Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Intervalo de año de publicación
1.
Circulation ; 143(18): 1809-1823, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33626882

RESUMEN

BACKGROUND: Coronary artery disease (CAD) is a multifactorial condition with both genetic and exogenous causes. The contribution of tissue-specific functional networks to the development of atherosclerosis remains largely unclear. The aim of this study was to identify and characterize central regulators and networks leading to atherosclerosis. METHODS: Based on several hundred genes known to affect atherosclerosis risk in mouse (as demonstrated in knockout models) and human (as shown by genome-wide association studies), liver gene regulatory networks were modeled. The hierarchical order and regulatory directions of genes within the network were based on Bayesian prediction models, as well as experimental studies including chromatin immunoprecipitation DNA-sequencing, chromatin immunoprecipitation mass spectrometry, overexpression, small interfering RNA knockdown in mouse and human liver cells, and knockout mouse experiments. Bioinformatics and correlation analyses were used to clarify associations between central genes and CAD phenotypes in both human and mouse. RESULTS: The transcription factor MAFF (MAF basic leucine zipper transcription factor F) interacted as a key driver of a liver network with 3 human genes at CAD genome-wide association studies loci and 11 atherosclerotic murine genes. Most importantly, expression levels of the low-density lipoprotein receptor (LDLR) gene correlated with MAFF in 600 CAD patients undergoing bypass surgery (STARNET [Stockholm-Tartu Atherosclerosis Reverse Network Engineering Task]) and a hybrid mouse diversity panel involving 105 different inbred mouse strains. Molecular mechanisms of MAFF were tested in noninflammatory conditions and showed positive correlation between MAFF and LDLR in vitro and in vivo. Interestingly, after lipopolysaccharide stimulation (inflammatory conditions), an inverse correlation between MAFF and LDLR in vitro and in vivo was observed. Chromatin immunoprecipitation mass spectrometry revealed that the human CAD genome-wide association studies candidate BACH1 (BTB domain and CNC homolog 1) assists MAFF in the presence of lipopolysaccharide stimulation with respective heterodimers binding at the MAF recognition element of the LDLR promoter to transcriptionally downregulate LDLR expression. CONCLUSIONS: The transcription factor MAFF was identified as a novel central regulator of an atherosclerosis/CAD-relevant liver network. MAFF triggered context-specific expression of LDLR and other genes known to affect CAD risk. Our results suggest that MAFF is a missing link between inflammation, lipid and lipoprotein metabolism, and a possible treatment target.


Asunto(s)
Aterosclerosis/metabolismo , Colesterol/metabolismo , Proteínas de Unión al ADN/metabolismo , Inflamación/metabolismo , Factor de Transcripción MafF/metabolismo , Proteínas Nucleares/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Noqueados
2.
J Lipid Res ; 56(1): 22-37, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25378658

RESUMEN

We performed silencing and overexpression studies of flavin containing monooxygenase (FMO) 3 in hyperlipidemic mouse models to examine its effects on trimethylamine N-oxide (TMAO) levels and atherosclerosis. Knockdown of hepatic FMO3 in LDL receptor knockout mice using an antisense oligonucleotide resulted in decreased circulating TMAO levels and atherosclerosis. Surprisingly, we also observed significant decreases in hepatic lipids and in levels of plasma lipids, ketone bodies, glucose, and insulin. FMO3 overexpression in transgenic mice, on the other hand, increased hepatic and plasma lipids. Global gene expression analyses suggested that these effects of FMO3 on lipogenesis and gluconeogenesis may be mediated through the PPARα and Kruppel-like factor 15 pathways. In vivo and in vitro results were consistent with the concept that the effects were mediated directly by FMO3 rather than trimethylamine/TMAO; in particular, overexpression of FMO3 in the human hepatoma cell line, Hep3B, resulted in significantly increased glucose secretion and lipogenesis. Our results indicate a major role for FMO3 in modulating glucose and lipid homeostasis in vivo, and they suggest that pharmacologic inhibition of FMO3 to reduce TMAO levels would be confounded by metabolic interactions.


Asunto(s)
Aterosclerosis/enzimología , Glucosa/metabolismo , Metabolismo de los Lípidos , Oxigenasas/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Dieta Occidental , Heces/química , Femenino , Regulación Enzimológica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Glucosa/biosíntesis , Homeostasis , Humanos , Insulina/sangre , Mucosa Intestinal/metabolismo , Factores de Transcripción de Tipo Kruppel , Lipogénesis , Lipoproteínas/sangre , Hígado/metabolismo , Metilaminas/metabolismo , Ratones , Oxigenasas/deficiencia , Oxigenasas/genética , PPAR alfa/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Factores de Transcripción/metabolismo
3.
J Lipid Res ; 56(1): 38-50, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25327529

RESUMEN

Exposure of endothelial cells (ECs) to agents such as oxidized glycerophospholipids (oxGPs) and cytokines, known to accumulate in atherosclerotic lesions, perturbs the expression of hundreds of genes in ECs involved in inflammatory and other biological processes. We hypothesized that microRNAs (miRNAs) are involved in regulating the inflammatory response in human aortic endothelial cells (HAECs) in response to oxGPs and interleukin 1ß (IL-1ß). Using next-generation sequencing and RT-quantitative PCR, we characterized the profile of expressed miRNAs in HAECs pre- and postexposure to oxGPs. Using this data, we identified miR-21-3p and miR-27a-5p to be induced 3- to 4-fold in response to oxGP and IL-1ß treatment compared with control treatment. Transient overexpression of miR-21-3p and miR-27a-5p resulted in the downregulation of 1,253 genes with 922 genes overlapping between the two miRNAs. Gene Ontology functional enrichment analysis predicted that the two miRNAs were involved in the regulation of nuclear factor κB (NF-κB) signaling. Overexpression of these two miRNAs leads to changes in p65 nuclear translocation. Using 3' untranslated region luciferase assay, we identified 20 genes within the NF-κB signaling cascade as putative targets of miRs-21-3p and -27a-5p, implicating these two miRNAs as modulators of NF-κB signaling in ECs.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Interleucina-1beta/farmacología , MicroARNs/genética , Fosfatidilcolinas/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Regiones no Traducidas 3'/genética , Transporte Activo de Núcleo Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Oxidación-Reducción , Fosfatidilcolinas/química , Análisis de Secuencia de ARN , Factor de Transcripción ReIA/genética , Factor de Necrosis Tumoral alfa/farmacología
4.
Mol Syst Biol ; 10: 730, 2014 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-24860088

RESUMEN

We profiled and analyzed 283 metabolites representing eight major classes of molecules including Lipids, Carbohydrates, Amino Acids, Peptides, Xenobiotics, Vitamins and Cofactors, Energy Metabolism, and Nucleotides in mouse liver of 104 inbred and recombinant inbred strains. We find that metabolites exhibit a wide range of variation, as has been previously observed with metabolites in blood serum. Using genome-wide association analysis, we mapped 40% of the quantified metabolites to at least one locus in the genome and for 75% of the loci mapped we identified at least one candidate gene by local expression QTL analysis of the transcripts. Moreover, we validated 2 of 3 of the significant loci examined by adenoviral overexpression of the genes in mice. In our GWAS results, we find that at significant loci the peak markers explained on average between 20 and 40% of variation in the metabolites. Moreover, 39% of loci found to be regulating liver metabolites in mice were also found in human GWAS results for serum metabolites, providing support for similarity in genetic regulation of metabolites between mice and human. We also integrated the metabolomic data with transcriptomic and clinical phenotypic data to evaluate the extent of co-variation across various biological scales.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Hígado/metabolismo , Metabolómica , Sitios de Carácter Cuantitativo/genética , Animales , Proteínas Sanguíneas/genética , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Polimorfismo de Nucleótido Simple
5.
Cell ; 151(3): 658-70, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-23101632

RESUMEN

Many common diseases have an important inflammatory component mediated in part by macrophages. Here we used a systems genetics strategy to examine the role of common genetic variation in macrophage responses to inflammatory stimuli. We examined genome-wide transcript levels in macrophages from 92 strains of the Hybrid Mouse Diversity Panel. We exposed macrophages to control media, bacterial lipopolysaccharide (LPS), or oxidized phospholipids. We performed association mapping under each condition and identified several thousand expression quantitative trait loci (eQTL), gene-by-environment interactions, and eQTL "hot spots" that specifically control LPS responses. We used siRNA knockdown of candidate genes to validate an eQTL hot spot in chromosome 8 and identified the gene 2310061C15Rik as a regulator of inflammatory responses in macrophages. We have created a public database where the data presented here can be used as a resource for understanding many common inflammatory traits that are modeled in the mouse and for the dissection of regulatory relationships between genes.


Asunto(s)
Interacción Gen-Ambiente , Inflamación/inmunología , Macrófagos/inmunología , Ratones/genética , Sitios de Carácter Cuantitativo , Animales , Células Cultivadas , Técnicas de Silenciamiento del Gen , Lipopolisacáridos/inmunología , Macrófagos/metabolismo , Masculino , Ratones/inmunología , Ratones Endogámicos , Especificidad de la Especie , Organismos Libres de Patógenos Específicos , Biología de Sistemas/métodos
6.
Proc Natl Acad Sci U S A ; 104(11): 4530-5, 2007 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-17360558

RESUMEN

The genetic factors contributing to the complex disorder of myocardial calcification are largely unknown. Using a mouse model, we fine-mapped the major locus (Dyscalc1) contributing to the dystrophic cardiac calcification (DCC) to an 840-kb interval containing 38 genes. We then identified the causal gene by using an approach integrating genetic segregation and expression array analyses to identify, on a global scale, cis-acting DNA variations that perturb gene expression. By studying two intercrosses, in which the DCC trait segregates, a single candidate gene (encoding the ATP-binding cassette transporter ABCC6) was identified. Transgenic complementation confirmed Abcc6 as the underlying causal gene for Dyscalc1. We demonstrate that in the cross, the expression of Abcc6 is highly correlated with the local mineralization regulatory system and the BMP2-Wnt signaling pathway known to be involved in the systemic regulation of calcification, suggesting potential pathways for the action of Abcc6 in DCC. Our results demonstrate the power of the integrative genomics in discovering causal genes and pathways underlying complex traits.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/fisiología , Calcinosis/genética , Regulación de la Expresión Génica , Genómica , Cardiopatías/genética , Miocardio/metabolismo , Animales , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Osteopontina/biosíntesis , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Wnt/metabolismo
7.
Nature ; 422(6929): 297-302, 2003 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-12646919

RESUMEN

Treating messenger RNA transcript abundances as quantitative traits and mapping gene expression quantitative trait loci for these traits has been pursued in gene-specific ways. Transcript abundances often serve as a surrogate for classical quantitative traits in that the levels of expression are significantly correlated with the classical traits across members of a segregating population. The correlation structure between transcript abundances and classical traits has been used to identify susceptibility loci for complex diseases such as diabetes and allergic asthma. One study recently completed the first comprehensive dissection of transcriptional regulation in budding yeast, giving a detailed glimpse of a genome-wide survey of the genetics of gene expression. Unlike classical quantitative traits, which often represent gross clinical measurements that may be far removed from the biological processes giving rise to them, the genetic linkages associated with transcript abundance affords a closer look at cellular biochemical processes. Here we describe comprehensive genetic screens of mouse, plant and human transcriptomes by considering gene expression values as quantitative traits. We identify a gene expression pattern strongly associated with obesity in a murine cross, and observe two distinct obesity subtypes. Furthermore, we find that these obesity subtypes are under the control of different loci.


Asunto(s)
Ratones/genética , Obesidad/genética , Sitios de Carácter Cuantitativo/genética , Transcripción Genética/genética , Zea mays/genética , Animales , Mapeo Cromosómico , Cromosomas Humanos Par 20/genética , Cromosomas de los Mamíferos/genética , Cruzamientos Genéticos , Femenino , Genómica/métodos , Humanos , Escala de Lod , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Análisis de Secuencia por Matrices de Oligonucleótidos , Linaje , Polimorfismo Genético/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA