Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Biomolecules ; 14(4)2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38672446

RESUMEN

Preclinical studies regarding the potential of liver X receptor (LXR) agonists to inhibit macrophage foam cell formation and the development of atherosclerotic lesions are generally executed in mice fed with Western-type diets enriched in cholesterol and fat. Here, we investigated whether LXR agonism remains anti-atherogenic under dietary conditions with a low basal hepatic lipogenesis rate. Hereto, atherosclerosis-susceptible male apolipoprotein E knockout mice were fed a low-fat diet with or without 10 mg/kg/day LXR agonist T0901317 supplementation for 8 weeks. Importantly, T0901317 significantly stimulated atherosclerosis susceptibility, despite an associated increase in the macrophage gene expression levels of cholesterol efflux transporters ABCA1 and ABCG1. The pro-atherogenic effect of T0901317 coincided with exacerbated hypercholesterolemia, hypertriglyceridemia, and a significant rise in hepatic triglyceride stores and macrophage numbers. Furthermore, T0901317-treated mice exhibited elevated plasma MCP-1 levels and monocytosis. In conclusion, these findings highlight that the pro-atherogenic hepatic effects of LXR agonism are dominant over the anti-atherogenic effects in macrophages in determining the overall atherosclerosis outcome under low-fat diet feeding conditions. A low-fat diet experimental setting, as compared to the commonly used high-fat-diet-based preclinical setup, thus appears more sensitive in uncovering the potential relevance of the off-target liver effects of novel anti-atherogenic therapeutic approaches that target macrophage LXR.


Asunto(s)
Apolipoproteínas E , Aterosclerosis , Bencenosulfonamidas , Fluorocarburos , Macrófagos , Animales , Masculino , Ratones , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Aterosclerosis/patología , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Hígado/metabolismo , Hígado/efectos de los fármacos , Hígado/patología , Receptores X del Hígado/agonistas , Receptores X del Hígado/metabolismo , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Triglicéridos/sangre , Triglicéridos/metabolismo
2.
Biomolecules ; 12(9)2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-36139044

RESUMEN

2-Hydroxypropyl-beta-cyclodextrin (2HPßCD) is able to bind and solubilize unesterified cholesterol and may therefore be able to reverse the deposition of cholesterol in macrophages within the aortic vessel wall, a hallmark of atherosclerotic cardiovascular disease. However, conflicting results regarding the potential of 2HPßCD to induce regression of established atherosclerotic lesions have been described. In the current study, we therefore also investigated the ability of 2HPßCD to stimulate cholesterol removal from macrophage foam cells in vitro and induce the regression of established atherosclerotic lesions in apolipoprotein E knockout (APOE KO) mice. In vitro studies using murine thioglycollate-elicited peritoneal macrophages verified that 2HPßCD is able to induce cholesterol efflux from macrophages in an ATP-binding cassette transporter-independent manner. Switching Western-type-diet-fed APOE KO mice with established atherosclerotic lesions back to a chow diet was associated with a reduction in the hypercholesterolemia extent and an increase in the absolute lesion size and plaque collagen-to-macrophage ratio. Importantly, parallel subcutaneous administration of 2HPßCD was not able to prevent the diet-switch-associated lesion growth or induce atherosclerosis regression. Although in our hands, 2HPßCD does effectively stimulate cellular cholesterol efflux from macrophages, we do not consider it worthwhile to further pursue 2HPßCD as therapeutic moiety in the atherosclerosis regression context.


Asunto(s)
Aterosclerosis , Tioglicolatos , 2-Hidroxipropil-beta-Ciclodextrina , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Aterosclerosis/metabolismo , Colesterol/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE
3.
Sci Rep ; 11(1): 16419, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34385562

RESUMEN

Total body upstream stimulatory factor 1 (USF1) deficiency in mice is associated with brown adipose tissue activation and a marked protection against the development of obesity and atherosclerotic lesions. Functional expression of USF1 has also been detected in monocytes and monocyte-derived macrophages. In the current study we therefore tested whether selective hematopoietic USF1 deficiency can also beneficially impact the development of atherosclerosis. For this purpose, LDL receptor knockout mice were transplanted with bone marrow from USF1 knockout mice or their wild-type littermate controls and subsequently fed a Western-type diet for 20 weeks to stimulate atherosclerotic lesion development. Strikingly, absence of USF1 function in bone marrow-derived cells was associated with exacerbated blood leukocyte (+ 100%; P < 0.01) and peritoneal leukocyte (+ 50%; P < 0.05) lipid loading and an increased atherosclerosis susceptibility (+ 31%; P < 0.05). These effects could be attributed to aggravated hyperlipidemia, i.e. higher plasma free cholesterol (+ 33%; P < 0.001) and cholesteryl esters (+ 39%; P < 0.001), and the development of hepatosteatosis. In conclusion, we have shown that hematopoietic USF1 deficiency is associated with an increased atherosclerosis susceptibility in LDL receptor knockout mice. These findings argue against a contribution of macrophage-specific USF1 deficiency to the previously described beneficial effect of total body USF1 deficiency on atherosclerosis susceptibility in mice.


Asunto(s)
Aterosclerosis/genética , Predisposición Genética a la Enfermedad , Receptores de LDL/genética , Factores Estimuladores hacia 5'/genética , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
J Nutr Biochem ; 89: 108564, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33321184

RESUMEN

Scavenger receptor BI (SR-BI) has been suggested to modulate adipocyte function. To uncover the potential relevance of SR-BI for the development of obesity and associated metabolic complications, we compared the metabolic phenotype of wild-type and SR-BI deficient mice fed an obesogenic diet enriched in fat. Both male and female SR-BI knockout mice gained significantly more weight as compared to their wild-type counterparts in response to 12 weeks high fat diet feeding (1.5-fold; P < .01 for genotype). Plasma free cholesterol levels were ~2-fold higher (P < .001) in SR-BI knockout mice of both genders, whilst plasma cholesteryl ester and triglyceride concentrations were only significantly elevated in males. Strikingly, the exacerbated obesity in SR-BI knockout mice was paralleled by a better glucose handling. In contrast, only SR-BI knockout mice developed atherosclerotic lesions in the aortic root, with a higher predisposition in females. Biochemical and histological studies in male mice revealed that SR-BI deficiency was associated with a reduced hepatic steatosis degree as evident from the 29% lower (P < .05) liver triglyceride levels. Relative mRNA expression levels of the glucose uptake transporter GLUT4 were increased (+47%; P < .05), whilst expression levels of the metabolic PPARgamma target genes CD36, HSL, ADIPOQ and ATGL were reduced 39%-58% (P < .01) in the context of unchanged PPARgamma expression levels in SR-BI knockout gonadal white adipose tissue. In conclusion, we have shown that SR-BI deficiency is associated with a decrease in adipocyte PPARgamma activity and a concomitant uncoupling of obesity development from hepatic steatosis and glucose intolerance development in high fat diet-fed mice.


Asunto(s)
Antígenos CD36/deficiencia , Hígado Graso/metabolismo , Intolerancia a la Glucosa/metabolismo , Obesidad/metabolismo , Adipocitos/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Aterosclerosis/metabolismo , Antígenos CD36/metabolismo , Colesterol/sangre , Ésteres del Colesterol/sangre , Dieta Alta en Grasa/efectos adversos , Femenino , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR gamma/genética , PPAR gamma/metabolismo , Receptores Depuradores de Clase B/metabolismo , Triglicéridos/sangre
5.
Atherosclerosis ; 281: 47-55, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30658191

RESUMEN

BACKGROUND AND AIMS: Stabilin-1 (STAB1) is a scavenger receptor expressed on alternatively activated macrophages and sinusoidal endothelial cells. Its ligands include oxidized low-density lipoprotein (LDL) and the extracellular matrix glycoprotein SPARC and it is present in both human and murine atherosclerotic lesions. We aimed to investigate the effect of specific deletion of STAB1 in bone marrow-derived cells, including macrophages on atherosclerotic lesion formation in mice. METHODS: Lethally irradiated hypercholesterolemic LDL receptor knockout mice received either wildtype (WT) or STAB1 knockout (STAB1 KO) bone marrow. Bone marrow transplanted mice were fed a Western-type diet for 9 weeks to induce atherosclerotic lesion formation. RESULTS: Interestingly, LDL receptor knockout mice reconstituted with STAB1 KO bone marrow showed increased body weight gain (two-way ANOVA: p < 0.001) and larger white adipocyte cell sizes (43% increase in cell area; p < 0.05) as compared to WT bone marrow transplanted mice, which correlated positively (r = 0.82; p < 0.001). This was paralleled by a significant increase in white adipose tissue relative mRNA expression levels of the adipokine leptin (+94% p < 0.05). Despite these changes, no differences in serum lipid levels, the extent of in vivo macrophage foam cell formation or circulating leukocyte concentrations were observed. Moreover, the size and composition of atherosclerotic lesions was not different between the two experimental groups. CONCLUSIONS: Bone marrow-specific Stabilin-1 deletion does not affect the susceptibility for atherosclerosis in mice. However, the increased body weight gain and adipocyte cell size highlight a potential role for leukocyte STAB1 in the development of metabolic disorders.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Moléculas de Adhesión Celular Neuronal/deficiencia , Células Espumosas/metabolismo , Receptores de LDL/deficiencia , Adipocitos Blancos/metabolismo , Adipocitos Blancos/patología , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Animales , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/patología , Moléculas de Adhesión Celular Neuronal/genética , Dieta Occidental , Modelos Animales de Enfermedad , Células Espumosas/patología , Hipercolesterolemia/complicaciones , Hipercolesterolemia/genética , Leptina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Placa Aterosclerótica , Receptores de LDL/genética , Aumento de Peso
7.
Br J Pharmacol ; 175(15): 3175-3183, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29774529

RESUMEN

BACKGROUND AND PURPOSE: Agonists for the liver X receptor (LXR) are considered promising therapeutic moieties in cholesterol-driven diseases by promoting cellular cholesterol efflux pathways. However, current clinical application of these agents is hampered by concomitant LXR-induced activation of a lipogenic transcriptional network, leading to hepatic steatosis. Recent studies have suggested that protein arginine methyltransferase 3 (PRMT3) may act as a selective co-activator of LXR activity. Here, we verified the hypothesis that PRMT3 inhibition selectively disrupts the ability of LXR to stimulate lipogenesis while maintaining its capacity to modulate macrophage cholesterol homeostasis. EXPERIMENTAL APPROACH: A combination of the LXR agonist T0901317 and palm oil was administered to C57BL/6 mice to maximally stimulate LXR and PRMT3 activity. PRMT3 activity was inhibited using the allosteric inhibitor SGC707. KEY RESULTS: Treatment with SGC707 did not negatively influence the T0901317/palm oil-induced up-regulation of the cholesterol efflux ATP-binding cassette transporter genes, ABCA1 and ABCG1, in peritoneal cells. In contrast, SGC707 treatment was associated with a significant decrease in the hepatic expression of the lipogenic gene fatty acid synthase (-64%). A similar trend was observed for stearoyl-coenzyme A desaturase and acetyl CoA carboxylase expression (-43%; -56%). This obstruction of lipogenic gene transcription coincided with a significant 2.3-fold decrease in liver triglyceride content as compared with the T0901317 and palm oil-treated control group. CONCLUSION AND IMPLICATIONS: We showed that inhibition of PRMT3 activity by SGC707 treatment selectively impairs LXR-driven transcription of hepatic lipogenic genes, while the positive effect of LXR stimulation on macrophage cholesterol efflux pathways is maintained.


Asunto(s)
Isoquinolinas/farmacología , Lipogénesis/efectos de los fármacos , Receptores X del Hígado/genética , Hígado/efectos de los fármacos , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Transportador 1 de Casete de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Animales , Colesterol/sangre , Hidrocarburos Fluorados/farmacología , Lipogénesis/genética , Hígado/metabolismo , Receptores X del Hígado/agonistas , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Aceite de Palma/farmacología , Sulfonamidas/farmacología , Transcripción Genética/efectos de los fármacos , Triglicéridos/sangre
8.
Atherosclerosis ; 274: 120-127, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29772480

RESUMEN

BACKGROUND AND AIMS: Proteoglycan 4 (Prg4) has a high structural similarity with the established atherosclerosis-modulating proteoglycan versican, but its role in atherogenesis is still unknown. Therefore, the impact of Prg4 deficiency on macrophage function in vitro and atherosclerosis susceptibility in vivo was investigated. METHODS: The presence and localization of Prg4 was studied in atherosclerotic lesions. Furthermore, the effect of Prg4 deficiency on macrophage foam cell formation, cholesterol efflux and lipopolysaccharide (LPS) response was determined. Finally, susceptibility for atherosclerotic lesion formation was investigated in bone marrow-specific Prg4 knockout (KO) mice. RESULTS: Prg4 mRNA expression was induced 91-fold (p<0.001) in murine initial atherosclerotic lesions and Prg4 protein co-localized with human lesional macrophages. Murine Prg4 KO macrophages showed increased foam cell formation (+2.1-fold, p<0.01). In parallel, the expression of the cholesterol efflux genes ATP-binding cassette transporter A1 and scavenger receptor type B1 was lower (-35%, p<0.05;-40%, p<0.05) in Prg4 KO macrophages. This translated into an impaired cholesterol efflux to high-density lipoprotein (-13%, p<0.001) and apolipoprotein A1 (-8%, p<0.05). Furthermore, Prg4 KO macrophages showed an impaired LPS-induced rise in TNFα secretion as compared to wild-type controls (-31%, p<0.001), indicating a reduced inflammatory response. Combined, these pro- and anti-atherogenic effects did not translate into a significant difference in atherosclerotic lesion formation upon bone marrow-specific deletion of Prg4 in low-density lipoprotein receptor KO mice. CONCLUSIONS: Prg4 is present in macrophages in both murine and human atherosclerotic lesions and critically influences macrophage function, but deletion of Prg4 in bone marrow-derived cells does not affect atherosclerotic lesion development.


Asunto(s)
Aterosclerosis/metabolismo , Células de la Médula Ósea/metabolismo , Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Células Espumosas/metabolismo , Macrófagos Peritoneales/metabolismo , Placa Aterosclerótica , Proteoglicanos/metabolismo , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/patología , Células Cultivadas , Colesterol/metabolismo , Modelos Animales de Enfermedad , Células Espumosas/efectos de los fármacos , Células Espumosas/patología , Células Espumosas/trasplante , Humanos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/patología , Macrófagos Peritoneales/trasplante , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Fenotipo , Proteoglicanos/deficiencia , Proteoglicanos/genética , Receptores Depuradores de Clase B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Atherosclerosis ; 251: 159-163, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27323229

RESUMEN

BACKGROUND AND AIMS: We explored the role of ATP-binding cassette transporter A1 (Abca1), in post-myocardial infarction (MI) cardiac injury. METHODS: In Abca1(-/-) mice, wild type (WT) mice, and WT mice transplanted with Abca1(-/-) or WT bone marrow, an MI was induced in vivo. Furthermore, an ex vivo MI was induced in isolated Abca1(-/-) and WT hearts. RESULTS: Twenty-four hours and two weeks after in vivo MI induction, MI size was reduced in Abca1(-/-) (-58%, p = 0.007; -59%, p = 0.03) compared to WT. Ex vivo MI induction showed no effect of Abca1(-/-) on infarct size. Interestingly, two weeks after MI, Abca1(-/-) mice showed higher circulating levels of B-cells (+3.0 fold, p = 0.02) and T-cells (+4.2 fold, p = 0.002) compared to WT. Bone marrow-specific Abca1(-/-) tended to reduce infarct size (-43%, p = 0.12), suggesting a detrimental role for hematopoietic Abca1 after MI. CONCLUSIONS: Although Abca1 has a protective role in atherosclerosis, it exerts detrimental effects on cardiac function after MI.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/deficiencia , Corazón/fisiopatología , Infarto del Miocardio/genética , Infarto del Miocardio/prevención & control , Animales , Aterosclerosis/metabolismo , Linfocitos B/citología , Trasplante de Médula Ósea , Femenino , Leucocitos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/metabolismo
10.
J Lipid Res ; 55(10): 2022-32, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25016380

RESUMEN

The melanocortin system is an important regulator of energy balance, and melanocortin 4 receptor (MC4R) deficiency is the most common monogenic cause of obesity. We investigated whether the relationship between melanocortin system activity and energy expenditure (EE) is mediated by brown adipose tissue (BAT) activity. Therefore, female APOE*3-Leiden.CETP transgenic mice were fed a Western-type diet for 4 weeks and infused intracerebroventricularly with the melanocortin 3/4 receptor (MC3/4R) antagonist SHU9119 or vehicle for 2 weeks. SHU9119 increased food intake (+30%) and body fat (+50%) and decreased EE by reduction in fat oxidation (-42%). In addition, SHU9119 impaired the uptake of VLDL-TG by BAT. In line with this, SHU9119 decreased uncoupling protein-1 levels in BAT (-60%) and induced large intracellular lipid droplets, indicative of severely disturbed BAT activity. Finally, SHU9119-treated mice pair-fed to the vehicle-treated group still exhibited these effects, indicating that MC4R inhibition impairs BAT activity independent of food intake. These effects were not specific to the APOE*3-Leiden.CETP background as SHU9119 also inhibited BAT activity in wild-type mice. We conclude that inhibition of central MC3/4R signaling impairs BAT function, which is accompanied by reduced EE, thereby promoting adiposity. We anticipate that activation of MC4R is a promising strategy to combat obesity by increasing BAT activity.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Hormonas Estimuladoras de los Melanocitos/farmacología , Receptor de Melanocortina Tipo 3/antagonistas & inhibidores , Receptor de Melanocortina Tipo 4/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Femenino , Ratones , Ratones Transgénicos , Oxidación-Reducción/efectos de los fármacos , Receptor de Melanocortina Tipo 3/genética , Receptor de Melanocortina Tipo 3/metabolismo , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo
11.
J Lipid Res ; 55(2): 180-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24285857

RESUMEN

Important players in triglyceride (TG) metabolism include the liver (production), white adipose tissue (WAT) (storage), heart and skeletal muscle (combustion to generate ATP), and brown adipose tissue (BAT) (combustion toward heat), the collective action of which determine plasma TG levels. Interestingly, recent evidence points to a prominent role of the hypothalamus in TG metabolism through innervating the liver, WAT, and BAT mainly via sympathetic branches of the autonomic nervous system. Here, we review the recent findings in the area of sympathetic control of TG metabolism. Various neuronal populations, such as neuropeptide Y (NPY)-expressing neurons and melanocortin-expressing neurons, as well as peripherally produced hormones (i.e., GLP-1, leptin, and insulin), modulate sympathetic outflow from the hypothalamus toward target organs and thereby influence peripheral TG metabolism. We conclude that sympathetic stimulation in general increases lipolysis in WAT, enhances VLDL-TG production by the liver, and increases the activity of BAT with respect to lipolysis of TG, followed by combustion of fatty acids toward heat. Moreover, the increased knowledge about the involvement of the neuroendocrine system in TG metabolism presented in this review offers new therapeutic options to fight hypertriglyceridemia by specifically modulating sympathetic nervous system outflow toward liver, BAT, or WAT.


Asunto(s)
Sistema Nervioso Simpático/fisiología , Triglicéridos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Humanos , Lipoproteínas/metabolismo , Hígado/metabolismo
12.
Diabetes ; 63(3): 880-91, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24270984

RESUMEN

Metformin is the first-line drug for the treatment of type 2 diabetes. Besides its well-characterized antihyperglycemic properties, metformin also lowers plasma VLDL triglyceride (TG). In this study, we investigated the underlying mechanisms in APOE*3-Leiden.CETP mice, a well-established model for human-like lipoprotein metabolism. We found that metformin markedly lowered plasma total cholesterol and TG levels, an effect mostly due to a decrease in VLDL-TG, whereas HDL was slightly increased. Strikingly, metformin did not affect hepatic VLDL-TG production, VLDL particle composition, and hepatic lipid composition but selectively enhanced clearance of glycerol tri[(3)H]oleate-labeled VLDL-like emulsion particles into brown adipose tissue (BAT). BAT mass and lipid droplet content were reduced in metformin-treated mice, pointing to increased BAT activation. In addition, both AMP-activated protein kinase α1 (AMPKα1) expression and activity and HSL and mitochondrial content were increased in BAT. Furthermore, therapeutic concentrations of metformin increased AMPK and HSL activities and promoted lipolysis in T37i differentiated brown adipocytes. Collectively, our results identify BAT as an important player in the TG-lowering effect of metformin by enhancing VLDL-TG uptake, intracellular TG lipolysis, and subsequent mitochondrial fatty acid oxidation. Targeting BAT might therefore be considered as a future therapeutic strategy for the treatment of dyslipidemia.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Hipoglucemiantes/farmacología , Lipoproteínas VLDL/metabolismo , Metformina/farmacología , Triglicéridos/sangre , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Células Cultivadas , Colesterol/sangre , Dislipidemias/tratamiento farmacológico , Femenino , Lipólisis/efectos de los fármacos , Lipoproteína Lipasa/fisiología , Ratones , Triglicéridos/metabolismo
13.
FASEB J ; 27(8): 3354-62, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23650188

RESUMEN

Mutations in apolipoprotein A5 (APOA5) have been associated with hypertriglyceridemia in humans and mice. This has been attributed to a stimulating role for APOA5 in lipoprotein lipase-mediated triglyceride hydrolysis and hepatic clearance of lipoprotein remnant particles. However, because of the low APOA5 plasma abundance, we investigated an additional signaling role for APOA5 in high-fat diet (HFD)-induced obesity. Wild-type (WT) and Apoa5(-/-) mice fed a chow diet showed no difference in body weight or 24-h food intake (Apoa5(-/-), 4.5±0.6 g; WT, 4.2±0.5 g), while Apoa5(-/-) mice fed an HFD ate more in 24 h (Apoa5(-/-), 2.8±0.4 g; WT, 2.5±0.3 g, P<0.05) and became more obese than WT mice. Also, intravenous injection of APOA5-loaded VLDL-like particles lowered food intake (VLDL control, 0.26±0.04 g; VLDL+APOA5, 0.11±0.07 g, P<0.01). In addition, the HFD-induced hyperphagia of Apoa5(-/-) mice was prevented by adenovirus-mediated hepatic overexpression of APOA5. Finally, intracerebroventricular injection of APOA5 reduced food intake compared to injection of the same mouse with artificial cerebral spinal fluid (0.40±0.11 g; APOA5, 0.23±0.08 g, P<0.01). These data indicate that the increased HFD-induced obesity of Apoa5(-/-) mice as compared to WT mice is at least partly explained by hyperphagia and that APOA5 plays a role in the central regulation of food intake.


Asunto(s)
Apolipoproteínas/deficiencia , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos/fisiología , Obesidad/fisiopatología , Animales , Apolipoproteína A-V , Apolipoproteínas/administración & dosificación , Apolipoproteínas/genética , Calorimetría , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Hígado Graso/genética , Hígado Graso/metabolismo , Hiperlipidemias/etiología , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Hiperfagia/etiología , Hiperfagia/genética , Hiperfagia/metabolismo , Inyecciones Intravenosas , Inyecciones Intraventriculares , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Obesidad/genética
14.
PLoS One ; 8(2): e55217, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23460782

RESUMEN

OBJECTIVE: Central neuropeptide Y (NPY) administration stimulates food intake in rodents. In addition, acute modulation of central NPY signaling increases hepatic production of very low-density lipoprotein (VLDL)-triglyceride (TG) in rats. As hypertriglyceridemia is an important risk factor for atherosclerosis, for which well-established mouse models are available, we set out to validate the effect of NPY on hepatic VLDL-TG production in mice, to ultimately investigate whether NPY, by increasing VLDL production, contributes to the development of atherosclerosis. RESEARCH DESIGN AND METHODS: Male C57Bl/6J mice received an intracerebroventricular (i.c.v.) cannula into the lateral (LV) or third (3V) ventricle of the brain. One week later, after a 4 h fast, the animals received an intravenous (i.v.) injection of Tran(35)S (100 µCi) followed by tyloxapol (500 mg/kg body weight; BW), enabling the study of hepatic VLDL-apoB and VLDL-TG production, respectively. Immediately after the i.v. injection of tyloxapol, the animals received either an i.c.v. injection of NPY (0.2 mg/kg BW in artificial cerebrospinal fluid; aCSF), synthetic Y1 receptor antagonist GR231118 (0.5 mg/kg BW in aCSF) or vehicle (aCSF), or an i.v. injection of PYY3-36 (0.5 mg/kg BW in PBS) or vehicle (PBS). RESULTS: Administration of NPY into both the LV and 3V increased food intake within one hour after injection (+164%, p<0.001 and +367%, p<0.001, respectively). NPY administration neither in the LV nor in the 3V affected hepatic VLDL-TG or VLDL-apoB production. Likewise, antagonizing central NPY signaling by either PYY3-36 or GR231118 administration did not affect hepatic VLDL production. CONCLUSION: In mice, as opposed to rats, acute central administration of NPY increases food intake without affecting hepatic VLDL production. These results are of great significance when extrapolating findings on the central regulation of hepatic VLDL production between species.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Lipoproteínas VLDL/biosíntesis , Hígado/metabolismo , Neuropéptido Y/administración & dosificación , Neuropéptido Y/farmacología , Anestesia , Animales , Ventrículos Cerebrales/efectos de los fármacos , Ventrículos Cerebrales/metabolismo , Estado de Conciencia/efectos de los fármacos , Inyecciones Intraventriculares , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Transducción de Señal/efectos de los fármacos
15.
PLoS One ; 7(11): e49152, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23133675

RESUMEN

OBJECTIVE: In addition to improve glucose intolerance, recent studies suggest that glucagon-like peptide-1 (GLP-1) receptor agonism also decreases triglyceride (TG) levels. The aim of this study was to evaluate the effect of GLP-1 receptor agonism on very-low-density lipoprotein (VLDL)-TG production and liver TG metabolism. EXPERIMENTAL APPROACH: The GLP-1 peptide analogues CNTO3649 and exendin-4 were continuously administered subcutaneously to high fat diet-fed APOE*3-Leiden transgenic mice. After 4 weeks, hepatic VLDL production, lipid content, and expression profiles of selected genes involved in lipid metabolism were determined. RESULTS: CNTO3649 and exendin-4 reduced fasting plasma glucose (up to -30% and -28% respectively) and insulin (-43% and -65% respectively). In addition, these agents reduced VLDL-TG production (-36% and -54% respectively) and VLDL-apoB production (-36% and -43% respectively), indicating reduced production of VLDL particles rather than reduced lipidation of apoB. Moreover, they markedly decreased hepatic content of TG (-39% and -55% respectively), cholesterol (-30% and -55% respectively), and phospholipids (-23% and -36% respectively), accompanied by down-regulation of expression of genes involved in hepatic lipogenesis (Srebp-1c, Fasn, Dgat1) and apoB synthesis (Apob). CONCLUSION: GLP-1 receptor agonism reduces VLDL production and hepatic steatosis in addition to an improvement of glycemic control. These data suggest that GLP-receptor agonists could reduce hepatic steatosis and ameliorate dyslipidemia in patients with type 2 diabetes mellitus.


Asunto(s)
Apolipoproteína E3/metabolismo , Hígado Graso/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Receptores de Glucagón/metabolismo , Animales , Apolipoproteína E3/genética , Apolipoproteínas B/metabolismo , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Dislipidemias/sangre , Exenatida , Hígado Graso/terapia , Receptor del Péptido 1 Similar al Glucagón , Insulina/metabolismo , Lipogénesis , Hígado/patología , Masculino , Ratones , Ratones Transgénicos , Péptidos/química , Péptidos/metabolismo , Ponzoñas/metabolismo
16.
Diabetes ; 60(12): 3132-40, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22028182

RESUMEN

OBJECTIVE: Insulin inhibits endogenous glucose production (EGP) and stimulates glucose uptake in peripheral tissues. Hypothalamic insulin signaling is required for the inhibitory effects of insulin on EGP. We examined the contribution of central insulin signaling on circulating insulin-stimulated tissue-specific glucose uptake. RESEARCH DESIGN AND METHODS: Tolbutamide, an inhibitor of ATP-sensitive K(+) channels (K(ATP) channels), or vehicle was infused into the lateral ventricle in the basal state and during hyperinsulinemic-euglycemic conditions in postabsorptive, chow-fed C57Bl/6J mice and in postabsorptive C57Bl/6J mice with diet-induced obesity. Whole-body glucose uptake was measured by d-[(14)C]glucose kinetics and tissue-specific glucose uptake by 2-deoxy-d-[(3)H]glucose uptake. RESULTS: During clamp conditions, intracerebroventricular administration of tolbutamide impaired the ability of insulin to inhibit EGP by ∼20%. In addition, intracerebroventricular tolbutamide diminished insulin-stimulated glucose uptake in muscle (by ∼59%) but not in heart or adipose tissue. In contrast, in insulin-resistant mice with diet-induced obesity, intracerebroventricular tolbutamide did not alter the effects of insulin during clamp conditions on EGP or glucose uptake by muscle. CONCLUSIONS: Insulin stimulates glucose uptake in muscle in part through effects via K(ATP) channels in the central nervous system, in analogy with the inhibitory effects of insulin on EGP. High-fat diet-induced obesity abolished the central effects of insulin on liver and muscle. These observations stress the role of central insulin resistance in the pathophysiology of diet-induced insulin resistance.


Asunto(s)
Transporte Biológico/efectos de los fármacos , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Glucosa/metabolismo , Insulina/farmacología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Corazón/efectos de los fármacos , Hipoglucemiantes/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Tolbutamida/farmacología
17.
J Lipid Res ; 52(9): 1712-22, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21700834

RESUMEN

Insulin signaling in the central nervous system (CNS) is required for the inhibitory effect of insulin on glucose production. Our aim was to determine whether the CNS is also involved in the stimulatory effect of circulating insulin on the tissue-specific retention of fatty acid (FA) from plasma. In wild-type mice, hyperinsulinemic-euglycemic clamp conditions stimulated the retention of both plasma triglyceride-derived FA and plasma albumin-bound FA in the various white adipose tissues (WAT) but not in other tissues, including brown adipose tissue (BAT). Intracerebroventricular (ICV) administration of insulin induced a similar pattern of tissue-specific FA partitioning. This effect of ICV insulin administration was not associated with activation of the insulin signaling pathway in adipose tissue. ICV administration of tolbutamide, a K(ATP) channel blocker, considerably reduced (during hyperinsulinemic-euglycemic clamp conditions) and even completely blocked (during ICV administration of insulin) WAT-specific retention of FA from plasma. This central effect of insulin was absent in CD36-deficient mice, indicating that CD36 is the predominant FA transporter in insulin-stimulated FA retention by WAT. In diet-induced insulin-resistant mice, these stimulating effects of insulin (circulating or ICV administered) on FA retention in WAT were lost. In conclusion, in insulin-sensitive mice, circulating insulin stimulates tissue-specific partitioning of plasma-derived FA in WAT in part through activation of K(ATP) channels in the CNS. Apparently, circulating insulin stimulates fatty acid uptake in WAT but not in BAT, directly and indirectly through the CNS.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Sistema Nervioso Central/metabolismo , Ácidos Grasos/metabolismo , Insulina/sangre , Canales KATP/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Dieta , Insulina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/fisiopatología , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA