Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Nature ; 478(7369): 404-7, 2011 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22012398

RESUMEN

Cardiovascular disease remains the leading cause of mortality in westernized countries, despite optimum medical therapy to reduce the levels of low-density lipoprotein (LDL)-associated cholesterol. The pursuit of novel therapies to target the residual risk has focused on raising the levels of high-density lipoprotein (HDL)-associated cholesterol in order to exploit its atheroprotective effects. MicroRNAs (miRNAs) have emerged as important post-transcriptional regulators of lipid metabolism and are thus a new class of target for therapeutic intervention. MicroRNA-33a and microRNA-33b (miR-33a/b) are intronic miRNAs whose encoding regions are embedded in the sterol-response-element-binding protein genes SREBF2 and SREBF1 (refs 3-5), respectively. These miRNAs repress expression of the cholesterol transporter ABCA1, which is a key regulator of HDL biogenesis. Recent studies in mice suggest that antagonizing miR-33a may be an effective strategy for raising plasma HDL levels and providing protection against atherosclerosis; however, extrapolating these findings to humans is complicated by the fact that mice lack miR-33b, which is present only in the SREBF1 gene of medium and large mammals. Here we show in African green monkeys that systemic delivery of an anti-miRNA oligonucleotide that targets both miR-33a and miR-33b increased hepatic expression of ABCA1 and induced a sustained increase in plasma HDL levels over 12 weeks. Notably, miR-33 antagonism in this non-human primate model also increased the expression of miR-33 target genes involved in fatty acid oxidation (CROT, CPT1A, HADHB and PRKAA1) and reduced the expression of genes involved in fatty acid synthesis (SREBF1, FASN, ACLY and ACACA), resulting in a marked suppression of the plasma levels of very-low-density lipoprotein (VLDL)-associated triglycerides, a finding that has not previously been observed in mice. These data establish, in a model that is highly relevant to humans, that pharmacological inhibition of miR-33a and miR-33b is a promising therapeutic strategy to raise plasma HDL and lower VLDL triglyceride levels for the treatment of dyslipidaemias that increase cardiovascular disease risk.


Asunto(s)
Chlorocebus aethiops , Regulación de la Expresión Génica/efectos de los fármacos , Lipoproteínas HDL/sangre , Lipoproteínas VLDL/sangre , Hígado/efectos de los fármacos , MicroARNs/antagonistas & inhibidores , Oligorribonucleótidos Antisentido/farmacología , Triglicéridos/sangre , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Células Cultivadas , Chlorocebus aethiops/sangre , Chlorocebus aethiops/genética , Chlorocebus aethiops/metabolismo , LDL-Colesterol/sangre , Silenciador del Gen , Células HEK293 , Humanos , Hígado/metabolismo , Masculino , MicroARNs/metabolismo , Factores de Tiempo
2.
Circ Res ; 115(10): 826-33, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25239141

RESUMEN

RATIONALE: Cholesterol esters (CE), especially cholesterol oleate, generated by hepatic and intestinal sterol O-acyltransferase 2 (SOAT2) play a critical role in cholesterol homeostasis. However, it is unknown whether the contribution of intestine-derived CE from SOAT2 would have similar effects in promoting atherosclerosis progression as for liver-derived CE. OBJECTIVE: To test whether, in low-density lipoprotein receptor null (LDLr(-/-)) mice, the conditional knockout of intestinal SOAT2 (SOAT2(SI-/SI-)) or hepatic SOAT2 (SOAT2(L-/L-)) would equally limit atherosclerosis development compared with the global deletion of SOAT2 (SOAT2(-/-)). METHODS AND RESULTS: SOAT2 conditional knockout mice were bred with LDLr(-/-) mice creating LDLr(-/-) mice with each of the specific SOAT2 gene deletions. All mice then were fed an atherogenic diet for 16 weeks. SOAT2(SI-/SI-)LDLr(-/-) and SOAT2(-/-)LDLr(-/-) mice had significantly lower levels of intestinal cholesterol absorption, more fecal sterol excretion, and lower biliary cholesterol levels. Analysis of plasma LDL showed that all mice with SOAT2 gene deletions had LDL CE with reduced percentages of cholesterol palmitate and cholesterol oleate. Each of the LDLr(-/-) mice with SOAT2 gene deletions had lower accumulations of total cholesterol and CE in the liver compared with control mice. Finally, aortic atherosclerosis development was significantly lower in all mice with global or tissue-restricted SOAT2 gene deletions. Nevertheless, SOAT2(-/-)LDLr(-/-) and SOAT2(L-/L-)LDLr(-/-) mice had less aortic CE accumulation and smaller aortic lesions than SOAT2(SI-/SI-)LDLr(-/-) mice. CONCLUSIONS: SOAT2-derived CE from both the intestine and liver significantly contribute to the development of atherosclerosis, although the CE from the hepatic enzyme appeared to promote more atherosclerosis development.


Asunto(s)
Aorta/metabolismo , Aterosclerosis/metabolismo , Ésteres del Colesterol/metabolismo , Absorción Intestinal/fisiología , Hígado/metabolismo , Esterol O-Aciltransferasa/deficiencia , Animales , Aorta/patología , Aterosclerosis/sangre , Aterosclerosis/patología , Ésteres del Colesterol/sangre , Femenino , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Esterol O-Aciltransferasa 2
4.
Arterioscler Thromb Vasc Biol ; 34(9): 1880-7, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24969772

RESUMEN

OBJECTIVE: Excessive caloric intake is associated with obesity and adipose tissue dysfunction. However, the role of dietary cholesterol in this process is unknown. The aim of this study was to determine whether increasing dietary cholesterol intake alters adipose tissue cholesterol content, adipocyte size, and endocrine function in nonhuman primates. APPROACH AND RESULTS: Age-matched, male African Green monkeys (n=5 per group) were assigned to 1 of 3 diets containing 0.002 (low [Lo]), 0.2 (medium [Med]), or 0.4 (high [Hi]) mg cholesterol/kcal. After 10 weeks of diet feeding, animals were euthanized for adipose tissue, liver, and plasma collection. With increasing dietary cholesterol, free cholesterol (FC) content and adipocyte size increased in a stepwise manner in visceral, but not in subcutaneous fat, with a significant association between visceral adipocyte size and FC content (r(2)=0.298; n=15; P=0.035). In visceral fat, dietary cholesterol intake was associated with (1) increased proinflammatory gene expression and macrophage recruitment, (2) decreased expression of genes involved in cholesterol biosynthesis and lipoprotein uptake, and (3) increased expression of proteins involved in FC efflux. CONCLUSIONS: Increasing dietary cholesterol selectively increases visceral fat adipocyte size, FC and macrophage content, and proinflammatory gene expression in nonhuman primates. Visceral fat cells seem to compensate for increased dietary cholesterol by limiting cholesterol uptake/synthesis and increasing FC efflux pathways.


Asunto(s)
Adipocitos/efectos de los fármacos , Colesterol en la Dieta/toxicidad , Grasa Intraabdominal/efectos de los fármacos , Grasa Subcutánea/efectos de los fármacos , Adipocitos/patología , Animales , Tamaño de la Célula/efectos de los fármacos , Chlorocebus aethiops , Colesterol/análisis , Regulación de la Expresión Génica/efectos de los fármacos , Hipertrofia , Inflamación/genética , Grasa Intraabdominal/química , Grasa Intraabdominal/patología , Lipoproteínas/metabolismo , Hígado/química , Masculino , Especificidad de Órganos , Grasa Subcutánea/química , Grasa Subcutánea/patología
5.
J Shoulder Elbow Surg ; 22(5): 681-6, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22981355

RESUMEN

BACKGROUND: More than one-quarter of Americans have hypercholesterolemia and/or are being treated with cholesterol-lowering medications. Given the systemic nature of hypercholesterolemia and remaining questions regarding its effect on tendons at a local level, we sought to assess the utility of small versus large animal model systems for translational studies by exploring the effect of hypercholesterolemia on supraspinatus tendon elastic mechanical properties in mice, rats, and monkeys. We hypothesized that stiffness and elastic modulus would be increased in tendons across species due to hypercholesterolemia. MATERIALS AND METHODS: Supraspinatus tendons from normal (control) and high-cholesterol (HC) mice, rats, and monkeys were used in this study. After dissection, tendons were geometrically measured and tensile tested with tissue strain measured optically. RESULTS: Overall, HC animals had significantly altered plasma lipid profiles. Biomechanical testing showed a significant increase in stiffness compared with control in HC mice and rats, as well as a nonsignificant trend for HC monkeys. Elastic modulus was also significantly increased in HC mice and monkeys, with HC rats showing a trend. CONCLUSIONS: The consistency of our findings across species and between small and large animals, combined with the fact that the aged mice were exposed to lifelong hypercholesterolemia (compared with rats and nonhuman primates, which were fed HC diets), suggests that these increased properties may be inherent to the effect of hypercholesterolemia on supraspinatus tendon rather than due to an effect of cumulative exposure time to the effects of HC. Further investigation is needed to confirm this concept.


Asunto(s)
Módulo de Elasticidad/fisiología , Hipercolesterolemia/fisiopatología , Tendones/fisiopatología , Animales , Fenómenos Biomecánicos , Chlorocebus aethiops , Modelos Animales de Enfermedad , Hipercolesterolemia/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley
6.
J Lipid Res ; 53(6): 1144-52, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22460046

RESUMEN

Acyl-CoA:cholesterol acyltransferase 2 (ACAT2) generates cholesterol esters (CE) for packaging into newly synthesized lipoproteins and thus is a major determinant of blood cholesterol levels. ACAT2 is expressed exclusively in the small intestine and liver, but the relative contributions of ACAT2 expression in these tissues to systemic cholesterol metabolism is unknown. We investigated whether CE derived from the intestine or liver would differentially affect hepatic and plasma cholesterol homeostasis. We generated liver-specific (ACAT2(L-/L-)) and intestine-specific (ACAT2(SI-/SI-)) ACAT2 knockout mice and studied dietary cholesterol-induced hepatic lipid accumulation and hypercholesterolemia. ACAT2(SI-/SI-) mice, in contrast to ACAT2(L-/L-) mice, had blunted cholesterol absorption. However, specific deletion of ACAT2 in the intestine generated essentially a phenocopy of the conditional knockout of ACAT2 in the liver, with reduced levels of plasma very low-density lipoprotein and hepatic CE, yet hepatic-free cholesterol does not build up after high cholesterol intake. ACAT2(L-/L-) and ACAT2(SI-/SI-) mice were equally protected from diet-induced hepatic CE accumulation and hypercholesterolemia. These results suggest that inhibition of intestinal or hepatic ACAT2 improves atherogenic hyperlipidemia and limits hepatic CE accumulation in mice and that depletion of intestinal ACAT2 is sufficient for most of the beneficial effects on cholesterol metabolism. Inhibitors of ACAT2 targeting either tissue likely would be beneficial for atheroprotection.


Asunto(s)
Colesterol/metabolismo , Dieta/efectos adversos , Técnicas de Inactivación de Genes , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Esterol O-Aciltransferasa/deficiencia , Esterol O-Aciltransferasa/genética , Alelos , Animales , Sistema Biliar/metabolismo , Colesterol/sangre , Ésteres del Colesterol/metabolismo , Femenino , Hipercolesterolemia/etiología , Hipercolesterolemia/metabolismo , Hipercolesterolemia/prevención & control , Absorción Intestinal , Intestino Delgado/metabolismo , Ratones , Especificidad de Órganos , Esterol O-Aciltransferasa 2
7.
Cell Metab ; 20(5): 910-918, 2014 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-25440061

RESUMEN

The LXR-regulated E3 ubiquitin ligase IDOL controls LDLR receptor stability independent of SREBP and PCSK9, but its relevance to plasma lipid levels is unknown. Here we demonstrate that the effects of the LXR-IDOL axis are both tissue and species specific. In mice, LXR agonist induces Idol transcript levels in peripheral tissues but not in liver, and does not change plasma LDL levels. Accordingly, Idol-deficient mice exhibit elevated LDLR protein levels in peripheral tissues, but not in the liver. By contrast, LXR activation in cynomolgus monkeys induces hepatic IDOL expression, reduces LDLR protein levels, and raises plasma LDL levels. Knockdown of IDOL in monkeys with an antisense oligonucleotide blunts the effect of LXR agonist on LDL levels. These results implicate IDOL as a modulator of plasma lipid levels in primates and support further investigation into IDOL inhibition as a potential strategy for LDL lowering in humans.


Asunto(s)
LDL-Colesterol/sangre , Hígado/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Células Cultivadas , LDL-Colesterol/metabolismo , Haplorrinos , Humanos , Receptores X del Hígado , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de LDL/metabolismo , Especificidad de la Especie
8.
PLoS One ; 9(6): e98953, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24901470

RESUMEN

The primary risk factor for atherosclerotic cardiovascular disease is LDL cholesterol, which can be reduced by increasing cholesterol excretion from the body. Fecal cholesterol excretion can be driven by a hepatobiliary as well as a non-biliary pathway known as transintestinal cholesterol efflux (TICE). We previously showed that chronic knockdown of the hepatic cholesterol esterifying enzyme sterol O-acyltransferase 2 (SOAT2) increased fecal cholesterol loss via TICE. To elucidate the initial events that stimulate TICE, C57Bl/6 mice were fed a high cholesterol diet to induce hepatic cholesterol accumulation and were then treated for 1 or 2 weeks with an antisense oligonucleotide targeting SOAT2. Within 2 weeks of hepatic SOAT2 knockdown (SOAT2HKD), the concentration of cholesteryl ester in the liver was reduced by 70% without a reciprocal increase in hepatic free cholesterol. The rapid mobilization of hepatic cholesterol stores resulted in a ∼ 2-fold increase in fecal neutral sterol loss but no change in biliary cholesterol concentration. Acute SOAT2HKD increased plasma cholesterol carried primarily in lipoproteins enriched in apoB and apoE. Collectively, our data suggest that acutely reducing SOAT2 causes hepatic cholesterol to be swiftly mobilized and packaged onto nascent lipoproteins that feed cholesterol into the TICE pathway for fecal excretion.


Asunto(s)
Colesterol/metabolismo , Heces/química , Esterol O-Aciltransferasa/metabolismo , Animales , Apolipoproteínas B/metabolismo , Apolipoproteínas E/metabolismo , Bilis/metabolismo , Colesterol/análisis , Colesterol/sangre , Ésteres del Colesterol/metabolismo , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Dieta Alta en Grasa , Femenino , Vesícula Biliar/metabolismo , Intestino Delgado/metabolismo , Lipoproteínas/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Oligonucleótidos Antisentido/farmacología , ARN Mensajero/metabolismo , Esterol O-Aciltransferasa/antagonistas & inhibidores , Esterol O-Aciltransferasa/genética , Esterol O-Aciltransferasa 2
9.
PLoS One ; 9(1): e84418, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24404162

RESUMEN

An effective way to reduce LDL cholesterol, the primary risk factor of atherosclerotic cardiovascular disease, is to increase cholesterol excretion from the body. Our group and others have recently found that cholesterol excretion can be facilitated by both hepatobiliary and transintestinal pathways. However, the lipoprotein that moves cholesterol through the plasma to the small intestine for transintestinal cholesterol efflux (TICE) is unknown. To test the hypothesis that hepatic very low-density lipoproteins (VLDL) support TICE, antisense oligonucleotides (ASO) were used to knockdown hepatic expression of microsomal triglyceride transfer protein (MTP), which is necessary for VLDL assembly. While maintained on a high cholesterol diet, Niemann-Pick C1-like 1 hepatic transgenic (L1Tg) mice, which predominantly excrete cholesterol via TICE, and wild type (WT) littermates were treated with control ASO or MTP ASO. In both WT and L1Tg mice, MTP ASO decreased VLDL triglyceride (TG) and cholesterol secretion. Regardless of treatment, L1Tg mice had reduced biliary cholesterol compared to WT mice. However, only L1Tg mice treated with MTP ASO had reduced fecal cholesterol excretion. Based upon these findings, we conclude that VLDL or a byproduct such as LDL can move cholesterol from the liver to the small intestine for TICE.


Asunto(s)
Colesterol/metabolismo , Hepatocitos/metabolismo , Lipoproteínas VLDL/metabolismo , Proteínas de Transporte de Membrana/genética , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Expresión Génica , Técnicas de Silenciamiento del Gen , Lipoproteínas/metabolismo , Lipoproteínas VLDL/sangre , Hígado/metabolismo , Masculino , Ratones , Ratones Transgénicos , Oligorribonucleótidos Antisentido/administración & dosificación , Oligorribonucleótidos Antisentido/genética , Receptores de LDL/metabolismo
10.
J Clin Invest ; 122(2): 558-68, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22214850

RESUMEN

Hypercholesterolemia is a major risk factor for atherosclerosis. It also is associated with platelet hyperactivity, which increases morbidity and mortality from cardiovascular disease. However, the mechanisms by which hypercholesterolemia produces a procoagulant state remain undefined. Atherosclerosis is associated with accumulation of oxidized lipoproteins within atherosclerotic lesions. Small quantities of oxidized lipoproteins are also present in the circulation of patients with coronary artery disease. We therefore hypothesized that hypercholesterolemia leads to elevated levels of oxidized LDL (oxLDL) in plasma and that this induces expression of the procoagulant protein tissue factor (TF) in monocytes. In support of this hypothesis, we report here that oxLDL induced TF expression in human monocytic cells and monocytes. In addition, patients with familial hypercholesterolemia had elevated levels of plasma microparticle (MP) TF activity. Furthermore, a high-fat diet induced a time-dependent increase in plasma MP TF activity and activation of coagulation in both LDL receptor-deficient mice and African green monkeys. Genetic deficiency of TF in bone marrow cells reduced coagulation in hypercholesterolemic mice, consistent with a major role for monocyte-derived TF in the activation of coagulation. Similarly, a deficiency of either TLR4 or TLR6 reduced levels of MP TF activity. Simvastatin treatment of hypercholesterolemic mice and monkeys reduced oxLDL, monocyte TF expression, MP TF activity, activation of coagulation, and inflammation, without affecting total cholesterol levels. Our results suggest that the prothrombotic state associated with hypercholesterolemia is caused by oxLDL-mediated induction of TF expression in monocytes via engagement of a TLR4/TLR6 complex.


Asunto(s)
Anticolesterolemiantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Hipercolesterolemia/sangre , Monocitos/metabolismo , Simvastatina/farmacología , Tromboplastina/metabolismo , Animales , Coagulación Sanguínea/fisiología , Células Cultivadas , Chlorocebus aethiops , Humanos , Lipoproteínas LDL/metabolismo , Masculino , Ratones , Receptores de LDL/genética , Receptores de LDL/metabolismo , Trombosis , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 6/genética , Receptor Toll-Like 6/metabolismo
11.
PLoS One ; 6(4): e19420, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21559365

RESUMEN

Genes involved in cholesterol biosynthesis and uptake are transcriptionally regulated in response to cellular sterol content in a coordinated manner. A number of these genes, including 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) and LDL receptor (LDLR), undergo alternative splicing, resulting in reductions of enzyme or protein activity. Here we demonstrate that cellular sterol depletion suppresses, and sterol loading induces, alternative splicing of multiple genes involved in the maintenance of cholesterol homeostasis including HMGCR and LDLR, the key regulators of cellular cholesterol biosynthesis and uptake, respectively. These changes were observed in both in vitro studies of the HepG2 human hepatoma derived cell line, as well as in vivo studies of St. Kitts vervets, also known as African green monkeys, a commonly used primate model for investigating cholesterol metabolism. These effects are mediated in part by sterol regulation of polypyrimidine tract binding protein 1 (PTBP1), since knock-down of PTBP1 eliminates sterol induced changes in alternative splicing of several of these genes. Single nucleotide polymorphisms (SNPs) that influence HMGCR and LDLR alternative splicing (rs3846662 and rs688, respectively), have been associated with variation in plasma LDL-cholesterol levels. Sterol-induced changes in alternative splicing are blunted in carriers of the minor alleles for each of these SNPs, indicating an interaction between genetic and non-genetic regulation of this process. Our results implicate alternative splicing as a novel mechanism of enhancing the robust transcriptional response to conditions of cellular cholesterol depletion or accumulation. Thus coordinated regulation of alternative splicing may contribute to cellular cholesterol homeostasis as well as plasma LDL levels.


Asunto(s)
Empalme Alternativo , Colesterol/metabolismo , Regulación de la Expresión Génica , Alelos , Animales , Línea Celular , Chlorocebus aethiops , Exones , Humanos , Hidroximetilglutaril-CoA Reductasas/metabolismo , Masculino , Polimorfismo de Nucleótido Simple , Receptores de LDL/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA