Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 39(5): 915-924, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30894011

RESUMEN

Objective- Aim of this study was to evaluate changes in LCAT (lecithin:cholesterol acyltransferase) concentration and activity in patients with an acute coronary syndrome, to investigate if these changes are related to the compromised capacity of HDL (high-density lipoprotein) to promote endothelial nitric oxide (NO) production, and to assess if rhLCAT (recombinant human LCAT) can rescue the defective vasoprotective HDL function. Approach and Results- Thirty ST-segment-elevation myocardial infarction (STEMI) patients were enrolled, and plasma was collected at hospital admission, 48 and 72 hours thereafter, at hospital discharge, and at 30-day follow-up. Plasma LCAT concentration and activity were measured and related to the capacity of HDL to promote NO production in cultured endothelial cells. In vitro studies were performed in which STEMI patients' plasma was added with rhLCAT and HDL vasoprotective activity assessed by measuring NO production in endothelial cells. The plasma concentration of the LCAT enzyme significantly decreases during STEMI with a parallel significant reduction in LCAT activity. HDL isolated from STEMI patients progressively lose the capacity to promote NO production by endothelial cells, and the reduction is related to decreased LCAT concentration. In vitro incubation of STEMI patients' plasma with rhLCAT restores HDL ability to promote endothelial NO production, possibly related to significant modification in HDL phospholipid classes. Conclusions- Impairment of cholesterol esterification may be a major factor in the HDL dysfunction observed during acute coronary syndrome. rhLCAT is able to restore HDL-mediated NO production in vitro, suggesting LCAT as potential therapeutic target for restoring HDL functionality in acute coronary syndrome.


Asunto(s)
Síndrome Coronario Agudo/sangre , Síndrome Coronario Agudo/fisiopatología , Lipoproteínas HDL/sangre , Fosfatidilcolina-Esterol O-Aciltransferasa/sangre , Infarto del Miocardio con Elevación del ST/sangre , Infarto del Miocardio con Elevación del ST/enzimología , Biomarcadores/sangre , Estudios de Cohortes , Femenino , Humanos , Masculino , Óxido Nítrico/metabolismo , Pronóstico , Infarto del Miocardio con Elevación del ST/diagnóstico por imagen , Sensibilidad y Especificidad , Esterol O-Aciltransferasa/sangre
2.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(9): 991-997, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29852278

RESUMEN

Proprotein convertase subtilisin/kexin 9 (PCSK9), a protein regulating the number of cell-surface LDL receptors (LDLR), circulates partially associated to plasma lipoproteins. How this interaction alters PCSK9 plasma levels is still unclear. In the present study, we took advantage of the availability of a large cohort of carriers of genetic HDL disorders to evaluate how HDL defects affect plasma PCSK9 levels and its distribution among lipoproteins. Plasma PCSK9 concentrations were determined by ELISA in carriers of mutations in LCAT, ABCA1, or APOAI genes, and lipoprotein distribution was analyzed by FPLC. Carriers of one or two mutations in the LCAT gene show plasma PCSK9 levels comparable to that of unaffected family controls (homozygotes, 159.4 ng/mL (124.9;243.3); heterozygotes, 180.3 ng/mL (127.6;251.5) and controls, 190.4 ng/mL (146.7;264.4); P for trend = 0.33). Measurement of PCSK9 in plasma of subjects carrying mutations in ABCA1 or APOAI genes confirmed normal values. When fractionated by FPLC, PCSK9 peaked in a region between LDL and HDL in control subjects. In carriers of all HDL defects, lipoprotein profile shows a strong reduction of HDL, but the distribution of PCSK9 was superimposable to that of controls. In conclusion, the present study demonstrates that in genetically determined low HDL states plasma PCSK9 concentrations and lipoprotein distribution are preserved, thus suggesting that HDL may not be involved in PCSK9 transport in plasma.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/sangre , Apolipoproteína A-I/sangre , Hipolipoproteinemias/sangre , Fosfatidilcolina-Esterol O-Aciltransferasa/sangre , Proproteína Convertasa 9/sangre , Transportador 1 de Casete de Unión a ATP/deficiencia , Transportador 1 de Casete de Unión a ATP/genética , Adulto , Anciano , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , Estudios de Casos y Controles , Femenino , Regulación de la Expresión Génica , Heterocigoto , Homocigoto , Humanos , Hipolipoproteinemias/genética , Hipolipoproteinemias/patología , Lipoproteínas HDL/sangre , Lipoproteínas HDL/genética , Lipoproteínas LDL/sangre , Lipoproteínas LDL/genética , Masculino , Persona de Mediana Edad , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Proproteína Convertasa 9/genética
3.
Circulation ; 138(10): 1000-1007, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-29748187

RESUMEN

BACKGROUND: Lecithin:cholesterol acyltransferase (LCAT) is the sole enzyme that esterifies cholesterol in plasma. Its role in the supposed protection from atherogenesis remains unclear because mutations in LCAT causing fish-eye disease (FED) or familial LCAT deficiency (FLD) have been reported to be associated with more or instead less carotid atherosclerosis, respectively. This discrepancy may be associated with the loss of cholesterol esterification on only apolipoprotein AI (FED) or on both apolipoprotein AI- and apolipoprotein B-containing lipoproteins (FLD), an aspect that has thus far not been investigated. METHODS: Seventy-four heterozygotes for LCAT mutations recruited from Italy and the Netherlands were assigned to FLD (n=33) or FED (n=41) groups and compared with 280 control subjects. Subclinical atherosclerosis was assessed with carotid intima-media thickness. RESULTS: Compared with control subjects, total cholesterol was lower by 16% (-32.9 mg/dL) and 7% (-14.9 mg/dL) and high-density lipoprotein cholesterol was lower by 29% (-16.7 mg/dL) and 36% (-20.7 mg/dL) in the FLD and FED groups, respectively. Subjects with FLD displayed a significant 18% lower low-density lipoprotein cholesterol compared with subjects with FED (101.9±35.0 versus 123.6±47.4 mg/dL; P=0.047) and control subjects (122.6±35.0 mg/dL; P=0.003). Remarkably, all 3 intima-media thickness parameters were lower in subjects with FLD compared with FED and control subjects (accounting for age, sex, body mass index, smoking, hypertension, family history of cardiovascular disease, and plasma lipids). After additional correction for nationality and ultrasonographic methods, average and maximum intima-media thickness remained significantly lower when subjects with FLD were compared with those with FED (0.59 versus 0.73 mm, P=0.003; and 0.87 versus 1.24 mm, P<0.001, respectively). In contrast, the common carotid intima-media thickness (corrected for age, sex, body mass index, smoking, hypertension, family history of cardiovascular disease, and plasma lipids) was higher in subjects with FED compared with control subjects (0.69 versus 0.65 mm; P=0.05), but this significance was lost after adjustment for nationality and ultrasonographic machine. CONCLUSIONS: In this head-to-head comparison, FLD and FED mutations were shown to be associated with decreased and increased atherosclerosis, respectively. We propose that this discrepancy is related to the capacity of LCAT to generate cholesterol esters on apolipoprotein B-containing lipoproteins. Although this capacity is lost in FLD, it is unaffected in FED. These results are important when considering LCAT as a target to decrease atherosclerosis.


Asunto(s)
Enfermedades de las Arterias Carótidas/etiología , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Mutación , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Adulto , Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Grosor Intima-Media Carotídeo , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Femenino , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Heterocigoto , Homocigoto , Humanos , Italia , Deficiencia de la Lecitina Colesterol Aciltransferasa/complicaciones , Deficiencia de la Lecitina Colesterol Aciltransferasa/diagnóstico , Deficiencia de la Lecitina Colesterol Aciltransferasa/enzimología , Masculino , Persona de Mediana Edad , Países Bajos , Fenotipo , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Medición de Riesgo , Factores de Riesgo
4.
Sci Rep ; 8(1): 2236, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29396407

RESUMEN

Recent evidence suggests that oxidative stress can play a role in the pathogenesis and the progression of prostate cancer (PCa). Reactive oxygen species (ROS) generation is higher in PCa cells compared to normal prostate epithelial cells and this increase is proportional to the aggressiveness of the phenotype. Since high density lipoproteins (HDL) are known to exert antioxidant activities, their ability to reduce ROS levels and the consequent impact on cell proliferation was tested in normal and PCa cell lines. HDL significantly reduced basal and H2O2-induced oxidative stress in normal, androgen receptor (AR)-positive and AR-null PCa cell lines. AR, scavenger receptor BI and ATP binding cassette G1 transporter were not involved. In addition, HDL completely blunted H2O2-induced increase of cell proliferation, through their capacity to prevent the H2O2-induced shift of cell cycle distribution from G0/G1 towards G2/M phase. Synthetic HDL, made of the two main components of plasma-derived HDL (apoA-I and phosphatidylcholine) and which are under clinical development as anti-atherosclerotic agents, retained the ability of HDL to inhibit ROS production in PCa cells. Collectively, HDL antioxidant activity limits cell proliferation induced by ROS in AR-positive and AR-null PCa cell lines, thus supporting a possible role of HDL against PCa progression.


Asunto(s)
Antioxidantes/farmacología , Apolipoproteína A-I/farmacología , Proliferación Celular/efectos de los fármacos , Lipoproteínas HDL/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosfatidilcolinas/farmacología , Neoplasias de la Próstata/patología , Antioxidantes/síntesis química , Apolipoproteína A-I/síntesis química , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Voluntarios Sanos , Humanos , Peróxido de Hidrógeno/metabolismo , Masculino , Células PC-3 , Fosfatidilcolinas/síntesis química , Interferencia de ARN , ARN Interferente Pequeño/genética
5.
J Lipid Res ; 58(5): 994-1001, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28351888

RESUMEN

The aim of this study was to evaluate the vasoprotective effects of HDL isolated from carriers of LCAT deficiency, which are characterized by a selective depletion of LpA-I:A-II particles and predominance of preß migrating HDL. HDLs were isolated from LCAT-deficient carriers and tested in vitro for their capacity to promote NO production and to inhibit vascular cell adhesion molecule-1 (VCAM-1) expression in cultured endothelial cells. HDLs from carriers were more effective than control HDLs in promoting eNOS activation with a gene-dose-dependent effect (PTrend = 0.048). As a consequence, NO production induced by HDL from carriers was significantly higher than that promoted by control HDL (1.63 ± 0.24-fold vs. 1.34 ± 0.07-fold, P = 0.031). HDLs from carriers were also more effective than control HDLs in inhibiting the expression of VCAM-1 (homozygotes, 65.0 ± 8.6%; heterozygotes, 53.1 ± 7.2%; controls, 44.4 ± 4.1%; PTrend = 0.0003). The increased efficiency of carrier HDL was likely due to the depletion in LpA-I:A-II particles. The in vitro findings might explain why carriers of LCAT deficiency showed flow-mediated vasodilation and plasma-soluble cell adhesion molecule concentrations comparable to controls, despite low HDL-cholesterol levels. These results indicate that selective depletion of apoA-II-containing HDL, as observed in carriers of LCAT deficiency, leads to an increased capacity of HDL to stimulate endothelial NO production, suggesting that changes in HDL apolipoprotein composition may be the target of therapeutic interventions designed to improve HDL functionality.


Asunto(s)
Apolipoproteína A-II/deficiencia , Apolipoproteína A-I/deficiencia , Células Endoteliales/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/patología , Lipoproteínas HDL/metabolismo , Adulto , Apolipoproteína A-I/metabolismo , Apolipoproteína A-II/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
6.
G Ital Nefrol ; 33(S68)2016.
Artículo en Italiano | MEDLINE | ID: mdl-27960011

RESUMEN

LCAT synthesizes most of the plasma cholesteryl esters, and plays a major role in HDL metabolism. Mutations in the LCAT gene cause two syndromes, familial LCAT deficiency (FLD) and fish-eye disease (FED), both characterized by severe alterations in plasma lipoprotein profile. Renal disease is the major cause of morbidity and mortality in FLD cases, but an established therapy is not currently available. The present therapy of LCAT deficiency is mainly aimed at correcting the dyslipidemia associated with the disease and at delaying evolution of chronic nephropathy. LCAT deficiency represents a candidate disease for enzyme replacement therapy. In vitro and in vivo studies proved the efficacy of recombinant human LCAT (rhLCAT) in correcting dyslipidemia, and rhLCAT is presently under development.


Asunto(s)
Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/terapia , Humanos , Deficiencia de la Lecitina Colesterol Aciltransferasa/diagnóstico
7.
Front Pharmacol ; 7: 2, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26834639

RESUMEN

Several lines of evidence suggest that, besides being a strong independent predictor of the occurrence of primary coronary events, a low plasma high density lipoprotein (HDL) cholesterol level is also associated with short- and long-term unfavorable prognosis in patients, who have recovered from a myocardial infarction, suggesting a direct detrimental effect of low HDL on post-ischemic myocardial function. Experiments performed in ex vivo and in vivo models of myocardial ischemia/reperfusion (I/R) injury have clearly shown that HDL are able to preserve cardiac function when given before ischemia or at reperfusion; the protective effects of HDL against I/R injury have been also confirmed in other tissues and organs, as brain and hind limb. HDL were shown to act on coronary endothelial cells, by limiting the increase of endothelium permeability and promoting vasodilation and neoangiogenesis, on white blood cells, by reducing their infiltration into the ischemic tissue and the release of pro-inflammatory and matrix-degrading molecules, and on cardiomyocytes, by preventing the activation of the apoptotic cascade. Synthetic HDL retains the cardioprotective activity of plasma-derived HDL and may become a useful adjunctive therapy to improve clinical outcomes in patients with acute coronary syndromes or undergoing coronary procedures.

8.
PLoS One ; 11(2): e0150083, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26919698

RESUMEN

Human familial lecithin:cholesterol acyltransferase (LCAT) deficiency (FLD) is characterized by low HDL, accumulation of an abnormal cholesterol-rich multilamellar particle called lipoprotein-X (LpX) in plasma, and renal disease. The aim of our study was to determine if LpX is nephrotoxic and to gain insight into the pathogenesis of FLD renal disease. We administered a synthetic LpX, nearly identical to endogenous LpX in its physical, chemical and biologic characteristics, to wild-type and Lcat-/- mice. Our in vitro and in vivo studies demonstrated an apoA-I and LCAT-dependent pathway for LpX conversion to HDL-like particles, which likely mediates normal plasma clearance of LpX. Plasma clearance of exogenous LpX was markedly delayed in Lcat-/- mice, which have low HDL, but only minimal amounts of endogenous LpX and do not spontaneously develop renal disease. Chronically administered exogenous LpX deposited in all renal glomerular cellular and matrical compartments of Lcat-/- mice, and induced proteinuria and nephrotoxic gene changes, as well as all of the hallmarks of FLD renal disease as assessed by histological, TEM, and SEM analyses. Extensive in vivo EM studies revealed LpX uptake by macropinocytosis into mouse glomerular endothelial cells, podocytes, and mesangial cells and delivery to lysosomes where it was degraded. Endocytosed LpX appeared to be degraded by both human podocyte and mesangial cell lysosomal PLA2 and induced podocyte secretion of pro-inflammatory IL-6 in vitro and renal Cxl10 expression in Lcat-/- mice. In conclusion, LpX is a nephrotoxic particle that in the absence of Lcat induces all of the histological and functional hallmarks of FLD and hence may serve as a biomarker for monitoring recombinant LCAT therapy. In addition, our studies suggest that LpX-induced loss of endothelial barrier function and release of cytokines by renal glomerular cells likely plays a role in the initiation and progression of FLD nephrosis.


Asunto(s)
Glomérulos Renales/efectos de los fármacos , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Lipoproteína X/toxicidad , Proteinuria/etiología , Animales , Apolipoproteína A-I/metabolismo , Células Cultivadas , Citoesqueleto/efectos de los fármacos , Citoesqueleto/ultraestructura , Células Endoteliales/metabolismo , Células Endoteliales/patología , Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica , Membrana Basal Glomerular/efectos de los fármacos , Membrana Basal Glomerular/patología , Mesangio Glomerular/citología , Mesangio Glomerular/metabolismo , Mesangio Glomerular/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-6/metabolismo , Glomérulos Renales/patología , Deficiencia de la Lecitina Colesterol Aciltransferasa/patología , Lipoproteína X/metabolismo , Lipoproteína X/farmacocinética , Lipoproteínas HDL/metabolismo , Lisosomas/metabolismo , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos C57BL , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Fosfolipasas A2/metabolismo , Pinocitosis , Podocitos/metabolismo , Podocitos/patología , Proteinuria/inducido químicamente , Proteinuria/genética , Proteinuria/patología
9.
J Atheroscler Thromb ; 23(2): 119-27, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26607351

RESUMEN

Lecithin:cholesterol acyltransferase (LCAT) is the only enzyme capable of esterifying cholesterol in plasma, thus determining the maturation of high-density lipoproteins. Because it maintains an unesterified cholesterol gradient between peripheral cells and extracellular acceptors, for a long time, LCAT has been considered as a key enzyme in reverse cholesterol transport. However, despite the fact that it has been more than 50 years since the identification of LCAT, the role of this enzyme in the pathogenesis of atherosclerosis is still debated. A number of studies have been conducted in different animal models, with contradictory results. Studies in humans, in particular in the general population, in subjects at high cardiovascular risk, and in carriers of genetic LCAT deficiency in an excellent model to evaluate the correlation between the reduction of LCAT activity and atherosclerosis also gave conflicting results. This review provides a comprehensive overview of the controversial findings obtained in animals and humans, strengthening the necessity of further investigation to establish how LCAT could be regulated in a promising therapeutic strategy to reduce cardiovascular risk.


Asunto(s)
Aterosclerosis/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/terapia , Humanos , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Factores de Riesgo
10.
Amyloid ; 22(3): 187-93, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26193960

RESUMEN

BACKGROUND: Among hereditary amyloidoses, apolipoprotein A-I (APO A-I) amyloidosis (Leu75Pro) is a rare, autosomal dominant condition in which renal, hepatic, and testicular involvement has been demonstrated. OBJECTIVE: To investigate vascular structural as well as functional alterations. METHODS: In 131 carriers of the amyloidogenic Leu75Pro APO A-I mutation (mean age 52 + 16 years, 56 women) and in 131 subjects matched for age, sex, body mass index and clinic blood pressure (BP), arterial stiffness (pulse wave velocity, PWV) and carotid intima-media thickness (IMT) were measured. RESULTS: By definition no differences for age, sex, body mass index, and BP were observed. Meanmax IMT (Mmax-IMT) in the common (CC), bifurcation (BIF) and internal (ICA) carotid artery were comparable in the two groups. After adjustment for high-density lipoprotein cholesterol and renal function differences between the two groups, a lower meanmax-IMT was observed in APO A-I Leu75Pro mutation carriers than in controls (CC Mmax-IMT 0.87 ± 0.21 versus 0.93 ± 0.2 mm, p = 0.07; BIF Mmax-IMT 1.19 ± 0.48 versus 1.36 ± 0.46 mm, p = 0.025; ICA Mmax-IMT 0.9 ± 0.37 versus 1.02 ± 0.35 mm, p = 0.028). On the other hand, aortic stiffness was significantly greater in patients with APO A-I amyloidosis than controls (PWV 11.5 ± 2.9 and 10.7 ± 2.3 m/s, p < 0.05), even after adjusting for confounders. CONCLUSIONS: In carriers of the amyloidogenic Leu75Pro APO A-I mutation, a significant increase in arterial stiffness is observed; on the contrary, carotid artery IMT is comparable to that of control subjects. These results may add significant information to the clinical features of this rare genetic disorder.


Asunto(s)
Amiloidosis/diagnóstico , Apolipoproteína A-I/genética , Arteria Carótida Común/patología , Mutación , Rigidez Vascular , Adulto , Anciano , Amiloidosis/diagnóstico por imagen , Amiloidosis/genética , Amiloidosis/patología , Presión Sanguínea , Índice de Masa Corporal , Arteria Carótida Común/diagnóstico por imagen , Arteria Carótida Común/metabolismo , Grosor Intima-Media Carotídeo/estadística & datos numéricos , Estudios de Casos y Controles , Femenino , Expresión Génica , Genes Dominantes , Humanos , Masculino , Persona de Mediana Edad , Análisis de la Onda del Pulso , Factores de Riesgo
11.
Vascul Pharmacol ; 74: 80-86, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26133666

RESUMEN

BACKGROUND: Fibrates and niacin are at present the most effective therapies to increase plasma levels of high density lipoprotein-cholesterol (HDL-C); to date, limited data are available on their effects on HDL protective functions. METHODS AND RESULTS: Within a multicenter, randomized, open-label, cross-over study, 37 patients with metabolic syndrome received 6weeks' treatment with fenofibrate or extended-release niacin (ER niacin), with a 4weeks' wash-out period. HDL ability to preserve endothelial cell homeostasis was assessed by incubating cultured endothelial cells with HDL isolated from patients at baseline and after each treatment. HDL isolated from patients at baseline were as effective as control HDL in inhibiting vascular cell adhesion molecule-1 (VCAM-1) expression, but less efficient in promoting endothelial cell nitric oxide (NO) release. Both fenofibrate and ER niacin increased HDL ability to inhibit TNFα-induced VCAM-1 expression (+7% and +11%, respectively). Fenofibrate and ER niacin also improved the impaired HDL ability to induce the expression of endothelial nitric oxide synthase and NO production (+10% and +8%, respectively). Interestingly, HDL isolated after treatment showed an ability to promote endothelial NO release similar to HDL isolated from controls. No differences were observed between the two drugs. With both drugs, HDL function was improved irrespective of baseline HDL-C levels. CONCLUSION: Treatment with fenofibrate or ER niacin in patients with metabolic syndrome not only increased HDL-C levels but also improved the endothelial protective effects of HDL.


Asunto(s)
HDL-Colesterol/sangre , Preparaciones de Acción Retardada/uso terapéutico , Células Endoteliales/efectos de los fármacos , Fenofibrato/uso terapéutico , Síndrome Metabólico/tratamiento farmacológico , Niacina/uso terapéutico , Sustancias Protectoras/uso terapéutico , Células Cultivadas , Estudios Cruzados , Células Endoteliales/metabolismo , Femenino , Humanos , Hipolipemiantes/uso terapéutico , Masculino , Síndrome Metabólico/sangre , Síndrome Metabólico/metabolismo , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
12.
Biochim Biophys Acta ; 1851(1): 13-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25068410

RESUMEN

Plasma high density lipoproteins (HDL) comprise a highly heterogeneous family of lipoprotein particles, differing in density, size, surface charge, and lipid and protein composition. Epidemiological studies have shown that plasma HDL level inversely correlates with atherosclerotic cardiovascular disease. The most relevant atheroprotective function of HDL is to promote the removal of cholesterol from macrophages within the arterial wall and deliver it to the liver for excretion in a process called reverse cholesterol transport. In addition, HDLs can contribute to the maintenance of endothelial cell homeostasis and have potent antioxidant properties. It has been long suggested that individual HDL subclasses may differ in terms of their functional properties, but which one is the good particle remains to be defined. Inherited HDL disorders are rare monogenic diseases due to mutations in genes encoding proteins involved in HDL metabolism. These disorders are not only characterized by extremely low or high plasma HDL levels but also by an abnormal HDL subclass distribution, and thus represent a unique tool to understand the relationship between plasma HDL concentration, HDL function, and HDL-mediated atheroprotection. This article is part of a Special Issue entitled Linking transcription to physiology in lipodomics.


Asunto(s)
Aterosclerosis/sangre , Aterosclerosis/metabolismo , Lipoproteínas HDL/sangre , Lipoproteínas HDL/metabolismo , Transporte Biológico , Células Endoteliales/metabolismo , Humanos
13.
Handb Exp Pharmacol ; 224: 593-615, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25523003

RESUMEN

The knowledge of an inverse relationship between plasma high-density lipoprotein cholesterol (HDL-C) concentrations and rates of cardiovascular disease has led to the concept that increasing plasma HDL-C levels would be protective against cardiovascular events. Therapeutic interventions presently available to correct the plasma lipid profile have not been designed to specifically act on HDL, but have modest to moderate effects on plasma HDL-C concentrations. Statins, the first-line lipid-lowering drug therapy in primary and secondary cardiovascular prevention, have quite modest effects on plasma HDL-C concentrations (2-10%). Fibrates, primarily used to reduce plasma triglyceride levels, also moderately increase HDL-C levels (5-15%). Niacin is the most potent available drug in increasing HDL-C levels (up to 30%), but its use is limited by side effects, especially flushing.The present chapter reviews the effects of established hypolipidemic drugs (statins, fibrates, and niacin) on plasma HDL-C levels and HDL subclass distribution, and on HDL functions, including cholesterol efflux capacity, endothelial protection, and antioxidant properties.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Hiperlipidemias/tratamiento farmacológico , Hipolipemiantes/uso terapéutico , Lipoproteínas HDL/sangre , Animales , Antioxidantes/metabolismo , Biomarcadores/sangre , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/etiología , HDL-Colesterol/sangre , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Hiperlipidemias/sangre , Hiperlipidemias/complicaciones , Lipoproteínas HDL/clasificación , Factores Protectores , Medición de Riesgo , Factores de Riesgo , Resultado del Tratamiento
14.
PLoS One ; 9(5): e95925, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24830642

RESUMEN

Mutations in the CETP gene resulting in defective CETP activity have been shown to cause remarkable elevations of plasma HDL-C levels, with the accumulation in plasma of large, buoyant HDL particles enriched in apolipoprotein E. Genetic CETP deficiency thus represents a unique tool to evaluate how structural alterations of HDL impact on HDL atheroprotective functions. Aim of the present study was to assess the ability of HDL obtained from CETP-deficient subjects to protect endothelial cells from the development of endothelial dysfunction. HDL isolated from one homozygous and seven heterozygous carriers of CETP null mutations were evaluated for their ability to down-regulate cytokine-induced cell adhesion molecule expression and to promote NO production in cultured endothelial cells. When compared at the same protein concentration, HDL and HDL3 from carriers proved to be as effective as control HDL and HDL3 in down-regulating cytokine-induced VCAM-1, while carrier HDL2 were more effective than control HDL2 in inhibiting VCAM-1 expression. On the other hand, HDL and HDL fractions from carriers of CETP deficiency were significantly less effective than control HDL and HDL fractions in stimulating NO production, due to a reduced eNOS activating capacity, likely because of a reduced S1P content. In conclusion, the present findings support the notion that genetic CETP deficiency, by affecting HDL particle structure, impacts on HDL vasculoprotective functions. Understanding of these effects might be important for predicting the outcomes of pharmacological CETP inhibition.


Asunto(s)
Proteínas de Transferencia de Ésteres de Colesterol/deficiencia , Errores Innatos del Metabolismo Lipídico/genética , Lipoproteínas HDL/química , Mutación , Óxido Nítrico Sintasa de Tipo III/genética , Proteínas de Transferencia de Ésteres de Colesterol/genética , Regulación hacia Abajo , Células Endoteliales/citología , Homocigoto , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación , Lipoproteínas/química , Óxido Nítrico/química , Molécula 1 de Adhesión Celular Vascular/metabolismo
15.
PLoS One ; 9(4): e95044, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24736652

RESUMEN

LCAT (lecithin:cholesterol acyltransferase) catalyzes the transacylation of a fatty acid of lecithin to cholesterol, generating a cholesteryl ester and lysolecithin. The knowledge of LCAT atomic structure and the identification of the amino acids relevant in controlling its structure and function are expected to be very helpful to understand the enzyme catalytic mechanism, as involved in HDL cholesterol metabolism. However - after an early report in the late '90 s - no recent advance has been made about LCAT three-dimensional structure. In this paper, we propose an LCAT atomistic model, built following the most up-to-date molecular modeling approaches, and exploiting newly solved crystallographic structures. LCAT shows the typical folding of the α/ß hydrolase superfamily, and its topology is characterized by a combination of α-helices covering a central 7-strand ß-sheet. LCAT presents a Ser/Asp/His catalytic triad with a peculiar geometry, which is shared with such other enzyme classes as lipases, proteases and esterases. Our proposed model was validated through different approaches. We evaluated the impact on LCAT structure of some point mutations close to the enzyme active site (Lys218Asn, Thr274Ala, Thr274Ile) and explained, at a molecular level, their phenotypic effects. Furthermore, we devised some LCAT modulators either designed through a de novo strategy or identified through a virtual high-throughput screening pipeline. The tested compounds were proven to be potent inhibitors of the enzyme activity.


Asunto(s)
Modelos Moleculares , Fosfatidilcolina-Esterol O-Aciltransferasa/química , Animales , Sitios de Unión , Catálisis , Activación Enzimática , Humanos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Mutación , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Conformación Proteica , Especificidad por Sustrato
16.
Biologicals ; 41(6): 446-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24140107

RESUMEN

Lecithin:cholesterol acyltransferase (LCAT) is the enzyme responsible for cholesterol esterification in plasma. Mutations in the LCAT gene leads to two rare disorders, familial LCAT deficiency and fish-eye disease, both characterized by severe hypoalphalipoproteinemia associated with several lipoprotein abnormalities. No specific treatment is presently available for genetic LCAT deficiency. In the present study, recombinant human LCAT was expressed and tested for its ability to correct the lipoprotein profile in LCAT deficient plasma. The results show that rhLCAT efficiently reduces the amount of unesterified cholesterol (-30%) and promotes the production of plasma cholesteryl esters (+210%) in LCAT deficient plasma. rhLCAT induces a marked increase in HDL-C levels (+89%) and induces the maturation of small preß-HDL into alpha-migrating particles. Moreover, the abnormal phospholipid-rich particles migrating in the LDL region were converted in normally sized LDL.


Asunto(s)
Deficiencia de la Lecitina Colesterol Aciltransferasa/sangre , Lipoproteínas/sangre , Fosfatidilcolina-Esterol O-Aciltransferasa/sangre , Proteínas Recombinantes/sangre , Western Blotting , Colesterol/sangre , Colesterol/metabolismo , Salud de la Familia , Células HEK293 , Humanos , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Lipoproteínas/metabolismo , Mutación , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Proteínas Recombinantes/metabolismo
17.
J Clin Lipidol ; 7(5): 414-22, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24079282

RESUMEN

BACKGROUND: The effectiveness of therapies that raise high-density lipoprotein cholesterol (HDL-C) to lower cardiovascular disease risk is currently under debate, and further research into the relationship between HDL-C and function is required. OBJECTIVE: o investigate whether 2 established HDL-C-raising therapies had differential effects on parameters of high-density lipoprotein (HDL) quality and function, such as HDL particle profile and cholesterol efflux capacity (CEC), in patients with dyslipidemia. METHODS AND RESULTS: Sixty-six patients with dyslipidemia, 24 with low HDL-C levels (<40 mg/dL) and 42 with normal HDL-C levels (40-59 mg/dL), were treated for 6 weeks with fenofibrate (160 mg/d) or extended-release (ER) niacin (0.5 g/d for 3 weeks, then 1 g/d) with 4 weeks of washout between treatments. Lipoprotein particle size distribution was determined using nuclear magnetic resonance, and pathway-specific serum CECs were assessed in J774 macrophages, hepatoma, and Chinese hamster ovary-human adenosine triphosphate-binding cassette transporter G1 cells. Comparable increases in HDL-C and apolipoprotein A-I levels were seen with fenofibrate and ER niacin. There was a shift toward larger HDL, predominantly to medium-size HDL particles for fenofibrate (+209%) and to large HDL particles for ER niacin (+221%). Minor changes in serum CECs were observed with fenofibrate and ER niacin for all the efflux pathways measured. Small increases in plasma cholesteryl ester transfer protein and lecithin: cholesterol acyltransferase concentrations, and decreases in cholesteryl ester transfer protein activity were seen with both drugs. CONCLUSIONS: Fenofibrate and ER niacin increased plasma HDL-C level similarly, but modulated HDL particle size distribution differently; however, these changes did not result in differential effects on serum CECs.


Asunto(s)
HDL-Colesterol/química , HDL-Colesterol/metabolismo , Dislipidemias/tratamiento farmacológico , Dislipidemias/metabolismo , Fenofibrato/farmacología , Niacina/administración & dosificación , Niacina/farmacología , Tamaño de la Partícula , Adolescente , Adulto , Anciano , Animales , Transporte Biológico/efectos de los fármacos , Células CHO , HDL-Colesterol/sangre , LDL-Colesterol/sangre , LDL-Colesterol/química , LDL-Colesterol/metabolismo , Cricetinae , Cricetulus , Dislipidemias/sangre , Dislipidemias/enzimología , Femenino , Fenofibrato/efectos adversos , Fenofibrato/uso terapéutico , Humanos , Hipolipemiantes/efectos adversos , Hipolipemiantes/farmacología , Hipolipemiantes/uso terapéutico , Masculino , Persona de Mediana Edad , Niacina/efectos adversos , Niacina/uso terapéutico , Fosfatidilcolina-Esterol O-Aciltransferasa/sangre , Seguridad , Triglicéridos/sangre , Adulto Joven
18.
Cardiovasc Res ; 100(1): 36-43, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23812298

RESUMEN

AIMS: The aim of the present study was to evaluate the high-density lipoprotein (HDL) structure and endothelial NO synthase (eNOS) activation capacity in ST-elevation myocardial infarction (STEMI) patients with different acute-phase inflammatory response (APR). METHODS AND RESULTS: Forty-five STEMI patients were stratified in quartiles according to the delta CRP level, calculated by subtracting the CRP value at admission from the CRP peak value (APR peak). The HDL structure and HDL capacity to stimulate NO production were evaluated at admission and at APR peak. STEMI patients with a low APR had a completely preserved HDL structure and HDL ability to activate eNOS and promote NO production, which did not change during STEMI. On the contrary, HDL from STEMI patients developing a significant APR had compromised ability to stimulate eNOS and promote NO production, and underwent a significant particle remodelling during STEMI. The defective capacity to stimulate NO production of HDL isolated from STEMI patients with high APR was explained, at least in part, by the reduced PON-1 and S1P content. The HDL ability to promote cell cholesterol efflux through different pathways was preserved in ACS patients independently of the inflammatory response. CONCLUSION: The present results extend previous studies reporting an impaired eNOS-activating capacity of HDL from ACS patients, showing that only a subset of patients undergoing STEMI, and in particular those developing an important inflammatory response, have circulating HDL defective in stimulating endothelial eNOS and NO production.


Asunto(s)
Síndrome Coronario Agudo/enzimología , Lipoproteínas HDL/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Anciano , Animales , Arildialquilfosfatasa/fisiología , Células Cultivadas , Colesterol/metabolismo , Activación Enzimática , Femenino , Humanos , Inflamación/enzimología , Lipoproteínas HDL/química , Masculino , Ratones , Persona de Mediana Edad , Óxido Nítrico/biosíntesis , Proteína Quinasa C beta/fisiología
19.
Biochem Pharmacol ; 85(4): 525-30, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23219857

RESUMEN

The administration of thrombolytic drugs is of proven benefit in a variety of clinical conditions requiring acute revascularization, including acute myocardial infarction (AMI), ischemic stroke, pulmonary embolism, and venous thrombosis. Generated plasmin can degrade non-target proteins, including apolipoprotein A-I (apoA-I), the major protein constituent of high-density lipoproteins (HDL). Aim of the present study was to compare the extent of apoA-I proteolytic degradation in AMI patients treated with two thrombolytic drugs, alteplase and the genetically engineered t-PA variant tenecteplase. ApoA-I degradation was evaluated in sera from 38 AMI patients treated with alteplase or tenecteplase. In vitro, apoA-I degradation was tested by incubating control sera or purified HDL with alteplase or tenecteplase at different concentrations (5-100 µg/ml). Treatment with alteplase and tenecteplase results in apoA-I proteolysis; the extent of apoA-I degradation was more pronounced in alteplase-treated patients than in tenecteplase-treated patients. In vitro, the extent of apoA-I proteolysis was higher in alteplase-treated sera than in tenecteplase-treated sera, in the whole drug concentration range. No direct effect of the two thrombolytic agents on apoA-I degradation was observed. In addition to apoA-I, apoA-IV was also degraded by the two thrombolytic agents and again proteolytic degradation was higher with alteplase than tenecteplase. In conclusion, this study indicates that both alteplase and tenecteplase cause plasmin-mediated proteolysis of apoA-I, with alteplase resulting in a greater apoA-I degradation than tenecteplase, potentially causing a transient impairment of HDL atheroprotective functions.


Asunto(s)
Apolipoproteína A-I/metabolismo , Fibrinolíticos/efectos adversos , Infarto del Miocardio/tratamiento farmacológico , Terapia Trombolítica/efectos adversos , Activador de Tejido Plasminógeno/efectos adversos , Anciano , Femenino , Fibrinolíticos/administración & dosificación , Fibrinolíticos/uso terapéutico , Humanos , Masculino , Persona de Mediana Edad , Tenecteplasa , Factores de Tiempo , Activador de Tejido Plasminógeno/administración & dosificación , Activador de Tejido Plasminógeno/uso terapéutico
20.
J Clin Lipidol ; 6(3): 244-50, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22658148

RESUMEN

BACKGROUND: Lecithin:cholesterol acyltransferase (LCAT) is responsible for cholesterol esterification in plasma. Mutations of LCAT gene cause familial LCAT deficiency, a metabolic disorder characterized by hypoalphalipoproteinemia. Apolipoprotein B (apoB) is the main protein component of very-low-density lipoproteins and low-density lipoprotein (LDL). Mutations of APOB gene cause familial hypobetalipoproteinemia, a codominant disorder characterized by low plasma levels of LDL cholesterol and apoB. OBJECTIVE: This was a genetic and biochemical analysis of an Italian kindred with hypobetalipoproteinemia whose proband presented with hypoalphalipoproteinemia and severe chronic kidney disease. METHODS: Plasma lipids and apolipoproteins, cholesterol esterification, and high-density lipoprotein (HDL) subclass distribution were analyzed. LCAT and APOB genes were sequenced. RESULTS: The proband had severe impairment of plasma cholesterol esterification and high preß-HDL content. He was heterozygote for the novel LCAT P406L variant, as were two other family members. The proband's wife and children presented with familial hypobetalipoproteinemia and were heterozygotes for the novel apoB H1401R variant. Cholesterol esterification rate of apoB H1401R carriers was reduced, likely attributable to the low amount of circulating LDL. After renal transplantation, proband's lipid profile, HDL subclass distribution, and plasma cholesterol esterification were almost at normal levels, suggesting a mild contribution of the LCAT P406L variant to his pretransplantation severe hypoalphalipoproteinemia and impairment of plasma cholesterol esterification. CONCLUSION: LCAT P406L variant had a mild effect on lipid profile, HDL subclass distribution, and plasma cholesterol esterification. ApoB H1401R variant was identified as possible cause of familial hypobetalipoproteinemia and resulted in a reduction of cholesterol esterification rate.


Asunto(s)
Apolipoproteínas B/genética , Hipobetalipoproteinemias/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Apolipoproteína A-I/sangre , HDL-Colesterol/sangre , Esterificación , Heterocigoto , Humanos , Hipobetalipoproteinemias/terapia , Italia , Trasplante de Riñón , Deficiencia de la Lecitina Colesterol Aciltransferasa/terapia , Masculino , Persona de Mediana Edad , Mutación Missense , Linaje , Análisis de Secuencia de ADN , Triglicéridos/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA