Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Proc Natl Acad Sci U S A ; 108(45): 18560-5, 2011 Nov 08.
Article in English | MEDLINE | ID: mdl-22042857

ABSTRACT

The circadian clock generates daily rhythms in mammalian liver processes, such as glucose and lipid homeostasis, xenobiotic metabolism, and regeneration. The mechanisms governing these rhythms are not well understood, particularly the distinct contributions of the cell-autonomous clock and central pacemaker to rhythmic liver physiology. Through microarray expression profiling in Met murine hepatocytes (MMH)-D3, we identified over 1,000 transcripts that exhibit circadian oscillations, demonstrating that the cell-autonomous clock can drive many rhythms, and that MMH-D3 is a valid circadian model system. The genes represented by these circadian transcripts displayed both cophasic and antiphasic organization within a protein-protein interaction network, suggesting the existence of competition for binding sites or partners by genes of disparate transcriptional phases. Multiple pathways displayed enrichment in MMH-D3 circadian transcripts, including the polyamine synthesis module of the glutathione metabolic pathway. The polyamine synthesis module, which is highly associated with cell proliferation and whose products are required for initiation of liver regeneration, includes enzymes whose transcripts exhibit circadian oscillations, such as ornithine decarboxylase and spermidine synthase. Metabolic profiling revealed that the enzymatic product of spermidine synthase, spermidine, cycles as well. Thus, the cell-autonomous hepatocyte clock can drive a significant amount of transcriptional rhythms and orchestrate physiologically relevant modules such as polyamine synthesis.


Subject(s)
Biogenic Polyamines/biosynthesis , Circadian Rhythm , Hepatocytes/cytology , Transcription, Genetic , Animals , Mice
2.
Arterioscler Thromb Vasc Biol ; 29(3): 348-55, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19122174

ABSTRACT

OBJECTIVE: We sought to determine the genetic factors contributing to atherosclerotic plaque size and cellular composition in the innominate artery, a murine model of advanced atherosclerosis. METHODS AND RESULTS: We examined genetic contributions to innominate atherosclerotic plaque size and cellular composition in an intercross between C57BL/6J.Apoe(-/-), a strain susceptible to aortic lesions, and C3H/HeJ.Apoe(-/-), a strain resistant to aortic lesions. Surprisingly, total innominate lesion size was similar in the two strains. Genetic analyses identified one novel locus on Chromosome 2 for innominate artery lesion size, a significant locus for fibrous cap thickness on Chromosome 15, and several suggestive loci for cellular composition, all distinct from loci influencing aortic lesions. The Chromosome 2 locus contains a candidate, CD44. We show that CD44 is expressed in the innominate artery and differs strikingly in expression between the parental strains. CONCLUSION: Multiple aspects of innominate lesion composition are genetically determined, but in a manner largely independent of the genetic contributions to aortic lesions.


Subject(s)
Atherosclerosis/genetics , Brachiocephalic Trunk/pathology , Chromosomes, Mammalian , Hyperlipidemias/genetics , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/pathology , Disease Models, Animal , Female , Gene Expression Profiling/methods , Genetic Linkage , Genetic Predisposition to Disease , Hyaluronan Receptors/genetics , Hyperlipidemias/complications , Hyperlipidemias/pathology , Lipids/blood , Lod Score , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Quantitative Trait Loci , Sex Factors , Species Specificity
3.
Circ Res ; 101(3): e11-30, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17641228

ABSTRACT

We report a combined genetic and genomic analysis of atherosclerosis in a cross between the strains C3H/HeJ and C57BL/6J on a hyperlipidemic apolipoprotein E-null background. We incorporated sex and sex-by-genotype interactions into our model selection procedure to identify 10 quantitative trait loci for lesion size, revealing a level of complexity greater than previously thought. Of the known risk factors for atherosclerosis, plasma triglyceride levels and plasma glucose to insulin ratios were particularly strongly, but negatively, associated with lesion size. We performed expression array analysis for 23,574 transcripts of the livers and adipose tissues of all 334 F2 mice and identified more than 10,000 expression quantitative trait loci that either mapped to the gene encoding the transcript, implying cis regulation, or to a separate locus, implying trans-regulation. The gene expression data allowed us to identify candidate genes that mapped to the atherosclerosis quantitative trait loci and for which the expression was regulated in cis. Genes highly correlated with lesions were enriched in certain known pathways involved in lesion development, including cholesterol metabolism, mitochondrial oxidative phosphorylation, and inflammation. Thus, global gene expression in peripheral tissues can reflect the systemic perturbations that contribute to atherosclerosis.


Subject(s)
Adipose Tissue/metabolism , Atherosclerosis/genetics , Gene Expression Regulation , Liver/metabolism , Metabolic Networks and Pathways/genetics , Quantitative Trait Loci , Animals , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cholesterol/metabolism , Crosses, Genetic , Diet, Atherogenic , Female , Gene Expression Profiling , Genetic Predisposition to Disease , Genotype , Hyperlipidemias/complications , Hyperlipidemias/genetics , Inflammation/genetics , Inflammation/metabolism , Lod Score , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Models, Genetic , Oxidative Phosphorylation , RNA, Messenger/biosynthesis , Risk Factors , Sex Characteristics
4.
Circ Res ; 100(8): 1200-7, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17379834

ABSTRACT

Paraoxonase 3 (PON3) is a member of the PON family, which includes PON1, PON2, and PON3. Recently, PON3 was shown to prevent the oxidation of low-density lipoprotein in vitro. To test the role of PON3 in atherosclerosis and related traits, 2 independent lines of human PON3 transgenic (Tg) mice on the C57BL/6J (B6) background were constructed. Human PON3 mRNA was detected in various tissues, including liver, lung, kidney, brain, adipose, and aorta, of both lines of Tg mice. The human PON3 mRNA levels in the livers of PON3 Tg mice were 4- to 7-fold higher as compared with the endogenous mouse Pon3 mRNA levels. Human PON3 protein and activity were detected in the livers of Tg mice as well. No significant differences in plasma total, high-density lipoprotein, and very-low-density lipoprotein/low-density lipoprotein cholesterol and triglyceride and glucose levels were observed between the PON3 Tg and non-Tg mice. Interestingly, atherosclerotic lesion areas were significantly smaller in both lines of male PON3 Tg mice as compared with the male non-Tg littermates on B6 background fed an atherogenic diet. When bred onto the low-density lipoprotein receptor knockout mouse background, the male PON3 Tg mice also exhibited decreased atherosclerotic lesion areas and decreased expression of monocyte chemoattractant protein-1 in the aorta as compared with the male non-Tg littermates. In addition, decreased adiposity and lower circulating leptin levels were observed in both lines of male PON3 Tg mice as compared with the male non-Tg mice. In an F2 cross, adipose Pon3 mRNA levels inversely correlated with adiposity and related traits. Our study demonstrates that elevated PON3 expression significantly decreases atherosclerotic lesion formation and adiposity in male mice. PON3 may play an important role in protection against obesity and atherosclerosis.


Subject(s)
Arteriosclerosis/enzymology , Arteriosclerosis/genetics , Esterases/genetics , Obesity/enzymology , Obesity/genetics , Animals , Aryldialkylphosphatase , Esterases/biosynthesis , Esterases/physiology , Female , Gene Expression Regulation, Enzymologic/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
5.
Arterioscler Thromb Vasc Biol ; 27(12): 2671-6, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17916774

ABSTRACT

OBJECTIVE: C3H/HeJ (C3H) mice are extremely resistant to atherosclerosis. To identify the genetic factors involved in lesion initiation, we studied a cross between C3H and the susceptible strain C57BL/6J (B6) on a hyperlipidemic (apolipoprotein E-null) background. METHODS AND RESULTS: Whereas a previous cross in mice fed a Western diet for 16 weeks revealed a very complex inheritance pattern with many significant lesion QTLs, the present cross, on a chow diet, revealed a single major locus on chromosome 9 (lod=5.0, Ath29*), and a suggestive locus on chromosome 4 (lod=2.6, Ath8). QTLs for plasma HDL, total cholesterol, and triglyceride levels were found on chromosome 1 over the ApoA2 gene. Neither of the lesion QTLs were associated with differences in plasma lipid levels or other systemic risk factors, consistent with the concept that genetic factors affecting cellular functions of the vessel wall are important determinants of atherosclerosis susceptibility. We generated a congenic strain for Ath29 and confirmed its contribution to lesion development. Toll-like receptor 4 (Tlr4), the lipopolysaccharide (LPS) receptor, is located in the Ath8 region and is known to be defective in C3H/HeJ mice. We constructed a congenic strain carrying a normal Tlr4 gene on the C3H Apoe-null background and found that the defective Tlr4 does not contribute significantly to lesion resistance during early lesion development. CONCLUSIONS: We identified one major QTL on chromosome 9, Ath29, for early lesion development in the BXH ApoE(-/-) cross fed on a chow diet and confirmed its contribution in congenic mice. We have also determined that Tlr4 on the C3H ApoE(-/-) background does not contribute to early lesion development. *Ath29 is referred to as Ath22 in Su et al 2006.


Subject(s)
Apolipoproteins E/metabolism , Atherosclerosis/genetics , Chromosome Mapping , Mice, Inbred C3H/genetics , Quantitative Trait Loci , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Crosses, Genetic , Dietary Fats/administration & dosage , Disease Models, Animal , Female , Genetic Predisposition to Disease , Lipids/blood , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Mutation , Risk Factors , Toll-Like Receptor 4/genetics
6.
BMC Genomics ; 7: 235, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16978413

ABSTRACT

BACKGROUND: Changes in gene expression are known to be responsible for phenotypic variation and susceptibility to diseases. Identification and annotation of the genomic sequence variants that cause gene expression changes is therefore likely to lead to a better understanding of the cause of disease at the molecular level. In this study we investigate the pattern of single nucleotide polymorphisms (SNPs) in genes for which the mRNA levels show cis-genetic linkage (gene expression quantitative trait loci mapping in cis, or cis-eQTLs) in segregating mouse populations. Such genes are expected to have polymorphisms near their physical location (cis-variations) that affect their mRNA levels by altering one or more of the cis-regulatory elements. This led us to characterize the SNPs in promoter (5 Kb upstream) and non-coding gene regions (introns and 5 Kb downstream) (cis-SNPs) and the effects they may have on putative transcription factor binding sites. RESULTS: We demonstrate that the cis-eQTL genes (CEGs) have a significantly higher frequency of cis-SNPs compared to non-CEGs (when both sets are taken from the non-IBD regions, i.e. regions not identical by descent). Most CEGs having cis-SNPs do not contain these SNPs in the phylogenetically conserved regions. In those CEGs that contain cis-SNPs in the phylogenetically conserved regions, enrichment of cis-SNPs occurs both within and outside of the conserved sequences. A higher fraction of CEGs are also seen to harbor cis-SNP that affect predicted transcription factor binding sites, a likely consequence of the higher cis-SNPs density in these genes. CONCLUSION: This present study provides the first genome-wide investigation of the putative cis-regulatory variations in a large set of genes whose levels of expression give rise to cis-linkage in segregating mammalian populations. Our results provide insights into the challenges that exist in identifying polymorphisms regulating gene expression using bioinformatic sequence analysis approaches. The data provided herein should benefit future investigations in this area.


Subject(s)
Gene Expression Regulation/genetics , Genetic Variation/genetics , Polymorphism, Single Nucleotide/genetics , Adipose Tissue/metabolism , Animals , Binding Sites/genetics , Brain/metabolism , Chromosomes, Mammalian/genetics , Female , Gene Expression Profiling , Genetic Linkage/genetics , Genetics, Population , Genotype , Liver/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Muscle, Skeletal/metabolism , Promoter Regions, Genetic/genetics , Quantitative Trait Loci/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/metabolism
8.
Genetics ; 185(3): 1059-68, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20439777

ABSTRACT

By treating the transcript abundance as a quantitative trait, gene expression can be mapped to local or distant genomic regions relative to the gene encoding the transcript. Local expression quantitative trait loci (eQTL) generally act in cis (that is, control the expression of only the contiguous structural gene), whereas distal eQTL act in trans. Distal eQTL are more difficult to identify with certainty due to the fact that significant thresholds are very high since all regions of the genome must be tested, and confounding factors such as batch effects can produce false positives. Here, we compare findings from two large genetic crosses between mouse strains C3H/HeJ and C57BL/6J to evaluate the reliability of distal eQTL detection, including "hotspots" influencing the expression of multiple genes in trans. We found that >63% of local eQTL and >18% of distal eQTL were replicable at a threshold of LOD > 4.3 between crosses and 76% of local and >24% of distal eQTL at a threshold of LOD > 6. Additionally, at LOD > 4.3 four tissues studied (adipose, brain, liver, and muscle) exhibited >50% preservation of local eQTL and >17% preservation of distal eQTL. We observed replicated distal eQTL hotspots between the crosses on chromosomes 9 and 17. Finally, >69% of local eQTL and >10% of distal eQTL were preserved in most tissues between sexes. We conclude that most local eQTL are highly replicable between mouse crosses, tissues, and sex as compared to distal eQTL, which exhibited modest replicability.


Subject(s)
DNA Replication , Gene Expression Profiling , Genome , Quantitative Trait Loci , Adipose Tissue/metabolism , Animals , Brain/metabolism , Chromosome Mapping , Female , Liver/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Muscles/metabolism , Organ Specificity , Phenotype , Sex Factors , Tissue Distribution
9.
Circ Cardiovasc Genet ; 2(6): 573-82, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20031637

ABSTRACT

BACKGROUND: Disruption of the elastic lamina, as an early indicator of aneurysm formation, and vascular calcification frequently occur together in atherosclerotic lesions of humans. METHODS AND RESULTS: We now report evidence of shared genetic basis for disruption of the elastic lamina (medial disruption) and medial calcification in an F(2) mouse intercross between C57BL/6J and C3H/HeJ on a hyperlipidemic apolipoprotein E (ApoE(-/-)) null BACKGROUND: gene, known to mediate myocardial calcification. Using transgenic complementation, we show that Abcc6 also contributes to aortic medial calcification. CONCLUSIONS: Our data indicate that calcification, though possibly contributory, does not always lead to medial disruption and that in addition to aneurysm formation, medial disruption may be the precursor to calcification.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/pathology , Calcinosis , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Aorta/pathology , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multidrug Resistance-Associated Proteins , Quantitative Trait Loci
10.
Endocrinology ; 150(3): 1235-49, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18974276

ABSTRACT

We previously used high-density expression arrays to interrogate a genetic cross between strains C3H/HeJ and C57BL/6J and observed thousands of differences in gene expression between sexes. We now report analyses of the molecular basis of these sex differences and of the effects of sex on gene expression networks. We analyzed liver gene expression of hormone-treated gonadectomized mice as well as XX male and XY female mice. Differences in gene expression resulted in large part from acute effects of gonadal hormones acting in adulthood, and the effects of sex chromosomes, apart from hormones, were modest. We also determined whether there are sex differences in the organization of gene expression networks in adipose, liver, skeletal muscle, and brain tissue. Although coexpression networks of highly correlated genes were largely conserved between sexes, some exhibited striking sex dependence. We observed strong body fat and lipid correlations with sex-specific modules in adipose and liver as well as a sexually dimorphic network enriched for genes affected by gonadal hormones. Finally, our analyses identified chromosomal loci regulating sexually dimorphic networks. This study indicates that gonadal hormones play a strong role in sex differences in gene expression. In addition, it results in the identification of sex-specific gene coexpression networks related to genetic and metabolic traits.


Subject(s)
Gene Regulatory Networks , Gonadal Hormones/physiology , Sex Characteristics , Animals , Apolipoproteins E/genetics , Crosses, Genetic , Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Female , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/physiology , Gonadal Hormones/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout
11.
Nat Genet ; 41(4): 415-23, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19270708

ABSTRACT

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.


Subject(s)
Carrier Proteins/genetics , Glutathione Peroxidase/genetics , Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Obesity/genetics , Abdomen/anatomy & histology , Adipose Tissue/anatomy & histology , Animals , Disease Models, Animal , Female , Gene Expression Profiling , Genetic Variation , Humans , Liver/physiology , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/anatomy & histology , Phenotype , Reproducibility of Results , Transcription, Genetic , Vesicular Transport Proteins
12.
PLoS One ; 3(10): e3583, 2008.
Article in English | MEDLINE | ID: mdl-18974833

ABSTRACT

A wealth of genetic associations for cardiovascular and metabolic phenotypes in humans has been accumulating over the last decade, in particular a large number of loci derived from recent genome wide association studies (GWAS). True complex disease-associated loci often exert modest effects, so their delineation currently requires integration of diverse phenotypic data from large studies to ensure robust meta-analyses. We have designed a gene-centric 50 K single nucleotide polymorphism (SNP) array to assess potentially relevant loci across a range of cardiovascular, metabolic and inflammatory syndromes. The array utilizes a "cosmopolitan" tagging approach to capture the genetic diversity across approximately 2,000 loci in populations represented in the HapMap and SeattleSNPs projects. The array content is informed by GWAS of vascular and inflammatory disease, expression quantitative trait loci implicated in atherosclerosis, pathway based approaches and comprehensive literature searching. The custom flexibility of the array platform facilitated interrogation of loci at differing stringencies, according to a gene prioritization strategy that allows saturation of high priority loci with a greater density of markers than the existing GWAS tools, particularly in African HapMap samples. We also demonstrate that the IBC array can be used to complement GWAS, increasing coverage in high priority CVD-related loci across all major HapMap populations. DNA from over 200,000 extensively phenotyped individuals will be genotyped with this array with a significant portion of the generated data being released into the academic domain facilitating in silico replication attempts, analyses of rare variants and cross-cohort meta-analyses in diverse populations. These datasets will also facilitate more robust secondary analyses, such as explorations with alternative genetic models, epistasis and gene-environment interactions.


Subject(s)
Cardiovascular Diseases/genetics , Genome-Wide Association Study/methods , Oligonucleotide Array Sequence Analysis/methods , Polymorphism, Single Nucleotide , Cardiovascular Diseases/ethnology , Concept Formation , Gene Frequency , Genome-Wide Association Study/instrumentation , Genotype , Humans , Population Groups/genetics , Quality Control , Research Design
13.
Proc Natl Acad Sci U S A ; 104(11): 4530-5, 2007 Mar 13.
Article in English | MEDLINE | ID: mdl-17360558

ABSTRACT

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.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/physiology , Calcinosis/genetics , Gene Expression Regulation , Genomics , Heart Diseases/genetics , Myocardium/metabolism , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Multidrug Resistance-Associated Proteins , Osteopontin/biosynthesis , Signal Transduction , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL