Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 151(1): 138-52, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-23021221

RESUMEN

Inflammation and macrophage foam cells are characteristic features of atherosclerotic lesions, but the mechanisms linking cholesterol accumulation to inflammation and LXR-dependent response pathways are poorly understood. To investigate this relationship, we utilized lipidomic and transcriptomic methods to evaluate the effect of diet and LDL receptor genotype on macrophage foam cell formation within the peritoneal cavities of mice. Foam cell formation was associated with significant changes in hundreds of lipid species and unexpected suppression, rather than activation, of inflammatory gene expression. We provide evidence that regulated accumulation of desmosterol underlies many of the homeostatic responses, including activation of LXR target genes, inhibition of SREBP target genes, selective reprogramming of fatty acid metabolism, and suppression of inflammatory-response genes, observed in macrophage foam cells. These observations suggest that macrophage activation in atherosclerotic lesions results from extrinsic, proinflammatory signals generated within the artery wall that suppress homeostatic and anti-inflammatory functions of desmosterol.


Asunto(s)
Aterosclerosis/inmunología , Colesterol/biosíntesis , Desmosterol/metabolismo , Células Espumosas/metabolismo , Metabolismo de los Lípidos , Transcriptoma , Animales , Aterosclerosis/metabolismo , Colesterol/análogos & derivados , Colesterol/metabolismo , Ácidos Grasos/metabolismo , Células Espumosas/inmunología , Técnicas de Silenciamiento del Gen , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de LDL/genética , Receptores de LDL/metabolismo , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo
2.
Cell ; 145(4): 584-95, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21565615

RESUMEN

Microglia and astrocytes play essential roles in the maintenance of homeostasis within the central nervous system, but mechanisms that control the magnitude and duration of responses to infection and injury remain poorly understood. Here, we provide evidence that 5-androsten-3ß,17ß-diol (ADIOL) functions as a selective modulator of estrogen receptor (ER)ß to suppress inflammatory responses of microglia and astrocytes. ADIOL and a subset of synthetic ERß-specific ligands, but not 17ß-estradiol, mediate recruitment of CtBP corepressor complexes to AP-1-dependent promoters, thereby repressing genes that amplify inflammatory responses and activate Th17 T cells. Reduction of ADIOL or ERß expression results in exaggerated inflammatory responses to TLR4 agonists. Conversely, the administration of ADIOL or synthetic ERß-specific ligands that promote CtBP recruitment prevents experimental autoimmune encephalomyelitis in an ERß-dependent manner. These findings provide evidence for an ADIOL/ERß/CtBP-transrepression pathway that regulates inflammatory responses in microglia and can be targeted by selective ERß modulators.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Receptor beta de Estrógeno/metabolismo , Inflamación/metabolismo , Microglía/metabolismo , Transducción de Señal , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , Oxidorreductasas de Alcohol/metabolismo , Androstenodiol/metabolismo , Animales , Astrocitos/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Encefalomielitis Autoinmune Experimental/prevención & control , Humanos , Ratones , Ratones Endogámicos C57BL , Enfermedades Neurodegenerativas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo
3.
Nature ; 498(7452): 118-22, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23719382

RESUMEN

Cholesterol is a structural component of the cell and is indispensable for normal cellular function, although its excess often leads to abnormal proliferation, migration, inflammatory responses and/or cell death. To prevent cholesterol overload, ATP-binding cassette (ABC) transporters mediate cholesterol efflux from the cells to apolipoprotein A-I (apoA-I) and the apoA-I-containing high-density lipoprotein (HDL). Maintaining efficient cholesterol efflux is essential for normal cellular function. However, the role of cholesterol efflux in angiogenesis and the identity of its local regulators are poorly understood. Here we show that apoA-I binding protein (AIBP) accelerates cholesterol efflux from endothelial cells to HDL and thereby regulates angiogenesis. AIBP- and HDL-mediated cholesterol depletion reduces lipid rafts, interferes with VEGFR2 (also known as KDR) dimerization and signalling and inhibits vascular endothelial growth factor-induced angiogenesis in vitro and mouse aortic neovascularization ex vivo. Notably, Aibp, a zebrafish homologue of human AIBP, regulates the membrane lipid order in embryonic zebrafish vasculature and functions as a non-cell-autonomous regulator of angiogenesis. aibp knockdown results in dysregulated sprouting/branching angiogenesis, whereas forced Aibp expression inhibits angiogenesis. Dysregulated angiogenesis is phenocopied in Abca1 (also known as Abca1a) Abcg1-deficient embryos, and cholesterol levels are increased in Aibp-deficient and Abca1 Abcg1-deficient embryos. Our findings demonstrate that secreted AIBP positively regulates cholesterol efflux from endothelial cells and that effective cholesterol efflux is critical for proper angiogenesis.


Asunto(s)
Proteínas Portadoras/metabolismo , Colesterol/metabolismo , Neovascularización Fisiológica/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Transportadoras de Casetes de Unión a ATP/deficiencia , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Transporte Biológico , Vasos Sanguíneos/embriología , Proteínas Portadoras/genética , Colesterol/análisis , Proteínas de Unión al ADN , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lipoproteínas HDL/metabolismo , Lípidos de la Membrana/metabolismo , Microdominios de Membrana/química , Microdominios de Membrana/metabolismo , Multimerización de Proteína , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra/embriología , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
4.
Circ Res ; 104(12): 1355-63, 2009 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-19461045

RESUMEN

Toll-like receptor (TLR)4 recognizes microbial pathogens, such as lipopolysaccharide, and mediates lipopolysaccharide-induced proinflammatory cytokine secretion, as well as microbial uptake by macrophages. In addition to exogenous pathogens, TLR4 recognizes modified self, such as minimally oxidized low-density lipoprotein (mmLDL). Here we report that mmLDL and its active components, cholesteryl ester hydroperoxides, induce TLR4-dependent fluid phase uptake typical of macropinocytosis. We show that mmLDL induced recruitment of spleen tyrosine kinase (Syk) to a TLR4 signaling complex, TLR4 phosphorylation, activation of a Vav1-Ras-Raf-MEK-ERK1/2 signaling cascade, phosphorylation of paxillin, and activation of Rac, Cdc42, and Rho. These mmLDL-induced and TLR4- and Syk-dependent signaling events and cytoskeletal rearrangements lead to enhanced uptake of small molecules, dextran, and, most importantly, both native and oxidized LDL, resulting in intracellular lipid accumulation. An intravenous injection of fluorescently labeled mmLDL in wild-type mice resulted in its rapid accumulation in circulating monocytes, which was significantly attenuated in TLR4-deficient mice. These data describe a novel mechanism leading to enhanced lipoprotein uptake in macrophages that would contribute to foam cell formation and atherosclerosis. These data also suggest that cholesteryl ester hydroperoxides are an endogenous ligand for TLR4. Because TLR4 is highly expressed on the surface of circulating monocytes in patients with chronic inflammatory conditions, and cholesteryl ester hydroperoxides are present in plasma, lipid uptake by monocytes in circulation may contribute to the pathological roles of monocytes in chronic inflammatory diseases.


Asunto(s)
Ésteres del Colesterol/metabolismo , Células Espumosas/metabolismo , Lipoproteínas LDL/metabolismo , Sistema de Señalización de MAP Quinasas , Pinocitosis , Receptor Toll-Like 4/metabolismo , Animales , Células COS , Chlorocebus aethiops , Ésteres del Colesterol/genética , Enfermedad Crónica , Citoesqueleto/genética , Citoesqueleto/metabolismo , Citoesqueleto/patología , Células Espumosas/patología , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ligandos , Lipoproteínas LDL/genética , Ratones , Ratones Mutantes , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/genética , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Quinasa Syk , Receptor Toll-Like 4/genética , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
5.
Nature ; 437(7059): 759-63, 2005 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-16127449

RESUMEN

Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) has essential roles in adipogenesis and glucose homeostasis, and is a molecular target of insulin-sensitizing drugs. Although the ability of PPAR-gamma agonists to antagonize inflammatory responses by transrepression of nuclear factor kappa B (NF-kappaB) target genes is linked to antidiabetic and antiatherogenic actions, the mechanisms remain poorly understood. Here we report the identification of a molecular pathway by which PPAR-gamma represses the transcriptional activation of inflammatory response genes in mouse macrophages. The initial step of this pathway involves ligand-dependent SUMOylation of the PPAR-gamma ligand-binding domain, which targets PPAR-gamma to nuclear receptor corepressor (NCoR)-histone deacetylase-3 (HDAC3) complexes on inflammatory gene promoters. This in turn prevents recruitment of the ubiquitylation/19S proteosome machinery that normally mediates the signal-dependent removal of corepressor complexes required for gene activation. As a result, NCoR complexes are not cleared from the promoter and target genes are maintained in a repressed state. This mechanism provides an explanation for how an agonist-bound nuclear receptor can be converted from an activator of transcription to a promoter-specific repressor of NF-kappaB target genes that regulate immunity and homeostasis.


Asunto(s)
Regulación hacia Abajo , Inflamación/genética , PPAR gamma/metabolismo , Proteínas Represoras/metabolismo , Proteína SUMO-1/metabolismo , Animales , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Histona Desacetilasas/metabolismo , Ligandos , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Ratones , Complejos Multiproteicos/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Proteínas Nucleares/metabolismo , Co-Represor 1 de Receptor Nuclear , Unión Proteica/efectos de los fármacos , Proteínas Inhibidoras de STAT Activados , Proteínas/metabolismo
6.
Artículo en Inglés | MEDLINE | ID: mdl-34083860

RESUMEN

PDT efficacy depends on the availability and dynamic interactions of photosensitizer, light, and oxygen. Tissue optical properties influence the delivered light dose and impact PDT outcome. In-vivo measurements of tissue optical properties and photosensitizer concentration enable determination of explicit and implicit dose factors affecting PDT and helps to understand the underlying biophysical mechanism of PDT. In this study, we measure tissue optical properties (absorption µa (λ) and scattering µs' (λ) coefficients) and PpIX concentration in tissue simulating liquid phantoms with a geometry that resembles anal canal. In-vivo light fluence rate and photosensitizer fluorescence of 405nm excitation light source were acquired using a dual-motor continuous wave transmittance spectroscopy system. We characterized the tissue optical properties correction factor of fluorescence signal using a series of tissue simulating phantoms with known PpIX concentrations and with absorption coefficient between 0.1 - 0.9 cm-1 and reduced scattering coefficient between 5 - 40 cm-1. The results demonstrated that our spectroscopy system could determine the distribution of tissue optical properties and PPIX concentration during anal PDT.

7.
Artículo en Inglés | MEDLINE | ID: mdl-34083859

RESUMEN

Tissue optical properties are crucial for determining the light dose delivered to the tumor. Two probes are compared: the two-catheter probe is based on transmittance measurement between one point source and one isotropic detector inside parallel catheters spaced at 0.5 cm along a 1-inch diameter transparent cylinder; and a 1-inch trans-rectal diffuse optical tomography (DOT) probe designed for prostate measurements, using a multiple fiber-array with source-detector separations of 1.4-10 mm. The two-catheter probe uses an empirical model for primary and scatter light fluence rates in the cylindrical cavity condition for anal PDT to determine optical properties along the source catheter using dual motors to move the source and detector along the catheters. The DOT probe uses finite element method (FEM) to obtain distribution of optical properties in 3D. Validations for the two probes were performed in liquid and solid phantoms. For each method, validation was performed in tissue-mimicking liquid phantoms for a range of known optical properties (µa between 0.05 and 0.9 cm-1 and µs' between 5.5 and 16.5 cm-1). To cross-check the two methods, solid phantoms were created of known optical properties at the University of Pennsylvania and sent for measurement to Princess Margaret Cancer Centre (PMH) to mimic realistic patient simulating conditions. Measurements were taken and optical properties were then recovered without knowing the expected values to cross-validate each probe. The results show modest agreement between the measured µa and µs'values, but high degree of agreement between the measured µeff performed independently using the two methods.

8.
J Lipid Res ; 51(5): 900-6, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20388921

RESUMEN

The liver X receptors LXRalpha and LXRbeta play critical roles in maintaining lipid homeostasis by functioning as transcription factors that regulate genetic networks controlling the transport, catabolism, and excretion of cholesterol. The studies described in this report examine the individual anti-atherogenic activity of LXRalpha and LXRbeta and determine the ability of each subtype to mediate the biological response to LXR agonists. Utilizing individual knockouts of LXRalpha and LXRbeta in the Ldlr(-/-) background, we demonstrate that LXRalpha has a dominant role in limiting atherosclerosis in vivo. Functional studies in macrophages indicate that LXRalpha is required for a robust response to LXR ligands, whereas LXRbeta functions more strongly as a repressor. Furthermore, selective knockout of LXRalpha in hematopoietic cells and rescue experiments indicate that the anti-atherogenic activity of this LXR subtype is not restricted to macrophages. These studies indicate that LXRalpha plays a selective role in limiting atherosclerosis in response to hyperlipidemia.


Asunto(s)
Aterosclerosis/metabolismo , Técnicas de Inactivación de Genes , Receptores Nucleares Huérfanos/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Animales , Células de la Médula Ósea/metabolismo , Susceptibilidad a Enfermedades/metabolismo , Regulación de la Expresión Génica , Receptores X del Hígado , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Receptores Nucleares Huérfanos/agonistas , Receptores Nucleares Huérfanos/deficiencia , Receptores Nucleares Huérfanos/genética
9.
Arterioscler Thromb Vasc Biol ; 27(4): 878-85, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17255537

RESUMEN

OBJECTIVE: Western-type high-fat/high-cholesterol diets used to induce atherogenesis in low-density lipoprotein (LDL) receptor-deficient mice also lead to obesity with concomitant metabolic complications, eg, hypertriglyceridemia, hyperinsulinemia, and insulin resistance. Our aim was to design a diet inducing atherosclerosis through moderate hypercholesterolemia without associated parameters of the metabolic syndrome. METHODS AND RESULTS: Male LDL receptor-deficient mice were fed regular chow (RC; 0.01% cholesterol/4.4% fat), cholesterol-enriched regular chow (HC; 1% cholesterol/4.4% fat), or Western diet (WD 0.06% cholesterol/21% milk fat) for 28 weeks. HC-feeding led to elevated plasma (approximately 20.7 mmol/L [800 mg/dL]) and LDL cholesterol and accelerated atherosclerosis. Plasma triglycerides were unaffected. Compared with RC-fed controls, HC-fed mice had normal body weight gain and normal fasting levels of glucose, free fatty acids, and insulin. In contrast, WD-fed mice were extremely hypercholesterolemic (>41.4 mmol/L), obese, hypertriglyceridemic, hyperinsulinemic, insulin resistant, and showed adverse health such as skin/fur abnormalities and hepatic steatosis. Although atherosclerotic surface areas in the entire aorta were similar in HC-fed and WD-fed mice, lesions in aortic origin cross sections were significantly larger in WD-fed mice. However, morphology was similar in lesions of equal size. CONCLUSIONS: The HC diet induced moderate hypercholesterolemia and extensive atherosclerosis and should be useful to study specific aspects of atherogenesis in the absence of confounding effects of the metabolic syndrome.


Asunto(s)
Aterosclerosis/etiología , Colesterol en la Dieta , Dieta Aterogénica , Hipercolesterolemia/etiología , Animales , Aorta/patología , Aterosclerosis/patología , Glucemia/metabolismo , Peso Corporal , Hipercolesterolemia/complicaciones , Insulina/metabolismo , Metabolismo de los Lípidos , Lípidos/sangre , Lipoproteínas/sangre , Hígado/metabolismo , Masculino , Síndrome Metabólico/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Tamaño de los Órganos
10.
J Clin Invest ; 114(11): 1564-76, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15578089

RESUMEN

PPARalpha, beta/delta, and gamma regulate genes involved in the control of lipid metabolism and inflammation and are expressed in all major cell types of atherosclerotic lesions. In vitro studies have suggested that PPARs exert antiatherogenic effects by inhibiting the expression of proinflammatory genes and enhancing cholesterol efflux via activation of the liver X receptor-ABCA1 (LXR-ABCA1) pathway. To investigate the potential importance of these activities in vivo, we performed a systematic analysis of the effects of PPARalpha, beta, and gamma agonists on foam-cell formation and atherosclerosis in male LDL receptor-deficient (LDLR(-/-)) mice. Like the PPARgamma agonist, a PPARalpha-specific agonist strongly inhibited atherosclerosis, whereas a PPARbeta-specific agonist failed to inhibit lesion formation. In concert with their effects on atherosclerosis, PPARalpha and PPARgamma agonists, but not the PPARbeta agonist, inhibited the formation of macrophage foam cells in the peritoneal cavity. Unexpectedly, PPARalpha and PPARgamma agonists inhibited foam-cell formation in vivo through distinct ABCA1-independent pathways. While inhibition of foam-cell formation by PPARalpha required LXRs, activation of PPARgamma reduced cholesterol esterification, induced expression of ABCG1, and stimulated HDL-dependent cholesterol efflux in an LXR-independent manner. In concert, these findings reveal receptor-specific mechanisms by which PPARs influence macrophage cholesterol homeostasis. In the future, these mechanisms may be exploited pharmacologically to inhibit the development of atherosclerosis.


Asunto(s)
Arteriosclerosis/metabolismo , Células Espumosas/fisiología , PPAR alfa/metabolismo , PPAR delta/metabolismo , PPAR gamma/metabolismo , PPAR-beta/metabolismo , Animales , Aorta/citología , Aorta/metabolismo , Aorta/patología , Arteriosclerosis/patología , Colesterol/metabolismo , Colesterol en la Dieta , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Humanos , Receptores X del Hígado , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Receptores Nucleares Huérfanos , PPAR alfa/agonistas , PPAR alfa/genética , PPAR delta/agonistas , PPAR delta/genética , PPAR gamma/agonistas , PPAR gamma/genética , PPAR-beta/agonistas , PPAR-beta/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Triglicéridos/metabolismo
11.
Arterioscler Thromb Vasc Biol ; 26(10): 2301-7, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16888235

RESUMEN

OBJECTIVE: The lungs of Abcg1-/- mice accumulate macrophage foam cells that contain high levels of unesterified and esterified cholesterol, consistent with a role for ABCG1 in facilitating the efflux of cholesterol from macrophages to high-density lipoprotein (HDL) and other exogenous sterol acceptors. Based on these observations, we investigated whether loss of ABCG1 affects foam cell deposition in the artery wall and the development of atherosclerosis. METHODS AND RESULTS: Bone marrow from wild-type or Abcg1-/- mice was transplanted into Ldlr-/- or ApoE-/- mice. After administration of a high-fat/high-cholesterol diet, plasma and tissue lipid levels and atherosclerotic lesion size were quantified and compared. Surprisingly, transplantation of Abcg1-/- bone marrow cells resulted in a significant reduction in lesion size in both mouse models, despite the fact that lipid levels increased in the lung, spleen, and kidney. Lesions of Ldlr-/- mice transplanted with Abcg1-/- cells contained increased numbers of apoptotic cells. Consistent with this observation, in vitro studies demonstrated that Abcg1-/- macrophages were more susceptible to oxidized low-density lipoprotein (ox-LDL)-dependent apoptosis than Abcg1+/+ cells. CONCLUSIONS: Diet-induced atherosclerosis is impaired when atherosclerotic-susceptible mice are transplanted with Abcg1-/- bone marrow. The demonstration that Abcg1-/- macrophages undergo accelerated apoptosis provides a mechanism to explain the decrease in the atherosclerotic lesions.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/prevención & control , Trasplante de Médula Ósea , Médula Ósea/metabolismo , Hiperlipidemias/metabolismo , Lipoproteínas/deficiencia , Receptores de LDL/deficiencia , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Apoptosis , Aterosclerosis/etiología , Grasas de la Dieta/administración & dosificación , Femenino , Silenciador del Gen , Hiperlipidemias/fisiopatología , Hiperlipidemias/cirugía , Lipoproteínas/genética , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
13.
EMBO Mol Med ; 5(9): 1443-57, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23964012

RESUMEN

Conjugated linoleic acid (CLA) has the unique property of inducing regression of pre-established murine atherosclerosis. Understanding the mechanism(s) involved may help identify endogenous pathways that reverse human atherosclerosis. Here, we provide evidence that CLA inhibits foam cell formation via regulation of the nuclear receptor coactivator, peroxisome proliferator-activated receptor (PPAR)-γ coactivator (PGC)-1α, and that macrophage PGC-1α plays a role in atheroprotection in vivo. PGC-1α was identified as a hub gene within a cluster in the aorta of the apoE(-/-) mouse in the CLA-induced regression model. PGC-1α was localized to macrophage/foam cells in the murine aorta where its expression was increased during CLA-induced regression. PGC-1α expression was also detected in macrophages in human atherosclerosis and was inversely linked to disease progression in patients with the disease. Deletion of PGC-1α in bone marrow derived macrophages promoted, whilst over expression of the gene inhibited foam cell formation. Importantly, macrophage specific deletion of PGC-1α accelerated atherosclerosis in the LDLR(-/-) mouse in vivo. These novel data support a functional role for PGC-1α in atheroprotection.


Asunto(s)
Aterosclerosis/fisiopatología , Células Espumosas/efectos de los fármacos , Ácidos Linoleicos Conjugados/metabolismo , Factores de Transcripción/metabolismo , Animales , Aorta/fisiopatología , Células Cultivadas , Eliminación de Gen , Expresión Génica , Humanos , Ratones , Ratones Noqueados , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma
14.
J Appl Physiol (1985) ; 115(11): 1694-704, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23990245

RESUMEN

Patients with obstructive sleep apnea, who experience episodic hypoxia and hypercapnia during sleep, often demonstrate increased inflammation, oxidative stress, and dyslipidemia. We hypothesized that sleep apnea patients would be predisposed to the development of atherosclerosis. To dissect the mechanisms involved, we developed an animal model in mice whereby we expose mice to intermittent hypoxia/hypercapnia (IHH) in normobaric environments. Two- to three-month-old low-density lipoprotein receptor deficient (Ldlr(-/-)) mice were fed a high-fat diet for 8 or 16 wk while being exposed to IHH for either 10 h/day or 24 h/day. Plasma lipid levels, pulmonary artery and aortic atherosclerotic lesions, and cardiac function were then assayed. Surprisingly, atherosclerosis in the aorta of IHH mice was similar compared with controls. However, in IHH mice, atherosclerosis was markedly increased in the trunk and proximal branches of the pulmonary artery of exposed mice; even though plasma cholesterol and triglycerides were lower than in controls. Hemodynamic analysis revealed that right ventricular maximum pressure and isovolumic relaxation constant were significantly increased in IHH exposed mice and left ventricular % fractional shortening was reduced. In conclusion, 1) Intermittent hypoxia/hypercapnia remarkably accelerated atherosclerotic lesions in the pulmonary artery of Ldlr(-/-) mice and 2) increased lesion formation in the pulmonary artery was associated with right and left ventricular dysfunction. These findings raise the possibility that patients with obstructive sleep apnea may be susceptible to atherosclerotic disease in the pulmonary vasculature, an observation that has not been previously recognized.


Asunto(s)
Aterosclerosis/patología , Hipercapnia/patología , Hipoxia/patología , Arteria Pulmonar/patología , Receptores de LDL/deficiencia , Disfunción Ventricular/patología , Animales , Aterosclerosis/sangre , Aterosclerosis/metabolismo , Colesterol/sangre , Modelos Animales de Enfermedad , Hemodinámica/fisiología , Hipercapnia/sangre , Hipercapnia/metabolismo , Hipoxia/sangre , Hipoxia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Arteria Pulmonar/metabolismo , Receptores de LDL/metabolismo , Apnea Obstructiva del Sueño/sangre , Apnea Obstructiva del Sueño/metabolismo , Apnea Obstructiva del Sueño/patología , Triglicéridos/sangre , Disfunción Ventricular/sangre , Disfunción Ventricular/metabolismo
15.
Cholesterol ; 2012: 564705, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22693663

RESUMEN

Cholesterol-fed zebrafish is an emerging animal model to study metabolic, oxidative, and inflammatory vascular processes relevant to pathogenesis of human atherosclerosis. Zebrafish fed a high-cholesterol diet (HCD) develop hypercholesterolemia and are characterized by profound lipoprotein oxidation and vascular lipid accumulation. Using optically translucent zebrafish larvae has the advantage of monitoring vascular pathology and assessing the efficacy of drug candidates in live animals. Thus, we investigated whether simvastatin and ezetimibe, the principal drugs used in management of hypercholesterolemia in humans, would also reduce cholesterol levels in HCD-fed zebrafish larvae. We found that ezetimibe was well tolerated by zebrafish and effectively reduced cholesterol levels in HCD-fed larvae. In contrast, simvastatin added to water was poorly tolerated by zebrafish larvae and, when added to food, had little effect on cholesterol levels in HCD-fed larvae. Combination of low doses of ezetimibe and simvastatin had an additive effect in reducing cholesterol levels in zebrafish. These results suggest that ezetimibe exerts in zebrafish a therapeutic effect similar to that in humans and that the hypercholesterolemic zebrafish can be used as a low-cost and informative model for testing new drug candidates and for investigating mechanisms of action for existing drugs targeting dyslipidemia.

16.
J Am Coll Cardiol ; 58(16): 1715-27, 2011 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-21982317

RESUMEN

OBJECTIVES: We sought to assess the in vivo importance of scavenger receptor (SR)-mediated uptake of oxidized low-density lipoprotein (OxLDL) in atherogenesis and to test the efficacy of human antibody IK17-Fab or IK17 single-chain Fv fragment (IK17-scFv), which lacks immunologic properties of intact antibodies other than the ability to inhibit uptake of OxLDL by macrophages, to inhibit atherosclerosis. BACKGROUND: The unregulated uptake of OxLDL by macrophage SR contributes to foam cell formation, but the importance of this pathway in vivo is uncertain. METHODS: Cholesterol-fed low-density lipoprotein receptor knockout (LDLR(-/-)) mice were treated with intraperitoneal infusion of human IK17-Fab (2.5 mg/kg) 3 times per week for 14 weeks. Because anti-human antibodies developed in these mice, LDLR(-/-)/low-density lipoprotein receptor Rag 1 double-knockout mice (lacking the ability to make immunoglobulins due to loss of T- and B-cell function) were treated with an adenoviral vector encoding adenovirus expressed (Adv)-IK17-scFv or control adenovirus-enhanced green fluorescent protein vector intravenously every 2 weeks for 16 weeks. RESULTS: In LDLR(-/-) mice, infusion of IK17-Fab was able to sustain IK17 plasma levels for the first 8 weeks, but these diminished afterward due to increasing murine anti-IK17 antibody titers. Despite this, after 14 weeks, a 29% decrease in en face atherosclerosis was noted compared with phosphate-buffered saline-treated mice. In LDLR(-/-)/low-density lipoprotein receptor Rag 1 double-knockout mice, sustained levels of plasma IK17-scFv was achieved by Adv-IK17-scFv-mediated hepatic expression, which led to a 46% reduction (p < 0.001) in en face atherosclerosis compared with adenovirus-enhanced green fluorescent protein vector. Importantly, peritoneal macrophages isolated from Adv-IK17-scFv treated mice had decreased lipid accumulation compared with adenovirus-enhanced green fluorescent protein-treated mice. CONCLUSIONS: These data support an important role for SR-mediated uptake of OxLDL in the pathogenesis of atherosclerosis and demonstrate that oxidation-specific antibodies reduce the progression of atherosclerosis, suggesting their potential in treating cardiovascular disease in humans.


Asunto(s)
Anticuerpos/química , Aterosclerosis/inmunología , Aterosclerosis/patología , Células Espumosas/metabolismo , Lipoproteínas LDL/metabolismo , Adenoviridae/metabolismo , Animales , Aterosclerosis/terapia , Progresión de la Enfermedad , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Humanos , Fragmentos de Inmunoglobulinas/química , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de LDL/genética , Proteínas Recombinantes/química
17.
Artículo en Inglés | MEDLINE | ID: mdl-16402897

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) alpha (alpha), beta/delta (beta/delta), and gamma (gamma) are members of the nuclear receptor superfamily, which also includes the estrogen, androgen, and glucocorticoid receptors. Recent evidence suggests that PPARs regulate genes involved in lipid metabolism, glucose homeostasis, and inflammation in various tissues; however, the mechanisms involved are not completely understood. Anti-diabetic drugs, called glitazones, can selectively activate PPARgamma, and hypolipidemic drugs, called fibrates, can weakly activate PPARalpha. Both classes of drugs can decrease insulin resistance and dyslipidemias, which also makes them attractive for treating the metabolic syndrome. The metabolic syndrome exhibits a constellation of risk factors for atherosclerosis that include obesity, insulin resistance, dyslipidemias, and hypertension. Interestingly, all three PPARs are present in macrophages and can therefore have a profound effect on several disease processes, including atherosclerosis. Macrophages are key players in atherosclerotic lesion development. Currently, the first line of defense in reducing the risk of atherosclerosis is aimed at lowering low-density lipoproteins (LDL) and raising high-density lipoproteins (HDL), but a large percentage of patients on statins still succumb to coronary artery disease. However, with the development of drugs selectively activating PPARs, a new arsenal of drugs specifically targeting to the macrophage/foam cell may potentially have a profound impact on how we treat cardiovascular disease.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Fármacos Cardiovasculares/uso terapéutico , Macrófagos/efectos de los fármacos , Receptores Activados del Proliferador del Peroxisoma/efectos de los fármacos , Receptores Activados del Proliferador del Peroxisoma/fisiología , Animales , Aterosclerosis/fisiopatología , Fármacos Cardiovasculares/farmacología , Humanos , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos
18.
J Biol Chem ; 281(44): 33066-77, 2006 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-16950764

RESUMEN

Unlike the livers of humans and mice, and most hepatoma cells, which accumulate triglycerides when treated with microsomal triglyceride transfer protein (MTP) inhibitors, L35 rat hepatoma cells do not express MTP and cannot secrete very low density lipoprotein (VLDL), yet they do not accumulate triglyceride. In these studies we show that transcriptional co-repression of the two lipid transfer proteins, liver fatty acid-binding protein (L-FABP) and MTP, which cooperatively shunt fatty acids into de novo synthesized glycerolipids and the transfer of lipids into VLDL, respectively, act together to maintain hepatic lipid homeostasis. FAO rat hepatoma cells express L-FABP and MTP and demonstrate the ability to assemble and secrete VLDL. In contrast, L35 cells, derived as a single cell clone from FAO cells, do not express L-FABP or MTP nor do they assemble and secrete VLDL. We used these hepatoma cells to elucidate how a conserved DR1 promoter element present in the promoters of L-FABP and MTP affects transcription, expression, and VLDL production. In FAO cells, the DR1 elements of both L-FABP and MTP promoters are occupied by peroxisome proliferator-activated receptor alpha-retinoid X receptor alpha (RXRalpha), with which PGC-1beta activates transcription. In contrast, in L35 cells the DR1 elements of both L-FABP and MTP promoters are occupied by chicken ovalbumin upstream promoter transcription factor II, and transcription is diminished. The combined findings indicate that peroxisome proliferator-activated receptor alpha-RXRalpha and PGC-1beta coordinately up-regulate L-FABP and MTP expression, by competing with chicken ovalbumin upstream promoter transcription factor II for the DR1 sites in the proximal promoters of each gene. Additional studies show that ablation of L-FABP prevents hepatic steatosis caused by treating mice with an MTP inhibitor. Our findings show that reducing both L-FABP and MTP is an effective means to reduce VLDL secretion without causing hepatic steatosis.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de Unión a Ácidos Grasos/genética , Lipoproteínas VLDL/metabolismo , Hígado/metabolismo , Transcripción Genética/genética , Animales , Apolipoproteínas B/metabolismo , Secuencia de Bases , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Línea Celular , Dimerización , Proteínas de Unión a Ácidos Grasos/deficiencia , Proteínas de Unión a Ácidos Grasos/metabolismo , Hígado Graso/genética , Hígado Graso/metabolismo , Expresión Génica/genética , Genes Reporteros/genética , Factor Nuclear 1-beta del Hepatocito/metabolismo , Masculino , Ratones , Ratones Noqueados , PPAR alfa/agonistas , PPAR alfa/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Mensajero/genética , Ratas , Receptores X Retinoide/agonistas , Receptores X Retinoide/metabolismo , Transcripción Genética/efectos de los fármacos
19.
Proc Natl Acad Sci U S A ; 102(31): 11029-34, 2005 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-16043712

RESUMEN

Resident macrophages (i.e., Kupffer cells) are derived from hematopoietic stem cells (HSCs) and are primarily responsible for the removal from plasma of oxidized forms of low-density lipoprotein (LDL). The therapeutic potential of Kupffer cell expression of a transgene encoding paraoxonase-1 (PON1), whose plasma activity correlates with the protection from atherosclerosis, was examined in mice rendered atherosclerosis-susceptible through genetic deletion of the LDL receptor. Mice having their bone marrow engrafted with HSCs expressing the PON1 transgene (PON1-Tg) driven by a macrophage-specific promoter were injected i.v. with saline (vehicle only) or with gadolinium chloride (GdCl(3)), an agent that rapidly causes Kupffer cell apoptosis. One month later, GdCl(3)-facilitated Kupffer cell apoptosis increased the hepatic expression of transgenic PON1 mRNA by 9-fold. After 12 weeks of being fed a cholesterol-enriched atherogenic diet, mice injected with GdCl(3) exhibited 50% reductions in both aortic sinus atherosclerotic lesions (P < 0.0097) and surface lesions of the abdominal aorta (P < 0.006). In contrast, mice receiving HSCs expressing the PON1-Tg but not treated with GdCl(3) showed no protection from atherosclerosis. In addition, mice engrafted with HSCs not expressing the PON1-Tg but injected with GdCl(3) also showed no protection from atherosclerosis. These findings, showing that GdCl(3)-enhanced hepatic expression of the PON1-Tg is essential for reducing atherosclerosis, indicate that Kupffer cells play an important role in atherogenesis. GdCl(3)-facilated replacement of Kupffer cells may enhance the efficacy of other HSC-based gene therapies.


Asunto(s)
Arteriosclerosis/terapia , Arildialquilfosfatasa/genética , Gadolinio/farmacología , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/enzimología , Animales , Arteriosclerosis/enzimología , Arteriosclerosis/genética , Arteriosclerosis/patología , Secuencia de Bases , Trasplante de Médula Ósea , ADN Complementario/genética , Expresión Génica , Terapia Genética , Macrófagos del Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de LDL/deficiencia , Receptores de LDL/genética
20.
J Lipid Res ; 45(12): 2161-73, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15489539

RESUMEN

The nuclear receptor superfamily is composed of transcription factors that positively and negatively regulate gene expression in response to the binding of a diverse array of lipid-derived hormones and metabolites. Intense efforts are currently being directed at defining the biological roles and mechanisms of action of liver X receptors (LXRs) and peroxisome proliferator-activated receptors (PPARs). LXRs have been found to play essential roles in the regulation of whole body cholesterol absorption and excretion, in the efflux of cholesterol from peripheral cells, and in the biosynthesis and metabolism of very low density lipoproteins. PPARs have been found to regulate diverse aspects of lipid metabolism, including fatty acid oxidation, fat cell development, lipoprotein metabolism, and glucose homeostasis. Intervention studies indicate that activation of PPARalpha, PPARgamma, and LXRs by specific synthetic ligands can inhibit the development of atherosclerosis in animal models. Here, we review recent studies that provide new insights into the mechanisms by which these subclasses of nuclear receptors act to systemically influence lipid and glucose metabolism and regulate gene expression within the artery wall.


Asunto(s)
Arteriosclerosis/metabolismo , Metabolismo de los Lípidos , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Arteriosclerosis/etiología , Proteínas de Unión al ADN , Humanos , Receptores X del Hígado , Receptores Nucleares Huérfanos , Receptores Activados del Proliferador del Peroxisoma/química , Receptores Citoplasmáticos y Nucleares/química , Factores de Transcripción/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA