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










Base de dados
Intervalo de ano de publicação
1.
Elife ; 4: e05607, 2015 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-26067236

RESUMO

To identify genetic and environmental factors contributing to the pathogenesis of non-alcoholic fatty liver disease, we examined liver steatosis and related clinical and molecular traits in more than 100 unique inbred mouse strains, which were fed a diet rich in fat and carbohydrates. A >30-fold variation in hepatic TG accumulation was observed among the strains. Genome-wide association studies revealed three loci associated with hepatic TG accumulation. Utilizing transcriptomic data from the liver and adipose tissue, we identified several high-confidence candidate genes for hepatic steatosis, including Gde1, a glycerophosphodiester phosphodiesterase not previously implicated in triglyceride metabolism. We confirmed the role of Gde1 by in vivo hepatic over-expression and shRNA knockdown studies. We hypothesize that Gde1 expression increases TG production by contributing to the production of glycerol-3-phosphate. Our multi-level data, including transcript levels, metabolite levels, and gut microbiota composition, provide a framework for understanding genetic and environmental interactions underlying hepatic steatosis.


Assuntos
Estudo de Associação Genômica Ampla , Hepatopatia Gordurosa não Alcoólica/veterinária , Diester Fosfórico Hidrolases/genética , Doenças dos Roedores/genética , Tecido Adiposo/patologia , Animais , Expressão Gênica , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Fígado/patologia , Camundongos Endogâmicos , Hepatopatia Gordurosa não Alcoólica/genética , Triglicerídeos/metabolismo
2.
J Cell Sci ; 128(12): 2236-48, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25956888

RESUMO

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.


Assuntos
Apoptose , Comunicação Autócrina , Autofagia , Biomarcadores/metabolismo , Endotélio Vascular/patologia , Fatores de Transcrição Forkhead/metabolismo , Mitocôndrias/patologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Western Blotting , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Endotélio Vascular/metabolismo , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Perfilação da Expressão Gênica , Humanos , Hipóxia/fisiopatologia , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Fosforilação , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
3.
Cell Metab ; 21(2): 334-347, 2015 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-25651185

RESUMO

Insulin resistance (IR) is a complex trait with multiple genetic and environmental components. Confounded by large differences between the sexes, environment, and disease pathology, the genetic basis of IR has been difficult to dissect. Here we examine IR and related traits in a diverse population of more than 100 unique male and female inbred mouse strains after feeding a diet rich in fat and refined carbohydrates. Our results show dramatic variation in IR among strains of mice and widespread differences between sexes that are dependent on genotype. We uncover more than 15 genome-wide significant loci and validate a gene, Agpat5, associated with IR. We also integrate plasma metabolite levels and global gene expression from liver and adipose tissue to identify metabolite quantitative trait loci (mQTL) and expression QTL (eQTL), respectively. Our results provide a resource for analysis of interactions between diet, sex, and genetic background in IR.


Assuntos
Resistência à Insulina/genética , 1-Acilglicerol-3-Fosfato O-Aciltransferase/genética , 1-Acilglicerol-3-Fosfato O-Aciltransferase/metabolismo , Animais , Dieta Hiperlipídica , Carboidratos da Dieta , Feminino , Variação Genética/genética , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA
4.
FASEB J ; 29(4): 1185-97, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25477283

RESUMO

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.


Assuntos
Arildialquilfosfatase/deficiência , Aterosclerose/enzimologia , Cálculos Biliares/enzimologia , Obesidade/enzimologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Arildialquilfosfatase/genética , Arildialquilfosfatase/metabolismo , Aterosclerose/etiologia , Aterosclerose/genética , Ácidos e Sais Biliares/metabolismo , Quimiocina CCL2/metabolismo , Colesterol na Dieta/administração & dosagem , Ácido Cólico/administração & dosagem , Dieta/efeitos adversos , Modelos Animais de Doenças , Feminino , Cálculos Biliares/etiologia , Cálculos Biliares/genética , Expressão Gênica , Predisposição Genética para Doença , Mediadores da Inflamação/metabolismo , Interleucina-6/metabolismo , Intestino Delgado/metabolismo , Rim/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Hepáticas/metabolismo , Obesidade/etiologia , Obesidade/genética
5.
PLoS One ; 9(1): e85341, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24489659

RESUMO

The CCAAT/enhancer binding proteins (C/EBPs) are transcription factors involved in hematopoietic cell development and induction of several inflammatory mediators. C/EBPε is expressed only in myeloid cells including monocytes/macrophages. Atherosclerosis is an inflammatory disorder of the vascular wall and circulating immune cells such as monocytes/macrophages. Mice deficient in the low density lipoprotein (LDL) receptor (Ldlr-/-) fed on a high cholesterol diet (HCD) show elevated blood cholesterol levels and are widely used as models to study human atherosclerosis. In this study, we generated Ldlr and Cebpe double-knockout (llee) mice and compared their atherogenic phenotypes to Ldlr single deficient (llEE) mice after HCD. Macrophages from llee mice have reduced lipid uptake by foam cells and impaired phagokinetic motility in vitro compared to macrophages from llEE mice. Also, compared to llEE mice, llee mice have alterations of lipid metabolism, and reduced atheroma and obesity, particularly the males. Peritoneal macrophages of llee male mice have reduced mRNA expression of FABP4, a fatty acid binding protein implicated in atherosclerosis. Overall, our study suggests that the myeloid specific factor C/EBPε is involved in systemic lipid metabolism and that silencing of C/EBPε could decrease the development of atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/deficiência , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Receptores de LDL/metabolismo , Animais , Aterosclerose/genética , Proteínas Estimuladoras de Ligação a CCAAT/genética , Movimento Celular/genética , Movimento Celular/fisiologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de LDL/deficiência , Receptores de LDL/genética
6.
Cell Metab ; 17(1): 141-52, 2013 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-23312289

RESUMO

Obesity is a highly heritable disease driven by complex interactions between genetic and environmental factors. Human genome-wide association studies (GWAS) have identified a number of loci contributing to obesity; however, a major limitation of these studies is the inability to assess environmental interactions common to obesity. Using a systems genetics approach, we measured obesity traits, global gene expression, and gut microbiota composition in response to a high-fat/high-sucrose (HF/HS) diet of more than 100 inbred strains of mice. Here we show that HF/HS feeding promotes robust, strain-specific changes in obesity that are not accounted for by food intake and provide evidence for a genetically determined set point for obesity. GWAS analysis identified 11 genome-wide significant loci associated with obesity traits, several of which overlap with loci identified in human studies. We also show strong relationships between genotype and gut microbiota plasticity during HF/HS feeding and identify gut microbial phylotypes associated with obesity.


Assuntos
Dieta Hiperlipídica , Mucosa Intestinal/microbiologia , Metagenoma , Obesidade/genética , Animais , Composição Corporal , Carboidratos da Dieta , Genoma , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Obesidade/patologia , Locos de Características Quantitativas
7.
Circulation ; 126(15): 1896-906, 2012 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-22952318

RESUMO

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.


Assuntos
Aterosclerose/genética , Doença da Artéria Coronariana/genética , Animais , Inibidor p16 de Quinase Dependente de Ciclina/genética , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética
8.
Physiol Genomics ; 44(17): 843-52, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22805347

RESUMO

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.


Assuntos
Apolipoproteína A-I/genética , HDL-Colesterol/sangue , HDL-Colesterol/genética , Dieta Hiperlipídica , Camundongos Congênicos/genética , Animais , Sequência de Bases , Mapeamento Cromossômico , Cruzamentos Genéticos , Loci Gênicos/genética , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutação/genética , Análise de Sequência de DNA , Especificidade da Espécie
9.
J Lipid Res ; 53(6): 1126-33, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22454476

RESUMO

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.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Colesterol/metabolismo , Receptores Nucleares Órfãos/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Fígado/metabolismo , Receptores X do Fígado , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo
10.
Biochim Biophys Acta ; 1821(3): 435-47, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21807117

RESUMO

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).


Assuntos
Histona Desacetilases/genética , Lipoproteínas HDL/metabolismo , Proteínas/genética , Tecido Adiposo/metabolismo , Análise de Variância , Animais , Colesterol/metabolismo , Cruzamentos Genéticos , Dieta Hiperlipídica , Feminino , Redes Reguladoras de Genes , Hibridização Genética , Fígado/metabolismo , Escore Lod , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Transcriptoma , Proteína 2 do Domínio Central WAP de Quatro Dissulfetos
11.
Physiol Genomics ; 44(1): 1-13, 2012 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-22010005

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental/etiologia , Diabetes Mellitus Experimental/genética , Predisposição Genética para Doença , Obesidade/complicações , Animais , Cruzamentos Genéticos , Diabetes Mellitus Experimental/patologia , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Predisposição Genética para Doença/genética , Técnicas Genéticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Obesos , Análise em Microsséries , Obesidade/genética , Polimorfismo de Nucleotídeo Único , Biologia de Sistemas/métodos
12.
Arterioscler Thromb Vasc Biol ; 31(1): 58-66, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20947826

RESUMO

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.


Assuntos
Antioxidantes/metabolismo , Doenças da Aorta/metabolismo , Aterosclerose/metabolismo , Colesterol/metabolismo , Lipoproteínas/sangue , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Doenças da Aorta/etiologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Doenças da Aorta/prevenção & controle , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/etiologia , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Transporte Biológico , Antígenos CD36/metabolismo , Modelos Animais de Doenças , Feminino , Células Espumosas/metabolismo , Regulação da Expressão Gênica , Lipogênese/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/deficiência , Fator 2 Relacionado a NF-E2/genética , Espécies Reativas de Oxigênio/metabolismo , Fatores Sexuais
13.
Arthritis Res Ther ; 12(3): R93, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20482780

RESUMO

INTRODUCTION: The purpose of this study was to evaluate the effects of L-4F, an apolipoprotein A-1 mimetic peptide, alone or with pravastatin, in apoE-/-Fas-/-C57BL/6 mice that spontaneously develop immunoglobulin G (IgG) autoantibodies, glomerulonephritis, osteopenia, and atherosclerotic lesions on a normal chow diet. METHODS: Female mice, starting at eight to nine weeks of age, were treated for 27 weeks with 1) pravastatin, 2) L-4F, 3) L-4F plus pravastatin, or 4) vehicle control, followed by disease phenotype assessment. RESULTS: In preliminary studies, dysfunctional, proinflammatory high-density lipoproteins (piHDL) were decreased six hours after a single L-4F, but not scrambled L-4F, injection in eight- to nine-week old mice. After 35 weeks, L-4F-treated mice, in the absence/presence of pravastatin, had significantly smaller lymph nodes and glomerular tufts (PL, LP<0.05), lower serum levels of IgG antibodies to double stranded DNA (dsDNA) (PL<0.05) and oxidized phospholipids (oxPLs) (PL, LP<0.005), and elevated total and vertebral bone mineral density (PL, LP<0.01) compared to vehicle controls. Although all treatment groups presented larger aortic root lesions compared to vehicle controls, enlarged atheromas in combination treatment mice had significantly less infiltrated CD68+ macrophages (PLP<0.01), significantly increased mean alpha-actin stained area (PLP<0.05), and significantly lower levels of circulating markers for atherosclerosis progression, CCL19 (PL, LP<0.0005) and VCAM-1 (PL<0.0002). CONCLUSIONS: L-4F treatment, alone or with pravastatin, significantly reduced IgG anti-dsDNA and IgG anti-oxPLs, proteinuria, glomerulonephritis, and osteopenia in a murine lupus model of accelerated atherosclerosis. Despite enlarged aortic lesions, increased smooth muscle content, decreased macrophage infiltration, and decreased pro-atherogenic chemokines in L-4F plus pravastatin treated mice suggest protective mechanisms not only on lupus-like disease, but also on potential plaque remodeling in a murine model of systemic lupus erythematosus (SLE) and accelerated atherosclerosis.


Assuntos
Apolipoproteína A-I/uso terapêutico , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/metabolismo , Peptídeos/uso terapêutico , Pravastatina/uso terapêutico , Animais , Apolipoproteína A-I/efeitos adversos , Apolipoproteína A-I/farmacologia , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Densidade Óssea/efeitos dos fármacos , Quimiocinas/sangue , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Imunoglobulina G/sangue , Lipídeos/sangue , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peptídeos/efeitos adversos , Peptídeos/farmacologia , Placa Aterosclerótica/induzido quimicamente , Placa Aterosclerótica/patologia , Pravastatina/efeitos adversos , Pravastatina/farmacologia , Receptor fas/genética , Receptor fas/metabolismo
14.
Diabetes ; 59(7): 1616-25, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20393148

RESUMO

OBJECTIVE: To identify metabolic derangements contributing to diabetes susceptibility in the leptin receptor-deficient obese C57BLKS/J-db/db (BKS-db) mouse strain. RESEARCH DESIGN AND METHODS: Young BKS-db mice were used to identify metabolic pathways contributing to the development of diabetes. Using the diabetes-resistant B6-db strain as a comparison, in vivo and in vitro approaches were applied to identify metabolic and molecular differences between the two strains. RESULTS: Despite higher plasma insulin levels, BKS-db mice exhibit lower lipogenic gene expression, rate of lipogenesis, hepatic triglyceride and glycogen content, and impaired insulin suppression of gluconeogenic genes. Hepatic insulin receptor substrate (IRS)-1 and IRS-2 expression and insulin-stimulated Akt-phosphorylation are decreased in BKS-db primary hepatocytes. Hyperinsulinemic-euglycemic clamp studies indicate that in contrast to hepatic insulin resistance, skeletal muscle is more insulin sensitive in BKS-db than in B6-db mice. We also demonstrate that elevated plasma triglyceride levels in BKS-db mice are associated with reduced triglyceride clearance due to lower lipase activities. CONCLUSIONS: Our study demonstrates the presence of metabolic derangements in BKS-db before the onset of beta-cell failure and identifies early hepatic insulin resistance as a component of the BKS-db phenotype. We propose that defects in hepatic insulin signaling contribute to the development of diabetes in the BKS-db mouse strain.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Hepatócitos/metabolismo , Resistência à Insulina/genética , Insulina/metabolismo , Fígado/metabolismo , Análise de Variância , Animais , Diabetes Mellitus Tipo 2/genética , Ácidos Graxos/metabolismo , Expressão Gênica , Gluconeogênese/genética , Hepatócitos/citologia , Insulina/genética , Lipase/metabolismo , Lipogênese/genética , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/genética , Obesidade/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética
15.
Genome Res ; 20(2): 281-90, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20054062

RESUMO

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.


Assuntos
Mapeamento Cromossômico/métodos , Estudo de Associação Genômica Ampla/métodos , Locos de Características Quantitativas/genética , Algoritmos , Animais , Ligação Genética , Lipoproteínas HDL/genética , Masculino , Camundongos , Camundongos Endogâmicos , Fenótipo
16.
Arterioscler Thromb Vasc Biol ; 30(1): 20-3, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19815815

RESUMO

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.


Assuntos
Apolipoproteína B-100/metabolismo , Apolipoproteína B-48/metabolismo , Aterosclerose/metabolismo , Quilomícrons/metabolismo , Receptores de Lipoproteínas/metabolismo , Ração Animal , Animais , Doenças da Aorta/epidemiologia , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Aterosclerose/epidemiologia , Aterosclerose/genética , Ácidos Graxos/metabolismo , Feminino , Predisposição Genética para Doença , Lipólise/fisiologia , Masculino , Camundongos , Camundongos Mutantes , Receptores de Lipoproteínas/genética , Fatores de Risco , Triglicerídeos/sangue
17.
Hum Mol Genet ; 19(4): 597-608, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19995791

RESUMO

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.


Assuntos
Hiperlipidemia Familiar Combinada/metabolismo , Lipídeos/sangue , Fatores Estimuladores Upstream/metabolismo , Animais , Colesterol/sangue , Modelos Animais de Doenças , Feminino , Humanos , Hiperlipidemia Familiar Combinada/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fatores Estimuladores Upstream/genética
18.
Diabetes ; 58(8): 1739-48, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19509018

RESUMO

OBJECTIVE: Insulin-resistant states, such as obesity and type 2 diabetes, contribute substantially to accelerated atherogenesis. Null mutations of myostatin (Mstn) are associated with increased muscle mass and decreased fat mass. In this study, we determined whether Mstn disruption could prevent the development of insulin resistance, proatherogenic dyslipidemia, and atherogenesis. RESEARCH DESIGN AND METHODS: C57BL/6 Ldlr(-/-) mice were cross-bred with C57BL/6 Mstn(-/-) mice for >10 generations to generate Mstn(-/-)/Ldlr(-/-) double-knockout mice. The effects of high-fat/high-cholesterol diet on body composition, plasma lipids, systemic and tissue-specific insulin sensitivity, hepatic steatosis, as well as aortic atheromatous lesion were characterized in Mstn(-/-)/Ldlr(-/-) mice in comparison with control Mstn(+/+)/Ldlr(-/-) mice. RESULTS: Compared with Mstn(+/+)/Ldlr(-/-) controls, Mstn(-/-)/ Ldlr(-/-) mice were resistant to diet-induced obesity, and had greatly improved insulin sensitivity, as indicated by 42% higher glucose infusion rate and 90% greater muscle [(3)H]-2-deoxyglucose uptake during hyperinsulinemic-euglycemic clamp. Mstn(-/-)/Ldlr(-/-) mice were protected against diet-induced hepatic steatosis and had 56% higher rate of hepatic fatty acid beta-oxidation than controls. Mstn(-/-)/Ldlr(-/-) mice also had 36% lower VLDL secretion rate and were protected against diet-induced dyslipidemia, as indicated by 30-60% lower VLDL and LDL cholesterol, free fatty acids, and triglycerides. Magnetic resonance angiography and en face analyses demonstrated 41% reduction in aortic atheromatous lesions in Ldlr(-/-) mice with Mstn deletion. CONCLUSIONS: Inactivation of Mstn protects against the development of insulin resistance, proatherogenic dyslipidemia, and aortic atherogenesis in Ldlr(-/-) mice. Myostatin may be a useful target for drug development for prevention and treatment of obesity and its associated type 2 diabetes and atherosclerosis.


Assuntos
Aterosclerose/genética , Aterosclerose/prevenção & controle , Dislipidemias/genética , Dislipidemias/prevenção & controle , Miostatina/genética , Receptores de LDL/genética , Tecido Adiposo/anatomia & histologia , Animais , Aterosclerose/etiologia , Glicemia/metabolismo , Composição Corporal , Dislipidemias/complicações , Deleção de Genes , Glucagon/sangue , Insulina/sangue , Lipídeos/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/anatomia & histologia
19.
Nat Genet ; 41(4): 415-23, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19270708

RESUMO

A principal task in dissecting the genetics of complex traits is to identify causal genes for disease phenotypes. We previously developed a method to infer causal relationships among genes through the integration of DNA variation, gene transcription and phenotypic information. Here we have validated our method through the characterization of transgenic and knockout mouse models of genes predicted to be causal for abdominal obesity. Perturbation of eight out of the nine genes, with Gas7, Me1 and Gpx3 being newly confirmed, resulted in significant changes in obesity-related traits. Liver expression signatures revealed alterations in common metabolic pathways and networks contributing to abdominal obesity and overlapped with a macrophage-enriched metabolic network module that is highly associated with metabolic traits in mice and humans. Integration of gene expression in the design and analysis of traditional F(2) intercross studies allows high-confidence prediction of causal genes and identification of pathways and networks involved.


Assuntos
Proteínas de Transporte/genética , Glutationa Peroxidase/genética , Glicoproteínas/genética , Proteínas do Tecido Nervoso/genética , Obesidade/genética , Abdome/anatomia & histologia , Tecido Adiposo/anatomia & histologia , Animais , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Variação Genética , Humanos , Fígado/fisiologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Músculo Esquelético/anatomia & histologia , Fenótipo , Reprodutibilidade dos Testes , Transcrição Gênica , Proteínas de Transporte Vesicular
20.
Endocrinology ; 149(5): 2208-18, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18239070

RESUMO

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.


Assuntos
Dieta Aterogênica , Hiperglicemia/prevenção & controle , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Adipocinas/sangue , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Apoptose/efeitos dos fármacos , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/prevenção & controle , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos , Ingestão de Alimentos/efeitos dos fármacos , Intolerância à Glucose/tratamento farmacológico , Hiperglicemia/etiologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Aumento de Peso/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...