Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Lipid Res ; 64(1): 100316, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36410424

RESUMO

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.


Assuntos
Anticolesterolemiantes , Humanos , Coelhos , Animais , Anticolesterolemiantes/farmacologia , Anticolesterolemiantes/uso terapêutico , Colesterol/metabolismo , Apolipoproteínas E/metabolismo , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , HDL-Colesterol
2.
Curr Atheroscler Rep ; 23(9): 51, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34235598

RESUMO

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.


Assuntos
Estenose da Valva Aórtica , Remoção de Componentes Sanguíneos , Doenças Cardiovasculares , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/prevenção & controle , Humanos , Lipoproteína(a) , Pró-Proteína Convertase 9 , Fatores de Risco
3.
Circulation ; 138(16): 1677-1692, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-29674325

RESUMO

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.


Assuntos
Adenilil Ciclases/deficiência , Aorta/enzimologia , Doenças da Aorta/prevenção & controle , Aterosclerose/prevenção & controle , Proteínas de Transferência de Ésteres de Colesterol/deficiência , Placa Aterosclerótica , Adenilil Ciclases/genética , Adiposidade , Animais , Aorta/patologia , Aorta/fisiopatologia , Doenças da Aorta/enzimologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Aterosclerose/enzimologia , Aterosclerose/genética , Aterosclerose/patologia , Sistema Nervoso Autônomo/fisiopatologia , Fatores Biológicos/metabolismo , Proliferação de Células , Proteínas de Transferência de Ésteres de Colesterol/genética , Dieta Hiperlipídica , Modelos Animais de Doenças , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Lipídeos/sangue , Lipólise , Macrófagos/enzimologia , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Pró-Proteína Convertase 9/genética , Prostaglandina-Endoperóxido Sintases/metabolismo , Transdução de Sinais , Vasodilatação , Aumento de Peso
4.
J Lipid Res ; 58(7): 1282-1291, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28515138

RESUMO

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.


Assuntos
HDL-Colesterol/química , HDL-Colesterol/metabolismo , Oxazolidinonas/farmacologia , Compostos de Sulfidrila/farmacologia , Amidas , Animais , Apolipoproteína A-I/metabolismo , Transporte Biológico/efeitos dos fármacos , Chlorocebus aethiops , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Ésteres , Células Hep G2 , Humanos , Masculino , Coelhos , Especificidade da Espécie
5.
Arterioscler Thromb Vasc Biol ; 34(2): 457-62, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24334872

RESUMO

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.


Assuntos
Estenose da Valva Aórtica/genética , Lipoproteínas HDL/genética , Polimorfismo de Nucleotídeo Único , Idoso , Animais , Valva Aórtica/diagnóstico por imagem , Valva Aórtica/fisiopatologia , Estenose da Valva Aórtica/sangue , Estenose da Valva Aórtica/diagnóstico por imagem , Estenose da Valva Aórtica/fisiopatologia , Estudos de Casos e Controles , Distribuição de Qui-Quadrado , HDL-Colesterol/sangue , HDL-Colesterol/genética , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/genética , Feminino , Marcadores Genéticos , Predisposição Genética para Doença , Células Hep G2 , Hepatócitos/metabolismo , Humanos , Lipoproteínas HDL/sangue , Modelos Logísticos , Macrófagos/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Paris , Fenótipo , Estudos Prospectivos , Quebeque , Fatores de Risco , Ultrassonografia
7.
J Vasc Res ; 50(5): 430-41, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24080574

RESUMO

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.


Assuntos
Miócitos de Músculo Liso/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ácido Palmítico/farmacologia , Calcificação Vascular , Acetofenonas/farmacologia , Animais , Aorta/metabolismo , Calcinose/etiologia , Transdiferenciação Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Masculino , Músculo Liso Vascular/citologia , NADPH Oxidases/antagonistas & inibidores , Fosforilação , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Varfarina/farmacologia
8.
Biochim Biophys Acta ; 1801(1): 42-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19761869

RESUMO

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.


Assuntos
Apolipoproteína C-I/metabolismo , Ésteres do Colesterol/metabolismo , Lipoproteínas HDL/metabolismo , Lipoproteínas LDL/metabolismo , Lipoproteínas/metabolismo , Sítios de Ligação , Células Cultivadas , Humanos , Lipoproteínas/sangue , Lipoproteínas HDL/sangue , Lipoproteínas LDL/sangue
9.
Circ Genom Precis Med ; 14(2): e003219, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33794646

RESUMO

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.


Assuntos
Adenilil Ciclases/genética , Amidas/uso terapêutico , Doenças Cardiovasculares/prevenção & controle , Ésteres/uso terapêutico , Medicina de Precisão , Compostos de Sulfidrila/uso terapêutico , Adenilil Ciclases/metabolismo , Biomarcadores/metabolismo , Doenças Cardiovasculares/genética , Colesterol/metabolismo , Proteínas de Transferência de Ésteres de Colesterol/química , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Genótipo , Humanos , Farmacogenética
10.
Cell Biochem Funct ; 28(6): 480-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20629037

RESUMO

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.


Assuntos
Antígenos CD36/metabolismo , Caveolina 1/metabolismo , Colesterol/metabolismo , Hepatócitos/metabolismo , Receptores Depuradores Classe B/metabolismo , Animais , Antígenos CD36/genética , Caveolina 1/genética , Células Cultivadas , Feminino , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Knockout , Receptores Depuradores Classe B/genética
11.
J Cell Biochem ; 108(4): 906-15, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19718657

RESUMO

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.


Assuntos
Caveolina 1/biossíntese , Regulação da Expressão Gênica , Lipoproteínas LDL/metabolismo , Lipoproteínas/química , Fígado/citologia , Fígado/metabolismo , Receptores de LDL/metabolismo , Transporte Biológico , Antígenos CD36/biossíntese , Antígenos CD36/metabolismo , Linhagem Celular , Sobrevivência Celular , Células Hep G2 , Humanos , Modelos Biológicos
12.
Arterioscler Thromb Vasc Biol ; 28(7): 1290-5, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18436808

RESUMO

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.


Assuntos
Antígenos CD36/metabolismo , Hepatócitos/metabolismo , Lipoproteínas LDL/metabolismo , Animais , Antígenos CD36/genética , Células Cultivadas , Ésteres do Colesterol/metabolismo , Hepatócitos/imunologia , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica , Receptores Depuradores Classe B/genética , Receptores Depuradores Classe B/metabolismo
13.
Expert Opin Investig Drugs ; 28(12): 1059-1079, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31752565

RESUMO

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.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Hipertrigliceridemia/tratamento farmacológico , Hipolipemiantes/farmacologia , Animais , Doenças Cardiovasculares/etiologia , Desenvolvimento de Medicamentos , Drogas em Investigação/administração & dosagem , Drogas em Investigação/farmacologia , Humanos , Hipertrigliceridemia/complicações , Hipolipemiantes/administração & dosagem , Índice de Gravidade de Doença , Triglicerídeos/sangue
14.
J Cell Biochem ; 105(6): 1374-85, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18980242

RESUMO

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.


Assuntos
Lipoproteínas HDL3/metabolismo , Lipoproteínas LDL/metabolismo , Osteoblastos/metabolismo , Morte Celular/fisiologia , Células Cultivadas , Humanos , Lipoproteínas LDL/farmacologia
15.
Free Radic Biol Med ; 44(4): 506-17, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18241787

RESUMO

Cardiovascular diseases have recently been noted as potential risk factors for osteoporosis development. Although it is poorly understood how these two pathologies are related, it is a known fact that oxidized low-density lipoproteins (OxLDL) constitute potential determinants for both of them. The current study investigated the metabolism of OxLDL by osteoblasts and its effect on osteoblastic viability. The results obtained show that OxLDL are internalized but not degraded by osteoblasts while they can selectively transfer their CE to these cells. It is also demonstrated that OxLDL induce proliferation at low concentrations but cell death at high concentrations. This reduction of osteoblast viability was associated with lysosomal membrane damage caused by OxLDL as demonstrated by acridine orange relocalization. Accordingly, chloroquine, an inhibitor of lysosomal activity, accentuated cell death induced by OxLDL. Finally, we demonstrate that osteoblasts have the capacity to oxidize LDL and thereby potentially increase the local concentration of OxLDL. Overall, the current study confirms the potential role of OxLDL in the development of osteoporosis given its influence on osteoblastic viability.


Assuntos
Lipoproteínas LDL/toxicidade , Osteoblastos/efeitos dos fármacos , Osteoporose/etiologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacologia , Lisossomos/efeitos dos fármacos , Camundongos , Osteoblastos/metabolismo , Oxirredução , Sais de Tetrazólio/metabolismo , Tiazóis/metabolismo
16.
Clin Pharmacol Ther ; 104(2): 244-256, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29761474

RESUMO

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.


Assuntos
Apolipoproteínas/sangue , Doenças Cardiovasculares/sangue , Dislipidemias/sangue , Mediadores da Inflamação/sangue , Inflamação/sangue , Lipídeos/sangue , Animais , Biomarcadores/sangue , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/epidemiologia , Dislipidemias/diagnóstico , Dislipidemias/epidemiologia , Microbioma Gastrointestinal , Marcadores Genéticos , Humanos , Inflamação/diagnóstico , Inflamação/epidemiologia , Metilaminas/sangue , Valor Preditivo dos Testes , Prognóstico , Fatores de Risco
17.
Atherosclerosis ; 258: 8-19, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28167355

RESUMO

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.


Assuntos
Aorta Torácica/enzimologia , Doenças da Aorta/enzimologia , Apolipoproteína A-I/metabolismo , Aterosclerose/enzimologia , Carbocianinas/química , Corantes Fluorescentes/química , Imagem Molecular/métodos , Peptídeo Hidrolases/metabolismo , Animais , Aorta Torácica/patologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Apolipoproteína A-I/química , Apolipoproteína B-100/genética , Apolipoproteína B-100/metabolismo , Aterosclerose/genética , Aterosclerose/patologia , Catepsinas/metabolismo , Linhagem Celular , Quimases/metabolismo , Modelos Animais de Doenças , Humanos , Cinética , Macrófagos/enzimologia , Metaloproteinase 12 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estabilidade Proteica , Proteólise , Receptores de LDL/deficiência , Receptores de LDL/genética , Espectrometria de Fluorescência , Espectroscopia de Luz Próxima ao Infravermelho , Tripsina/metabolismo
18.
Int J Biochem Cell Biol ; 38(7): 1160-70, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16427800

RESUMO

In blood circulation, low density lipoproteins (LDL) can undergo modification, such as oxidation, and become key factors in the development of atherosclerosis. Although the liver is the major organ involved in the elimination of oxidized LDL (oxLDL), the identity of the receptor(s) involved remains to be defined. Our work aims to clarify the role of the scavenger receptor class B type I (SR-BI) in the hepatic metabolism of mildly and standardly oxLDL as well as the relative contribution of parenchymal (hepatocytes) and nonparenchymal liver cells with a special emphasis on CE-selective uptake. The association of native LDL and mildly or standardly oxLDL labeled either in proteins or in cholesteryl esters (CE) was measured on primary cultures of mouse hepatocytes from normal and SR-BI knock-out (KO) mice. These in vitro assays demonstrated that hepatocytes are able to mediate CE-selective uptake from both LDL and oxLDL and that SR-BI KO hepatocytes have a 60% reduced ability to selectively take CE from LDL but not towards mildly or standardly oxLDL. When lipoproteins were injected in the mouse inferior vena cava, parenchymal and nonparenchymal liver cells accumulated more CE than proteins from native, mildly and standardly oxLDL, indicating that selective uptake of CE from these lipoproteins occurs in vivo in these two cell types. The parenchymal cells contribute near 90% of the LDL-CE selective uptake and SR-BI for 60% of this pathway. Nonparenchymal cells capture mainly standardly oxLDL while parenchymal and nonparenchymal cells equally take up mildly oxLDL. An 82% reduction of standardly oxLDL-CE selective uptake by the nonparenchymal cells of SR-BI KO mice allowed emphasizing the contribution of SR-BI in hepatic metabolism of standardly oxLDL. However, SR-BI is not responsible for mildly oxLDL metabolism. Thus, SR-BI is involved in LDL- and standardly oxLDL-CE selective uptake in parenchymal and nonparenchymal cells, respectively.


Assuntos
Ésteres do Colesterol/metabolismo , Ésteres do Colesterol/farmacocinética , LDL-Colesterol/farmacocinética , Hepatócitos/metabolismo , Lipoproteínas LDL/farmacocinética , Receptores Depuradores Classe B/metabolismo , Animais , Lipoproteínas LDL/metabolismo , Camundongos , Camundongos Endogâmicos/metabolismo , Camundongos Knockout , Oxirredução
19.
Int J Cardiol ; 215: 364-71, 2016 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-27128563

RESUMO

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.


Assuntos
Estenose da Valva Aórtica/fisiopatologia , Colesterol/administração & dosagem , Hipercolesterolemia/fisiopatologia , Hipertrofia Ventricular Esquerda/tratamento farmacológico , Peptidomiméticos/administração & dosagem , Disfunção Ventricular Esquerda/tratamento farmacológico , Animais , Estenose da Valva Aórtica/induzido quimicamente , Apoptose/efeitos dos fármacos , Células Cultivadas , Colesterol/efeitos adversos , Modelos Animais de Doenças , Humanos , Hipercolesterolemia/induzido quimicamente , Hipertrofia Ventricular Esquerda/fisiopatologia , Lipoproteínas HDL/química , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Peptidomiméticos/farmacologia , Coelhos , Disfunção Ventricular Esquerda/fisiopatologia
20.
PLoS One ; 9(1): e85922, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24465790

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental/complicações , Produtos Finais de Glicação Avançada/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais , Calcificação Vascular/etiologia , Calcificação Vascular/metabolismo , Animais , Antioxidantes/farmacologia , Aorta/metabolismo , Aorta/patologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Artéria Femoral/efeitos dos fármacos , Artéria Femoral/patologia , Artéria Femoral/fisiopatologia , Hemodinâmica/efeitos dos fármacos , Masculino , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Oxirredução/efeitos dos fármacos , Compostos de Piridínio , Ratos , Ratos Wistar , Receptor para Produtos Finais de Glicação Avançada , Transdução de Sinais/efeitos dos fármacos , Calcificação Vascular/patologia , Calcificação Vascular/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA