Apo(a) secretion was also reduced by siRNA-mediated knockdown of APOB. CONCLUSIONS: Our findings explain the coupling of apo(a) and Lp(a)-apoB production observed in human metabolic studies using stable isotopes as well as the ability of agents that inhibit apoB biosynthesis to lower Lp(a) levels.


Asunto(s)
Apolipoproteína B-100/metabolismo , Apolipoproteínas A/metabolismo , Hepatocitos/metabolismo , Lipoproteína(a)/metabolismo , Apolipoproteína B-100/química , Apolipoproteínas A/química , Apolipoproteínas A/genética , Sitios de Unión/genética , Células Hep G2 , Humanos , Kringles/genética , Lipoproteína(a)/química , Lisina/química , Redes y Vías Metabólicas , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
3.
J Lipid Res ; 63(12): 100306, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36309064

RESUMEN

Lipoprotein(a) [Lp(a)] concentrations are regulated by the LPA gene mainly via the large kringle IV-type 2 (KIV-2) copy number variation and multiple causal variants. Early studies suggested an effect of long pentanucleotide repeat (PNR) alleles (10 and 11 repeats, PNR10 and PNR11) in the LPA promoter on gene transcription and found an association with lower Lp(a). Subsequent in vitro studies showed no effects on mRNA transcription, but the association with strongly decreased Lp(a) remained consistent. We investigated the isolated and combined effect of PNR10, PNR11, and the frequent splice site variant KIV-2 4925G>A on Lp(a) concentrations in the Cooperative Health Research in the Region of Augsburg F4 study by multiple quantile regression in single-SNP and joint models. Data on Lp(a), apolipoprotein(a) Western blot isoforms, and variant genotypes were available for 2,858 individuals. We found a considerable linkage disequilibrium between KIV-2 4925G>A and the alleles PNR10 and PNR11. In single-variant analysis adjusted for age, sex, and the shorter apo(a) isoform, we determined that both PNR alleles were associated with a highly significant Lp(a) decrease (PNR10: ß = -14.43 mg/dl, 95% CI: -15.84, -13.02, P = 3.33e-84; PNR11: ß = -17.21 mg/dl, 95% CI: -20.19, -14.23, P = 4.01e-29). However, a joint model, adjusting the PNR alleles additionally for 4925G>A, abolished the effect on Lp(a) (PNR10: ß = +0.44 mg/dl, 95% CI: -1.73, 2.60, P = 0.69; PNR11: ß = -1.52 mg/dl, 95% CI: -6.05, 3.00, P = 0.51). Collectively, we conclude that the previously reported Lp(a) decrease observed in pentanucleotide alleles PNR10 or PNR11 carriers results from a linkage disequilibrium with the frequent splicing mutation KIV-2 4925G>A.


Asunto(s)
Variaciones en el Número de Copia de ADN , Kringles , Humanos , Apoproteína(a)/genética , Kringles/genética , Apolipoproteínas A/genética , Lipoproteína(a)/genética , Repeticiones de Microsatélite
4.
J Lipid Res ; 60(1): 186-199, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30413653

RESUMEN

Lipoprotein (a) [Lp(a)] concentrations are among the strongest genetic risk factors for cardiovascular disease and present pronounced interethnic and interindividual differences. Approximately 90% of Lp(a) variance is controlled by the LPA gene, which contains a 5.6-kb-large copy number variation [kringle IV type 2 (KIV-2) repeat] that generates >40 protein isoforms. Variants within the KIV-2 region are not called in common sequencing projects, leaving up to 70% of the LPA coding region currently unaddressed. To completely assess the variability in LPA, we developed a sequencing strategy for this region and report here the first map of genetic variation in the KIV-2 region, a comprehensively evaluated ultradeep sequencing protocol, and an easy-to-use variant analysis pipeline. We sequenced 123 Central-European individuals and reanalyzed public data of 2,504 individuals from 26 populations. We found 14 different loss-of-function and splice-site mutations, as well as >100, partially even common, missense variants. Some coding variants were frequent in one population but absent in others. This provides novel candidates to explain the large ethnic and individual differences in Lp(a) concentrations. Importantly, our approach and pipeline are also applicable to other similar copy number variable regions, allowing access to regions that are not captured by common genome sequencing.


Asunto(s)
Variaciones en el Número de Copia de ADN , Genómica , Kringles/genética , Lipoproteína(a)/química , Lipoproteína(a)/genética , Polimorfismo de Nucleótido Simple , Humanos , Mutación
5.
Biochem Biophys Res Commun ; 508(1): 130-137, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30471863

RESUMEN

Current clinical treatments for ocular neovascularization are characterized by high possibility of damaging healthy tissues and high recurrence rates. It is necessary to develop new treatment methods to control neovascularization with a stable and effective effect. Kringle1 domain of hepatocyte growth factor (HGFK1) has anti-angiogenesis activity. Here, we established oxygen-induced retinopathy (OIR) model to study if using adeno-associated virus (AAV) as a delivery system to overexpression HGFK1 in retinal cells could benefit retinal neovascularization. We show that, overexpressed exogenous gene was mainly expressed in the inner and outer nuclear layer of the retina. Compared with control mice, the mice pretreated with rAAV-HGFK1 at P3 showed relatively normal vascular branches examined by fluorescence fundus angiography. Subsequent H&E staining and immunohistochemical staining of CD31 of the eye tissue sections showed that the mice received rAAV-HGFK1 had a relatively normal distribution of vascular endothelial cells. Additionally, immunohistochemical staining indicated a lower expression of VEGF in the eye tissues of rAAV-HGFK1 treated OIR mice. Further in vitro studies showed that HGFK1 could inhibit the proliferation but promote the apoptosis of bovine retinal microvascular endothelial cells (BRECs) under the presence of VEGF. Moreover, HGFK1 could inhibit VEGF induced ERK activation but promote p38 activation in BRECs. Therefore, we propose that intravitreal injection of rAAV-HGFK1 might be used to improve the retinal neovascularization and HGFK1 may function through regulating VEGF signaling pathway to inhibit neovascularization.


Asunto(s)
Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Neovascularización Retiniana/prevención & control , Animales , Apoptosis , Bovinos , Proliferación Celular , Células Cultivadas , Dependovirus/genética , Modelos Animales de Enfermedad , Terapia Genética/métodos , Factor de Crecimiento de Hepatocito/química , Humanos , Kringles/genética , Ratones , Ratones Endogámicos C57BL , Oxígeno/administración & dosificación , Retina/metabolismo , Neovascularización Retiniana/genética , Neovascularización Retiniana/metabolismo , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Transfección , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/administración & dosificación
6.
Lipids Health Dis ; 17(1): 111, 2018 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-29747697

RESUMEN

BACKGROUND: Lipoprotein (a) [Lp(a)], which is genetically determined by the LPA gene kringle IV type 2 (KIV-2) repeat copy number, has previously been reported in different populations. However, it is uncertain if the same occurs in the Chinese Han population. This study explored the correlation of Lp(a) mass or particle concentration with KIV-2 repeat copy number and application for coronary atherosclerotic heart disease (CAHD) risk assessment. METHODS: A cross-sectional study including 884 subjects was conducted. The Lp(a) level and routine risk factors of CAHD were compared. The KIV-2 copy number distribution, relationship with Lp(a), and assessment for CAHD risk were explored. RESULTS: The mean of Lp(a) mass or particle concentration in the CAHD group was higher than that in the non-CAHD group, while the KIV-2 copy number in the CAHD group was lower. Lp(a) had auxiliary values in gauging the type of plaque and was significantly higher in the soft-plaque group than that in the other two groups (200 mg/L [21.5 nmol/L], 166 mg/L [18.6 nmol/L], 149 mg/L [17.1 nmol/L], respectively, P < 0.05). Kappa test indicated divergence for the same individual using two Lp(a) concentrations (kappa value was 0.536 [< 0.75]). Elevated Lp(a) was an independent CAHD risk factor, whatever mass or particle concentration, and large KIV-2 copy number was a protective factor. CONCLUSION: Lp(a) level and small KIV-2 copy number are risk factors for CAHD in the Chinese Han population; furthermore, elevated Lp(a) may gauge the type of coronary plaque.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Enfermedad Coronaria/genética , Dosificación de Gen/genética , Lipoproteína(a)/sangre , Pueblo Asiatico , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/patología , Enfermedad Coronaria/sangre , Enfermedad Coronaria/patología , Estudios Transversales , Femenino , Genotipo , Humanos , Kringles/genética , Lipoproteína(a)/genética , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Secuencias Repetitivas de Aminoácido/genética , Factores de Riesgo
7.
Eur Heart J ; 38(23): 1823-1831, 2017 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-28444229

RESUMEN

AIMS: Lp(a) concentrations represent a major cardiovascular risk factor and are almost entirely controlled by one single locus (LPA). However, many genetic factors in LPA governing the enormous variance of Lp(a) levels are still unknown. Since up to 70% of the LPA coding sequence are located in a difficult to access hypervariable copy number variation named KIV-2, we hypothesized that it may contain novel functional variants with pronounced effects on Lp(a) concentrations. We performed a large scale mutation analysis in the KIV-2 using an extreme phenotype approach. METHODS AND RESULTS: We compiled an discovery set of 123 samples showing discordance between LPA isoform phenotype and Lp(a) concentrations and controls. Using ultra-deep sequencing, we identified a splice site variant (G4925A) in preferential association with the smaller LPA isoforms. Follow-up in a European general population (n = 2892) revealed an exceptionally high carrier frequency of 22.1% in the general population. The variant explains 20.6% of the Lp(a) variance in carriers of low molecular weight (LMW) apo(a) isoforms (P = 5.75e-38) and reduces Lp(a) concentrations by 31.3 mg/dL. Accordingly the odds ratio for cardiovascular disease was reduced from 1.39 [95% confidence interval (CI): 1.17-1.66, P = 1.89e-04] for wildtype LMW individuals to 1.19 [95%CI: 0.92; 1.56, P = 0.19] in LMW individuals who were additionally positive for G4925A. Functional studies point towards a reduction of splicing efficiency by this novel variant. CONCLUSION: A highly frequent but until now undetected variant in the LPA KIV-2 region is strongly associated with reduced Lp(a) concentrations and reduced cardiovascular risk in LMW individuals.


Asunto(s)
Enfermedades Cardiovasculares/genética , Kringles/genética , Lipoproteína(a)/genética , Adulto , Anciano , Variaciones en el Número de Copia de ADN/genética , Femenino , Genotipo , Humanos , Desequilibrio de Ligamiento/genética , Lipoproteína(a)/metabolismo , Masculino , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Riesgo
8.
Proc Natl Acad Sci U S A ; 111(21): 7630-5, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24821807

RESUMEN

The zymogen prothrombin is proteolytically converted by factor Xa to the active protease thrombin in a reaction that is accelerated >3,000-fold by cofactor Va. This physiologically important effect is paradigmatic of analogous cofactor-dependent reactions in the coagulation and complement cascades, but its structural determinants remain poorly understood. Prothrombin has three linkers connecting the N-terminal Gla domain to kringle-1 (Lnk1), the two kringles (Lnk2), and kringle-2 to the C-terminal protease domain (Lnk3). Recent developments indicate that the linkers, and particularly Lnk2, confer on the zymogen significant flexibility in solution and enable prothrombin to sample alternative conformations. The role of this flexibility in the context of prothrombin activation was tested with several deletions. Removal of Lnk2 in almost its entirety (ProTΔ146-167) drastically reduces the enhancement of thrombin generation by cofactor Va from >3,000-fold to 60-fold because of a significant increase in the rate of activation in the absence of cofactor. Deletion of Lnk2 mimics the action of cofactor Va and offers insights into how prothrombin is activated at the molecular level. The crystal structure of ProTΔ146-167 reveals a contorted architecture where the domains are not vertically stacked, kringle-1 comes within 9 Å of the protease domain, and the Gla-domain primed for membrane binding comes in contact with kringle-2. These findings broaden our molecular understanding of a key reaction of the blood coagulation cascade where cofactor Va enhances activation of prothrombin by factor Xa by compressing Lnk2 and morphing prothrombin into a conformation similar to the structure of ProTΔ146-167.


Asunto(s)
Factor Va/metabolismo , Factor Xa/metabolismo , Kringles/genética , Modelos Moleculares , Protrombina/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Cromatografía de Afinidad , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Clonación Molecular , Cricetinae , Cricetulus , Cristalografía por Rayos X , Electroforesis en Gel de Poliacrilamida , Humanos , Hidrólisis , Cinética , Espectrometría de Masas , Datos de Secuencia Molecular , Conformación Proteica , Protrombina/química , Análisis de Secuencia de Proteína
9.
J Lipid Res ; 57(7): 1111-25, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26637279

RESUMEN

Levels of lipoprotein (a) [Lp(a)], a complex between an LDL-like lipid moiety containing one copy of apoB, and apo(a), a plasminogen-derived carbohydrate-rich hydrophilic protein, are primarily genetically regulated. Although stable intra-individually, Lp(a) levels have a skewed distribution inter-individually and are strongly impacted by a size polymorphism of the LPA gene, resulting in a variable number of kringle IV (KIV) units, a key motif of apo(a). The variation in KIV units is a strong predictor of plasma Lp(a) levels resulting in stable plasma levels across the lifespan. Studies have demonstrated pronounced differences across ethnicities with regard to Lp(a) levels and some of this difference, but not all of it, can be explained by genetic variations across ethnic groups. Increasing evidence suggests that age, sex, and hormonal impact may have a modest modulatory influence on Lp(a) levels. Among clinical conditions, Lp(a) levels are reported to be affected by kidney and liver diseases.


Asunto(s)
Apolipoproteínas A/genética , Apolipoproteínas B/genética , Kringles/genética , Lipoproteína(a)/genética , Factores de Edad , Apolipoproteínas A/sangre , Apolipoproteínas B/sangre , Etnicidad/genética , Femenino , Variación Genética , Humanos , Lipoproteína(a)/sangre , Masculino , Polimorfismo de Nucleótido Simple , Factores de Riesgo , Caracteres Sexuales
10.
J Ind Microbiol Biotechnol ; 41(6): 989-96, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24682857

RESUMEN

Recently, as a new non-immunoglobulin-based protein scaffold, a human kringle domain was successfully engineered toward biologically functional agonists and antagonists. In this study, the fed-batch cultivation conditions were optimized for enhanced production of an Fc-fused kringle domain (KD548-Fc) in Pichia pastoris. Fed-batch cultivations were performed in 5-l laboratory-scale bioreactors, and in order to find the optimal conditions for high-level production of KD548-Fc, several parameters including the initial carbon source (glycerol) concentration, temperature, and pH were investigated. When cells were cultivated at pH 4.0 and 25 °C with 9.5 % glycerol in the initial medium, the highest production yield (635 mg/l) was achieved with high productivity (7.2 mg/l/h). Furthermore, functional KD548-Fc was successfully purified from the culture broth using a simple purification procedure with high purity and recovery yield.


Asunto(s)
Kringles/genética , Pichia/genética , Reactores Biológicos , Medios de Cultivo , Fermentación , Glicerol/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Fragmentos Fc de Inmunoglobulinas/genética , Pichia/crecimiento & desarrollo , Pichia/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/aislamiento & purificación , Temperatura
11.
Protein Expr Purif ; 88(1): 41-6, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23220455

RESUMEN

TAL6003 is an engineered form of human plasmin under development for thrombolytic therapy. TAL6003 (38.2kDa) contains nine disulfide bonds distributed within and between its two functional domains, a 25.2 kDa serine protease domain linked to a 13.0 kDa kringle I domain; kringles 2-5 present in native human plasmin have been deleted from this plasmin molecule. TAL6003 is expressed as its zymogen in Escherichia coli and is harvested in inclusion bodies. Following solubilization of inclusion bodies with urea as the chaotrope and glutathione as the reducing agent, this zymogen is refolded by dilution to a final concentration of 0.5mg/ml, with a yield of 48% (relative to total zymogen). Refolded TAL6003 zymogen is filtered, diafiltered, and filtered again prior to capture and purification on SP Sepharose, which is highly effective in removing host-cell protein. Subsequent affinity purification on ECH-Lysine Sepharose serves to capture polypeptide chains containing correctly refolded kringle 1 domain, which is the locus of the molecule's lysine-binding site, and to further eliminate host-cell protein. TAL6003 zymogen eluted from the ECH-Lysine Sepharose column is activated to TAL6003 with streptokinase, with an activity yield of approximately 80%. Proteolytically active TAL6003 is stripped of streptokinase by passage through an anion-exchange (Q) membrane and is then affinity purified on Benzamidine Sepharose, which serves to remove unreacted TAL6003 zymogen and proteolytically degraded TAL6003. An ultrafiltration/diafiltration step, to concentrate and to formulate TAL6003, completes the purification process. The final product exhibited a specific activity of close to unity and high purity by several relevant criteria.


Asunto(s)
Fibrinolisina/química , Fibrinolisina/aislamiento & purificación , Péptido Hidrolasas/química , Péptido Hidrolasas/aislamiento & purificación , Secuencia de Aminoácidos , Clonación Molecular , Escherichia coli , Fibrinolisina/biosíntesis , Expresión Génica , Humanos , Cuerpos de Inclusión/química , Cuerpos de Inclusión/metabolismo , Kringles/genética , Lisina/química , Lisina/genética , Péptido Hidrolasas/biosíntesis , Pliegue de Proteína
12.
J Mol Evol ; 74(5-6): 319-31, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22752046

RESUMEN

Protochordate genomes enable a prevalence of hemostasis evolution. Broad searches were performed for homologs of human serine proteases of hemostasis on the genomes of Branchiostoma floridae, Saccoglossus kowalevskii, and Strongylocentrotus purpuratus. Sequences were analyzed by multiple bioinformatic tools. The survey revealed numerous homologous components. Amphioxus was rich in some serine proteases not accompanied by gamma-carboxyglutamic or kringle domains similar more to thrombin than to other coagulation factors. The serine proteases found in amphioxus exhibited the attributes similar to those of thrombin by phylogeny relationships, sequence conservation, gene synteny, spatial structure, and ligand docking. A few plasminogen- and plasminogen activators-like proteases with kringles were also present. Those serine proteases demonstrated the greatest proximity rather to plasminogen or plasminogen activators than to thrombin. Searching for homologs of serine protease hemostatic factors in acorn worm and sea urchin revealed several components similar to those found in amphioxus. Hypothetically, the common ancestor of chordates had three separate serine proteases that evolved independently into immunoglobulin-like and kringle proteases in lancelets, and prothrombin, plasminogen activators, and plasminogen in vertebrates. Ancestral proteases evolved in vertebrates into hemostasis factors after merging the proper N-terminal domains and duplications.


Asunto(s)
Cordados/metabolismo , Equinodermos/genética , Evolución Molecular , Genoma/genética , Hemostasis/genética , Trombina/genética , Secuencia de Aminoácidos , Animales , Coagulación Sanguínea , Cromosomas Humanos/genética , Humanos , Kringles/genética , Ligandos , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Filogenia , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Sintenía/genética , Trombina/química
13.
N Engl J Med ; 361(26): 2518-28, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20032323

RESUMEN

BACKGROUND: An increased level of Lp(a) lipoprotein has been identified as a risk factor for coronary artery disease that is highly heritable. The genetic determinants of the Lp(a) lipoprotein level and their relevance for the risk of coronary disease are incompletely understood. METHODS: We used a novel gene chip containing 48,742 single-nucleotide polymorphisms (SNPs) in 2100 candidate genes to test for associations in 3145 case subjects with coronary disease and 3352 control subjects. Replication was tested in three independent populations involving 4846 additional case subjects with coronary disease and 4594 control subjects. RESULTS: Three chromosomal regions (6q26-27, 9p21, and 1p13) were strongly associated with the risk of coronary disease. The LPA locus on 6q26-27 encoding Lp(a) lipoprotein had the strongest association. We identified a common variant (rs10455872) at the LPA locus with an odds ratio for coronary disease of 1.70 (95% confidence interval [CI], 1.49 to 1.95) and another independent variant (rs3798220) with an odds ratio of 1.92 (95% CI, 1.48 to 2.49). Both variants were strongly associated with an increased level of Lp(a) lipoprotein, a reduced copy number in LPA (which determines the number of kringle IV-type 2 repeats), and a small Lp(a) lipoprotein size. Replication studies confirmed the effects of both variants on the Lp(a) lipoprotein level and the risk of coronary disease. A meta-analysis showed that with a genotype score involving both LPA SNPs, the odds ratios for coronary disease were 1.51 (95% CI, 1.38 to 1.66) for one variant and 2.57 (95% CI, 1.80 to 3.67) for two or more variants. After adjustment for the Lp(a) lipoprotein level, the association between the LPA genotype score and the risk of coronary disease was abolished. CONCLUSIONS: We identified two LPA variants that were strongly associated with both an increased level of Lp(a) lipoprotein and an increased risk of coronary disease. Our findings provide support for a causal role of Lp(a) lipoprotein in coronary disease.


Asunto(s)
Enfermedad Coronaria/genética , Predisposición Genética a la Enfermedad , Lipoproteína(a)/genética , Polimorfismo de Nucleótido Simple , Estudios de Casos y Controles , Enfermedad Coronaria/sangre , Marcadores Genéticos , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Kringles/genética , Funciones de Verosimilitud , Lipoproteína(a)/sangre , Lipoproteína(a)/química , Infarto del Miocardio/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Regresión , Factores de Riesgo
14.
Atherosclerosis ; 349: 17-35, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35606073

RESUMEN

High lipoprotein(a) [Lp(a)] concentrations are one of the most important genetically determined risk factors for cardiovascular disease. Lp(a) concentrations are an enigmatic trait largely controlled by one single gene (LPA) that contains a complex interplay of several genetic elements with many surprising effects discussed in this review. A hypervariable coding copy number variation (the kringle IV type-2 repeat, KIV-2) generates >40 apolipoprotein(a) protein isoforms and determines the median Lp(a) concentrations. Carriers of small isoforms with up to 22 kringle IV domains have median Lp(a) concentrations up to 5 times higher than those with large isoforms (>22 kringle IV domains). The effect of the apo(a) isoforms are, however, modified by many functional single nucleotide polymorphisms (SNPs) distributed over the complete range of allele frequencies (<0.1% to >20%) with very pronounced effects on Lp(a) concentrations. A complex interaction is present between the apo(a) isoforms and LPA SNPs, with isoforms partially masking the effect of functional SNPs and, vice versa, SNPs lowering the Lp(a) concentrations of affected isoforms. This picture is further complicated by SNP-SNP interactions, a poorly understood role of other polymorphisms such as short tandem repeats and linkage structures that are poorly captured by common R2 values. A further layer of complexity derives from recent findings that several functional SNPs are located in the KIV-2 repeat and are thus not accessible to conventional sequencing and genotyping technologies. A critical impact of the ancestry on correlation structures and baseline Lp(a) values becomes increasingly evident. This review provides a comprehensive overview on the complex genetic architecture of the Lp(a) concentrations in plasma, a field that has made tremendous progress with the introduction of new technologies. Understanding the genetics of Lp(a) might be a key to many mysteries of Lp(a) and booster new ideas on the metabolism of Lp(a) and possible interventional targets.


Asunto(s)
Kringles , Lipoproteína(a) , Apolipoproteínas A/genética , Apoproteína(a)/genética , Variaciones en el Número de Copia de ADN , Kringles/genética , Lipoproteína(a)/genética , Polimorfismo de Nucleótido Simple , Isoformas de Proteínas/genética
15.
Semin Thromb Hemost ; 37(7): 810-3, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22187404

RESUMEN

Increased levels of lipoprotein(a) (Lp[a])are known independent risk factor for atherosclerosis, heart disease, and stroke in adults. Even in children it could be shown that elevated levels of Lp(a) are an independent risk factor for symptomatic thromboembolism. The aim of this work was to describe the methods used for evaluating Lp(a) phenotypes, to link them to Lp(a) plasma concentrations, and to establish age-dependent reference values in children. Lp(a) plasma concentrations were measured with enzyme-linked immunosorbent assay technique in parallel to agarose gel electrophoresis and subsequent anti-apolipoprotein(a) immunoblotting. We included 184 pediatric patients with stroke or venous thromboembolism, and 150 healthy age-matched controls in this study. In the control children we could find a mean Lp(a) concentration of 3 mg/dL for children 1 to 12 months of age, and in subjects 1.2 to 18 years of age, the mean Lp(a) concentration was 10 mg/dL. Using percentile classification the upper percentile cut-offs were as follows: age 3 to 6 months: 14 mg/dL; 6.1 to 12 months: 15 mg/dL; 1.1 to 9 years: 22 mg/dL; and 9.1 to 18 years: 30 mg/dL, respectively. In the present study we have established age-dependent reference values of plasma Lp(a) concentrations. The latter will help to harmonize international pediatric studies and to further evaluate the role of elevated Lp(a) in childhood vascular disease.


Asunto(s)
Apolipoproteínas A/sangre , Lipoproteína(a)/sangre , Adolescente , Niño , Preescolar , Ensayo de Inmunoadsorción Enzimática , Cardiopatías/genética , Humanos , Lactante , Kringles/genética , Lipoproteína(a)/genética , Fenotipo , Valores de Referencia , Factores de Riesgo , Accidente Cerebrovascular/genética , Tromboembolia Venosa/genética , Población Blanca
16.
Biotechnol Lett ; 33(3): 503-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21120587

RESUMEN

An expression cassette containing kringle 2 and serine protease domains (K2S), tissue plasminogen activator (tPA), together with a signal sequence derived from Leishmania tarentolae and two fragments of the small subunit ribosomal RNA locus, was introduced into L. tarentolae. The transfected cells produced recombinant K2S (rK2S) protein extracellularly with serine protease activity. Expression and enzyme activity of rK2S in the supernatant was 930 i.u./ml. The specific activity of purified rK2S was 7.4 U/mg of protein. Replacement of the human signal sequence tPA with the signal sequence derived from Leishmania increased the secretion of recombinant protein up to 30 times.


Asunto(s)
Leishmania/metabolismo , Proteínas Recombinantes/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Humanos , Kringles/genética , Leishmania/genética , Modelos Genéticos , ARN Ribosómico/genética , Proteínas Recombinantes/genética , Serina Proteasas/genética , Serina Proteasas/metabolismo , Activador de Tejido Plasminógeno/genética
17.
Pharm Biol ; 49(6): 653-7, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21554008

RESUMEN

CONTEXT: The kringle 2 plus serine protease domains (K2S) of human tissue plasminogen activator (tPA) is an efficacious thrombolytic drug, which has been used to treat heart attacks and strokes by breaking up the clots that cause them. It has nine disulfide bridges, which are needed for proper folding and be the bottleneck in improving the production in the Escherichia coli system. So far, few reports have described the production of soluble active K2S from E. coli. OBJECTIVE: To achieve high-level expression of active K2S in the E. coli system. MATERIALS AND METHODS: The DNA fragment coding for K2S was fused with the E. coli disulfide isomerase DsbC. The constructed fusion protein was expressed in E. coli, and then purified with the Ni(2+)-chelating affinity chromatography. K2S was released by cleavage with Factor Xa protease, and the thrombolytic activity was determined using the fibrin plate assay. RESULTS: The fusion protein DsbC-K2S was found in the culture supernatant of recombinant E. coli as a soluble form of ~40%. The result of fibrinolysis fibrin plate assay showed that the purified recombinant K2S exhibited significant fibrinolysis activity in vitro. DISCUSSION AND CONCLUSION: These works provided a novel approach for the production of active K2S in E. coli without the requirements of in vitro refolding process, and might establish a significant foundation for the following production of K2S.


Asunto(s)
Fibrinólisis/efectos de los fármacos , Kringles/genética , Proteínas Recombinantes/biosíntesis , Activador de Tejido Plasminógeno/genética , Cromatografía de Afinidad , Escherichia coli/genética , Escherichia coli/metabolismo , Fibrinolíticos/farmacología , Humanos , Plásmidos/genética , Plásmidos/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Proteínas Recombinantes/farmacología
18.
J Am Coll Cardiol ; 78(5): 437-449, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34325833

RESUMEN

BACKGROUND: Lipoprotein(a) (Lp(a)) concentrations are a major independent risk factor for coronary artery disease (CAD) and are mainly determined by variation in LPA. Up to 70% of the LPA coding sequence is located in the hypervariable kringle IV type 2 (KIV-2) region. It is hardly accessible by conventional technologies, but may contain functional variants. OBJECTIVES: This study sought to investigate the new, very frequent splicing variant KIV-2 4733G>A on Lp(a) and CAD. METHODS: We genotyped 4733G>A in the GCKD (German Chronic Kidney Disease) study (n = 4,673) by allele-specific polymerase chain reaction, performed minigene assays, identified proxy single nucleotide polymorphisms and used them to characterize its effect on CAD by survival analysis in UK Biobank (n = 440,234). Frequencies in ethnic groups were assessed in the 1000 Genomes Project. RESULTS: The 4733G>A variant (38.2% carrier frequency) was found in most isoform sizes. It reduces allelic expression without abolishing protein production, lowers Lp(a) by 13.6 mg/dL (95% CI: 12.5-14.7; P < 0.0001) and is the strongest variance-explaining factor after the smaller isoform. Splicing of minigenes was modified. Compound heterozygosity (4.6% of the population) for 4733G>A and 4925G>A, another KIV-2 splicing mutation, reduces Lp(a) by 31.8 mg/dL and most importantly narrows the interquartile range by 9-fold (from 42.1 to 4.6 mg/dL) when compared to the wild type. In UK Biobank 4733G>A alone and compound heterozygosity with 4925G>A reduced HR for CAD by 9% (95% CI: 7%-11%) and 12% (95% CI: 7%-16%) (both P < 0.001). Frequencies in ethnicities differ notably. CONCLUSIONS: Functional variants in the previously inaccessible LPA KIV-2 region cooperate in determining Lp(a) variance and CAD risk. Even a moderate but lifelong genetic Lp(a) reduction translates to a noticeable CAD risk reduction.


Asunto(s)
Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/genética , Kringles/genética , Lipoproteína(a)/sangre , Lipoproteína(a)/genética , Variación Genética , Humanos , Lipoproteína(a)/fisiología , Estudios Prospectivos
19.
J Struct Biol ; 169(3): 349-59, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19800007

RESUMEN

The solution structure of the complex containing the isolated kringle 2 domain of human plasminogen (K2(Pg)) and VEK-30, a 30-amino acid residue internal peptide from a streptococcal M-like plasminogen (Pg) binding protein (PAM), has been determined by multinuclear high-resolution NMR. Complete backbone and side-chain assignments were obtained from triple-resonance experiments, after which structure calculations were performed and ultimately refined by restrained molecular simulation in water. We find that, in contrast with the dimer of complexes observed in the asymmetric unit of the crystal, global correlation times and buoyant molecular weight determinations of the complex and its individual components showed the monomeric nature of all species in solution. The NMR-derived structure of K2(Pg) in complex with VEK-30 presents a folding pattern typical of other kringle domains, while bound VEK-30 forms an end-to-end alpha-helix (residues 6-27) in the complex. Most of the VEK-30/K2(Pg) interactions in solution occur between a single face of the alpha-helix of VEK-30 and the lysine binding site (LBS) of K2(Pg). The canonical LBS of K2(Pg), consisting of Asp54, Asp56, Trp60, Arg69, and Trp70 (kringle numbering), interacts with an internal pseudo-lysine of VEK-30, comprising side-chains of Arg17, His18, and Glu20. Site-specific mutagenesis analysis confirmed that the electrostatic field formed by the N-terminal anionic residues of the VEK-30 alpha-helix, viz., Asp7, and the non-conserved cationic residues of K2(Pg), viz., Lys43 and Arg55, play additional important roles in the docking of VEK-30 to K2(Pg). Structural analysis and kringle sequence alignments revealed several important features related to exosite binding that provide a structural rationale for the high specificity and affinity of VEK-30 for K2(Pg).


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Plasminógeno/química , Plasminógeno/metabolismo , Proteínas Bacterianas/genética , Proteínas Portadoras/genética , Dicroismo Circular , Humanos , Kringles/genética , Kringles/fisiología , Espectroscopía de Resonancia Magnética , Mutagénesis Sitio-Dirigida , Plasminógeno/genética , Unión Proteica/genética , Unión Proteica/fisiología , Resonancia por Plasmón de Superficie , Ultracentrifugación
20.
Intern Emerg Med ; 15(7): 1239-1245, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31993950

RESUMEN

Increasing evidence shows an association between high lipoprotein(a) [Lp(a)] levels and atherothrombotic diseases. Lp(a) trait is largely controlled by kringle-IV type 2 (KIV-2) size polymorphism in LPA gene, encoding for apo(a). Environmental factors are considered to determinate minor phenotypic variability in Lp(a) levels. In the present study, we investigated the possible gene-environment interaction between KIV-2 polymorphism and Mediterranean diet adherence or fish weekly intake in determining Lp(a) levels. We evaluated Lp(a), KIV-2 polymorphism, fish intake and Mediterranean diet adherence in 452 subjects [median age (range) 66 (46-80)years] from Montignoso Heart and Lung Project (MEHLP) population. In subjects with high KIV-2 repeats number, influence of Mediterranean diet adherence in reducing Lp(a) levels was observed (p = 0.049). No significant difference in subjects with low KIV-2 repeats according to diet was found. Moreover, in high-KIV-2-repeat subjects, we observed a trend towards influence of fish intake on reducing Lp(a) levels (p = 0.186). At multivariate linear regression analysis, high adherence to Mediterranean diet remains a significant and independent determinant of lower Lp(a) levels (ß = - 64.97, standard error = 26.55, p = 0.015). In conclusion, this study showed that only subjects with high KIV-2 repeats can take advantage to lower Lp(a) levels from correct nutritional habits and, in particular, from Mediterranean diet.


Asunto(s)
Dieta Mediterránea , Interacción Gen-Ambiente , Lipoproteína(a)/genética , Lipoproteína(a)/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Femenino , Peces , Genotipo , Humanos , Italia , Kringles/genética , Masculino , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA