Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 128(12): 2236-48, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25956888

RESUMEN

Autocrine VEGF is necessary for endothelial survival, although the cellular mechanisms supporting this function are unknown. Here, we show that--even after full differentiation and maturation--continuous expression of VEGF by endothelial cells is needed to sustain vascular integrity and cellular viability. Depletion of VEGF from the endothelium results in mitochondria fragmentation and suppression of glucose metabolism, leading to increased autophagy that contributes to cell death. Gene-expression profiling showed that endothelial VEGF contributes to the regulation of cell cycle and mitochondrial gene clusters, as well as several--but not all--targets of the transcription factor FOXO1. Indeed, VEGF-deficient endothelium in vitro and in vivo showed increased levels of FOXO1 protein in the nucleus and cytoplasm. Silencing of FOXO1 in VEGF-depleted cells reversed expression profiles of several of the gene clusters that were de-regulated in VEGF knockdown, and rescued both cell death and autophagy phenotypes. Our data suggest that endothelial VEGF maintains vascular homeostasis through regulation of FOXO1 levels, thereby ensuring physiological metabolism and endothelial cell survival.


Asunto(s)
Apoptosis , Comunicación Autocrina , Autofagia , Biomarcadores/metabolismo , Endotelio Vascular/patología , Factores de Transcripción Forkhead/metabolismo , Mitocondrias/patología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Western Blotting , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Endotelio Vascular/metabolismo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Humanos , Hipoxia/fisiopatología , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Fosforilación , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
2.
FASEB J ; 29(4): 1185-97, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25477283

RESUMEN

We report the engineering and characterization of paraoxonase-3 knockout mice (Pon3KO). The mice were generally healthy but exhibited quantitative alterations in bile acid metabolism and a 37% increased body weight compared to the wild-type mice on a high fat diet. PON3 was enriched in the mitochondria-associated membrane fraction of hepatocytes. PON3 deficiency resulted in impaired mitochondrial respiration, increased mitochondrial superoxide levels, and increased hepatic expression of inflammatory genes. PON3 deficiency did not influence atherosclerosis development on an apolipoprotein E null hyperlipidemic background, but it did lead to a significant 60% increase in atherosclerotic lesion size in Pon3KO mice on the C57BL/6J background when fed a cholate-cholesterol diet. On the diet, the Pon3KO had significantly increased plasma intermediate-density lipoprotein/LDL cholesterol and bile acid levels. They also exhibited significantly elevated levels of hepatotoxicity markers in circulation, a 58% increase in gallstone weight, a 40% increase in hepatic cholesterol level, and increased mortality. Furthermore, Pon3KO mice exhibited decreased hepatic bile acid synthesis and decreased bile acid levels in the small intestine compared with wild-type mice. Our study suggests a role for PON3 in the metabolism of lipid and bile acid as well as protection against atherosclerosis, gallstone disease, and obesity.


Asunto(s)
Arildialquilfosfatasa/deficiencia , Aterosclerosis/enzimología , Cálculos Biliares/enzimología , Obesidad/enzimología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Arildialquilfosfatasa/genética , Arildialquilfosfatasa/metabolismo , Aterosclerosis/etiología , Aterosclerosis/genética , Ácidos y Sales Biliares/metabolismo , Quimiocina CCL2/metabolismo , Colesterol en la Dieta/administración & dosificación , Ácido Cólico/administración & dosificación , Dieta/efectos adversos , Modelos Animales de Enfermedad , Femenino , Cálculos Biliares/etiología , Cálculos Biliares/genética , Expresión Génica , Predisposición Genética a la Enfermedad , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Intestino Delgado/metabolismo , Riñón/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Hepáticas/metabolismo , Obesidad/etiología , Obesidad/genética
3.
Nat Genet ; 37(11): 1224-33, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16200066

RESUMEN

Forward genetic approaches to identify genes involved in complex traits such as common human diseases have met with limited success. Fine mapping of linkage regions and validation of positional candidates are time-consuming and not always successful. Here we detail a hybrid procedure to map loci involved in complex traits that leverages the strengths of forward and reverse genetic approaches. By integrating genotypic and expression data in a segregating mouse population, we show how clusters of expression quantitative trait loci linking to regions of the genome accurately reflect the underlying perturbation to the transcriptional network induced by DNA variations in genes that control the complex traits. By matching patterns of gene expression in a segregating population with expression responses induced by single-gene perturbation experiments, we show how genes controlling clusters of expression and clinical quantitative trait loci can be mapped directly. We demonstrate the utility of this approach by identifying 5-lipoxygenase as underlying previously identified quantitative trait loci in an F(2) cross between strains C57BL/6J and DBA/2J and showing that it has pleiotropic effects on body fat, lipid levels and bone density.


Asunto(s)
Araquidonato 5-Lipooxigenasa/genética , Densidad Ósea/genética , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Obesidad/genética , Animales , Cruzamientos Genéticos , Femenino , Genoma , Genotipo , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Modelos Biológicos , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , PPAR gamma/genética , Sitios de Carácter Cuantitativo
4.
Biochim Biophys Acta ; 1821(3): 435-47, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21807117

RESUMEN

We report a systems genetic analysis of high density lipoprotein (HDL) levels in an F2 intercross between inbred strains CAST/EiJ and C57BL/6J. We previously showed that there are dramatic differences in HDL metabolism in a cross between these strains, and we now report co-expression network analysis of HDL that integrates global expression data from liver and adipose with relevant metabolic traits. Using data from a total of 293 F2 intercross mice, we constructed weighted gene co-expression networks and identified modules (subnetworks) associated with HDL and clinical traits. These were examined for genes implicated in HDL levels based on large human genome-wide associations studies (GWAS) and examined with respect to conservation between tissue and sexes in a total of 9 data sets. We identify genes that are consistently ranked high by association with HDL across the 9 data sets. We focus in particular on two genes, Wfdc2 and Hdac3, that are located in close proximity to HDL QTL peaks where causal testing indicates that they may affect HDL. Our results provide a rich resource for studies of complex metabolic interactions involving HDL. This article is part of a Special Issue entitled Advances in High Density Lipoprotein Formation and Metabolism: A Tribute to John F. Oram (1945-2010).


Asunto(s)
Histona Desacetilasas/genética , Lipoproteínas HDL/metabolismo , Proteínas/genética , Tejido Adiposo/metabolismo , Análisis de Varianza , Animales , Colesterol/metabolismo , Cruzamientos Genéticos , Dieta Alta en Grasa , Femenino , Redes Reguladoras de Genes , Hibridación Genética , Hígado/metabolismo , Escala de Lod , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Transcriptoma , Proteína 2 de Dominio del Núcleo de Cuatro Disulfuros WAP
5.
Circulation ; 126(15): 1896-906, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-22952318

RESUMEN

BACKGROUND: The human 9p21.3 chromosome locus has been shown to be an independent risk factor for atherosclerosis in multiple large-scale genome-wide association studies, but the underlying mechanism remains unknown. We set out to investigate the potential role of the 9p21.3 locus neighboring genes, including Mtap, the 2 isoforms of Cdkn2a, p16Ink4a and p19Arf, and Cdkn2b, in atherosclerosis using knockout mice models. METHODS AND RESULTS: Gene-targeted mice for neighboring genes, including Mtap, Cdkn2a, p19Arf, and Cdkn2b, were each bred to mice carrying the human APO*E3 Leiden transgene that sensitizes the mice for atherosclerotic lesions through elevated plasma cholesterol. We found that the mice heterozygous for Mtap developed larger lesions compared with wild-type mice (49623±21650 versus 18899±9604 µm(2) per section [mean±SD]; P=0.01), with morphology similar to that of wild-type mice. The Mtap heterozygous mice demonstrated changes in metabolic and methylation profiles and CD4(+) cell counts. The Cdkn2a knockout mice had smaller lesions compared with wild-type and heterozygous mice, and there were no significant differences in lesion size in p19Arf and Cdkn2b mutants compared with wild type. We observed extensive, tissue-specific compensatory regulation of the Cdkn2a and Cdkn2b genes among the various knockout mice, making the effects on atherosclerosis difficult to interpret. CONCLUSIONS: Mtap plays a protective role against atherosclerosis, whereas Cdkn2a appears to be modestly proatherogenic. However, no relation was found between the 9p21 genotype and the transcription of 9p21 neighboring genes in primary human aortic vascular cells in vitro. There is extensive compensatory regulation in the highly conserved 9p21 orthologous region in mice.


Asunto(s)
Aterosclerosis/genética , Enfermedad de la Arteria Coronaria/genética , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética
6.
Genome Res ; 20(2): 281-90, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20054062

RESUMEN

Systems genetics relies on common genetic variants to elucidate biologic networks contributing to complex disease-related phenotypes. Mice are ideal model organisms for such approaches, but linkage analysis has been only modestly successful due to low mapping resolution. Association analysis in mice has the potential of much better resolution, but it is confounded by population structure and inadequate power to map traits that explain less than 10% of the variance, typical of mouse quantitative trait loci (QTL). We report a novel strategy for association mapping that combines classic inbred strains for mapping resolution and recombinant inbred strains for mapping power. Using a mixed model algorithm to correct for population structure, we validate the approach by mapping over 2500 cis-expression QTL with a resolution an order of magnitude narrower than traditional QTL analysis. We also report the fine mapping of metabolic traits such as plasma lipids. This resource, termed the Hybrid Mouse Diversity Panel, makes possible the integration of multiple data sets and should prove useful for systems-based approaches to complex traits and studies of gene-by-environment interactions.


Asunto(s)
Mapeo Cromosómico/métodos , Estudio de Asociación del Genoma Completo/métodos , Sitios de Carácter Cuantitativo/genética , Algoritmos , Animales , Ligamiento Genético , Lipoproteínas HDL/genética , Masculino , Ratones , Ratones Endogámicos , Fenotipo
7.
Nat Genet ; 30(1): 110-6, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11753387

RESUMEN

Familial combined hyperlipidemia (FCHL, MIM-144250) is a common, multifactorial and heterogeneous dyslipidemia predisposing to premature coronary artery disease and characterized by elevated plasma triglycerides, cholesterol, or both. We identified a mutant mouse strain, HcB-19/Dem (HcB-19), that shares features with FCHL, including hypertriglyceridemia, hypercholesterolemia, elevated plasma apolipoprotein B and increased secretion of triglyceride-rich lipoproteins. The hyperlipidemia results from spontaneous mutation at a locus, Hyplip1, on distal mouse chromosome 3 in a region syntenic with a 1q21-q23 FCHL locus identified in Finnish, German, Chinese and US families. We fine-mapped Hyplip1 to roughly 160 kb, constructed a BAC contig and sequenced overlapping BACs to identify 13 candidate genes. We found substantially decreased mRNA expression for thioredoxin interacting protein (Txnip). Sequencing of the critical region revealed a Txnip nonsense mutation in HcB-19 that is absent in its normolipidemic parental strains. Txnip encodes a cytoplasmic protein that binds and inhibits thioredoxin, a major regulator of cellular redox state. The mutant mice have decreased CO2 production but increased ketone body synthesis, suggesting that altered redox status down-regulates the citric-acid cycle, sparing fatty acids for triglyceride and ketone body production. These results reveal a new pathway of potential clinical significance that contributes to plasma lipid metabolism.


Asunto(s)
Proteínas Portadoras/genética , Clonación Molecular , Hiperlipidemia Familiar Combinada/genética , Animales , Animales Congénicos , Dióxido de Carbono/metabolismo , Proteínas Portadoras/metabolismo , Cromosomas Artificiales Bacterianos/genética , Cromosomas Humanos Par 1/genética , Ciclo del Ácido Cítrico/genética , Codón/genética , Codón sin Sentido , Mapeo Contig , Cósmidos/genética , Cricetinae , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Exones/genética , Ácidos Grasos/metabolismo , Haplotipos/genética , Humanos , Células Híbridas , Hiperlipidemia Familiar Combinada/metabolismo , Cuerpos Cetónicos/biosíntesis , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Oxidación-Reducción , Tiorredoxinas/antagonistas & inhibidores , Triglicéridos/sangre
8.
J Lipid Res ; 53(6): 1126-33, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22454476

RESUMEN

The liver X receptor (LXR) signaling pathway is an important modulator of atherosclerosis, but the relative importance of the two LXRs in atheroprotection is incompletely understood. We show here that LXRα, the dominant LXR isotype expressed in liver, plays a particularly important role in whole-body sterol homeostasis. In the context of the ApoE(-/-) background, deletion of LXRα, but not LXRß, led to prominent increases in atherosclerosis and peripheral cholesterol accumulation. However, combined loss of LXRα and LXRß on the ApoE(-/-) background led to an even more severe cholesterol accumulation phenotype compared to LXRα(-/-)ApoE(-/-) mice, indicating that LXRß does contribute to reverse cholesterol transport (RCT) but that this contribution is quantitatively less important than that of LXRα. Unexpectedly, macrophages did not appear to underlie the differential phenotype of LXRα(-/-)ApoE(-/-) and LXRß(-/-)ApoE(-/-) mice, as in vitro assays revealed no difference in the efficiency of cholesterol efflux from isolated macrophages. By contrast, in vivo assays of RCT using exogenously labeled macrophages revealed a marked defect in fecal sterol efflux in LXRα(-/-)ApoE(-/-) mice. Mechanistically, this defect was linked to a specific requirement for LXRα(-/-) in the expression of hepatic LXR target genes involved in sterol transport and metabolism. These studies reveal a previously unrecognized requirement for hepatic LXRα for optimal reverse cholesterol transport in mice.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Colesterol/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Animales , Transporte Biológico , Línea Celular , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Hígado/metabolismo , Receptores X del Hígado , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo
9.
Physiol Genomics ; 44(17): 843-52, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22805347

RESUMEN

Epidemiological studies show that high HDL-cholesterol (HDLc) decreases the risk of cardiovascular disease. To map genes controlling lipid metabolism, particularly HDLc levels, we screened the plasma lipids of 36 AcB/BcA RC mouse strains subjected to either a normal or a high-fat/cholesterol diet. Strains BcA68 and AcB65 showed deviant HDLc plasma levels compared with the parental A/J and C57BL/6J strains; they were thus selected to generate informative F2 crosses. Linkage analyses in the AcB65 strain identified a locus on chromosome 4 (Hdlq78) responsible for high post-high fat diet HDLc levels. This locus has been previously associated at genome-wide significance to two regions in the human genome. A second linkage analysis in strain BcA68 identified linkage in the vicinity of a gene cluster known to control HDLc levels. Sequence analysis of these candidates identified a de novo, loss-of-function mutation in the ApoA1 gene of BcA68 that prematurely truncates the ApoA1 protein. The possibility of dissecting the specific effects of this new ApoA1 deficiency in the context of isogenic controls makes the BcA68 mouse a valuable new tool.


Asunto(s)
Apolipoproteína A-I/genética , HDL-Colesterol/sangre , HDL-Colesterol/genética , Dieta Alta en Grasa , Ratones Congénicos/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Cruzamientos Genéticos , Sitios Genéticos/genética , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Mutación/genética , Análisis de Secuencia de ADN , Especificidad de la Especie
10.
Physiol Genomics ; 44(1): 1-13, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22010005

RESUMEN

Inbred strains of mice are strikingly different in susceptibility to obesity-driven diabetes. For instance, deficiency in leptin receptor (db/db) leads to hyperphagia and obesity in both C57BL/6 and DBA/2 mice, but only on the DBA/2 background do the mice develop beta-cell loss leading to severe diabetes, while C57BL/6 mice are relatively resistant. To further investigate the genetic factors predisposing to diabetes, we have studied leptin receptor-deficient offspring of an F2 cross between C57BL/6J (db/+) males and DBA/2J females. The results show that the genetics of diabetes susceptibility are enormously complex and a number of quantitative trait loci (QTL) contributing to diabetes-related traits were identified, notably on chromosomes 4, 6, 7, 9, 10, 11, 12, and 19. The Chr. 4 locus is likely due to a disruption of the Zfp69 gene in C57BL/6J mice. To identify candidate genes and to model coexpression networks, we performed global expression array analysis in livers of the F2 mice. Expression QTL (eQTL) were identified and used to prioritize candidate genes at clinical trait QTL. In several cases, clusters of eQTLs colocalized with clinical trait QTLs, suggesting a common genetic basis. We constructed coexpression networks for both 5 and 12 wk old mice and identified several modules significantly associated with clinical traits. One module in 12 wk old mice was associated with several measures of hepatic fat content as well as with other lipid- and diabetes-related traits. These results add to the understanding of the complex genetic interactions contributing to obesity-induced diabetes.


Asunto(s)
Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/genética , Predisposición Genética a la Enfermedad , Obesidad/complicaciones , Animales , Cruzamientos Genéticos , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Técnicas Genéticas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Obesos , Análisis por Micromatrices , Obesidad/genética , Polimorfismo de Nucleótido Simple , Biología de Sistemas/métodos
11.
Hum Mol Genet ; 19(4): 597-608, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19995791

RESUMEN

Upstream transcription factor 1 (USF1) has been associated with familial combined hyperlipidemia, the metabolic syndrome, and related conditions, but the mechanisms involved are unknown. In this study, we report validation of Usf1 as a causal gene of cholesterol homeostasis, insulin sensitivity and body composition in mouse models using several complementary approaches and identify associated pathways and gene expression network modules. Over-expression of human USF1 in both transgenic mice and mice with transient liver-specific over-expression influenced metabolic trait phenotypes, including obesity, total cholesterol level, LDL/VLDL cholesterol and glucose/insulin ratio. Additional analyses of trait and hepatic gene expression data from an F2 population derived from C57BL/6J and C3H/HeJ strains in which there is a naturally occurring variation in Usf1 expression supported a causal role for Usf1 for relevant metabolic traits. Gene network and pathway analyses of the liver gene expression signatures in the F2 population and the hepatic over-expression model suggested the involvement of Usf1 in immune responses and metabolism, including an Igfbp2-centered module. In all three mouse model settings, notable sex specificity was observed, consistent with human studies showing differences in association with USF1 gene polymorphisms between sexes.


Asunto(s)
Hiperlipidemia Familiar Combinada/metabolismo , Lípidos/sangre , Factores Estimuladores hacia 5'/metabolismo , Animales , Colesterol/sangre , Modelos Animales de Enfermedad , Femenino , Humanos , Hiperlipidemia Familiar Combinada/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores Estimuladores hacia 5'/genética
12.
Arterioscler Thromb Vasc Biol ; 31(1): 58-66, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20947826

RESUMEN

OBJECTIVE: To test the hypothesis that NF-E2-related factor 2 (Nrf2) expression plays an antiatherogenic role by its vascular antioxidant and anti-inflammatory properties. METHODS AND RESULTS: Nrf2 is an important transcription factor that regulates the expression of phase 2 detoxifying enzymes and antioxidant genes. Its expression in vascular cells appears to be an important factor in the protection against vascular oxidative stress and inflammation. We developed Nrf2 heterozygous (HET) and homozygous knockout (KO) mice on an apolipoprotein (apo) E-null background by sequential breeding, resulting in Nrf2(-/-), apoE(-/-) (KO), Nrf2(-/+), apoE(-/-) (HET) and Nrf2(+/+), and apoE(-/-) wild-type littermates. KO mice exhibited decreased levels of antioxidant genes with evidence of increased reactive oxygen species generation compared with wild-type controls. Surprisingly, KO males exhibited 47% and 53% reductions in the degree of aortic atherosclerosis compared with HET or wild-type littermates, respectively. Decreased atherosclerosis in KO mice correlated with lower plasma total cholesterol in a sex-dependent manner. KO mice also had a decreased hepatic cholesterol content and a lower expression of lipogenic genes, suggesting that hepatic lipogenesis could be reduced. In addition, KO mice exhibited atherosclerotic plaques characterized by a lesser macrophage component and decreased foam cell formation in an in vitro lipid-loading assay. This was associated with a lower rate of cholesterol influx, mediated in part by decreased expression of the scavenger receptor CD36. CONCLUSIONS: Nrf2 expression unexpectedly promotes atherosclerotic lesion formation in a sex-dependent manner, most likely by a combination of systemic metabolic and local vascular effects.


Asunto(s)
Antioxidantes/metabolismo , Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Colesterol/metabolismo , Lipoproteínas/sangre , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Transporte Biológico , Antígenos CD36/metabolismo , Modelos Animales de Enfermedad , Femenino , Células Espumosas/metabolismo , Regulación de la Expresión Génica , Lipogénesis/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Factor 2 Relacionado con NF-E2/deficiencia , Factor 2 Relacionado con NF-E2/genética , Especies Reactivas de Oxígeno/metabolismo , Factores Sexuales
13.
Arterioscler Thromb Vasc Biol ; 30(1): 20-3, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19815815

RESUMEN

OBJECTIVE: The risk of atherosclerosis in the setting of chylomicronemia has been a topic of debate. In this study, we examined susceptibility to atherosclerosis in Gpihbp1-deficient mice (Gpihbp1(-/-)), which manifest severe chylomicronemia as a result of defective lipolysis. METHODS AND RESULTS: Gpihbp1(-/-) mice on a chow diet have plasma triglyceride and cholesterol levels of 2812+/-209 and 319+/-27 mg/dL, respectively. Even though nearly all of the lipids were contained in large lipoproteins (50 to 135 nm), the mice developed progressive aortic atherosclerosis. In other experiments, we found that both Gpihbp1-deficient "apo-B48-only" mice and Gpihbp1-deficient "apo-B100-only" mice manifest severe chylomicronemia. Thus, GPIHBP1 is required for the processing of both apo-B48- and apo-B100-containing lipoproteins. CONCLUSIONS: Chylomicronemia causes atherosclerosis in mice. Also, we found that GPIHBP1 is required for the lipolytic processing of both apo-B48- and apo-B100-containing lipoproteins.


Asunto(s)
Apolipoproteína B-100/metabolismo , Apolipoproteína B-48/metabolismo , Aterosclerosis/metabolismo , Quilomicrones/metabolismo , Receptores de Lipoproteína/metabolismo , Alimentación Animal , Animales , Enfermedades de la Aorta/epidemiología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Aterosclerosis/epidemiología , Aterosclerosis/genética , Ácidos Grasos/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Lipólisis/fisiología , Masculino , Ratones , Ratones Mutantes , Receptores de Lipoproteína/genética , Factores de Riesgo , Triglicéridos/sangre
14.
Endocrinology ; 149(5): 2208-18, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18239070

RESUMEN

Subclinical inflammation is a recently discovered phenomenon in type 2 diabetes. Elevated cytokines impair beta-cell function and survival. A recent clinical trial shows that blocking IL-1beta signaling by IL-1 receptor antagonist (IL-1Ra) improves beta-cell secretory function in patients with type 2 diabetes. In the present study, we provide further mechanisms of the protective role of IL-1Ra on the beta-cell. IL-1Ra prevented diabetes in vivo in C57BL/6J mice fed a high-fat/high-sucrose diet (HFD) for 12 wk; it improved glucose tolerance and insulin secretion. High-fat diet treatment increased serum levels of free fatty acids and of the adipokines resistin and leptin, which were reduced by IL-1Ra treatment. In addition, IL-1Ra counteracted adiponectin levels, which were decreased by high-fat feeding. Studies on isolated islets revealed that IL-1Ra specifically acted on the beta-cell. IL-1Ra protected islets from HFD treated animals from beta-cell apoptosis, induced beta-cell proliferation, and improved glucose-stimulated insulin secretion. Insulin mRNA was reduced in islets from mice fed a HFD but normalized in the IL-1Ra group. Our results show that IL-1Ra improves beta-cell survival and function, and support the potential role for IL-1Ra in the treatment of diabetes.


Asunto(s)
Dieta Aterogénica , Hiperglucemia/prevención & control , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Adipoquinas/sangre , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Apoptosis/efectos de los fármacos , Recuento de Células , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/prevención & control , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Ingestión de Alimentos/efectos de los fármacos , Intolerancia a la Glucosa/tratamiento farmacológico , Hiperglucemia/etiología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/fisiología , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Aumento de Peso/efectos de los fármacos
15.
Biochim Biophys Acta ; 1764(8): 1363-71, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16876491

RESUMEN

Using mass spectrometry, we have recently reported on molecular masses of the apolipoproteins associated with porcine and equine HDL. In addition to obtaining accurate masses for the various apolipoproteins, we also were able to detect mass variations due to post-translational modifications. In the present study, we have used these same approaches to characterize the apolipoproteins in two inbred mouse strains, C57BL/6 and BALB/c. Comparing our molecular mass data with calculated values for molecular weight, we were able to identify the correct sequences for several of the major apolipoproteins. Analyses were carried out on the apolipoproteins of ultracentrifugally isolated HDL. Prior to analyses by electrospray ionization mass spectrometry (ESI-MS), the apolipoproteins were separated either by size exclusion or reverse phase chromatography. The molecular masses of apoA-I, proapoA-I, apoA-II, proapoA-II, apoC-I and apoC-III were obtained. Comparing the values obtained for the two strains, differences in the molecular masses of apoA-I, apoA-II and apoC-III were observed. In this study, post-translationally modified apolipoproteins, involving loss of amino acids from both the N- and C-termini, oxidation of methionine residues and possible acylation, were noted following reverse-phase separation. Further analyses by tandem mass spectrometry (MSMS) done on the tryptic digests of apolipoproteins separated by reverse phase chromatography enabled us to confirm sequence differences between the two strains, to verify selected apoA-I sequences that had been entered into the GenBank and to identify which methionines in apoA-I, apoC-III and apoE had been converted to methionine sulfoxides.


Asunto(s)
Apolipoproteínas/química , Lipoproteínas HDL/química , Secuencia de Aminoácidos , Animales , Apolipoproteínas/genética , Apolipoproteínas/aislamiento & purificación , Lipoproteínas HDL/genética , Lipoproteínas HDL/aislamiento & purificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Peso Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/aislamiento & purificación , Procesamiento Proteico-Postraduccional , Especificidad de la Especie , Espectrometría de Masa por Ionización de Electrospray , Tripsina
16.
Circ Res ; 92(11): 1262-7, 2003 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-12738753

RESUMEN

Apolipoprotein A-II (apoA-II) is a major protein on high-density lipoprotein (HDL) particles, and in mice, its levels are associated with triglyceride and glucose metabolism. In particular, transgenic mice overexpressing apoA-II exhibit hypertriglyceridemia, increased body fat, and insulin resistance, whereas apoA-II-null mice have decreased triglycerides and increased insulin sensitivity. Given the phenotypic overlap between familial combined hyperlipidemia (FCH) and apoA-II transgenic mice, we investigated the relationship of apoA-II to this disorder. Despite having lower HDL-cholesterol (HDL-C), FCH subjects had higher apoA-II levels compared with unaffected relatives (P<0.00016). Triglyceride and HDL-C levels were significant predictors of apoA-II, demonstrating that apoA-II variation is associated with several FCH-related traits. After adjustment for multiple covariates, there was evidence for the heritability of apoA-II levels (h2=0.15; P<0.02) in this sample. A genome scan for apoA-II levels identified significant evidence (LOD=3.1) for linkage to a locus on chromosome 1q41, coincident with a suggestive linkage for triglycerides (LOD score=1.4). Thus, this locus may have pleiotropic effects on apoA-II and FCH traits. Our results demonstrate that apoA-II is biochemically and genetically associated with FCH and may serve as a useful marker for understanding the mechanism by which FCH develops.


Asunto(s)
Apolipoproteína A-II/sangre , Predisposición Genética a la Enfermedad , Hiperlipidemia Familiar Combinada/sangre , Hiperlipidemia Familiar Combinada/genética , Adulto , Ligamiento Genético , Humanos , Hiperlipidemia Familiar Combinada/diagnóstico , Lípidos/sangre , Persona de Mediana Edad
17.
Circulation ; 106(4): 484-90, 2002 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-12135950

RESUMEN

BACKGROUND: Serum paraoxonase (PON1), an enzyme carried on HDL, inhibits LDL oxidation, and in human population studies, low PON1 activity is associated with atherosclerosis. In addition, PON1 knockout mice are more susceptible to lipoprotein oxidation and atherosclerosis. To evaluate whether PON1 protects against atherosclerosis and lipid oxidation in a dose-dependent manner, we generated and studied human PON1 transgenic mice. METHODS AND RESULTS: Human PON1 transgenic mice were produced by using bacterial artificial chromosome genomic clones. The mice had 2- to 4-fold increased plasma PON1 levels, but plasma cholesterol levels were unchanged. Atherosclerotic lesions were significantly reduced in the transgenic mice when both dietary and apoE-null mouse models were used. HDL isolated from the transgenic mice also protected against LDL oxidation more effectively. CONCLUSIONS: Our results indicate that PON1 protects against atherosclerosis in a dose-dependent manner and suggest that it may be a potential target for developing therapeutic agents for the treatment of cardiovascular disease.


Asunto(s)
Antioxidantes , Arteriosclerosis/prevención & control , Esterasas/genética , Animales , Apolipoproteínas E/genética , Arteriosclerosis/enzimología , Arteriosclerosis/metabolismo , Arteriosclerosis/patología , Arildialquilfosfatasa , Citoprotección , Esterasas/fisiología , Humanos , Lipoproteínas LDL/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , ARN Mensajero/análisis
18.
J Clin Endocrinol Metab ; 90(8): 4816-23, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15928243

RESUMEN

CONTEXT: Haplotypes comprising six single nucleotide polymorphisms (SNPs) (intron 7 to intron 9) of the lipoprotein lipase (LPL) gene appear to influence risk for atherosclerosis and insulin resistance in Mexican-Americans. OBJECTIVE: Based on rodent studies, we hypothesized that these haplotypes are in linkage disequilibrium with functional variants in the 3' untranslated region of LPL, which is encoded by exon 10, and that these variants influence phenotype by altering LPL expression. DESIGN: We sequenced exon 10 in subjects with divergent insulin sensitivity and divergent haplotypes. We also sequenced the other common LPL haplotypes. Variants identified by sequencing were genotyped in a large, family-based population along with the six SNPs spanning intron 7 to intron 9. We tested the potential functional significance of variation in exon 10 by evaluating association of haplotypes with post-heparin plasma LPL activity. SETTING: The study took place within the general community, with the Mexican-American Coronary Artery Disease Project cohort. PARTICIPANTS: Participants included 847 subjects from 163 families. MAIN OUTCOME MEASURES: We determined LPL haplogenotype and post-heparin plasma LPL activity. RESULTS: Exon 10 sequencing identified 15 variants. Thirteen of these variants were genotyped in large-scale along with the six SNPs spanning intron 7 to intron 9. LPL haplotypes and their relative frequencies in Mexican-Americans were determined. The fourth most common haplotype based on 19 SNPs (haplotype 19-4) was associated with increased LPL activity as well as multiple phenotypes related to the metabolic syndrome. CONCLUSIONS: These results support the possibility that variation in the 3' untranslated region of LPL affects LPL expression and activity, consequently influencing risk of atherosclerosis and insulin resistance, and provides important tools for further dissection of LPL regulation.


Asunto(s)
Regiones no Traducidas 3'/genética , Arteriosclerosis/genética , Heparina/metabolismo , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Adulto , Arteriosclerosis/etnología , Exones , Femenino , Haplotipos , Humanos , Resistencia a la Insulina/genética , Intrones , Desequilibrio de Ligamiento , Lipoproteína Lipasa/sangre , Masculino , Americanos Mexicanos/genética , Fenotipo , Polimorfismo de Nucleótido Simple , Factores de Riesgo
19.
Arterioscler Thromb Vasc Biol ; 22(1): 121-6, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11788471

RESUMEN

C57BL/6J mice are susceptible to atherosclerosis when fed a diet consisting of fat, cholesterol, and taurocholate. The susceptibility to diet-induced atherosclerosis is linked to a reduction in plasma high density lipoprotein (HDL). Diet-induced reduction of plasma HDL shows a physiological and a genetic correlation with repression of cholesterol-7-alpha-hydroxylase, the liver-specific enzyme that regulates the conversion of cholesterol into bile acids. To examine the hypothesis that the repression of cholesterol-7-alpha-hydroxylase is responsible for initiating the metabolic alterations leading to the formation of atherosclerosis and gallstones, we determined whether constitutive transgenic expression of cholesterol-7-alpha-hydroxylase in C57BL/6J mice would confer resistance to these 2 common human diseases. When fed the atherogenic diet, nontransgenic littermates, but not cholesterol-7-alpha-hydroxylase transgenic mice, accumulated cholesterol and cholesterol esters in their livers and plasma. Although the atherogenic diet caused a marked decrease in plasma HDL cholesterol in nontransgenic mice, HDL levels in transgenic mice remained relatively unchanged. Moreover, the ability of cholesterol-7-alpha-hydroxylase transgenic mice to maintain cholesterol and lipoprotein homeostasis completely prevented the formation of atherosclerosis and gallstones. These data establish the integral role that cholesterol-7-alpha-hydroxylase has in maintaining hepatic cholesterol homeostasis and, thus, in the susceptibility to the formation of gallstones and atherosclerosis.


Asunto(s)
Arteriosclerosis/metabolismo , Colelitiasis/metabolismo , Colesterol 7-alfa-Hidroxilasa/biosíntesis , HDL-Colesterol/metabolismo , Colesterol/metabolismo , Hígado/metabolismo , Animales , Arteriosclerosis/etiología , Arteriosclerosis/prevención & control , Colelitiasis/etiología , Colelitiasis/prevención & control , Colesterol/sangre , Colesterol 7-alfa-Hidroxilasa/genética , Dieta Aterogénica , Susceptibilidad a Enfermedades , Represión Enzimática , Femenino , Vesícula Biliar/metabolismo , Homeostasis , Lipoproteínas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Caracteres Sexuales
20.
Arterioscler Thromb Vasc Biol ; 24(10): 1928-34, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15331434

RESUMEN

OBJECTIVE: We previously reported the mapping of a quantitative trait locus (QTL) on chromosome 15 contributing to hyperlipidemia in a cross between inbred strains MRL/MpJ (MRL) and BALB/cJ (BALB). Using marker-assisted breeding, we constructed a congenic strain in which chromosome 15 interval from MRL is placed on the genetic background of BALB. The congenic allowed us to confirm the QTL result and to further characterize the properties and location of the underlying gene. METHODS AND RESULTS: On chow and high-fat (atherogenic) diets, the congenic mice exhibited higher levels of plasma triglycerides and cholesterol than BALB mice. In response to the atherogenic diet, the congenic mice but not BALB mice exhibited a dramatic approximately 30-fold increase in atherogenic lesions accompanied by approximately 2-fold decrease in high-density lipoprotein cholesterol levels. With respect to atherosclerotic lesions and some lipid parameters, this chromosome 15 gene, designated Hyplip2, exhibited dominant inheritance. Expression array analyses suggested that Hyplip2 may influence inflammatory and bile acid synthesis pathways. Finally, we demonstrated the usefulness of subcongenic strains to narrow the locus (50 Mbp) with the goal of positionally cloning Hyplip2. CONCLUSIONS: Our data demonstrate that the Hyplip2 gene significantly contributes to combined hyperlipidemia and increased atherosclerosis in mice.


Asunto(s)
Arteriosclerosis/genética , Hiperlipidemias/genética , Animales , Animales Congénicos , Arteriosclerosis/metabolismo , Mapeo Cromosómico/métodos , Cruzamientos Genéticos , Dieta Aterogénica , Genes/fisiología , Hiperlipidemias/metabolismo , Lípidos/sangre , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos BALB C , Ratones Endogámicos MRL lpr , Sitios de Carácter Cuantitativo/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA