Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Lipid Res ; 64(1): 100316, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36410424

RESUMEN

The large HDL particles generated by administration of cholesteryl ester transfer protein inhibitors (CETPi) remain poorly characterized, despite their potential importance in the routing of cholesterol to the liver for excretion, which is the last step of the reverse cholesterol transport. Thus, the effects of the CETPi dalcetrapib and anacetrapib on HDL particle composition were studied in rabbits and humans. The association of rabbit HDL to the LDL receptor (LDLr) in vitro was also evaluated. New Zealand White rabbits receiving atorvastatin were treated with dalcetrapib or anacetrapib. A subset of patients from the dal-PLAQUE-2 study treated with dalcetrapib or placebo were also studied. In rabbits, dalcetrapib and anacetrapib increased HDL-C by more than 58% (P < 0.01) and in turn raised large apo E-containing HDL by 66% (P < 0.001) and 59% (P < 0.01), respectively. Additionally, HDL from CETPi-treated rabbits competed with human LDL for binding to the LDLr on HepG2 cells more than control HDL (P < 0.01). In humans, dalcetrapib increased concentrations of large HDL particles (+69%, P < 0.001) and apo B-depleted plasma apo E (+24%, P < 0.001), leading to the formation of apo E-containing HDL (+47%, P < 0.001) devoid of apo A-I. Overall, in rabbits and humans, CETPi increased large apo E-containing HDL particle concentration, which can interact with hepatic LDLr. The catabolism of these particles may depend on an adequate level of LDLr to contribute to reverse cholesterol transport.


Asunto(s)
Anticolesterolemiantes , Humanos , Conejos , Animales , Anticolesterolemiantes/farmacología , Anticolesterolemiantes/uso terapéutico , Colesterol/metabolismo , Apolipoproteínas E/metabolismo , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , HDL-Colesterol
2.
Curr Atheroscler Rep ; 23(9): 51, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34235598

RESUMEN

PURPOSE OF REVIEW: The purpose of this article is to review current evidence for lipoprotein (a) (Lp(a)) as a risk factor for multiple cardiovascular (CV) disease phenotypes, provide a rationale for Lp(a) lowering to reduce CV risk, identify therapies that lower Lp(a) levels that are available clinically and under investigation, and discuss future directions. RECENT FINDINGS: Mendelian randomization and epidemiological studies have shown that elevated Lp(a) is an independent and causal risk factor for atherosclerosis and major CV events. Lp(a) is also associated with non-atherosclerotic endpoints such as venous thromboembolism and calcific aortic valve disease. It contributes to residual CV risk in patients receiving standard-of-care LDL-lowering therapy. Plasma Lp(a) levels present a skewed distribution towards higher values and vary widely between individuals and according to ethnic background due to genetic variants in the LPA gene, but remain relatively constant throughout a person's life. Thus, elevated Lp(a) (≥50 mg/dL) is a prevalent condition affecting >20% of the population but is still underdiagnosed. Treatment guidelines have begun to advocate measurement of Lp(a) to identify patients with very high levels that have a family history of premature CVD or elevated Lp(a). Lipoprotein apheresis (LA) efficiently lowers Lp(a) and was recently associated with a reduction of incident CV events. Statins have neutral or detrimental effects on Lp(a), while PCSK9 inhibitors significantly reduce its level by up to 30%. Specific lowering of Lp(a) with antisense oligonucleotides (ASO) shows good safety and strong efficacy with up to 90% reductions. The ongoing CV outcomes study Lp(a)HORIZON will provide a first answer as to whether selective Lp(a) lowering with ASO reduces the risk of major CV events. Given the recently established association between Lp(a) level and CV risk, guidelines now recommend Lp(a) measurement in specific clinical conditions. Accordingly, Lp(a) is a current target for drug development to reduce CV risk in patients with elevated levels, and lowering Lp(a) with ASO represents a promising avenue.


Asunto(s)
Estenosis de la Válvula Aórtica , Eliminación de Componentes Sanguíneos , Enfermedades Cardiovasculares , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/prevención & control , Humanos , Lipoproteína(a) , Proproteína Convertasa 9 , Factores de Riesgo
3.
Circ Genom Precis Med ; 14(2): e003219, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33794646

RESUMEN

Following the neutral results of the dal-OUTCOMES trial, a genome-wide study identified the rs1967309 variant in the adenylate cyclase type 9 (ADCY9) gene on chromosome 16 as being associated with the risk of future cardiovascular events only in subjects taking dalcetrapib, a CETP (cholesterol ester transfer protein) modulator. Homozygotes for the minor A allele (AA) were protected from recurrent cardiovascular events when treated with dalcetrapib, while homozygotes for the major G allele (GG) had increased risk. Here, we present the current state of knowledge regarding the impact of rs1967309 in ADCY9 on clinical observations and biomarkers in dalcetrapib trials and the effects of mouse ADCY9 gene inactivation on cardiovascular physiology. Finally, we present our current model of the interaction between dalcetrapib and ADCY9 gene variants in the arterial wall macrophage, based on the intracellular role of CETP in the transfer of complex lipids from endoplasmic reticulum membranes to lipid droplets. Briefly, the concept is that dalcetrapib would inhibit CETP-mediated transfer of cholesteryl esters, resulting in a progressive inhibition of cholesteryl ester synthesis and free cholesterol accumulation in the endoplasmic reticulum. Reduced ADCY9 activity, by paradoxically leading to higher cyclic AMP levels and in turn increased cellular cholesterol efflux, could impart cardiovascular protection in rs1967309 AA patients. The ongoing dal-GenE trial recruited 6145 patients with the protective AA genotype and will provide a definitive answer to whether dalcetrapib will be protective in this population.


Asunto(s)
Adenilil Ciclasas/genética , Amidas/uso terapéutico , Enfermedades Cardiovasculares/prevención & control , Ésteres/uso terapéutico , Medicina de Precisión , Compuestos de Sulfhidrilo/uso terapéutico , Adenilil Ciclasas/metabolismo , Biomarcadores/metabolismo , Enfermedades Cardiovasculares/genética , Colesterol/metabolismo , Proteínas de Transferencia de Ésteres de Colesterol/química , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Genotipo , Humanos , Farmacogenética
4.
Expert Opin Investig Drugs ; 28(12): 1059-1079, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31752565

RESUMEN

Introduction: Elevated triglyceride (TG) level is a prevalent condition in the general population and in patients with cardiovascular (CV) risk even under statin therapy. Severe hypertriglyceridemia (HTG) puts patients at risk for acute pancreatitis. Several TG-lowering drugs failed in clinical trials, but subgroup analyses suggest that high-risk patients, such as those with atherogenic dyslipidemia or diabetes, benefit from TG lowering.Areas covered: We review advances for TG-lowering drugs in clinical development. These include selective PPARα modulators, omega-3 fatty acid formulations that have been approved for severe HTG, and inhibitors of apolipoprotein C-III, angiopoietin-like-3 or microsomal transfer protein. Lessons learned from the success of the phase 3 trial REDUCE-IT with high-dose icosapent ethyl are also reviewed.Expert opinion: We believe that TG-lowering therapies are coming of age as they will allow to treat patients with high CV risk and moderate HTG, including T2D subjects, as well as patients with severe HTG or even homozygous familial hypercholesterolemia, all of which being 'optimally' treated with a statin. More studies on the impact of therapy on quality of life in patients with severe HTG should be conducted with the help of patient registries.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Hipertrigliceridemia/tratamiento farmacológico , Hipolipemiantes/farmacología , Animales , Enfermedades Cardiovasculares/etiología , Desarrollo de Medicamentos , Drogas en Investigación/administración & dosificación , Drogas en Investigación/farmacología , Humanos , Hipertrigliceridemia/complicaciones , Hipolipemiantes/administración & dosificación , Índice de Severidad de la Enfermedad , Triglicéridos/sangre
5.
Clin Pharmacol Ther ; 104(2): 244-256, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29761474

RESUMEN

Cardiovascular diseases (CVD) are the first cause of death in the world. CVD risk is influenced by multiple factors, some nonmodifiable such as age, sex, and genetic background, and others modifiable. Great progress has been made over the last decades in the identification of biomarkers of incident or recurrent CV risk and surrogate endpoints of CV outcomes. We present the current state of knowledge for CV biomarkers in plasma including lipids, apolipoproteins, inflammation-related, and emerging omics-based biomarkers. Clinically validated surrogate endpoints for CV outcomes include plasma low-density lipoprotein-cholesterol reduction, and plasma triglyceride reduction is a likely relevant surrogate endpoint. High-density lipoprotein-cholesterol is not a validated surrogate endpoint, but is a useful biomarker of CV risk. CV risk biomarkers of interest include apolipoprotein B and non-HDL-cholesterol, lipoprotein (a), C-reactive protein, and recently, genetic and protein-based risk scores and gut microbiota-derived trimethylamine oxide levels.


Asunto(s)
Apolipoproteínas/sangre , Enfermedades Cardiovasculares/sangre , Dislipidemias/sangre , Mediadores de Inflamación/sangre , Inflamación/sangre , Lípidos/sangre , Animales , Biomarcadores/sangre , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/epidemiología , Dislipidemias/diagnóstico , Dislipidemias/epidemiología , Microbioma Gastrointestinal , Marcadores Genéticos , Humanos , Inflamación/diagnóstico , Inflamación/epidemiología , Metilaminas/sangre , Valor Predictivo de las Pruebas , Pronóstico , Factores de Riesgo
6.
Circulation ; 138(16): 1677-1692, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-29674325

RESUMEN

BACKGROUND: Pharmacogenomic studies have shown that ADCY9 genotype determines the effects of the CETP (cholesteryl ester transfer protein) inhibitor dalcetrapib on cardiovascular events and atherosclerosis imaging. The underlying mechanisms responsible for the interactions between ADCY9 and CETP activity have not yet been determined. METHODS: Adcy9-inactivated ( Adcy9Gt/Gt) and wild-type (WT) mice, that were or not transgenic for the CETP gene (CETPtg Adcy9Gt/Gt and CETPtg Adcy9WT), were submitted to an atherogenic protocol (injection of an AAV8 [adeno-associated virus serotype 8] expressing a PCSK9 [proprotein convertase subtilisin/kexin type 9] gain-of-function variant and 0.75% cholesterol diet for 16 weeks). Atherosclerosis, vasorelaxation, telemetry, and adipose tissue magnetic resonance imaging were evaluated. RESULTS: Adcy9Gt/Gt mice had a 65% reduction in aortic atherosclerosis compared to WT ( P<0.01). CD68 (cluster of differentiation 68)-positive macrophage accumulation and proliferation in plaques were reduced in Adcy9Gt/Gt mice compared to WT animals ( P<0.05 for both). Femoral artery endothelial-dependent vasorelaxation was improved in Adcy9Gt/Gt mice (versus WT, P<0.01). Selective pharmacological blockade showed that the nitric oxide, cyclooxygenase, and endothelial-dependent hyperpolarization pathways were all responsible for the improvement of vasodilatation in Adcy9Gt/Gt ( P<0.01 for all). Aortic endothelium from Adcy9Gt/Gt mice allowed significantly less adhesion of splenocytes compared to WT ( P<0.05). Adcy9Gt/Gt mice gained more weight than WT with the atherogenic diet; this was associated with an increase in whole body adipose tissue volume ( P<0.01 for both). Feed efficiency was increased in Adcy9Gt/Gt compared to WT mice ( P<0.01), which was accompanied by prolonged cardiac RR interval ( P<0.05) and improved nocturnal heart rate variability ( P=0.0572). Adcy9 inactivation-induced effects on atherosclerosis, endothelial function, weight gain, adipose tissue volume, and feed efficiency were lost in CETPtg Adcy9Gt/Gt mice ( P>0.05 versus CETPtg Adcy9WT). CONCLUSIONS: Adcy9 inactivation protects against atherosclerosis, but only in the absence of CETP activity. This atheroprotection may be explained by decreased macrophage accumulation and proliferation in the arterial wall, and improved endothelial function and autonomic tone.


Asunto(s)
Adenilil Ciclasas/deficiencia , Aorta/enzimología , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Proteínas de Transferencia de Ésteres de Colesterol/deficiencia , Placa Aterosclerótica , Adenilil Ciclasas/genética , Adiposidad , Animales , Aorta/patología , Aorta/fisiopatología , Enfermedades de la Aorta/enzimología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/enzimología , Aterosclerosis/genética , Aterosclerosis/patología , Sistema Nervioso Autónomo/fisiopatología , Factores Biológicos/metabolismo , Proliferación Celular , Proteínas de Transferencia de Ésteres de Colesterol/genética , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Células Endoteliales/patología , Lípidos/sangre , Lipólisis , Macrófagos/enzimología , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , Proproteína Convertasa 9/genética , Prostaglandina-Endoperóxido Sintasas/metabolismo , Transducción de Señal , Vasodilatación , Aumento de Peso
8.
J Lipid Res ; 58(7): 1282-1291, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28515138

RESUMEN

Inhibition of cholesteryl ester transfer protein (CETP) increases HDL cholesterol (HDL-C) levels. However, the circulating CETP level varies and the impact of its inhibition in species with high CETP levels on HDL structure and function remains poorly characterized. This study investigated the effects of dalcetrapib and anacetrapib, the two CETP inhibitors (CETPis) currently being tested in large clinical outcome trials, on HDL particle subclass distribution and cholesterol efflux capacity of serum in rabbits and monkeys. New Zealand White rabbits and vervet monkeys received dalcetrapib and anacetrapib. In rabbits, CETPis increased HDL-C, raised small and large α-migrating HDL, and increased ABCA1-induced cholesterol efflux. In vervet monkeys, although anacetrapib produced similar results, dalcetrapib caused opposite effects because the LDL-C level was increased by 42% and HDL-C decreased by 48% (P < 0.01). The levels of α- and preß-HDL were reduced by 16% (P < 0.001) and 69% (P < 0.01), resulting in a decrease of the serum cholesterol efflux capacity. CETPis modulate the plasma levels of mature and small HDL in vivo and consequently the cholesterol efflux capacity. The opposite effects of dalcetrapib in different species indicate that its impact on HDL metabolism could vary greatly according to the metabolic environment.


Asunto(s)
HDL-Colesterol/química , HDL-Colesterol/metabolismo , Oxazolidinonas/farmacología , Compuestos de Sulfhidrilo/farmacología , Amidas , Animales , Apolipoproteína A-I/metabolismo , Transporte Biológico/efectos de los fármacos , Chlorocebus aethiops , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Ésteres , Células Hep G2 , Humanos , Masculino , Conejos , Especificidad de la Especie
9.
Atherosclerosis ; 258: 8-19, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28167355

RESUMEN

BACKGROUND AND AIMS: The potential benefits of high-density lipoproteins (HDL) against atherosclerosis are attributed to its major protein component, apolipoprotein A-I (apoA-I). Most of the apoA-I in the vascular wall appears to be in its lipid-poor form. The latter, however, is subjected to degradation by proteases localized in atherosclerotic plaques, which, in turn, has been shown to negatively impact its atheroprotective functions. Here, we report the development and in vivo use of a bioactivatable near-infrared full-length apoA-I-Cy5.5 fluorescent probe for the assessment of apoA-I-degrading proteolytic activities. METHODS: Fluorescence quenching was obtained by saturation of Cy5.5 fluorophore molecules on apoA-I protein. ApoA-I cleavage led to near-infrared fluorescence enhancement. In vitro proteolysis of the apoA-I probe by a variety of proteases including serine, cysteine, and metalloproteases resulted in an up to 11-fold increase in fluorescence (n = 5, p ≤ 0.05). RESULTS: We detected activation of the probe in atherosclerotic mice aorta sections using in situ zymography and showed that broad-spectrum protease inhibitors protected the probe from degradation, resulting in decreased fluorescence (-54%, n = 6 per group, p ≤ 0.0001). In vivo, the injected probe showed stronger fluorescence emission in the aorta of human apoB transgenic Ldlr-/- atherosclerotic mice (ATX) as compared to wild-type mice. In vivo observations were confirmed by ex vivo aorta imaging quantification where a 10-fold increase in fluorescent signal in ATX mice (p ≤ 0.05 vs. control mice) was observed. CONCLUSIONS: The use of this probe in different applications may help to assess new molecular mechanisms of atherosclerosis and may improve current HDL-based therapies by enhancing apoA-I functionality.


Asunto(s)
Aorta Torácica/enzimología , Enfermedades de la Aorta/enzimología , Apolipoproteína A-I/metabolismo , Aterosclerosis/enzimología , Carbocianinas/química , Colorantes Fluorescentes/química , Imagen Molecular/métodos , Péptido Hidrolasas/metabolismo , Animales , Aorta Torácica/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteína A-I/química , Apolipoproteína B-100/genética , Apolipoproteína B-100/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Catepsinas/metabolismo , Línea Celular , Quimasas/metabolismo , Modelos Animales de Enfermedad , Humanos , Cinética , Macrófagos/enzimología , Metaloproteinasa 12 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Estabilidad Proteica , Proteolisis , Receptores de LDL/deficiencia , Receptores de LDL/genética , Espectrometría de Fluorescencia , Espectroscopía Infrarroja Corta , Tripsina/metabolismo
10.
Int J Cardiol ; 215: 364-71, 2016 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-27128563

RESUMEN

OBJECTIVES: High-density lipoprotein (HDL) infusions induce rapid improvement of experimental atherosclerosis in rabbits but their effect on ventricular function remains unknown. We aimed to evaluate the effects of the HDL mimetic peptide CER-522 on left ventricular diastolic dysfunction (LVDD). METHODS: Rabbits were fed with a cholesterol- and vitamin D2-enriched diet until mild aortic valve stenosis and hypercholesterolemia-induced LV hypertrophy and LVDD developed. Animals then received saline or 10 or 30mg/kg CER-522 infusions 6 times over 2weeks. We performed serial echocardiograms and LV histology to evaluate the effects of CER-522 therapy on LVDD. RESULTS: LVDD was reduced by CER-522 as shown by multiple parameters including early filling mitral deceleration time, deceleration rate, Em/Am ratio, E/Em ratio, pulmonary venous velocities, and LVDD score. These findings were associated with reduced macrophages (RAM-11 positive cells) in the pericoronary area and LV, and decreased levels of apoptotic cardiomyocytes in CER-522-treated rabbits. CER-522 treatment also resulted in decreased atheromatous plaques and internal elastic lamina area in coronary arteries. CONCLUSIONS: CER-522 improves LVDD in rabbits, with reductions of LV macrophage accumulation, cardiomyocyte apoptosis, coronary atherosclerosis and remodelling.


Asunto(s)
Estenosis de la Válvula Aórtica/fisiopatología , Colesterol/administración & dosificación , Hipercolesterolemia/fisiopatología , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Peptidomiméticos/administración & dosificación , Disfunción Ventricular Izquierda/tratamiento farmacológico , Animales , Estenosis de la Válvula Aórtica/inducido químicamente , Apoptosis/efectos de los fármacos , Células Cultivadas , Colesterol/efectos adversos , Modelos Animales de Enfermedad , Humanos , Hipercolesterolemia/inducido químicamente , Hipertrofia Ventricular Izquierda/fisiopatología , Lipoproteínas HDL/química , Macrófagos/citología , Macrófagos/efectos de los fármacos , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Peptidomiméticos/farmacología , Conejos , Disfunción Ventricular Izquierda/fisiopatología
11.
Atherosclerosis ; 236(2): 277-85, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25108619

RESUMEN

OBJECTIVE: Excessive neointima formation often occurs after arterial injury. Interleukin-1ß (IL-1ß) is a potent pleiotropic cytokine that has been shown to regulate neointimal proliferation. We investigated the effects of the IL-1ß modulator gevokizumab in a rat carotid denudation model. METHODS: Sprague-Dawley rats were subjected to balloon denudation of the right carotid artery and were then randomized to receive a single subcutaneous infusion immediately after balloon injury of saline (control group, n = 13) or gevokizumab (gevokizumab groups, n = 15 in each group: 1, 10 and 50 mg/kg). We evaluated the treatment effects on carotid intima-media thickness (IMT) using ultrasonography, on endothelial regrowth using Evans Blue staining and on inflammatory response using histology. We also assessed the effects of IL-1ß and gevokizumab on human umbilical vein endothelial cells (HUVEC) and rat smooth muscle cells. RESULTS: We found that carotid IMT, in the proximal part of the denuded artery at day 28, was decreased by gevokizumab 1 mg/kg compared with controls. Neointima area and the intima/media area ratio were both reduced in the gevokizumab 1 mg/kg-treated group. Gevokizumab at the 1 mg/kg dose also improved endothelial regrowth. No effect was observed with gevokizumab 10 or 50 mg/kg. Gevokizumab also decreased the inflammatory effect of IL-1ß in in vitro cell experiments and protected HUVECs from IL-1ß's deleterious effects on cell migration, apoptosis and proliferation. CONCLUSION: A single administration of gevokizumab 1 mg/kg improves endothelial regrowth and reduces neointima formation in rats following carotid denudation, at least in part through its beneficial effects on endothelial cells.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Neointima/prevención & control , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/farmacología , Aorta/citología , Apoptosis/efectos de los fármacos , Traumatismos de las Arterias Carótidas/diagnóstico por imagen , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/diagnóstico por imagen , Arteria Carótida Común/patología , Grosor Intima-Media Carotídeo , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Endotelio Vascular/fisiopatología , Perfilación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-1beta/farmacología , Interleucina-1beta/fisiología , Masculino , Miocitos del Músculo Liso/efectos de los fármacos , Neointima/tratamiento farmacológico , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Regeneración , Vasculitis/tratamiento farmacológico , Vasculitis/prevención & control
12.
PLoS One ; 9(1): e85922, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24465790

RESUMEN

Advanced-glycation end products (AGEs) were recently implicated in vascular calcification, through a process mediated by RAGE (receptor for AGEs). Although a correlation between AGEs levels and vascular calcification was established, there is no evidence that reducing in vivo AGEs deposition or inhibiting AGEs-RAGE signaling pathways can decrease medial calcification. We evaluated the impact of inhibiting AGEs formation by pyridoxamine or elimination of AGEs by alagebrium on diabetic medial calcification. We also evaluated if the inhibition of AGEs-RAGE signaling pathways can prevent calcification. Rats were fed a high fat diet during 2 months before receiving a low dose of streptozotocin. Then, calcification was induced with warfarin. Pyridoxamine was administered at the beginning of warfarin treatment while alagebrium was administered 3 weeks after the beginning of warfarin treatment. Results demonstrate that AGEs inhibitors prevent the time-dependent accumulation of AGEs in femoral arteries of diabetic rats. This effect was accompanied by a reduced diabetes-accelerated calcification. Ex vivo experiments showed that N-methylpyridinium, an agonist of RAGE, induced calcification of diabetic femoral arteries, a process inhibited by antioxidants and different inhibitors of signaling pathways associated to RAGE activation. The physiological importance of oxidative stress was demonstrated by the reduction of femoral artery calcification in diabetic rats treated with apocynin, an inhibitor of reactive oxygen species production. We demonstrated that AGE inhibitors prevent or limit medial calcification. We also showed that diabetes-accelerated calcification is prevented by antioxidants. Thus, inhibiting the association of AGE-RAGE or the downstream signaling reduced medial calcification in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Productos Finales de Glicación Avanzada/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal , Calcificación Vascular/etiología , Calcificación Vascular/metabolismo , Animales , Antioxidantes/farmacología , Aorta/metabolismo , Aorta/patología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Arteria Femoral/efectos de los fármacos , Arteria Femoral/patología , Arteria Femoral/fisiopatología , Hemodinámica/efectos de los fármacos , Masculino , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Oxidación-Reducción/efectos de los fármacos , Compuestos de Piridinio , Ratas , Ratas Wistar , Receptor para Productos Finales de Glicación Avanzada , Transducción de Señal/efectos de los fármacos , Calcificación Vascular/patología , Calcificación Vascular/fisiopatología
13.
Arterioscler Thromb Vasc Biol ; 34(2): 457-62, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24334872

RESUMEN

OBJECTIVE: Studies have shown that high-density lipoprotein (HDL)-raising compounds induce regression of aortic valve stenosis (AVS) in animal models. However, whether patients with AVS have an impaired HDL metabolism is unknown. APPROACH AND RESULTS: A total of 1435 single nucleotide polymorphisms in genes associated with HDL cholesterol levels (in or around GALNT2, LPL, ABCA1, APOA5, SCARB1, LIPC, CETP, LCAT, LIPG, APOC4, and PLTP) were genotyped in 382 patients with echocardiography-confirmed AVS (aortic jet velocity ≥2.5 m/s) and 401 controls. After control for multiple testing, none of the genetic variants showed a positive association with case/control status (adjusted P≥0.05 for all single nucleotide polymorphisms tested). In a subsample of this cohort, HDL cholesterol levels, apolipoprotein AI levels, lecithin-cholesterol acyltransferase activity, pre-ß-HDL, HDL size, and 4 parameters of cholesterol efflux capacity were measured in apolipoprotein B-depleted serum samples from 86 patients with and 86 patients without AVS. Cholesterol efflux capacity was measured using J774 macrophages with and without stimulation of ATP-binding cassette A-1 expression by cAMP, and HepG2 hepatocytes for scavenger receptor class B type 1-mediated efflux. None of these parameters were different between cases and controls. However, compared with patients without coronary artery disease, sera from patients with coronary artery disease had lower HDL cholesterol levels, scavenger receptor class B type 1-mediated efflux, and HDL size (P≤0.003), independently of the presence or absence of AVS. CONCLUSIONS: Results of the present study suggest that, based on HDL genetics and HDL functionality, HDL metabolism does not seem to predict the risk of AVS. Because of our limited sample size, additional studies are needed to confirm these findings.


Asunto(s)
Estenosis de la Válvula Aórtica/genética , Lipoproteínas HDL/genética , Polimorfismo de Nucleótido Simple , Anciano , Animales , Válvula Aórtica/diagnóstico por imagen , Válvula Aórtica/fisiopatología , Estenosis de la Válvula Aórtica/sangre , Estenosis de la Válvula Aórtica/diagnóstico por imagen , Estenosis de la Válvula Aórtica/fisiopatología , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , HDL-Colesterol/sangre , HDL-Colesterol/genética , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/genética , Femenino , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Lipoproteínas HDL/sangre , Modelos Logísticos , Macrófagos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Paris , Fenotipo , Estudios Prospectivos , Quebec , Factores de Riesgo , Ultrasonografía
14.
J Vasc Res ; 50(5): 430-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24080574

RESUMEN

BACKGROUND: Aortic medial calcification is a cellular-regulated process leading to arterial stiffness. Although epidemiological studies have suggested an association between the saturation of fatty acids (FA) and arterial stiffness, there is no evidence that saturated FA can induce arterial calcification. This study investigated the capacity of palmitic acid (PA) to induce medial calcification and the signaling pathway(s) implicated in this process. METHODS: Rat aortic segments and vascular smooth muscle cells (VSMC) were exposed to calcification medium supplemented with PA. In vivo, rats were treated with warfarin to induce calcification and fed a PA-enriched diet. RESULTS: In vitro and ex vivo, palmitate increases calcification and ROS production. Palmitate increases extracellular-signal-regulated kinase (ERK1/2) phosphorylation and osteogenic gene expression. Inhibition of NADPH oxidase with apocynin or an siRNA prevents these effects. ERK1/2 inhibition attenuates the amplification of osteogenic gene expression and calcification induced by palmitate. In vivo, a PA-enriched diet amplified medial calcification and pulse wave velocity (PWV). These effects are mediated by ROS production as indicated by the inhibition of calcification and PWV normalization in rats concomitantly treated with apocynin. CONCLUSION: ROS induction by palmitate leads to ERK1/2 phosphorylation and subsequently induces the osteogenic differentiation of VSMC. © 2013 S. Karger AG, Basel.


Asunto(s)
Miocitos del Músculo Liso/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ácido Palmítico/farmacología , Calcificación Vascular , Acetofenonas/farmacología , Animales , Aorta/metabolismo , Calcinosis/etiología , Transdiferenciación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Músculo Liso Vascular/citología , NADPH Oxidasas/antagonistas & inhibidores , Fosforilación , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Warfarina/farmacología
15.
Future Cardiol ; 8(4): 513-31, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22871191

RESUMEN

CETP is the target of CETP inhibitors such as anacetrapib and the modulator dalcetrapib. Both molecules have entered Phase III clinical trials, with the ultimate goal of reducing cardiovascular events by raising HDL cholesterol. At the 600-mg dose selected for the dal-OUTCOMES study, dalcetrapib is expected to inhibit CETP activity by approximately 30% and raise HDL-C by approximately 30% with limited effects on LDL cholesterol. Importantly, dalcetrapib does not raise blood pressure or aldosterone levels, two effects previously associated with the CETP inhibitor torcetrapib. Dalcetrapib has been well tolerated at the 600-mg dose. In the dal-PLAQUE atherosclerosis imaging study, dalcetrapib reduced the enlargement of total vessel area over time. In May 2012, following the results of the second interim analysis of dal-OUTCOMES, the Data and Safety Monitoring Board recommended stopping the study owing to a lack of clinically significant benefit, which was followed by Roche's (Basel, Switzerland) decision to terminate the study and the dalcetrapib program (dal-HEART). Contrary to anacetrapib, a potent CETP inhibitor that markedly increases HDL cholesterol and significantly reduces LDL cholesterol, dalcetrapib has allowed us to test the hypothesis that an isolated, moderate elevation in HDL cholesterol prevents cardiovascular events.


Asunto(s)
Anticolesterolemiantes/farmacología , Proteínas de Transferencia de Ésteres de Colesterol/efectos de los fármacos , HDL-Colesterol/sangre , Compuestos de Sulfhidrilo/farmacología , Amidas , Animales , Anticolesterolemiantes/administración & dosificación , Anticolesterolemiantes/química , Anticolesterolemiantes/farmacocinética , Enfermedades Cardiovasculares/prevención & control , Ensayos Clínicos Fase II como Asunto , Quimioterapia Combinada , Ésteres , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Hipolipemiantes/farmacología , Hipolipemiantes/uso terapéutico , Síndrome Metabólico/sangre , Niacina/farmacología , Niacina/uso terapéutico , Oxazolidinonas/farmacología , Placa Aterosclerótica/prevención & control , Recurrencia , Compuestos de Sulfhidrilo/administración & dosificación , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/farmacocinética
16.
Cell Biochem Funct ; 28(6): 480-9, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20629037

RESUMEN

In non-hepatic cells, scavenger receptor class B type I (SR-BI), cluster of differentiation 36 (CD36), and caveolin-1 were described as mediators of cholesterol efflux, the first step of reverse cholesterol transport (RCT). Stable transformants of HepG2 cells overexpressing SR-BI, CD36, or caveolin-1 were generated, as well as cells overexpressing both caveolin-1 and SR-BI or caveolin-1 and CD36 in order to address the effect of caveolin-1 on both receptor activities. These cells were analyzed for their ability to efflux cholesterol to HDL(3). Our results show that overexpressing SR-BI, CD36, or caveolin-1 increases cholesterol efflux by 106, 92, and 48%, respectively. Moreover, the dual overexpressions of caveolin-1 and SR-BI or caveolin-1 and CD36 lead to a more prominent increase in cholesterol efflux. Studies were also conducted with primary cultures of SR-BI knockout (KO), CD36 KO, and SR-BI/CD36 double-KO (dKO) mice. SR-BI KO and SR-BI/CD36 dKO hepatic cells show 41 and 56% less cholesterol efflux, respectively, than normal hepatic cells. No significant difference was observed between the efflux of normal and CD36 KO cells. The difference between the role of human and murine CD36 correlated with the absence of CD36 dimers in mouse caveolae/rafts. Overall, our results show that SR-BI is clearly involved in cholesterol efflux in mouse and human hepatic cells, while CD36 plays a significant role in human cells.


Asunto(s)
Antígenos CD36/metabolismo , Caveolina 1/metabolismo , Colesterol/metabolismo , Hepatocitos/metabolismo , Receptores Depuradores de Clase B/metabolismo , Animales , Antígenos CD36/genética , Caveolina 1/genética , Células Cultivadas , Femenino , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Noqueados , Receptores Depuradores de Clase B/genética
17.
Biochim Biophys Acta ; 1801(1): 42-8, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19761869

RESUMEN

Plasma cholesterol from low- and high-density lipoproteins (LDL and HDL) are cleared from the circulation by specific receptors that either totally degrade lipoproteins as the LDL receptor or selectively take up their cholesteryl esters (CE) like the scavenger receptor class B type I (SR-BI). The aim of the present study was to define the effect of apoC-I on the uptake of LDL and HDL(3) by HepG2 cells. In experiments conducted with exogenously added purified apoC-I, no significant effect was observed on lipoprotein-protein association and degradation; however, LDL- and HDL(3)-CE selective uptake was significantly reduced in a dose-dependent manner. This study also shows that apoC-I has the ability to associate with HepG2 cells and with LDL and HDL(3). Moreover, pre-incubation of HepG2 cells with apoC-I reduces HDL(3)-CE selective uptake and pre-incubation of LDL and HDL(3) with apoC-I decreases their CE selective uptake by HepG2 cells. Thus, apoC-I can accomplish its inhibitory effect on SR-BI activity by either binding to SR-BI or lipoproteins. We conclude that by reducing hepatic lipoprotein-CE selective uptake, apoC-I has an atherogenic character.


Asunto(s)
Apolipoproteína C-I/metabolismo , Ésteres del Colesterol/metabolismo , Lipoproteínas HDL/metabolismo , Lipoproteínas LDL/metabolismo , Lipoproteínas/metabolismo , Sitios de Unión , Células Cultivadas , Humanos , Lipoproteínas/sangre , Lipoproteínas HDL/sangre , Lipoproteínas LDL/sangre
18.
J Cell Biochem ; 108(4): 906-15, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19718657

RESUMEN

Oxidized LDL (OxLDL) that are positively associated with the risk of developing cardiovascular diseases are ligands of scavenger receptor-class B type I (SR-BI) and cluster of differentiation-36 (CD36) which can be found in caveolae. The contribution of these receptors in human hepatic cell is however unknown. The HepG2 cell, a human hepatic parenchymal cell model, expresses these receptors and is characterized by a very low level of caveolin-1. Our aim was to define the contribution of human CD36, SR-BI, and caveolin-1 in the metabolism of OxLDL in HepG2 cells and conversely the effects of OxLDL on the levels/localization of these receptors. By comparing mildly (M)- and heavily (H)-OxLDL metabolism between control HepG2 cells and HepG2 cells overexpressing CD36, SR-BI, or caveolin-1, we found that (1) CD36 increases M- and H-OxLDL-protein uptake; (2) SR-BI drives M-OxLDL through a degradation pathway at the expense of the cholesterol ester (CE) selective uptake pathway; (3) caveolin-1 increases M- and H-OxLDL-protein uptake and decreases CE selective uptake from M-OxLDL. Also, incubation with M- or H-OxLDL decreases the levels of SR-BI and LDL-receptor in control HepG2 cells which can be overcome by caveolin-1 expression. In addition, OxLDL move CD36 from low to high buoyant density membrane fractions, as well as caveolin-1 in cells overexpressing this protein. Thus, hepatic caveolin-1 expression has significant effects on OxLDL metabolism and on lipoprotein receptor levels.


Asunto(s)
Caveolina 1/biosíntesis , Regulación de la Expresión Génica , Lipoproteínas LDL/metabolismo , Lipoproteínas/química , Hígado/citología , Hígado/metabolismo , Receptores de LDL/metabolismo , Transporte Biológico , Antígenos CD36/biosíntesis , Antígenos CD36/metabolismo , Línea Celular , Supervivencia Celular , Células Hep G2 , Humanos , Modelos Biológicos
19.
J Cell Biochem ; 105(6): 1374-85, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18980242

RESUMEN

Oxidized low density lipoproteins (OxLDL) are known to promote atherosclerosis, but it is only recently that OxLDL have been associated with alterations of the functions of bone-forming osteoblasts and osteoporosis. Although high density lipoproteins (HDL) are recognized for their anti-atherogenic action, there is less information about their ability to protect against osteoporosis. Therefore, we investigated the capacity of HDL3 to prevent the cell death induced by OxLDL in human osteoblastic cells. Simultaneous exposure of the cells to HDL3 and OxLDL abolished the reduction of cell viability monitored by MTT activity measurement and the induction of apoptosis determined by annexin V staining indicating that HDL3 prevent the apoptosis of osteoblasts induced by OxLDL. This protection correlated with the displacement by HDL3 of OxLDL association to osteoblasts, signifying that OxLDL binding and/or internalization are/is necessary for their cytotoxic effects. We also found that exposition of osteoblastic cells to HDL3 prior to incubation with OxLDL reduced cell death and preserved the lysosomal integrity. This protection was correlated with an increase of SR-BI expression, a modification of OxLDL metabolism with less global uptake of OxLDL and greater selective uptake of cholesterol from OxLDL. These results strongly suggest that, as for atherosclerosis, HDL may exert beneficial actions on bone metabolism.


Asunto(s)
Lipoproteínas HDL3/metabolismo , Lipoproteínas LDL/metabolismo , Osteoblastos/metabolismo , Muerte Celular/fisiología , Células Cultivadas , Humanos , Lipoproteínas LDL/farmacología
20.
Arterioscler Thromb Vasc Biol ; 28(7): 1290-5, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18436808

RESUMEN

OBJECTIVE: The cluster of differentiation-36 (CD36) is a multifunctional protein which is recognized for its in vitro ability to take up oxidized low-density lipoproteins (oxLDL) in macrophages and is therefore considered atherogenic. It also binds LDL. Our objective was to define the physiological role of CD36 in both native LDL and oxLDL metabolism in mice. METHODS AND RESULTS: Clearance studies of labeled LDL and oxLDL were conducted in wild-type, CD36 knockout (KO), scavenger receptor class B, type I (SR-BI) KO, and SR-BI/CD36 double KO mice. We found that CD36 impedes the disappearance of native LDL and favors that of oxLDL. This was confirmed by association and degradation assays with primary cultures of hepatic cells from wild-type and CD36 KO mice. In addition, our in vivo work indicates that neither SR-BI nor CD36 plays a significant role in cholesteryl esters (CE) selective uptake (SU) from oxLDL, whereas CD36, in absence of SR-BI, can selectively take CE from LDL. CONCLUSIONS: Our investigation showed for the first time that CD36 plays a significant role in oxLDL uptake in vivo in the mouse. As CD36 also retards LDL clearance, its atherogenic character may also relate to its negative effect on LDL catabolism.


Asunto(s)
Antígenos CD36/metabolismo , Hepatocitos/metabolismo , Lipoproteínas LDL/metabolismo , Animales , Antígenos CD36/genética , Células Cultivadas , Ésteres del Colesterol/metabolismo , Hepatocitos/inmunología , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Receptores Depuradores de Clase B/genética , Receptores Depuradores de Clase B/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...