Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 42(2): 127-144, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34911361

RESUMEN

OBJECTIVE: Nobiletin is a dietary flavonoid that improves insulin resistance and atherosclerosis in mice with metabolic dysfunction. Dysregulation of intestinal lipoprotein metabolism contributes to atherogenesis. The objective of the study was to determine if nobiletin targets the intestine to improve metabolic dysregulation in both male and female mice. Approach and Results: Triglyceride-rich lipoprotein (TRL) secretion, intracellular triglyceride kinetics, and intestinal morphology were determined in male and female LDL (low-density lipoprotein) receptor knockout (Ldlr-/-), and male wild-type mice fed a standard laboratory diet or high-fat, high-cholesterol (HFHC) diet ± nobiletin using an olive oil gavage, radiotracers, and electron microscopy. Nobiletin attenuated postprandial TRL levels in plasma and enhanced TRL clearance. Nobiletin reduced fasting jejunal triglyceride accumulation through accelerated TRL secretion and lower jejunal fatty acid synthesis with no impact on fatty acid oxidation. Fasting-refeeding experiments revealed that nobiletin led to higher levels of phosphorylated AKT (protein kinase B) and FoxO1 (forkhead box O1) and normal Srebf1c expression indicating increased insulin sensitivity. Intestinal length and weight were diminished by HFHC feeding and restored by nobiletin. Both fasting and postprandial plasma GLP-1 (glucagon-like peptide-1; and likely GLP-2) were elevated in response to nobiletin. Treatment with a GLP-2 receptor antagonist, GLP-2(3-33), reduced villus length in HFHC-fed mice but did not impact TRL secretion in any diet group. In contrast to males, nobiletin did not improve postprandial lipid parameters in female mice. CONCLUSIONS: Nobiletin opposed the effects of the HFHC diet by normalizing intestinal de novo lipogenesis through improved insulin sensitivity. Nobiletin prevents postprandial lipemia because the enhanced TRL clearance more than compensates for increased TRL secretion.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Flavonas/farmacología , Hiperlipidemias/prevención & control , Metabolismo de los Lípidos/efectos de los fármacos , Sustancias Protectoras/farmacología , Animales , Femenino , Flavonas/uso terapéutico , Hiperlipidemias/sangre , Hiperlipidemias/metabolismo , Intestinos/efectos de los fármacos , Intestinos/metabolismo , Masculino , Ratones Endogámicos C57BL , Periodo Posprandial , Sustancias Protectoras/uso terapéutico , Triglicéridos/sangre , Triglicéridos/metabolismo
2.
J Lipid Res ; 61(3): 387-402, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31964763

RESUMEN

Obesity, dyslipidemia, and insulin resistance, the increasingly common metabolic syndrome, are risk factors for CVD and type 2 diabetes that warrant novel therapeutic interventions. The flavonoid nobiletin displays potent lipid-lowering and insulin-sensitizing properties in mice with metabolic dysfunction. However, the mechanisms by which nobiletin mediates metabolic protection are not clearly established. The central role of AMP-activated protein kinase (AMPK) as an energy sensor suggests that AMPK is a target of nobiletin. We tested the hypothesis that metabolic protection by nobiletin required phosphorylation of AMPK and acetyl-CoA carboxylase (ACC) in mouse hepatocytes, in mice deficient in hepatic AMPK (Ampkß1-/-), in mice incapable of inhibitory phosphorylation of ACC (AccDKI), and in mice with adipocyte-specific AMPK deficiency (iß1ß2AKO). We fed mice a high-fat/high-cholesterol diet with or without nobiletin. Nobiletin increased phosphorylation of AMPK and ACC in primary mouse hepatocytes, which was associated with increased FA oxidation and attenuated FA synthesis. Despite loss of ACC phosphorylation in Ampkß1-/- hepatocytes, nobiletin suppressed FA synthesis and enhanced FA oxidation. Acute injection of nobiletin into mice did not increase phosphorylation of either AMPK or ACC in liver. In mice fed a high-fat diet, nobiletin robustly prevented obesity, hepatic steatosis, dyslipidemia, and insulin resistance, and it improved energy expenditure in Ampkß1-/-, AccDKI, and iß1ß2AKO mice to the same extent as in WT controls. Thus, the beneficial metabolic effects of nobiletin in vivo are conferred independently of hepatic or adipocyte AMPK activation. These studies further underscore the therapeutic potential of nobiletin and begin to clarify possible mechanisms.


Asunto(s)
Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Antioxidantes/farmacología , Citrus/química , Flavonas/farmacología , Sustancias Protectoras/farmacología , Proteínas Quinasas Activadas por AMP/deficiencia , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Antioxidantes/química , Dieta Alta en Grasa/efectos adversos , Flavonas/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sustancias Protectoras/química
3.
Annu Rev Pharmacol Toxicol ; 57: 223-244, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-27575716

RESUMEN

Proprotein convertase subtilisin/kexin type-9 (PCSK9) is a secreted zymogen expressed primarily in the liver. PCSK9 circulates in plasma, binds to cell surface low-density lipoprotein (LDL) receptors, is internalized, and then targets the receptors to lysosomal degradation. Studies of naturally occurring PCSK9 gene variants that caused extreme plasma LDL cholesterol (LDL-C) deviations and altered atherosclerosis risk unleashed a torrent of biological and pharmacological research. Rapid progress in understanding the physiological regulation of PCSK9 was soon translated into commercially available biological inhibitors of PCSK9 that reduced LDL-C levels and likely also cardiovascular outcomes. Here we review the swift evolution of PCSK9 from novel gene to drug target, to animal and human testing, and finally to outcome trials and clinical applications. In addition, we explore how the genetics-guided path to PCSK9 inhibitor development exemplifies a new paradigm in pharmacology. Finally, we consider some potential challenges as PCSK9 inhibition becomes established in the clinic.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Dislipidemias/tratamiento farmacológico , Inhibidores de PCSK9 , Proproteína Convertasa 9/fisiología , Animales , LDL-Colesterol/antagonistas & inhibidores , LDL-Colesterol/sangre , Sistemas de Liberación de Medicamentos/tendencias , Descubrimiento de Drogas/tendencias , Dislipidemias/sangre , Dislipidemias/enzimología , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/metabolismo , Humanos , Hipercolesterolemia/sangre , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/enzimología
4.
Curr Opin Lipidol ; 30(1): 1-9, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30586346

RESUMEN

PURPOSE OF REVIEW: Bempedoic acid has emerged as a potent inhibitor of ATP-citrate lyase (ACLY), a target for the reduction of LDL cholesterol (LDL-C). We review the impact of bempedoic acid treatment on lipoprotein metabolism and atherosclerosis in preclinical models and patients with hypercholesterolemia. RECENT FINDINGS: The liver-specific activation of bempedoic acid inhibits ACLY, a key enzyme linking glucose catabolism to lipogenesis by catalyzing the formation of acetyl-CoA from mitochondrial-derived citrate for de novo synthesis of fatty acids and cholesterol. Adenosine monophosphate-activated protein kinase activation by bempedoic acid is not required for its lipid-regulating effects in vivo. Mendelian randomization of large human study cohorts has validated ACLY inhibition as a target for LDL-C lowering and atheroprotection. In rodents, bempedoic acid decreases plasma cholesterol and triglycerides, and prevents hepatic steatosis. In apolipoprotein E-deficient (Apoe) mice, LDL receptor-deficient (Ldlr) mice and LDLR-deficient miniature pigs, bempedoic acid reduces LDL-C and attenuates atherosclerosis. LDLR expression and activity are increased in primary human hepatocytes and in Apoe mouse liver treated with bempedoic acid suggesting a mechanism for LDL-C lowering, although additional pathways are likely involved. Phase 2 and 3 clinical trials revealed that bempedoic acid effectively lowers LDL-C as monotherapy, combined with ezetimibe, added to statin therapy and in statin-intolerant hypercholesterolemic patients. Treatment does not affect plasma concentrations of triglyceride or other lipoproteins. SUMMARY: The LDL-C-lowering and attenuated atherosclerosis in animal models and reduced LDL-C in hypercholesterolemic patients has validated ACLY inhibition as a therapeutic strategy. Positive results from phase 3 long-term cardiovascular outcome trials in high-risk patients are required for bempedoic acid to be approved for prevention of atherosclerosis.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Ácidos Dicarboxílicos/farmacología , Ácidos Grasos/farmacología , Lipoproteínas/metabolismo , ATP Citrato (pro-S)-Liasa/antagonistas & inhibidores , Animales , Ácidos Dicarboxílicos/uso terapéutico , Ácidos Grasos/uso terapéutico , Humanos , Terapia Molecular Dirigida , Triglicéridos/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 38(5): 1178-1190, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29449335

RESUMEN

OBJECTIVE: Bempedoic acid (BemA; ETC-1002) is a novel drug that targets hepatic ATP-citrate lyase to reduce cholesterol biosynthesis. In phase 2 studies, BemA lowers elevated low-density lipoprotein cholesterol (LDL-C) in hypercholesterolemic patients. In the present study, we tested the ability of BemA to decrease plasma cholesterol and LDL-C and attenuate atherosclerosis in a large animal model of familial hypercholesterolemia. APPROACH AND RESULTS: Gene targeting has been used to generate Yucatan miniature pigs heterozygous (LDLR+/-) or homozygous (LDLR-/-) for LDL receptor deficiency (ExeGen). LDLR+/- and LDLR-/- pigs were fed a high-fat, cholesterol-containing diet (34% kcal fat; 0.2% cholesterol) and orally administered placebo or BemA for 160 days. In LDLR+/- pigs, compared with placebo, BemA decreased plasma cholesterol and LDL-C up to 40% and 61%, respectively. In LDLR-/- pigs, in which plasma cholesterol and LDL-C were 5-fold higher than in LDLR+/- pigs, BemA decreased plasma cholesterol and LDL-C up to 27% and 29%, respectively. Plasma levels of triglycerides and high-density lipoprotein cholesterol, fasting glucose and insulin, and liver lipids were unaffected by treatment in either genotype. In the aorta of LDLR+/- pigs, BemA robustly attenuated en face raised lesion area (-58%) and left anterior descending coronary artery cross-sectional lesion area (-40%). In LDLR-/- pigs, in which lesions were substantially more advanced, BemA decreased aortic lesion area (-47%) and left anterior descending coronary artery lesion area (-48%). CONCLUSIONS: In a large animal model of LDLR deficiency and atherosclerosis, long-term treatment with BemA reduces LDL-C and attenuates the development of aortic and coronary atherosclerosis in both LDLR+/- and LDLR-/- miniature pigs.


Asunto(s)
Anticolesterolemiantes/farmacología , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , LDL-Colesterol/sangre , Enfermedad de la Arteria Coronaria/prevención & control , Ácidos Dicarboxílicos/farmacología , Ácidos Grasos/farmacología , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Receptores de LDL/deficiencia , Animales , Animales Modificados Genéticamente , Anticolesterolemiantes/farmacocinética , Enfermedades de la Aorta/sangre , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/patología , Biomarcadores/sangre , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/patología , Ácidos Dicarboxílicos/farmacocinética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Ácidos Grasos/farmacocinética , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/genética , Masculino , Fenotipo , Placa Aterosclerótica , Receptores de LDL/genética , Porcinos , Porcinos Enanos
6.
Curr Opin Lipidol ; 29(2): 87-94, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29369832

RESUMEN

PURPOSE OF REVIEW: Regression, or reversal, of atherosclerosis has become an important clinical objective. The development of consistent models of murine atherosclerosis regression has accelerated this field of research. The purpose of this review is to highlight recent mouse studies that reveal molecular mechanisms as well as therapeutics targeted for regression. RECENT FINDINGS: Atherosclerosis regression does not involve the same mechanisms as progression in reverse order. Distinct molecular processes within the plaque characterize regression. These processes remained elusive until the advent of murine regression models including aortic transplant, the Reversa mouse, gene complementation and dietary intervention. Studies revealed that depletion of plaque macrophages is a quintessential characteristic of regression, driven by reduced monocyte recruitment into plaques, increased egress of macrophages from plaques and reduced macrophage proliferation. In addition, regression results in polarization of remaining plaque macrophages towards an anti-inflammatory phenotype, smaller necrotic cores and promotion of an organized fibrous cap. Furthermore, type 1 diabetes hinders plaque regression, and several therapeutic interventions show promise in slowing plaque progression or inducing regression. SUMMARY: Mouse models of atherosclerosis regression have accelerated our understanding of the molecular mechanisms governing lesion resolution. These insights will be valuable in identifying therapeutic targets aimed at atherosclerosis regression.


Asunto(s)
Aterosclerosis , Animales , Animales Modificados Genéticamente , Aterosclerosis/complicaciones , Aterosclerosis/genética , Aterosclerosis/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Ratones
7.
J Lipid Res ; 59(9): 1714-1728, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30008441

RESUMEN

Obesity and its associated metabolic dysfunction and cardiovascular disease risk represent a leading cause of adult morbidity worldwide. Currently available pharmacological therapies for obesity have had limited success in reversing existing obesity and metabolic dysregulation. Previous prevention studies demonstrated that the citrus flavonoids, naringenin and nobiletin, protect against obesity and metabolic dysfunction in Ldlr-/- mice fed a high-fat cholesterol-containing (HFHC) diet. However, their effects in an intervention model are unknown. In this report, we show that, in Ldlr-/- mice with diet-induced obesity, citrus flavonoid supplementation to a HFHC diet reversed existing obesity and adipocyte size and number through enhanced energy expenditure and increased hepatic fatty acid oxidation. Caloric intake was unaffected and no evidence of white adipose tissue browning was observed. Reversal of adiposity was accompanied by improvements in hyperlipidemia, insulin sensitivity, hepatic steatosis, and a modest reduction in blood monocytes. Together, this resulted in atherosclerotic lesions that were unchanged in size, but characterized by reduced macrophage content, consistent with a more stable plaque phenotype. These studies further suggest potential therapeutic utility of citrus flavonoids, especially in the context of existing obesity, metabolic dysfunction, and cardiovascular disease.


Asunto(s)
Aterosclerosis/complicaciones , Citrus/química , Flavonoides/farmacología , Síndrome Metabólico/complicaciones , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Receptores de LDL/deficiencia , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Animales , Peso Corporal/efectos de los fármacos , Colesterol/metabolismo , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/efectos de los fármacos , Flavonoides/uso terapéutico , Hiperlipidemias/complicaciones , Resistencia a la Insulina , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Obesidad/metabolismo , Obesidad/patología
8.
Arterioscler Thromb Vasc Biol ; 37(4): 647-656, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28153881

RESUMEN

OBJECTIVE: Bempedoic acid (ETC-1002, 8-hydroxy-2,2,14,14-tetramethylpentadecanedioic acid) is a novel low-density lipoprotein cholesterol-lowering compound. In animals, bempedoic acid targets the liver where it inhibits cholesterol and fatty acid synthesis through inhibition of ATP-citrate lyase and through activation of AMP-activated protein kinase. In this study, we tested the hypothesis that bempedoic acid would prevent diet-induced metabolic dysregulation, inflammation, and atherosclerosis. APPROACH AND RESULTS: Ldlr-/- mice were fed a high-fat, high-cholesterol diet (42% kcal fat, 0.2% cholesterol) supplemented with bempedoic acid at 0, 3, 10 and 30 mg/kg body weight/day. Treatment for 12 weeks dose-dependently attenuated diet-induced hypercholesterolemia, hypertriglyceridemia, hyperglycemia, hyperinsulinemia, fatty liver and obesity. Compared to high-fat, high-cholesterol alone, the addition of bempedoic acid decreased plasma triglyceride (up to 64%) and cholesterol (up to 50%) concentrations, and improved glucose tolerance. Adiposity was significantly reduced with treatment. In liver, bempedoic acid prevented cholesterol and triglyceride accumulation, which was associated with increased fatty acid oxidation and reduced fatty acid synthesis. Hepatic gene expression analysis revealed that treatment significantly increased expression of genes involved in fatty acid oxidation while suppressing inflammatory gene expression. In full-length aorta, bempedoic acid markedly suppressed cholesteryl ester accumulation, attenuated the expression of proinflammatory M1 genes and attenuated the iNos/Arg1 ratio. Treatment robustly attenuated atherosclerotic lesion development in the aortic sinus by 44%, with beneficial changes in morphology, characteristic of earlier-stage lesions. CONCLUSIONS: Bempedoic acid effectively prevents plasma and tissue lipid elevations and attenuates the onset of inflammation, leading to the prevention of atherosclerotic lesion development in a mouse model of metabolic dysregulation.


Asunto(s)
ATP Citrato (pro-S)-Liasa/antagonistas & inhibidores , Aterosclerosis/prevención & control , Ácidos Dicarboxílicos/farmacología , Dieta Alta en Grasa , Dislipidemias/prevención & control , Inhibidores Enzimáticos/farmacología , Ácidos Grasos/farmacología , Inflamación/prevención & control , Hígado/efectos de los fármacos , Obesidad/prevención & control , Receptores de LDL/deficiencia , ATP Citrato (pro-S)-Liasa/metabolismo , Animales , Aterosclerosis/sangre , Aterosclerosis/enzimología , Aterosclerosis/genética , Biomarcadores/sangre , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Modelos Animales de Enfermedad , Dislipidemias/sangre , Dislipidemias/enzimología , Dislipidemias/genética , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Inflamación/sangre , Inflamación/enzimología , Inflamación/genética , Mediadores de Inflamación/sangre , Insulina/sangre , Lípidos/sangre , Hígado/enzimología , Masculino , Ratones Noqueados , Obesidad/sangre , Obesidad/enzimología , Obesidad/genética , Fenotipo , Receptores de LDL/genética , Factores de Tiempo
9.
Curr Opin Lipidol ; 28(2): 193-200, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28059952

RESUMEN

PURPOSE OF REVIEW: ATP-citrate lyase (ACLY) has re-emerged as a drug target for LDL cholesterol (LDL-C) lowering. We review ACLY as a therapeutic strategy, its genetics, its molecular and cellular biology, and also its inhibition. RECENT FINDINGS: ACLY is a critical enzyme linking glucose catabolism to lipogenesis by providing acetyl-CoA from mitochondrial citrate for fatty acid and cholesterol biosynthesis. Human genetic variants have been associated with enhanced growth and survival of several cancers, and with attenuated plasma triglyceride responses to dietary fish oil. In mice, liver-specific Acly deficiency protects from hepatic steatosis and dyslipidemia, whereas adipose tissue-specific Acly deletion has no phenotype, supporting therapeutic inhibition of ACLY. A lipid-regulating compound, bempedoic acid, was discovered to potently inhibit ACLY, and in animal models, it prevents dyslipidemia and attenuates atherosclerosis. Phase 2 clinical trials revealed that bempedoic acid effectively lowers LDL-C as monotherapy, combined with ezetimibe, added to statin therapy and in statin-intolerant hypercholesterolemic patients. SUMMARY: The efficacy of bempedoic acid as an LDL-C-lowering agent has validated ACLY inhibition as a therapeutic strategy. Positive results of phase 3 patient studies, together with long-term cardiovascular disease outcome trials, are required to establish ACLY as a major new target in cardiovascular medicine.


Asunto(s)
ATP Citrato (pro-S)-Liasa/genética , ATP Citrato (pro-S)-Liasa/metabolismo , Dislipidemias/tratamiento farmacológico , Dislipidemias/enzimología , Terapia Molecular Dirigida/métodos , ATP Citrato (pro-S)-Liasa/antagonistas & inhibidores , Animales , Ácidos Dicarboxílicos/metabolismo , Dislipidemias/genética , Dislipidemias/patología , Inhibidores Enzimáticos/farmacología , Ácidos Grasos/metabolismo , Humanos
10.
Annu Rev Nutr ; 36: 275-99, 2016 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-27146015

RESUMEN

Citrus flavonoids are polyphenolic compounds with significant biological properties. This review summarizes recent advances in understanding the ability of citrus flavonoids to modulate lipid metabolism, other metabolic parameters related to the metabolic syndrome, and atherosclerosis. Citrus flavonoids, including naringenin, hesperitin, nobiletin, and tangeretin, have emerged as potential therapeutics for the treatment of metabolic dysregulation. Epidemiological studies reveal an association between the intake of citrus flavonoid-containing foods and a decreased incidence of cardiovascular disease. Studies in cell culture and animal models, as well as a limited number of clinical studies, reveal the lipid-lowering, insulin-sensitizing, antihypertensive, and anti-inflammatory properties of citrus flavonoids. In animal models, supplementation of rodent diets with citrus flavonoids prevents hepatic steatosis, dyslipidemia, and insulin resistance primarily through inhibition of hepatic fatty acid synthesis and increased fatty acid oxidation. Citrus flavonoids blunt the inflammatory response in metabolically important tissues including liver, adipose, kidney, and the aorta. The mechanisms underlying flavonoid-induced metabolic regulation have not been completely established, although several potential targets have been identified. In mouse models, citrus flavonoids show marked suppression of atherogenesis through improved metabolic parameters as well as through direct impact on the vessel wall. Recent studies support a role for citrus flavonoids in the treatment of dyslipidemia, insulin resistance, hepatic steatosis, obesity, and atherosclerosis. Larger human studies examining dose, bioavailability, efficacy, and safety are required to promote the development of these promising therapeutic agents.


Asunto(s)
Aterosclerosis/prevención & control , Citrus/química , Suplementos Dietéticos , Flavonoides/uso terapéutico , Hiperlipidemias/dietoterapia , Hipolipemiantes/uso terapéutico , Lipoproteínas/metabolismo , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Fármacos Antiobesidad/uso terapéutico , Antioxidantes/uso terapéutico , Aterosclerosis/epidemiología , Aterosclerosis/etiología , Diabetes Mellitus Tipo 2/dietoterapia , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Humanos , Hiperlipidemias/inmunología , Hiperlipidemias/metabolismo , Hiperlipidemias/fisiopatología , Resistencia a la Insulina , Lipoproteínas/sangre , Enfermedad del Hígado Graso no Alcohólico/dietoterapia , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Sobrepeso/dietoterapia , Sobrepeso/inmunología , Sobrepeso/metabolismo , Sobrepeso/fisiopatología , Factores de Riesgo
11.
Arterioscler Thromb Vasc Biol ; 36(12): 2439-2445, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27765764

RESUMEN

OBJECTIVE: Next-generation sequencing technology is transforming our understanding of heterozygous familial hypercholesterolemia, including revision of prevalence estimates and attribution of polygenic effects. Here, we examined the contributions of monogenic and polygenic factors in patients with severe hypercholesterolemia referred to a specialty clinic. APPROACH AND RESULTS: We applied targeted next-generation sequencing with custom annotation, coupled with evaluation of large-scale copy number variation and polygenic scores for raised low-density lipoprotein cholesterol in a cohort of 313 individuals with severe hypercholesterolemia, defined as low-density lipoprotein cholesterol >5.0 mmol/L (>194 mg/dL). We found that (1) monogenic familial hypercholesterolemia-causing mutations detected by targeted next-generation sequencing were present in 47.3% of individuals; (2) the percentage of individuals with monogenic mutations increased to 53.7% when copy number variations were included; (3) the percentage further increased to 67.1% when individuals with extreme polygenic scores were included; and (4) the percentage of individuals with an identified genetic component increased from 57.0% to 92.0% as low-density lipoprotein cholesterol level increased from 5.0 to >8.0 mmol/L (194 to >310 mg/dL). CONCLUSIONS: In a clinically ascertained sample with severe hypercholesterolemia, we found that most patients had a discrete genetic basis detected using a comprehensive screening approach that includes targeted next-generation sequencing, an assay for copy number variations, and polygenic trait scores.


Asunto(s)
Hiperlipoproteinemia Tipo II/genética , Herencia Multifactorial , Mutación , Adulto , Anciano , Biomarcadores/sangre , LDL-Colesterol/sangre , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Femenino , Dosificación de Gen , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Herencia , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/diagnóstico , Masculino , Persona de Mediana Edad , Ontario , Fenotipo , Valor Predictivo de las Pruebas , Factores de Riesgo , Índice de Severidad de la Enfermedad
12.
Arterioscler Thromb Vasc Biol ; 35(1): 213-21, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25395619

RESUMEN

OBJECTIVE: Estrogen deficiency is linked with increased low-density lipoprotein (LDL) cholesterol. The hormone receptor mediating this effect is unknown. G-protein estrogen receptor (GPER) is a recently recognized G-protein-coupled receptor that is activated by estrogens. We recently identified a common hypofunctional missense variant of GPER, namely P16L. However, the role of GPER in LDL metabolism is unknown. Therefore, we examined the association of the P16L genotype with plasma LDL cholesterol level. Furthermore, we studied the role of GPER in regulating expression of the LDL receptor and proprotein convertase subtilisin kexin type 9. APPROACH AND RESULTS: Our discovery cohort was a genetically isolated population of Northern European descent, and our validation cohort consisted of normal, healthy women aged 18 to 56 years from London, Ontario. In addition, we examined the effect of GPER on the regulation of proprotein convertase subtilisin kexin type 9 and LDL receptor expression by the treatment with the GPER agonist, G1. In the discovery cohort, GPER P16L genotype was associated with a significant increase in LDL cholesterol (mean±SEM): 3.18±0.05, 3.25±0.08, and 4.25±0.33 mmol/L, respectively, in subjects with CC (homozygous for P16), CT (heterozygotes), and TT (homozygous for L16) genotypes (P<0.05). In the validation cohort (n=339), the GPER P16L genotype was associated with a similar increase in LDL cholesterol: 2.17±0.05, 2.34±0.06, and 2.42±0.16 mmol/L, respectively, in subjects with CC, CT, and TT genotypes (P<0.05). In the human hepatic carcinoma cell line, the GPER agonist, G1, mediated a concentration-dependent increase in LDL receptor expression, blocked by either pretreatment with the GPER antagonist G15 or by shRNA-mediated GPER downregulation. G1 also mediated a GPER- and concentration-dependent decrease in proprotein convertase subtilisin kexin type 9 expression. CONCLUSIONS: GPER activation upregulates LDL receptor expression, probably at least, in part, via proprotein convertase subtilisin kexin type 9 downregulation. Furthermore, humans carrying the hypofunctional P16L genetic variant of GPER have increased plasma LDL cholesterol. In aggregate, these data suggest an important role of GPER in the regulation of LDL receptor expression and consequently LDL metabolism.


Asunto(s)
LDL-Colesterol/sangre , Mutación Missense , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Adolescente , Adulto , Anciano , Canadá , Relación Dosis-Respuesta a Droga , Femenino , Regulación de la Expresión Génica , Frecuencia de los Genes , Genética de Población , Células Hep G2 , Heterocigoto , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Proproteína Convertasa 9 , Proproteína Convertasas/genética , Proproteína Convertasas/metabolismo , Interferencia de ARN , Receptores de Estrógenos/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores de LDL/genética , Receptores de LDL/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Transfección , Población Blanca/genética , Adulto Joven
13.
Arterioscler Thromb Vasc Biol ; 34(1): 52-60, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24158519

RESUMEN

OBJECTIVE: The peroxisome proliferator-activated receptor (PPAR) δ regulates systemic lipid homeostasis and inflammation. However, the ability of PPARδ agonists to improve the pathology of pre-established lesions and whether PPARδ activation is atheroprotective in the setting of insulin resistance have not been reported. Here, we examine whether intervention with a selective PPARδ agonist corrects metabolic dysregulation and attenuates aortic inflammation and atherosclerosis. APPROACH AND RESULTS: Low-density lipoprotein receptor knockout mice were fed a chow or a high-fat, high-cholesterol (HFHC) diet (42% fat, 0.2% cholesterol) for 4 weeks. For a further 8 weeks, the HFHC group was fed either HFHC or HFHC plus GW1516 (3 mg/kg per day). GW1516 significantly attenuated pre-established fasting hyperlipidemia, hyperglycemia, and hyperinsulinemia, as well as glucose and insulin intolerance. GW1516 intervention markedly reduced aortic sinus lesions and lesion macrophages, whereas smooth muscle α-actin was unchanged and collagen deposition enhanced. In aortae, GW1516 increased the expression of the PPARδ-specific gene Adfp but not PPARα- or γ-specific genes. GW1516 intervention decreased the expression of aortic proinflammatory M1 cytokines, increased the expression of the anti-inflammatory M2 cytokine Arg1, and attenuated the iNos/Arg1 ratio. Enhanced mitogen-activated protein kinase signaling, known to induce inflammatory cytokine expression in vitro, was enhanced in aortae of HFHC-fed mice. Furthermore, the HFHC diet impaired aortic insulin signaling through Akt and forkhead box O1, which was associated with elevated endoplasmic reticulum stress markers CCAAT-enhancer-binding protein homologous protein and 78kDa glucose regulated protein. GW1516 intervention normalized mitogen-activated protein kinase activation, insulin signaling, and endoplasmic reticulum stress. CONCLUSIONS: Intervention with a PPARδ agonist inhibits aortic inflammation and attenuates the progression of pre-established atherosclerosis.


Asunto(s)
Antiinflamatorios/farmacología , Aortitis/prevención & control , Aterosclerosis/prevención & control , Resistencia a la Insulina , PPAR delta/agonistas , Receptores de LDL/deficiencia , Tiazoles/farmacología , Animales , Aortitis/sangre , Aortitis/etiología , Aortitis/genética , Aortitis/patología , Aterosclerosis/sangre , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/patología , Biomarcadores/sangre , Glucemia/metabolismo , Colesterol en la Dieta , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Dislipidemias/sangre , Dislipidemias/tratamiento farmacológico , Dislipidemias/genética , Dislipidemias/metabolismo , Mediadores de Inflamación/metabolismo , Insulina/sangre , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR delta/metabolismo , Receptores de LDL/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
14.
J Lipid Res ; 55(7): 1254-66, 2014 07.
Artículo en Inglés | MEDLINE | ID: mdl-24864274

RESUMEN

PPARδ regulates systemic lipid homeostasis and inflammation, but its role in hepatic lipid metabolism remains unclear. Here, we examine whether intervening with a selective PPARδ agonist corrects hepatic steatosis induced by a high-fat, cholesterol-containing (HFHC) diet. Ldlr(-/-) mice were fed a chow or HFHC diet (42% fat, 0.2% cholesterol) for 4 weeks. For an additional 8 weeks, the HFHC group was fed HFHC or HFHC plus GW1516 (3 mg/kg/day). GW1516-intervention significantly attenuated liver TG accumulation by induction of FA ß-oxidation and attenuation of FA synthesis. In primary mouse hepatocytes, GW1516 treatment stimulated AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC) phosphorylation in WT hepatocytes, but not AMPKß1(-/-) hepatocytes. However, FA oxidation was only partially reduced in AMPKß1(-/-) hepatocytes, suggesting an AMPK-independent contribution to the GW1516 effect. Similarly, PPARδ-mediated attenuation of FA synthesis was partially due to AMPK activation, as GW1516 reduced lipogenesis in WT hepatocytes but not AMPKß1(-/-) hepatocytes. HFHC-fed animals were hyperinsulinemic and exhibited selective hepatic insulin resistance, which contributed to elevated fasting FA synthesis and hyperglycemia. GW1516 intervention normalized fasting hyperinsulinemia and selective hepatic insulin resistance and attenuated fasting FA synthesis and hyperglycemia. The HFHC diet polarized the liver toward a proinflammatory M1 state, which was reversed by GW1516 intervention. Thus, PPARδ agonist treatment inhibits the progression of preestablished hepatic steatosis.


Asunto(s)
Grasas de la Dieta/efectos adversos , Ácidos Grasos/biosíntesis , Hígado Graso/metabolismo , Resistencia a la Insulina , Lipogénesis/efectos de los fármacos , PPAR delta/metabolismo , Receptores de LDL/metabolismo , Animales , Grasas de la Dieta/farmacología , Ácidos Grasos/genética , Hígado Graso/inducido químicamente , Hígado Graso/genética , Hígado Graso/patología , Lipogénesis/genética , Ratones , Ratones Noqueados , Oxidación-Reducción/efectos de los fármacos , PPAR delta/genética , Receptores de LDL/genética
16.
Curr Opin Lipidol ; 24(2): 171-7, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23481229

RESUMEN

PURPOSE OF REVIEW: Therapeutic strategies to alleviate the growing epidemic of insulin-resistant syndromes (obesity and type 2 diabetes) as well as the conferred cardiovascular disease risk remain sparse. The peroxisome proliferator-activated receptor δ (PPARδ) has emerged as a versatile regulator of lipid homeostasis and inflammatory signaling, making it an attractive therapeutic target for the treatment and prevention of type 2 diabetes and atherosclerosis. RECENT FINDINGS: PPARδ activation regulates lipid homeostasis and inflammatory signaling in a variety of cell types, conferring protection from metabolic disease and atherosclerosis. Specifically, PPARδ activation in the liver stimulates glucose utilization and inhibits gluconeogenesis, which improves insulin resistance and hyperglycemia. In macrophages, PPARδ-specific activation with synthetic agonists inhibits VLDL-induced triglyceride accumulation and inflammation. In mice, PPARδ agonists halt the progression of atherosclerosis and stabilize existing lesions by promoting an anti-inflammatory milieu within the diseased macrovasculature. In humans, PPARδ activation improves insulin sensitivity and reduces atherogenic dyslipidemia via a mechanism complementary to statin monotherapy. SUMMARY: Recent advances in the understanding of PPARδ reveal that activation of this receptor represents a multifaceted therapeutic strategy for the prevention and treatment of insulin-resistant syndromes and atherosclerosis.


Asunto(s)
Inflamación/patología , Hígado/metabolismo , PPAR delta/metabolismo , Animales , Antiinflamatorios/farmacología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Aterosclerosis/patología , Ensayos Clínicos como Asunto , Dislipidemias/tratamiento farmacológico , Dislipidemias/metabolismo , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Inflamación/metabolismo , Resistencia a la Insulina , Hígado/patología , Macrófagos/metabolismo , Ratones , PPAR delta/agonistas , Tiazoles/farmacología
17.
Curr Opin Lipidol ; 24(1): 34-40, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23254473

RESUMEN

PURPOSE OF REVIEW: Citrus flavonoids are polyphenolic compounds with powerful biological properties. This review aims to summarize recent advances towards understanding the ability of citrus flavonoids to regulate lipid metabolism and other metabolic parameters relevant to the metabolic syndrome, type 2 diabetes and cardiovascular disease. RECENT FINDINGS: Citrus flavonoids, including naringenin, hesperidin, nobiletin and tangeretin, have emerged as promising therapeutic agents for the treatment of metabolic dysregulation. Epidemiological studies report that intake of citrus flavonoid-containing foods attenuates cardiovascular diseases. Experimental and a limited number of clinical studies reveal lipid-lowering, insulin-sensitizing, antihypertensive and anti-inflammatory properties. In animal models, citrus flavonoid supplements prevent hepatic steatosis, dyslipidemia and insulin sensitivity primarily through inhibition of hepatic fatty acid synthesis and increased fatty acid oxidation. Citrus flavonoids blunt the inflammatory response in metabolically important tissues including liver, adipose tissue, kidney and the aorta. The mechanisms underlying flavonoid-induced metabolic regulation have not been completely established. In mouse models, citrus flavonoids show marked suppression of atherogenesis through improved metabolic parameters and also through direct impact on the vessel wall. SUMMARY: These recent studies suggest an important role of citrus flavonoids in the treatment of dyslipidemia, insulin resistance, hepatic steatosis, obesity and atherosclerosis. The favorable outcomes are achieved through multiple mechanisms. Human studies focussed on dose, bioavailability, efficacy and safety are required to propel the use of these promising therapeutic agents into the clinical arena.


Asunto(s)
Citrus/metabolismo , Flavanonas/metabolismo , Metabolismo de los Lípidos , Fitoterapia , Animales , Antiinflamatorios/metabolismo , Antiinflamatorios/farmacología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Dislipidemias/tratamiento farmacológico , Dislipidemias/metabolismo , Flavanonas/farmacología , Flavonas/metabolismo , Flavonas/farmacología , Hesperidina/metabolismo , Hesperidina/farmacología , Humanos , Hipolipemiantes/metabolismo , Hipolipemiantes/farmacología , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Oxidación-Reducción
18.
J Lipid Res ; 54(3): 711-724, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23269394

RESUMEN

Obesity-associated chronic inflammation contributes to metabolic dysfunction and propagates atherosclerosis. Recent evidence suggests that increased dietary cholesterol exacerbates inflammation in adipose tissue and liver, contributing to the proatherogenic milieu. The ability of the citrus flavonoid naringenin to prevent these cholesterol-induced perturbations is unknown. To assess the ability of naringenin to prevent the amplified inflammatory response and atherosclerosis induced by dietary cholesterol, male Ldlr⁻/⁻ mice were fed either a cholesterol-enriched high-fat or low-fat diet supplemented with 3% naringenin for 12 weeks. Naringenin, through induction of hepatic fatty acid (FA) oxidation and attenuation of FA synthesis, prevented hepatic steatosis, hepatic VLDL overproduction, and hyperlipidemia induced by both cholesterol-rich diets. Naringenin attenuated hepatic macrophage infiltration and inflammation stimulated by dietary cholesterol. Insulin resistance, adipose tissue expansion, and inflammation were alleviated by naringenin. Naringenin attenuated the cholesterol-induced formation of both foam cells and expression of inflammatory markers in peritoneal macrophages. Naringenin significantly decreased atherosclerosis and inhibited the formation of complex lesions, which was associated with normalized aortic lipids and a reversal of aortic inflammation. We demonstrate that in mice fed cholesterol-enriched diets, naringenin attenuates peripheral and systemic inflammation, leading to protection from atherosclerosis. These studies offer a therapeutically relevant alternative for the prevention of cholesterol-induced metabolic dysregulation.


Asunto(s)
Aterosclerosis/inducido químicamente , Aterosclerosis/prevención & control , Colesterol/efectos adversos , Flavanonas/uso terapéutico , Inflamación/inducido químicamente , Inflamación/prevención & control , Animales , Hígado Graso/inducido químicamente , Hígado Graso/prevención & control , Flavonoides/metabolismo , Resistencia a la Insulina , Masculino , Ratones , Ratones Noqueados , Receptores de LDL/genética , Receptores de LDL/metabolismo
19.
Arterioscler Thromb Vasc Biol ; 32(12): 2919-28, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23023367

RESUMEN

OBJECTIVE: Hypertriglyceridemia is an important risk factor for cardiovascular disease. Elevated plasma very low-density lipoprotein (VLDL) puts insulin-resistant patients at risk for atherosclerosis. VLDL readily induces macrophage lipid accumulation and inflammatory responses, for which targeted therapeutic strategies remain elusive. We examined the ability of VLDL to induce macrophage foam cells and the inflammatory response and sought to define the cell signaling cascades involved. We further examined the potential of peroxisome proliferator-activated receptor (PPAR) δ activation to attenuate both VLDL-stimulated lipid accumulation and cytokine expression. METHODS AND RESULTS: THP-1 macrophages exposed to VLDL displayed significant triglyceride accumulation, which was attenuated by PPARδ activation. PPARδ agonists stimulated a transcriptional program resulting in inhibition of lipoprotein lipase activity, activation of fatty acid uptake, and enhanced ß-oxidation. VLDL-treated macrophages significantly increased the expression of activator protein 1 associated cytokines interleukin-1ß, macrophage inflammatory protein 1α, and intercellular adhesion molecule-1. VLDL treatment significantly increased the phosphorylation of both extracellular signal-related kinase 1 and 2 and p38. VLDL reduced AKT phosphorylation as well as its downstream effector forkhead box protein O1, concomitant with increased nuclear forkhead box protein O1. Cells treated with PPARδ agonists were completely resistant to VLDL-induced expression of inflammatory cytokines, mediated by normalization of mitogen-activated protein kinase (MAPK)(erk) and AKT/forkhead box protein O1 signaling. CONCLUSIONS: The combined PPARδ-mediated reductions of lipid accumulation and inflammatory cytokine expression suggest a novel macrophage-targeted therapeutic option in treating atherosclerosis.


Asunto(s)
Células Espumosas/metabolismo , Células Espumosas/patología , Inflamación/inducido químicamente , Lipoproteínas VLDL/efectos adversos , Macrófagos/metabolismo , Macrófagos/patología , PPAR delta/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Ácidos Grasos/metabolismo , Células Espumosas/efectos de los fármacos , Humanos , Inflamación/metabolismo , Inflamación/patología , Ligandos , Lipoproteínas VLDL/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , PPAR delta/agonistas , Transducción de Señal/efectos de los fármacos , Triglicéridos/metabolismo
20.
Arterioscler Thromb Vasc Biol ; 31(8): 1916-26, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21597005

RESUMEN

OBJECTIVE: Earlier studies have suggested that a common genetic architecture underlies the clinically heterogeneous polygenic Fredrickson hyperlipoproteinemia (HLP) phenotypes defined by hypertriglyceridemia (HTG). Here, we comprehensively analyzed 504 HLP-HTG patients and 1213 normotriglyceridemic controls and confirmed that a spectrum of common and rare lipid-associated variants underlies this heterogeneity. METHODS AND RESULTS: First, we demonstrated that genetic determinants of plasma lipids and lipoproteins, including common variants associated with plasma triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C) from the Global Lipids Genetics Consortium were associated with multiple HLP-HTG phenotypes. Second, we demonstrated that weighted risk scores composed of common TG-associated variants were distinctly increased across all HLP-HTG phenotypes compared with controls; weighted HDL-C and LDL-C risk scores were also increased, although to a less pronounced degree with some HLP-HTG phenotypes. Interestingly, decomposition of HDL-C and LDL-C risk scores revealed that pleiotropic variants (those jointly associated with TG) accounted for the greatest difference in HDL-C and LDL-C risk scores. The APOE E2/E2 genotype was significantly overrepresented in HLP type 3 versus other phenotypes. Finally, rare variants in 4 genes accumulated equally across HLP-HTG phenotypes. CONCLUSIONS: HTG susceptibility and phenotypic heterogeneity are both influenced by accumulation of common and rare TG-associated variants.


Asunto(s)
Hipertrigliceridemia/sangre , Hipertrigliceridemia/genética , Lípidos/sangre , Lípidos/genética , Adulto , Anciano , Alelos , Apolipoproteína E2/genética , Estudios de Casos y Controles , HDL-Colesterol/sangre , HDL-Colesterol/genética , LDL-Colesterol/sangre , LDL-Colesterol/genética , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Hiperlipoproteinemia Tipo IV/sangre , Hiperlipoproteinemia Tipo IV/genética , Masculino , Persona de Mediana Edad , Herencia Multifactorial , Fenotipo , Factores de Riesgo , Triglicéridos/sangre , Triglicéridos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA