Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
JHEP Rep ; 6(1): 100902, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38074507

RESUMEN

Background & Aims: Non-alcoholic fatty liver disease (NAFLD) is characterised by the accumulation of lipid droplets (LDs) within hepatocytes. Perilipin 2 (PLIN2) is the most abundant protein in hepatic LDs and its expression correlates with intracellular lipid accumulation. A recently discovered PLIN2 coding variant, Ser251Pro (rs35568725), was found to promote the accumulation of small LDs in embryonic kidney cells. In this study, we investigate the role of PLIN2-Ser251Pro (PLIN2-Pro251) on hepatic LD metabolism in vivo and research the metabolic phenotypes associated with this variant in humans. Methods: For our animal model, we used Plin2 knockout mice in which we expressed either human PLIN2-Pro251 (Pro251 mice) or wild-type human PLIN2-Ser251 (Ser251 mice) in a hepatocyte-specific manner. We fed both cohorts a lipogenic high-fat, high-cholesterol, high-fructose diet for 12 weeks. Results: Pro251 mice were associated with reduced liver triglycerides (TGs) and had lower mRNA expression of fatty acid synthase and diacylglycerol O-acyltransferase-2 compared with Ser251 mice. Moreover, Pro251 mice had a reduction of polyunsaturated fatty acids-TGs and reduced expression of epoxygenase genes. For our human study, we analysed the Penn Medicine BioBank, the Million Veteran Program, and UK Biobank. Across these databases, the minor allele frequency of PLIN2-Pro251 was approximately 5%. There was no association with the clinical diagnosis of NAFLD, however, there was a trend toward reduced liver fat in PLIN2-Pro251 carriers by MRI-spectroscopy in UK Biobank subjects. Conclusions: In mice lacking endogenous Plin2, expression of human PLIN2-Pro251 attenuated high-fat, high-fructose, high-cholesterol, diet-induced hepatic steatosis compared with human wild-type PLIN2-Ser251. Moreover, Pro251 mice had lower polyunsaturated fatty acids-TGs and epoxygenase genes expression, suggesting less liver oxidative stress. In humans, PLIN2-Pro251 is not associated with NAFLD. Impact and Implications: Lipid droplet accumulation in hepatocytes is the distinctive characteristic of non-alcoholic fatty liver disease. Perilipin 2 (PLIN2) is the most abundant protein in hepatic lipid droplets; however, little is known on the role of a specific polymorphism PLIN2-Pro251 on hepatic lipid droplet metabolism. PLIN2-Pro251 attenuates liver triglycerides accumulation after a high-fat-high-glucose-diet. PLIN2-Pro251 may be a novel lipid droplet protein target for the treatment of liver steatosis.

2.
Alzheimers Res Ther ; 14(1): 194, 2022 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-36572909

RESUMEN

BACKGROUND: Alzheimer's disease (AD) shares risk factors with cardiovascular disease (CVD) and dysregulated cholesterol metabolism is a mechanism common to both diseases. Cholesterol efflux capacity (CEC) is an ex vivo metric of plasma high-density lipoprotein (HDL) function and inversely predicts incident CVD independently of other risk factors. Cholesterol pools in the central nervous system (CNS) are largely separate from those in blood, and CNS cholesterol excess may promote neurodegeneration. CEC of cerebrospinal fluid (CSF) may be a useful measure of CNS cholesterol trafficking. We hypothesized that subjects with AD and mild cognitive impairment (MCI) would have reduced CSF CEC compared with Cognitively Normal (CN) and that CSF apolipoproteins apoA-I, apoJ, and apoE might have associations with CSF CEC. METHODS: We retrieved CSF and same-day ethylenediaminetetraacetic acid (EDTA) plasma from 108 subjects (40 AD; 18 MCI; and 50 CN) from the Center for Neurodegenerative Disease Research biobank at the Perelman School of Medicine, University of Pennsylvania. For CSF CEC assays, we used N9 mouse microglial cells and SH-SY5Y human neuroblastoma cells, and the corresponding plasma assay used J774 cells. Cells were labeled with [3H]-cholesterol for 24 h, had ABCA1 expression upregulated for 6 h, were exposed to 33 µl of CSF, and then were incubated for 2.5 h. CEC was quantified as percent [3H]-cholesterol counts in medium of total counts medium+cells, normalized to a pool sample. ApoA-I, ApoJ, ApoE, and cholesterol were also measured in CSF. RESULTS: We found that CSF CEC was significantly lower in MCI compared with controls and was poorly correlated with plasma CEC. CSF levels of ApoJ/Clusterin were also significantly lower in MCI and were significantly associated with CSF CEC. While CSF ApoA-I was also associated with CSF CEC, CSF ApoE had no association with CSF CEC. CSF CEC is significantly and positively associated with CSF Aß. Taken together, ApoJ/Clusterin may be an important determinant of CSF CEC, which in turn could mitigate risk of MCI and AD risk by promoting cellular efflux of cholesterol or other lipids. In contrast, CSF ApoE does not appear to play a role in determining CSF CEC.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Cardiovasculares , Neuroblastoma , Enfermedades Neurodegenerativas , Humanos , Ratones , Animales , Clusterina , Enfermedad de Alzheimer/líquido cefalorraquídeo , Apolipoproteína A-I , Apolipoproteínas E/líquido cefalorraquídeo , Colesterol
3.
Artículo en Inglés | MEDLINE | ID: mdl-35381375

RESUMEN

Adenosine triphosphate-binding cassette transporter subfamily A member 7 (ABCA7) performs incompletely understood biochemical functions that affect pathogenesis of Alzheimer's disease. ABCA7 is most similar in primary structure to ABCA1, the protein that mediates cell lipid efflux and formation of high-density lipoprotein (HDL). Lipid metabolic labeling/tracer efflux assays were employed to investigate lipid efflux in BHK-ABCA7(low expression), BHK-ABCA7(high expression) and BHK-ABCA1 cells. Shotgun lipid mass spectrometry was used to determine lipid composition of HDL synthesized by BHK-ABCA7 and BHK-ABCA1 cells. BHK-ABCA7(low) cells exhibited significant efflux only of choline-phospholipid and phosphatidylinositol. BHK-ABCA7(high) cells had significant cholesterol and choline-phospholipid efflux to apolipoprotein (apo) A-I, apo E, the 18A peptide, HDL, plasma and cerebrospinal fluid and significant efflux of sphingosine-lipid, serine-lipid (which is composed of phosphatidylserine and phosphatidylethanolamine in BHK cells) and phosphatidylinositol to apo A-I. In efflux assays to apo A-I, after adjustment to choline-phospholipid, ABCA7-mediated efflux removed ~4 times more serine-lipid and phosphatidylinositol than ABCA1-mediated efflux, while ABCA1-mediated efflux removed ~3 times more cholesterol than ABCA7-mediated efflux. Shotgun lipidomic analysis revealed that ABCA7-HDL had ~20 mol% less phosphatidylcholine and 3-5 times more serine-lipid and phosphatidylinositol than ABCA1-HDL, while ABCA1-HDL contained only ~6 mol% (or ~1.1 times) more cholesterol than ABCA7-HDL. The discrepancy between the tracer efflux assays and shotgun lipidomics with respect to cholesterol may be explained by an underestimate of ABCA7-mediated cholesterol efflux in the former approach. Overall, these results suggest that ABCA7 lacks specificity for phosphatidylcholine and releases significantly but not dramatically less cholesterol in comparison with ABCA1.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Apolipoproteína A-I , Transportadoras de Casetes de Unión a ATP/metabolismo , Apolipoproteína A-I/metabolismo , Colesterol/metabolismo , Colina , Lipoproteínas HDL/metabolismo , Fosfatidilcolinas/metabolismo , Fosfatidilinositoles , Fosfolípidos/metabolismo , Serina
4.
Molecules ; 26(22)2021 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-34833954

RESUMEN

High-density lipoprotein cholesterol (HDL-C) is thought to be atheroprotective yet some patients with elevated HDL-C levels develop cardiovascular disease, possibly due to the presence of dysfunctional HDL. We aimed to assess the metabolic fate of circulating HDL particles in patients with high HDL-C with and without coronary artery disease (CAD) using in vivo dual labeling of its cholesterol and protein moieties. We measured HDL apolipoprotein (apo) A-I, apoA-II, free cholesterol (FC), and cholesteryl ester (CE) kinetics using stable isotope-labeled tracers (D3-leucine and 13C2-acetate) as well as ex vivo cholesterol efflux to HDL in subjects with (n = 6) and without (n = 6) CAD that had HDL-C levels >90th percentile. Healthy controls with HDL-C within the normal range (n = 6) who underwent the same procedures were used as the reference. Subjects with high HDL-C with and without CAD had similar plasma lipid levels and similar apoA-I, apoA-II, HDL FC, and CE pool sizes with no significant differences in fractional clearance rates (FCRs) or production rates (PRs) of these components between groups. Subjects with high HDL-C with and without CAD also had similar basal and cAMP-stimulated ex vivo cholesterol efflux to HDL. When all subjects were considered (n = 18), unstimulated non-ABCA1-mediated efflux (but not ABCA1-specific efflux) was correlated positively with apoA-I production (r = 0.552, p = 0.017) and HDL FC and CE pool sizes, and negatively with the fractional clearance rate of FC (r = -0.759, p = 4.1 × 10-4) and CE (r = -0.652, p = 4.57 × 10-3). Our data are consistent with the concept that ex vivo non-ABCA1 efflux capacity may correlate with slower in vivo turnover of HDL cholesterol moieties. The use of a dual labeling protocol provided for the first time the opportunity to assess the association of ex vivo cholesterol efflux capacity with in vivo HDL cholesterol metabolic parameters.


Asunto(s)
HDL-Colesterol/sangre , HDL-Colesterol/metabolismo , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/metabolismo , Adulto , Anciano , Apolipoproteína A-I/metabolismo , Femenino , Humanos , Lípidos/sangre , Lipoproteínas HDL/metabolismo , Masculino , Persona de Mediana Edad
5.
PLoS Genet ; 17(9): e1009802, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34543263

RESUMEN

Triglyceride-rich lipoproteins (TRLs) are circulating reservoirs of fatty acids used as vital energy sources for peripheral tissues. Lipoprotein lipase (LPL) is a predominant enzyme mediating triglyceride (TG) lipolysis and TRL clearance to provide fatty acids to tissues in animals. Physiological and human genetic evidence support a primary role for LPL in hydrolyzing TRL TGs. We hypothesized that endothelial lipase (EL), another extracellular lipase that primarily hydrolyzes lipoprotein phospholipids may also contribute to TRL metabolism. To explore this, we studied the impact of genetic EL loss-of-function on TRL metabolism in humans and mice. Humans carrying a loss-of-function missense variant in LIPG, p.Asn396Ser (rs77960347), demonstrated elevated plasma TGs and elevated phospholipids in TRLs, among other lipoprotein classes. Mice with germline EL deficiency challenged with excess dietary TG through refeeding or a high-fat diet exhibited elevated TGs, delayed dietary TRL clearance, and impaired TRL TG lipolysis in vivo that was rescued by EL reconstitution in the liver. Lipidomic analyses of postprandial plasma from high-fat fed Lipg-/- mice demonstrated accumulation of phospholipids and TGs harboring long-chain polyunsaturated fatty acids (PUFAs), known substrates for EL lipolysis. In vitro and in vivo, EL and LPL together promoted greater TG lipolysis than either extracellular lipase alone. Our data positions EL as a key collaborator of LPL to mediate efficient lipolysis of TRLs in humans and mice.


Asunto(s)
Lipasa/metabolismo , Lipólisis , Lipoproteínas/metabolismo , Triglicéridos/metabolismo , Animales , Dieta Alta en Grasa , Humanos , Lipasa/genética , Liposomas , Ratones , Mutación Missense , Periodo Posprandial , Triglicéridos/sangre
6.
J Alzheimers Dis ; 74(2): 563-578, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32065798

RESUMEN

BACKGROUND: Basic research has implicated intracellular cholesterol in neurons, microglia, and astrocytes in the pathogenesis of Alzheimer's disease (AD), but there is presently no assay to access intracellular cholesterol in neural cells in living people in the context of AD. OBJECTIVE: To devise and characterize an assay that can access intracellular cholesterol and cholesterol efflux in neural cells in living subjects. METHODS: We modified the protocol for high-density lipoprotein cholesterol efflux capacity (CEC) from macrophages, a biomarker that accesses cholesterol in macrophages in atherosclerosis. To measure cerebrospinal fluid (CSF) CECs from neurons, microglia, and astrocytes, CSF was exposed to, correspondingly, neuronal, microglial, and astrocytic cholesterol source cells. Human neuroblastoma SH-SY5Y, mouse microglial N9, and human astroglial A172 cells were used as the cholesterol source cells. CSF samples were screened for contamination with blood. CSF CECs were measured in a small cohort of 22 individuals. RESULTS: CSF CECs from neurons, microglia, and astrocytes were moderately to moderately strongly correlated with CSF concentrations of cholesterol, apolipoprotein A-I, apolipoprotein E, and clusterin (Pearson's r = 0.53-0.86), were in poor agreement with one another regarding CEC of the CSF samples (Lin's concordance coefficient rc = 0.71-0.76), and were best predicted by models consisting of, correspondingly, CSF phospholipid (R2 = 0.87, p < 0.0001), CSF apolipoprotein A-I and clusterin (R2 = 0.90, p < 0.0001), and CSF clusterin (R2 = 0.62, p = 0.0005). CONCLUSION: Characteristics of the CSF CEC metrics suggest a potential for independent association with AD and provision of fresh insight into the role of cholesterol in AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/epidemiología , Astrocitos/metabolismo , Colesterol/líquido cefalorraquídeo , Microglía/metabolismo , Neuronas/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/diagnóstico , Biomarcadores/líquido cefalorraquídeo , Línea Celular Tumoral , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas
7.
J Lipid Res ; 58(4): 752-762, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28167703

RESUMEN

Reverse cholesterol transport (RCT) is thought to be an atheroprotective function of HDL, and macrophage-specific RCT in mice is inversely associated with atherosclerosis. We developed a novel method using 3H-cholesterol nanoparticles to selectively trace macrophage-specific RCT in vivo in humans. Use of 3H-cholesterol nanoparticles was initially tested in mice to assess the distribution of tracer and response to interventions known to increase RCT. Thirty healthy subjects received 3H-cholesterol nanoparticles intravenously, followed by blood and stool sample collection. Tracer counts were assessed in plasma, nonHDL, HDL, and fecal fractions. Data were analyzed by using multicompartmental modeling. Administration of 3H-cholesterol nanoparticles preferentially labeled macrophages of the reticuloendothelial system in mice, and counts were increased in mice treated with a liver X receptor agonist or reconstituted HDL, as compared with controls. In humans, tracer disappeared from plasma rapidly after injection of nanoparticles, followed by reappearance in HDL and nonHDL fractions. Counts present as free cholesterol increased rapidly and linearly in the first 240 min after nadir; counts in cholesteryl ester increased steadily over time. Estimates of fractional transfer rates of key RCT steps were obtained. These results support the use of 3H-cholesterol nanoparticles as a feasible approach for the measurement of macrophage RCT in vivo in humans.


Asunto(s)
Aterosclerosis/sangre , HDL-Colesterol/sangre , Colesterol/sangre , Lipoproteínas HDL/metabolismo , Adolescente , Adulto , Anciano , Animales , Aterosclerosis/patología , Transporte Biológico/genética , Colesterol/química , Colesterol/genética , HDL-Colesterol/química , HDL-Colesterol/aislamiento & purificación , Heces/química , Femenino , Humanos , Lipoproteínas HDL/aislamiento & purificación , Hígado/metabolismo , Hígado/patología , Receptores X del Hígado/agonistas , Receptores X del Hígado/sangre , Macrófagos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Nanopartículas/administración & dosificación , Nanopartículas/química
8.
J Lipid Res ; 58(4): 731-741, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28137768

RESUMEN

Phospholipid transfer protein (PLTP) may affect macrophage reverse cholesterol transport (mRCT) through its role in the metabolism of HDL. Ex vivo cholesterol efflux capacity and in vivo mRCT were assessed in PLTP deletion and PLTP overexpression mice. PLTP deletion mice had reduced HDL mass and cholesterol efflux capacity, but unchanged in vivo mRCT. To directly compare the effects of PLTP overexpression and deletion on mRCT, human PLTP was overexpressed in the liver of wild-type animals using an adeno-associated viral (AAV) vector, and control and PLTP deletion animals were injected with AAV-null. PLTP overexpression and deletion reduced plasma HDL mass and cholesterol efflux capacity. Both substantially decreased ABCA1-independent cholesterol efflux, whereas ABCA1-dependent cholesterol efflux remained the same or increased, even though preß HDL levels were lower. Neither PLTP overexpression nor deletion affected excretion of macrophage-derived radiocholesterol in the in vivo mRCT assay. The ex vivo and in vivo assays were modified to gauge the rate of cholesterol efflux from macrophages to plasma. PLTP activity did not affect this metric. Thus, deviations in PLTP activity from the wild-type level reduce HDL mass and ex vivo cholesterol efflux capacity, but not the rate of macrophage cholesterol efflux to plasma or in vivo mRCT.


Asunto(s)
HDL-Colesterol/sangre , Colesterol/sangre , Lipoproteínas HDL/sangre , Proteínas de Transferencia de Fosfolípidos/genética , Animales , Transporte Biológico/genética , Dependovirus/genética , Regulación de la Expresión Génica , Lipoproteínas de Alta Densidad Pre-beta/biosíntesis , Lipoproteínas de Alta Densidad Pre-beta/sangre , Lipoproteínas de Alta Densidad Pre-beta/genética , Humanos , Lipoproteínas HDL/genética , Hígado/metabolismo , Macrófagos/metabolismo , Ratones , Proteínas de Transferencia de Fosfolípidos/biosíntesis , Eliminación de Secuencia
9.
Hum Mol Genet ; 25(14): 3042-3054, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27270415

RESUMEN

We report an individual who presented with severe neurodevelopmental delay and an intractable infantile-onset seizure disorder. Exome sequencing identified a homozygous single nucleotide change that abolishes a splice donor site in the ARV1 gene (c.294 + 1G > A homozygous). This variant completely prevented splicing in minigene assays, and resulted in exon skipping and an in-frame deletion of 40 amino acids in primary human fibroblasts (NP_073623.1: p.(Lys59_Asn98del). The p.(Lys59_Asn98del) and previously reported p.(Gly189Arg) ARV1 variants were evaluated for protein expression and function. The p.(Gly189Arg) variant partially rescued the temperature-dependent growth defect in arv1Δ yeast, while p.(Lys59-Asn98del) completely failed to rescue at restrictive temperature. In contrast to wild type human ARV1, neither variant expressed detectable levels of protein in mammalian cells. Mice with a neuronal deletion of Arv1 recapitulated the human phenotype, exhibiting seizures and a severe survival defect in adulthood. Our data support ARV1 deficiency as a cause of autosomal recessive epileptic encephalopathy.


Asunto(s)
Proteínas Portadoras/genética , Predisposición Genética a la Enfermedad , Proteínas de la Membrana/genética , Espasmos Infantiles/genética , Exones/genética , Femenino , Genotipo , Humanos , Lactante , Mutación , Linaje , Fenotipo , Sitios de Empalme de ARN/genética , Espasmos Infantiles/fisiopatología
10.
Lipids Health Dis ; 10: 186, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-22018327

RESUMEN

BACKGROUND: The plasma lipoprotein response of F1B Golden-Syrian hamsters fed diets high in very long chain (VLC) n-3 polyunsaturated fatty acids (PUFA) is paradoxical to that observed in humans. This anomaly is attributed, in part, to low lipoprotein lipase activity and is dependent on cholesterol status. To further elucidate the mechanism(s) for these responses, hamsters were fed diets containing supplemental fish oil (VLC n-3 PUFA) or safflower oil (n-6 PUFA) (both 10% [w/w]) and either cholesterol-supplemented (0.1% cholesterol [w/w]) or cholesterol-depleted (0.01% cholesterol [w/w] and 10 days prior to killing fed 0.15% lovastatin+2% cholestyramine [w/w]). RESULTS: Cholesterol-supplemented hamsters fed fish oil, relative to safflower oil, had higher non-high density lipoprotein (HDL) cholesterol and triglyceride concentrations (P < 0.001) which were associated with lower hepatic low density lipoprotein (LDL) receptor, sterol regulatory element binding protein (SREBP)-1c and acyl-CoA: cholesterol acyl transferase-2 (ACAT) mRNA and protein (p < 0.05), and higher hepatic apolipoprotein (apo) B-100 and apo E protein levels. In contrast, cholesterol-depleted hamsters fed fish oil, relative to safflower oil, had lower non-HDL cholesterol and triglyceride concentrations (P < 0.001) which were associated with lower hepatic SREBP-1c (p < 0.05) but not apo B-100, apo E or ACAT-2 mRNA or protein levels. Independent of cholesterol status, fish oil fed hamsters had lower HDL cholesterol concentrations (p < 0.001), which were associated with lower hepatic apoA-I protein levels (p < 0.05). CONCLUSION: These data suggest disturbing cholesterol homeostasis in F1B hamsters alters their response to dietary fatty acids, which is reflected in altered plasma lipoprotein patterns and regulation of genes associated with their metabolism.


Asunto(s)
Colesterol/sangre , Ácidos Grasos Omega-3/uso terapéutico , Ácidos Grasos Omega-6/uso terapéutico , Hipercolesterolemia/dietoterapia , Lipoproteínas/sangre , Acetil-CoA C-Acetiltransferasa/genética , Acetil-CoA C-Acetiltransferasa/metabolismo , Animales , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Colesterol en la Dieta/efectos adversos , Cricetinae , Aceites de Pescado/uso terapéutico , Regulación de la Expresión Génica , Homeostasis , Hipercolesterolemia/sangre , Hipercolesterolemia/metabolismo , Hígado/metabolismo , Masculino , Mesocricetus , ARN Mensajero/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Aceite de Cártamo/uso terapéutico , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
11.
Circulation ; 124(12): 1382-90, 2011 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-21859969

RESUMEN

BACKGROUND: The role of hepatic ATP-binding cassette transporter 1 (ABCA1) in maintaining plasma high density lipoprotein cholesterol (HDL-C) levels is well established, but its role in reverse cholesterol transport (RCT) is unclear. Probucol is a compound that reduces HDL-C levels but also reduces atherosclerosis in animal models and xanthomas in humans. The aim of the present study was to test the hypothesis that probucol inhibits hepatic ABCA1 activity, thereby reducing HDL-C levels but promoting RCT from macrophages. METHODS AND RESULTS: Wild-type (WT) C57BL/6 mice and scavenger receptor class B type I (SR-BI) knockout mice were fed a chow diet containing 0.5% probucol or normal chow for 2 weeks. In WT mice, probucol, despite decreasing HDL-C by >80%, effectively maintained macrophage RCT. In SR-BI knockout mice, probucol also substantially reduced HDL-C but significantly increased macrophage RCT. Furthermore, probucol significantly enhanced the excretion of HDL-derived cholesterol into feces in both WT and SR-BI knockout mice. Probucol inhibited ABCA1-dependent cholesterol efflux from mouse primary hepatocytes, and this effect was shown to be responsible for the effect of probucol on increasing the fecal excretion of HDL-derived cholesterol in vivo. CONCLUSIONS: We demonstrate that pharmacological inhibition of hepatic ABCA1 activity with probucol reduced HDL-C levels but promoted RCT through diversion of HDL-derived cholesterol from efflux back into plasma instead to excretion in the bile. These results explain the beneficial effects of probucol on atherosclerosis and xanthomas despite its HDL-lowering effects and suggest that inactivation of hepatic ABCA1 leads to increased RCT despite reducing plasma HDL-C levels.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Aterosclerosis/metabolismo , HDL-Colesterol/sangre , Hepatocitos/metabolismo , Macrófagos/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Animales , Anticolesterolemiantes/farmacología , Aterosclerosis/prevención & control , Sistema Biliar/metabolismo , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Células Cultivadas , HDL-Colesterol/farmacocinética , Modelos Animales de Enfermedad , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Humanos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Probucol/farmacología , Xantomatosis/metabolismo , Xantomatosis/prevención & control
12.
Arterioscler Thromb Vasc Biol ; 31(6): 1276-82, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21441141

RESUMEN

OBJECTIVE: Peroxisome proliferator-activated receptor-α (PPARα) activation has been shown in vitro to increase macrophage cholesterol efflux, the initial step in reverse cholesterol transport (RCT). However, it remains unclear whether PPARα activation promotes macrophage RCT in vivo. METHODS AND RESULTS: We demonstrated that a specific potent PPARα agonist GW7647 inhibited atherosclerosis and promoted macrophage RCT in hypercholesterolemic mice expressing the human apolipoprotein A-I (apoA-I) gene. We compared the effect of GW7647 on RCT in human apoA-I transgenic (hA-ITg) mice with wild-type mice and showed that the PPARα agonist promoted RCT in hA-ITg mice to a much greater extent than in wild-type mice, indicating that human apoA-I expression is important for PPARα-induced RCT. We further investigated the dependence of the macrophage PPARα-liver X receptor (LXR) pathway on the promotion of RCT by GW7647. Primary murine macrophages lacking PPARα or LXR abolished the ability of GW7647 to promote RCT in hA-ITg mice. In concert, the PPARα agonist promoted cholesterol efflux and ATP binding cassette transporter A1/G1 expression in primary macrophages, and this was also by the PPARα-LXR pathway. CONCLUSION: Our observations demonstrate that a potent PPARα agonist promotes macrophage RCT in vivo in a manner that is enhanced by human apoA-I expression and dependent on both macrophage PPARα and LXR expression.


Asunto(s)
Colesterol/metabolismo , Macrófagos/metabolismo , Receptores Nucleares Huérfanos/fisiología , PPAR alfa/fisiología , Transducción de Señal , Animales , Apolipoproteína A-I/fisiología , Aterosclerosis/prevención & control , Transporte Biológico , Butiratos/farmacología , Humanos , Receptores X del Hígado , Ratones , PPAR alfa/agonistas , Compuestos de Fenilurea/farmacología
13.
Metabolism ; 59(10): 1491-501, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20197195

RESUMEN

Cholesterol status and dietary fat alter several metabolic pathways reflected in lipoprotein profiles. To assess plasma lipoprotein response and mechanisms by which cholesterol and dietary fat type regulate expression of genes involved in lipoprotein metabolism, we developed an experimental model system using F1B hamsters fed diets (12 weeks) enriched in 10% (wt/wt) coconut, olive, or safflower oil with either high cholesterol (0.1%; cholesterol supplemented) or low cholesterol coupled with cholesterol-lowering drugs 10 days before killing (0.01% cholesterol, 0.15% lovastatin, 2% cholestyramine; cholesterol depleted). Irrespective of dietary fat, cholesterol depletion, relative to supplementation, resulted in lower plasma non-high-density lipoprotein (non-HDL) and HDL cholesterol, and triglyceride concentrations (all Ps < .05). In the liver, these differences were associated with higher sterol regulatory element binding protein-2, low-density lipoprotein receptor, 3-hydroxy-3-methylglutaryl coenzyme A reductase, and 7α-hydroxylase messenger RNA (mRNA) levels; higher scavenger receptor B1 and apolipoprotein A-I mRNA and protein levels; lower apolipoprotein E protein levels; and in intestine, modestly lower sterol transporters adenosine triphosphate-binding cassette (ABC) A1, ABCG5, and ABCG8 mRNA levels. Irrespective of cholesterol status, coconut oil, relative to olive and safflower oils, resulted in higher non-HDL cholesterol and triglyceride concentrations (both Ps < .05) and modestly higher sterol regulatory element binding protein-2 mRNA levels. These data suggest that, in F1B hamsters, differences in plasma lipoprotein profiles in response to cholesterol depletion are associated with changes in the expression of genes involved in cholesterol metabolism, whereas the effect of dietary fat type on gene expression was modest, which limits the usefulness of the experimental animal model.


Asunto(s)
Colesterol/metabolismo , Grasas de la Dieta/farmacología , Homeostasis/efectos de los fármacos , Trastornos del Metabolismo de los Lípidos/metabolismo , Lipoproteínas/metabolismo , Animales , Cricetinae , Grasas de la Dieta/clasificación , Modelos Animales de Enfermedad , Eritrocitos/metabolismo , Ácidos Grasos/metabolismo , Homeostasis/fisiología , Mucosa Intestinal/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Trastornos del Metabolismo de los Lípidos/genética , Trastornos del Metabolismo de los Lípidos/fisiopatología , Hígado/metabolismo , Masculino , Metaboloma/efectos de los fármacos , ARN Mensajero/metabolismo
14.
Circulation ; 120(2): 160-9, 2009 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-19564558

RESUMEN

BACKGROUND: Lecithin:cholesterol acyltransferase (LCAT) catalyzes the formation of plasma cholesteryl ester, plays a key role in high-density lipoprotein metabolism, and has been believed to be critical in the process of reverse cholesterol transport (RCT). METHODS AND RESULTS: The role of LCAT in RCT from macrophages was quantified with a validated assay involving intraperitoneal injection in mice of (3)H-cholesterol-labeled J774 macrophages and monitoring the appearance of tracer in plasma, liver, bile, and feces. Human LCAT overexpression in human apolipoprotein A-I transgenic mice substantially increased plasma high-density lipoprotein cholesterol levels but surprisingly did not increase macrophage RCT. Even in the setting of coexpression of scavenger receptor BI or cholesteryl ester transfer protein, both of which promoted the transfer of LCAT-derived high-density lipoprotein cholesterol ester to the liver, LCAT overexpression still had no effect on RCT. Serum from LCAT-overexpressing mice had reduced ability to promote cholesterol efflux from macrophages ex vivo via ABCA1. To determine the effect of LCAT deficiency on macrophage RCT, LCAT(-/-) and LCAT(+/-) mice were compared with wild-type mice. Despite extremely low plasma levels of high-density lipoprotein cholesterol, LCAT-deficient mice had only a 50% reduction in RCT. LCAT(+/-) mice had normal RCT despite a significant reduction in high-density lipoprotein cholesterol. Serum from LCAT-deficient mice had increased ability to promote ABCA1-mediated cholesterol efflux from macrophages ex vivo. CONCLUSIONS: These results demonstrate that LCAT overexpression does not promote an increased rate of macrophage RCT. Although LCAT activity does become rate limiting in the context of complete LCAT deficiency, RCT is reduced by only 50% even in the absence of LCAT. These data suggest that macrophage RCT may not be as dependent on LCAT activity as has previously been believed.


Asunto(s)
Colesterol/metabolismo , Macrófagos/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Transporte Biológico/fisiología , Antígenos CD36/metabolismo , Línea Celular , Células Cultivadas , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , HDL-Colesterol/metabolismo , Femenino , Humanos , Inyecciones Intraperitoneales , Macrófagos/citología , Ratones , Ratones Noqueados , Ratones Transgénicos
15.
Circulation ; 119(8): 1135-45, 2009 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-19221221

RESUMEN

BACKGROUND: Inflammation is proposed to impair reverse cholesterol transport (RCT), a major atheroprotective function of high-density lipoprotein (HDL). The present study presents the first integrated functional evidence that inflammation retards numerous components of RCT. METHODS AND RESULTS: We used subacute endotoxemia in the rodent macrophage-to-feces RCT model to assess the effects of inflammation on RCT in vivo and performed proof of concept experimental endotoxemia studies in humans. Endotoxemia (3 mg/kg SC) reduced (3)H-cholesterol movement from macrophage to plasma and (3)H-cholesterol associated with HDL fractions. At 48 hours, bile and fecal counts were markedly reduced consistent with downregulation of hepatic expression of ABCG5, ABCG8, and ABCB11 biliary transporters. Low-dose lipopolysaccharide (0.3 mg/kg SC) also reduced bile and fecal counts, as well as expression of biliary transporters, but in the absence of effects on plasma or liver counts. In vitro, lipopolysaccharide impaired (3)H-cholesterol efflux from human macrophages to apolipoprotein A-I and serum coincident with reduced expression of the cholesterol transporter ABCA1. During human (3 ng/kg; n=20) and murine endotoxemia (3 mg/kg SC), ex vivo macrophage cholesterol efflux to acute phase HDL was attenuated. CONCLUSIONS: We provide the first in vivo evidence that inflammation impairs RCT at multiple steps in the RCT pathway, particularly cholesterol flux through liver to bile and feces. Attenuation of RCT and HDL efflux function, independent of HDL cholesterol levels, may contribute to atherosclerosis in chronic inflammatory states including obesity, metabolic syndrome, and type 2 diabetes.


Asunto(s)
Colesterol/metabolismo , Endotoxemia/patología , Adolescente , Adulto , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/prevención & control , Transporte Biológico Activo/fisiología , Línea Celular , Endotoxemia/metabolismo , Femenino , Humanos , Inflamación/metabolismo , Inflamación/patología , Lipoproteínas HDL/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Adulto Joven
16.
Arterioscler Thromb Vasc Biol ; 29(1): 40-6, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19008531

RESUMEN

OBJECTIVE: Apolipoprotein F (ApoF) is a protein component of several lipoprotein classes including HDL. It is also known as lipid transfer inhibitor protein (LTIP) based on its ability to inhibit lipid transfer between lipoproteins ex vivo. We sought to investigate the role of ApoF in HDL metabolism. METHODS AND RESULTS: Adeno-associated viruses (AAV) based on serotype 8, were used to overexpress either murine or human ApoF in mice. Overexpression of murine ApoF significantly reduced total cholesterol levels by 28% (P<0.001), HDL by 27% (P<0.001), and phospholipid levels by 19% (P<0.001). Overexpression of human ApoF had similar effects. Human ApoF was nearly exclusively HDL-associated in mice. In agreement with this finding, greater than 90% of the ApoF in human plasma was found on HDL(3), with only a small amount on LDL. Overexpression of mouse ApoF accelerated the plasma clearance of [(3)H]-cholesteryl ether labeled HDL. Plasma from mice overexpressing ApoF showed improved macrophage cholesterol efflux on a per HDL-C basis. CONCLUSIONS: ApoF overexpression reduces HDL cholesterol levels in mice by increasing clearance of HDL-CE. ApoF may be an important determinant of HDL metabolism and reverse cholesterol transport.


Asunto(s)
Apolipoproteínas/genética , HDL-Colesterol/sangre , Alanina Transaminasa/sangre , Animales , Apolipoproteínas/sangre , Médula Ósea/fisiología , Línea Celular , Colesterol/sangre , Colesterol/metabolismo , Clonación Molecular , Dependovirus/genética , Regulación de la Expresión Génica , Humanos , Riñón/embriología , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfolípidos/sangre , Plásmidos , Triglicéridos/sangre
17.
Atherosclerosis ; 204(2): 418-23, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18990393

RESUMEN

OBJECTIVE: The mechanism by which non-nucleoside reverse transcriptase inhibitors (NNRTIs) increase HDL cholesterol (HDL-C) in HIV+ patients and the benefits of this with respect to cardiovascular risk are not known. Studies were conducted to test the hypothesis that NNRTIs have a beneficial effect on HDL-C and reverse cholesterol transport (RCT). METHODS: LDLr-/- and hA-I transgenic mice were fed a Western diet containing either nevirapine (20mg/kg per day), efavirenz (10mg/kg per day), or diet alone. hA-I transgenic mice underwent a study to measure RCT (measured by excretion of macrophage [(3)H]-cholesterol into HDL and feces) at 8 weeks. RESULTS: LDLr-/- and hA-I transgenic mice treated with nevirapine and efavirenz had a significant increase in HDL-C level (up to 23% in hA-I transgenic) at 4 weeks. However, there was no difference in HDL levels beyond 4 weeks of treatment. At 4 weeks, the FPLC profile of hA-I transgenic mice showed an increase in large HDL. hApoA-I transgenic mice treated with efavirenz for 4 weeks had increased expression of human apoA-I in liver and an increased human apoA-I production rate. Incubation of plasma from hA-I transgenic mice treated for 4 weeks with [(3)H]-cholesterol-labeled macrophages revealed increased cholesterol efflux to plasma from mice treated with efavirenz and nevirapine. Following injection of hA-I transgenic mice treated for 8 weeks with [(3)H]-cholesterol-labeled macrophages, RCT was increased in the efavirenz (p=0.01) group and trended towards an increase in the nevirapine (p=0.15) group. CONCLUSION: Nevirapine and efavirenz transiently increased HDL-C in LDLr-/- and hA-I transgenic mice fed a Western diet that was associated with increased apoA-I production. An increase in RCT in hA-I transgenic mice at 8 weeks despite no difference in HDL levels indicates that these drugs affect additional factors in the RCT pathway that enhance cholesterol efflux from the macrophage and peripheral tissues to plasma and delivery to liver for excretion. These results suggest that treatment with NNRTIs has a beneficial effect on cholesterol efflux and RCT.


Asunto(s)
Fármacos Anti-VIH/farmacología , Benzoxazinas/farmacología , HDL-Colesterol/metabolismo , Colesterol/metabolismo , Macrófagos/efectos de los fármacos , Nevirapina/farmacología , Inhibidores de la Transcriptasa Inversa/farmacología , Alquinos , Animales , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Transporte Biológico , Línea Celular , Colesterol/sangre , HDL-Colesterol/sangre , Ciclopropanos , Grasas de la Dieta/metabolismo , Femenino , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores de LDL/deficiencia , Receptores de LDL/genética , Factores de Tiempo , Regulación hacia Arriba
18.
Biochim Biophys Acta ; 1781(3): 97-104, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18252207

RESUMEN

Acyl-CoA:diacylglycerol acyltransferases (DGATs) are enzymes that catalyze the formation of triglyceride (TG) from acyl-CoA and diacylglycerol. Two DGATs have been identified which belong to two distinct gene families and both are ubiquitously expressed. DGAT2 knockout mice are lipopenic and die shortly after birth. In the current study, wild type mice were treated with increasing doses (25-60 mg/kg twice weekly) of a DGAT2 gene-specific antisense oligonucleotide (ASO). Treatment resulted in a dose dependent decrease in hepatic DGAT2 gene expression (up to 80%) which was associated with a 40% decrease in hepatic DGAT2 activity and a 45% decrease in hepatic TG. Decreased levels of DGAT2 resulted in a significant dose dependent decrease in VLDL TG secretion (up to 52%) and reduced plasma TG, total cholesterol, and ApoB. Similar results were obtained when DGAT1 KO mice were treated with the DGAT2 ASO. Treatment of ob/ob mice with the DGAT2 ASO resulted in significant decreases in weight gain (10%), adipose weight (25%) and hepatic TG content (80%). Our findings indicate that the majority of TG destined for secretion by liver is synthesized by DGAT2 and suggests that DGAT2 may be a therapeutic target for treatment of hypertriglyceridemia, hepatic steatosis and obesity.


Asunto(s)
Apolipoproteínas B/metabolismo , Diacilglicerol O-Acetiltransferasa/deficiencia , Lipoproteínas VLDL/metabolismo , Oligonucleótidos Antisentido/farmacología , Triglicéridos/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/enzimología , Animales , Diacilglicerol O-Acetiltransferasa/genética , Diacilglicerol O-Acetiltransferasa/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Modelos Biológicos , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Aumento de Peso/efectos de los fármacos
19.
J Lipid Res ; 48(11): 2453-62, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17761631

RESUMEN

Cholesterol efflux occurs by different pathways, including transport mediated by specific proteins. We determined the effect of enriching cells with free cholesterol (FC) on the release of FC to human serum. Loading Fu5AH cells with FC had no effect on fractional efflux, whereas enriching mouse peritoneal macrophages (MPMs) resulted in a doubling of fractional efflux. Efflux from cholesterol-normal MPM and Fu5AH cells to 15 human sera correlated well with HDL parameters. However, these relationships were reduced or lost with cholesterol-loaded MPMs. Using macrophages from scavenger receptor class B type I (SR-BI)-, ABCA1-, and ABCG1-knockout mice, together with inhibitors of SR-BI- and ABCA1-mediated efflux, we were able to quantitate efflux upon loading macrophages with excess cholesterol and to establish the contributions of the various efflux pathways in cholesterol-normal and -enriched cells. The removal of ABCA1 had essentially no effect on the total efflux when cell cholesterol levels were normal. However, in cholesterol-enriched cells, the removal of ABCA1 reduced efflux by 50%. Approximately 20% of the efflux stimulated by FC-loading MPM is attributable to ABCG1. The SR-BI contribution to efflux was small. Another pathway that is present in all cells is aqueous diffusion. Our studies demonstrate that this mechanism is one of the major contributors to efflux, particularly in cholesterol-normal cells.


Asunto(s)
Colesterol/metabolismo , Macrófagos Peritoneales/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/fisiología , Animales , Humanos , Lipoproteínas/fisiología , Lipoproteínas HDL/sangre , Ratones , Ratones Noqueados , Receptores Depuradores de Clase B/fisiología
20.
Circulation ; 116(11): 1267-73, 2007 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-17709636

RESUMEN

BACKGROUND: Cholesteryl ester transfer protein (CETP) transfers cholesteryl esters from high-density lipoproteins to apolipoprotein (apo) B-containing lipoproteins and in humans plays an important role in lipoprotein metabolism. However, the role that CETP plays in mediation of reverse cholesterol transport (RCT) remains unclear. We used a validated in vivo assay of macrophage RCT to test the effect of CETP expression in mice (which naturally lack CETP) on macrophage RCT, including in mice that lack the low-density lipoprotein receptor or the scavenger receptor class B, type I. METHOD AND RESULTS: A vector based on adeno-associated virus serotype 8 (AAV8) with a liver-specific thyroglobulin promoter was used to stably express human CETP in livers of mice and was compared with an AAV8-lacZ control vector. The RCT assay was performed 4 weeks after vector injection and involved the intraperitoneal injection of acetylated low-density lipoprotein cholesterol-loaded and 3H-cholesterol-labeled J774 macrophages in mice with plasma sampling at several time points, liver and bile sampling at 48 hours, and continuous fecal collection to measure 3H-sterol as an integrated readout of macrophage RCT. In apobec-1-null mice, CETP expression reduced plasma high-density lipoprotein cholesterol levels but significantly increased fecal 3H-sterol excretion. In low-density lipoprotein receptor/apobec-1 double-null mice, CETP expression reduced high-density lipoprotein cholesterol levels and had no effect on fecal 3H-sterol excretion. Finally, in scavenger receptor class B, type I-null mice, CETP expression reduced high-density lipoprotein cholesterol levels and significantly increased fecal 3H-sterol excretion. CONCLUSION: The present results demonstrate that CETP expression promotes macrophage RCT in mice, that this effect is dependent on the low-density lipoprotein receptor, and that CETP expression restores to normal the impaired RCT in mice deficient in scavenger receptor class B, type I.


Asunto(s)
Proteínas de Transferencia de Ésteres de Colesterol/biosíntesis , Colesterol/metabolismo , Regulación de la Expresión Génica/fisiología , Macrófagos/metabolismo , Animales , Transporte Biológico/fisiología , Colesterol/genética , Proteínas de Transferencia de Ésteres de Colesterol/genética , Humanos , Metabolismo de los Lípidos/fisiología , Masculino , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...