Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 165(1): 111-124, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-26972052

RESUMO

Normal platelet function is critical to blood hemostasis and maintenance of a closed circulatory system. Heightened platelet reactivity, however, is associated with cardiometabolic diseases and enhanced potential for thrombotic events. We now show gut microbes, through generation of trimethylamine N-oxide (TMAO), directly contribute to platelet hyperreactivity and enhanced thrombosis potential. Plasma TMAO levels in subjects (n > 4,000) independently predicted incident (3 years) thrombosis (heart attack, stroke) risk. Direct exposure of platelets to TMAO enhanced sub-maximal stimulus-dependent platelet activation from multiple agonists through augmented Ca(2+) release from intracellular stores. Animal model studies employing dietary choline or TMAO, germ-free mice, and microbial transplantation collectively confirm a role for gut microbiota and TMAO in modulating platelet hyperresponsiveness and thrombosis potential and identify microbial taxa associated with plasma TMAO and thrombosis potential. Collectively, the present results reveal a previously unrecognized mechanistic link between specific dietary nutrients, gut microbes, platelet function, and thrombosis risk.


Assuntos
Plaquetas/metabolismo , Microbioma Gastrointestinal , Metilaminas/metabolismo , Trombose/metabolismo , Animais , Cálcio/metabolismo , Lesões das Artérias Carótidas/patologia , Ceco/microbiologia , Cloretos , Colina/metabolismo , Dieta , Feminino , Compostos Férricos , Vida Livre de Germes , Humanos , Metilaminas/sangue , Camundongos , Camundongos Endogâmicos C57BL , Trombose/patologia
2.
Mol Syst Biol ; 17(1): e9684, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33417276

RESUMO

To elucidate the contributions of specific lipid species to metabolic traits, we integrated global hepatic lipid data with other omics measures and genetic data from a cohort of about 100 diverse inbred strains of mice fed a high-fat/high-sucrose diet for 8 weeks. Association mapping, correlation, structure analyses, and network modeling revealed pathways and genes underlying these interactions. In particular, our studies lead to the identification of Ifi203 and Map2k6 as regulators of hepatic phosphatidylcholine homeostasis and triacylglycerol accumulation, respectively. Our analyses highlight mechanisms for how genetic variation in hepatic lipidome can be linked to physiological and molecular phenotypes, such as microbiota composition.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Fígado Gorduroso/genética , Glucose/efeitos adversos , Resistência à Insulina/genética , MAP Quinase Quinase 6/genética , Proteínas Nucleares/genética , Animais , Modelos Animais de Doenças , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Variação Genética , Lipidômica , Masculino , Camundongos , Fosfatidilcolinas/metabolismo , Triglicerídeos/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 41(1): 220-233, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33086870

RESUMO

OBJECTIVE: Previous studies have shown that deficiency of M-CSF (macrophage colony-stimulating factor; or CSF1 [colony stimulating factor 1]) dramatically reduces atherosclerosis in hyperlipidemic mice. We characterize the underlying mechanism and investigate the relevant sources of CSF1 in lesions. Approach and Results: We quantitatively assessed the effects of CSF1 deficiency on macrophage proliferation and apoptosis in atherosclerotic lesions. Staining of aortic lesions with markers of proliferation, Ki-67 and bromodeoxyuridine, revealed around 40% reduction in CSF1 heterozygous (Csf1+/-) as compared with WT (wild type; Csf1+/+) mice. Similarly, staining with a marker of apoptosis, activated caspase-3, revealed a 3-fold increase in apoptotic cells in Csf1+/- mice. Next, we determined the cellular sources of CSF1 contributing to lesion development. Cell-specific deletions of Csf1 in smooth muscle cells using SM22α-Cre (smooth muscle protein 22-alpha-Cre) reduced lesions by about 40%, and in endothelial cells, deletions with Cdh5-Cre (VE-cadherin-Cre) reduced lesions by about 30%. Macrophage-specific deletion with LysM-Cre (lysozyme M-Cre), on the other hand, did not significantly reduce lesions size. Transplantation of Csf1 null (Csf1-/-) mice bone marrow into Csf1+/+ mice reduced lesions by about 35%, suggesting that CSF1 from hematopoietic cells other than macrophages contributes to atherosclerosis. None of the cell-specific knockouts affected circulating CSF1 levels, and only the smooth muscle cell deletions had any effect on the percentage monocytes in the circulation. Also, Csf1+/- mice did not exhibit significant differences in Ly6Chigh/Ly6Clow monocytes as compared with Csf1+/+. CONCLUSIONS: CSF1 contributes to both macrophage proliferation and survival in lesions. Local CSF1 production by smooth muscle cell and endothelial cell rather than circulating CSF1 is the primary driver of macrophage expansion in atherosclerosis.


Assuntos
Apoptose , Aterosclerose/metabolismo , Proliferação de Células , Células Endoteliais/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Aorta/metabolismo , Aorta/patologia , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Caderinas/genética , Caderinas/metabolismo , Modelos Animais de Doenças , Células Endoteliais/patologia , Feminino , Fator Estimulador de Colônias de Macrófagos/deficiência , Fator Estimulador de Colônias de Macrófagos/genética , Macrófagos/patologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transdução de Sinais
4.
Eur Heart J ; 42(9): 919-933, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33532862

RESUMO

AIMS: While most patients with myocardial infarction (MI) have underlying coronary atherosclerosis, not all patients with coronary artery disease (CAD) develop MI. We sought to address the hypothesis that some of the genetic factors which establish atherosclerosis may be distinct from those that predispose to vulnerable plaques and thrombus formation. METHODS AND RESULTS: We carried out a genome-wide association study for MI in the UK Biobank (n∼472 000), followed by a meta-analysis with summary statistics from the CARDIoGRAMplusC4D Consortium (n∼167 000). Multiple independent replication analyses and functional approaches were used to prioritize loci and evaluate positional candidate genes. Eight novel regions were identified for MI at the genome wide significance level, of which effect sizes at six loci were more robust for MI than for CAD without the presence of MI. Confirmatory evidence for association of a locus on chromosome 1p21.3 harbouring choline-like transporter 3 (SLC44A3) with MI in the context of CAD, but not with coronary atherosclerosis itself, was obtained in Biobank Japan (n∼165 000) and 16 independent angiography-based cohorts (n∼27 000). Follow-up analyses did not reveal association of the SLC44A3 locus with CAD risk factors, biomarkers of coagulation, other thrombotic diseases, or plasma levels of a broad array of metabolites, including choline, trimethylamine N-oxide, and betaine. However, aortic expression of SLC44A3 was increased in carriers of the MI risk allele at chromosome 1p21.3, increased in ischaemic (vs. non-diseased) coronary arteries, up-regulated in human aortic endothelial cells treated with interleukin-1ß (vs. vehicle), and associated with smooth muscle cell migration in vitro. CONCLUSIONS: A large-scale analysis comprising ∼831 000 subjects revealed novel genetic determinants of MI and implicated SLC44A3 in the pathophysiology of vulnerable plaques.


Assuntos
Doença da Artéria Coronariana , Infarto do Miocárdio , Doença da Artéria Coronariana/genética , Células Endoteliais , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Humanos , Japão , Infarto do Miocárdio/genética , Polimorfismo de Nucleotídeo Único/genética , Fatores de Risco
5.
Arterioscler Thromb Vasc Biol ; 38(9): 2016-2027, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30026271

RESUMO

Objective- The objective of this study was to determine the basis of resistance to atherosclerosis of inbred mouse strain BALB/cJ. Approach and Results- BALB/cJ mice carry a naturally occurring null mutation of the gene encoding the transcription factor Zhx2, and genetic analyses suggested that this may confer resistance to atherosclerosis. On a hyperlipidemic low-density lipoprotein receptor null background, BALB/cJ mice carrying the mutant allele for Zhx2 exhibited up to a 10-fold reduction in lesion size as compared with an isogenic strain carrying the wild-type allele. Several lines of evidence, including bone marrow transplantation studies, indicate that this effect of Zhx2 is mediated, in part, by monocytes/macrophages although nonbone marrow-derived pathways are clearly involved as well. Both in culture and in atherosclerotic lesions, macrophages from Zhx2 null mice exhibited substantially increased apoptosis. Zhx2 null macrophages were also enriched for M2 markers. Effects of Zhx2 on proliferation and other bone marrow-derived cells, such as lymphocytes, were at most modest. Expression microarray analyses identified >1000 differentially expressed transcripts between Zhx2 wild-type and null macrophages. To identify the global targets of Zhx2, we performed ChIP-seq (chromatin immunoprecipitation sequencing) studies with the macrophage cell line RAW264.7. The ChIP-seq peaks overlapped significantly with gene expression and together suggested roles for transcriptional repression and apoptosis. Conclusions- A mutation of Zhx2 carried in BALB/cJ mice is responsible in large part for its relative resistance to atherosclerosis. Our results indicate that Zhx2 promotes macrophage survival and proinflammatory functions in atherosclerotic lesions, thereby contributing to lesion growth.


Assuntos
Apoptose , Aterosclerose/fisiopatologia , Proteínas de Homeodomínio/fisiologia , Macrófagos/fisiologia , Fatores de Transcrição/fisiologia , Dedos de Zinco/fisiologia , Animais , Proliferação de Células , Modelos Animais de Doenças , Expressão Gênica , Proteínas de Homeodomínio/genética , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fatores de Transcrição/genética , Dedos de Zinco/genética
6.
Curr Opin Lipidol ; 29(5): 404-410, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30156571

RESUMO

PURPOSE OF REVIEW: Sex differences are pervasive in metabolic and cardiovascular traits, yet they have often been ignored in human and animal model research. Sex differences can arise from reversible hormonal effects, from irreversible organizational (developmental) processes, and from gene expression differences from the X and Y chromosomes. We briefly review our current understanding of the impact of these factors in metabolic traits and disorders, with an emphasis on the recent literature. RECENT FINDINGS: Novel sex differences continue to be identified for metabolic and cardiovascular traits. For example, it is now clear that gut microbiota tend to differ between men and women, with potentially large implications for disease susceptibility. Also, tissue-specific gene regulation differs between men and women, contributing to differential metabolism. These new insights will open up personalized therapeutic avenues for cardiometabolic diseases. SUMMARY: Sex differences in body fat distribution, glucose homeostasis, insulin signaling, ectopic fat accumulation, and lipid metabolism during normal growth and in response to hormonal or nutritional imbalance are mediated partly through sex hormones and the sex chromosome complement. Most of these differences are mediated in a tissue-specific manner. Important future goals are to better understand the interactions between genetic variation and sex differences, and to bring an understanding of sex differences into clinical practice.


Assuntos
Doenças Cardiovasculares/fisiopatologia , Doenças Metabólicas/fisiopatologia , Caracteres Sexuais , Animais , Doenças Cardiovasculares/metabolismo , Humanos , Doenças Metabólicas/metabolismo
7.
J Lipid Res ; 59(7): 1164-1174, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29739864

RESUMO

Elevated hepatic ceramide levels have been implicated in both insulin resistance (IR) and hepatic steatosis. To understand the factors contributing to hepatic ceramide levels in mice of both sexes, we have quantitated ceramides in a reference population of mice, the Hybrid Mouse Diversity Panel that has been previously characterized for a variety of metabolic syndrome traits. We observed significant positive correlations between Cer(d18:1/16:0) and IR/hepatic steatosis, consistent with previous findings, although the relationship broke down between sexes, as females were less insulin resistant, but had higher Cer(d18:1/16:0) levels than males. The sex difference was due in part to testosterone-mediated repression of ceramide synthase 6. One ceramide species, Cer(d18:1/20:0), was present at higher levels in males and was associated with IR only in males. Clear evidence of gene-by-sex and gene-by-diet interactions was observed, including sex-specific genome-wide association study results. Thus, our studies show clear differences in how hepatic ceramides are regulated between the sexes, which again suggests that the physiological roles of certain hepatic ceramides differ between the sexes.


Assuntos
Ceramidas/metabolismo , Dieta , Resistência à Insulina/genética , Resistência à Insulina/fisiologia , Fígado/metabolismo , Caracteres Sexuais , Animais , Ceramidas/biossíntese , Feminino , Fígado/efeitos dos fármacos , Masculino , Camundongos , Testosterona/farmacologia
8.
Genome Res ; 25(10): 1558-69, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26260972

RESUMO

Genetics provides a potentially powerful approach to dissect host-gut microbiota interactions. Toward this end, we profiled gut microbiota using 16s rRNA gene sequencing in a panel of 110 diverse inbred strains of mice. This panel has previously been studied for a wide range of metabolic traits and can be used for high-resolution association mapping. Using a SNP-based approach with a linear mixed model, we estimated the heritability of microbiota composition. We conclude that, in a controlled environment, the genetic background accounts for a substantial fraction of abundance of most common microbiota. The mice were previously studied for response to a high-fat, high-sucrose diet, and we hypothesized that the dietary response was determined in part by gut microbiota composition. We tested this using a cross-fostering strategy in which a strain showing a modest response, SWR, was seeded with microbiota from a strain showing a strong response, A×B19. Consistent with a role of microbiota in dietary response, the cross-fostered SWR pups exhibited a significantly increased response in weight gain. To examine specific microbiota contributing to the response, we identified various genera whose abundance correlated with dietary response. Among these, we chose Akkermansia muciniphila, a common anaerobe previously associated with metabolic effects. When administered to strain A×B19 by gavage, the dietary response was significantly blunted for obesity, plasma lipids, and insulin resistance. In an effort to further understand host-microbiota interactions, we mapped loci controlling microbiota composition and prioritized candidate genes. Our publicly available data provide a resource for future studies.


Assuntos
Microbioma Gastrointestinal/genética , Animais , Dieta , Dieta Hiperlipídica , Meio Ambiente , Feminino , Estudo de Associação Genômica Ampla , Hereditariedade , Masculino , Camundongos , Camundongos Endogâmicos , Obesidade/microbiologia , RNA Ribossômico 16S , Sacarose/metabolismo
9.
J Lipid Res ; 58(1): 178-187, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27811231

RESUMO

The etiology of nonalcoholic fatty liver disease is complex and influenced by factors such as obesity, insulin resistance, hyperlipidemia, and sex. We now report a study on sex difference in hepatic steatosis in the context of genetic variation using a population of inbred strains of mice. While male mice generally exhibited higher concentration of hepatic TG levels on a high-fat high-sucrose diet, sex differences showed extensive interaction with genetic variation. Differences in percentage body fat were the best predictor of hepatic steatosis among the strains and explained about 30% of the variation in both sexes. The difference in percent gonadal fat and HDL explained 9.6% and 6.7% of the difference in hepatic TGs between the sexes, respectively. Genome-wide association mapping of hepatic TG revealed some striking differences in genetic control of hepatic steatosis between females and males. Gonadectomy increased the hepatic TG to body fat percentage ratio among male, but not female, mice. Our data suggest that the difference between the sexes in hepatic TG can be partly explained by differences in body fat distribution, plasma HDL, and genetic regulation. Future studies are required to understand the molecular interactions between sex, genetics, and the environment.


Assuntos
Fígado Gorduroso/genética , Lipoproteínas HDL/genética , Hepatopatia Gordurosa não Alcoólica/genética , Obesidade/genética , Triglicerídeos/genética , Animais , Dieta Hiperlipídica , Fígado Gorduroso/sangue , Fígado Gorduroso/patologia , Feminino , Estudo de Associação Genômica Ampla , Hormônios/genética , Hormônios/metabolismo , Hiperlipidemias/sangue , Hiperlipidemias/genética , Hiperlipidemias/patologia , Resistência à Insulina/genética , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Hepatopatia Gordurosa não Alcoólica/sangue , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade/sangue , Obesidade/patologia , Polimorfismo de Nucleotídeo Único/genética , Caracteres Sexuais
10.
J Lipid Res ; 57(6): 925-42, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27099397

RESUMO

The Hybrid Mouse Diversity Panel (HMDP) is a collection of approximately 100 well-characterized inbred strains of mice that can be used to analyze the genetic and environmental factors underlying complex traits. While not nearly as powerful for mapping genetic loci contributing to the traits as human genome-wide association studies, it has some important advantages. First, environmental factors can be controlled. Second, relevant tissues are accessible for global molecular phenotyping. Finally, because inbred strains are renewable, results from separate studies can be integrated. Thus far, the HMDP has been studied for traits relevant to obesity, diabetes, atherosclerosis, osteoporosis, heart failure, immune regulation, fatty liver disease, and host-gut microbiota interactions. High-throughput technologies have been used to examine the genomes, epigenomes, transcriptomes, proteomes, metabolomes, and microbiomes of the mice under various environmental conditions. All of the published data are available and can be readily used to formulate hypotheses about genes, pathways and interactions.


Assuntos
Doenças Cardiovasculares/genética , Modelos Animais de Doenças , Doenças Metabólicas/genética , Transcriptoma/genética , Animais , Aterosclerose/genética , Doenças Cardiovasculares/patologia , Estudo de Associação Genômica Ampla , Insuficiência Cardíaca/genética , Humanos , Hibridização Genética , Resistência à Insulina/genética , Doenças Metabólicas/patologia , Camundongos , Microbiota/genética , Obesidade/genética , Osteoporose/genética , Locos de Características Quantitativas/genética
11.
Circ Res ; 111(5): 516-20, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22811557

RESUMO

RATIONALE: Mutations of the orphan transporter ABCC6 (ATP-binding cassette, subfamily C, member 6) cause the connective tissue disorder pseudoxanthoma elasticum. ABCC6 was thought to be located on the plasma membrane of liver and kidney cells. OBJECTIVE: Mouse systems genetics and bioinformatics suggested that ABCC6 deficiency affects mitochondrial gene expression. We therefore tested whether ABCC6 associates with mitochondria. METHODS AND RESULTS: We found ABCC6 in crude mitochondrial fractions and subsequently pinpointed its localization to the purified mitochondria-associated membrane fraction. Cell-surface biotinylation in hepatocytes confirmed that ABCC6 is intracellular. Abcc6-knockout mice demonstrated mitochondrial abnormalities and decreased respiration reserve capacity. CONCLUSIONS: Our finding that ABCC6 localizes to the mitochondria-associated membrane has implications for its mechanism of action in normal and diseased states.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Calcinose/metabolismo , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Pseudoxantoma Elástico/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Biotinilação , Calcinose/genética , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/metabolismo , Fracionamento Celular , Respiração Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Genes Mitocondriais/fisiologia , Hepatócitos/citologia , Hepatócitos/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos , Pseudoxantoma Elástico/genética
12.
PLoS Genet ; 7(3): e1002023, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21483757

RESUMO

The idea that most morphological adaptations can be attributed to changes in the cis-regulation of gene expression levels has been gaining increasing acceptance, despite the fact that only a handful of such cases have so far been demonstrated. Moreover, because each of these cases involves only one gene, we lack any understanding of how natural selection may act on cis-regulation across entire pathways or networks. Here we apply a genome-wide test for selection on cis-regulation to two subspecies of the mouse Mus musculus. We find evidence for lineage-specific selection at over 100 genes involved in diverse processes such as growth, locomotion, and memory. These gene sets implicate candidate genes that are supported by both quantitative trait loci and a validated causality-testing framework, and they predict a number of phenotypic differences, which we confirm in all four cases tested. Our results suggest that gene expression adaptation is widespread and that these adaptations can be highly polygenic, involving cis-regulatory changes at numerous functionally related genes. These coordinated adaptations may contribute to divergence in a wide range of morphological, physiological, and behavioral phenotypes.


Assuntos
Adaptação Biológica/genética , Evolução Molecular , Regulação da Expressão Gênica , Herança Multifatorial/genética , Locos de Características Quantitativas/genética , Animais , Bases de Dados Genéticas , Perfilação da Expressão Gênica , Crescimento e Desenvolvimento/genética , Locomoção/genética , Memória , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/genética , Fenótipo , Seleção Genética
13.
Nat Genet ; 37(11): 1224-33, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16200066

RESUMO

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.


Assuntos
Araquidonato 5-Lipoxigenase/genética , Densidade Óssea/genética , Perfilação da Expressão Gênica , Predisposição Genética para Doença , Obesidade/genética , Animais , Cruzamentos Genéticos , Feminino , Genoma , Genótipo , Lipídeos/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Modelos Biológicos , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , PPAR gama/genética , Locos de Características Quantitativas
14.
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
15.
PLoS Pathog ; 7(12): e1002469, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22241984

RESUMO

Anthrax is a potentially fatal disease resulting from infection with Bacillus anthracis. The outcome of infection is influenced by pathogen-encoded virulence factors such as lethal toxin (LT), as well as by genetic variation within the host. To identify host genes controlling susceptibility to anthrax, a library of congenic mice consisting of strains with homozygous chromosomal segments from the LT-responsive CAST/Ei strain introgressed on a LT-resistant C57BL/6 (B6) background was screened for response to LT. Three congenic strains containing CAST/Ei regions of chromosome 11 were identified that displayed a rapid inflammatory response to LT similar to, but more severe than that driven by a LT-responsive allele of the inflammasome constituent NRLP1B. Importantly, increased response to LT in congenic mice correlated with greater resistance to infection by the Sterne strain of B. anthracis. The genomic region controlling the inflammatory response to LT was mapped to 66.36-74.67 Mb on chromosome 11, a region that encodes the LT-responsive CAST/Ei allele of Nlrp1b. However, known downstream effects of NLRP1B activation, including macrophage pyroptosis, cytokine release, and leukocyte infiltration could not fully explain the response to LT or the resistance to B. anthracis Sterne in congenic mice. Further, the exacerbated response in congenic mice is inherited in a recessive manner while the Nlrp1b-mediated response to LT is dominant. Finally, congenic mice displayed increased responsiveness in a model of sepsis compared with B6 mice. In total, these data suggest that allelic variation of one or more chromosome 11 genes in addition to Nlrp1b controls the severity of host response to multiple inflammatory stimuli and contributes to resistance to B. anthracis Sterne. Expression quantitative trait locus analysis revealed 25 genes within this region as high priority candidates for contributing to the host response to LT.


Assuntos
Alelos , Antraz/genética , Bacillus anthracis , Cromossomos de Mamíferos/genética , Variação Genética , Imunidade Inata/genética , Animais , Antraz/imunologia , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Cromossomos de Mamíferos/imunologia , Inflamação/genética , Inflamação/imunologia , Camundongos , Locos de Características Quantitativas/imunologia
16.
Cell Host Microbe ; 31(6): 1038-1053.e10, 2023 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-37279756

RESUMO

The microbes and microbial pathways that influence host inflammatory disease progression remain largely undefined. Here, we show that variation in atherosclerosis burden is partially driven by gut microbiota and is associated with circulating levels of uric acid (UA) in mice and humans. We identify gut bacterial taxa spanning multiple phyla, including Bacillota, Fusobacteriota, and Pseudomonadota, that use multiple purines, including UA as carbon and energy sources anaerobically. We identify a gene cluster that encodes key steps of anaerobic purine degradation and that is widely distributed among gut-dwelling bacteria. Furthermore, we show that colonization of gnotobiotic mice with purine-degrading bacteria modulates levels of UA and other purines in the gut and systemically. Thus, gut microbes are important drivers of host global purine homeostasis and serum UA levels, and gut bacterial catabolism of purines may represent a mechanism by which gut bacteria influence health.


Assuntos
Microbioma Gastrointestinal , Humanos , Animais , Camundongos , Homeostase , Purinas/metabolismo , Bactérias/genética , Bactérias/metabolismo , Ácido Úrico/metabolismo
17.
Elife ; 112022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-35072627

RESUMO

Obesity has repeatedly been linked to reorganization of the gut microbiome, yet to this point obesity therapeutics have been targeted exclusively toward the human host. Here, we show that gut microbe-targeted inhibition of the trimethylamine N-oxide (TMAO) pathway protects mice against the metabolic disturbances associated with diet-induced obesity (DIO) or leptin deficiency (Lepob/ob). Small molecule inhibition of the gut microbial enzyme choline TMA-lyase (CutC) does not reduce food intake but is instead associated with alterations in the gut microbiome, improvement in glucose tolerance, and enhanced energy expenditure. We also show that gut microbial CutC inhibition is associated with reorganization of host circadian control of both phosphatidylcholine and energy metabolism. This study underscores the relationship between microbe and host metabolism and provides evidence that gut microbe-derived trimethylamine (TMA) is a key regulator of the host circadian clock. This work also demonstrates that gut microbe-targeted enzyme inhibitors have potential as anti-obesity therapeutics.


Assuntos
Colina/análogos & derivados , Ritmo Circadiano/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Obesidade/metabolismo , Animais , Colina/administração & dosagem , Colina/metabolismo , Dieta Hiperlipídica , Inibidores Enzimáticos/farmacologia , Leptina/deficiência , Liases/efeitos dos fármacos , Masculino , Metilaminas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/genética , Obesidade/microbiologia
18.
PLoS Biol ; 6(5): e107, 2008 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-18462017

RESUMO

Genetic variants that are associated with common human diseases do not lead directly to disease, but instead act on intermediate, molecular phenotypes that in turn induce changes in higher-order disease traits. Therefore, identifying the molecular phenotypes that vary in response to changes in DNA and that also associate with changes in disease traits has the potential to provide the functional information required to not only identify and validate the susceptibility genes that are directly affected by changes in DNA, but also to understand the molecular networks in which such genes operate and how changes in these networks lead to changes in disease traits. Toward that end, we profiled more than 39,000 transcripts and we genotyped 782,476 unique single nucleotide polymorphisms (SNPs) in more than 400 human liver samples to characterize the genetic architecture of gene expression in the human liver, a metabolically active tissue that is important in a number of common human diseases, including obesity, diabetes, and atherosclerosis. This genome-wide association study of gene expression resulted in the detection of more than 6,000 associations between SNP genotypes and liver gene expression traits, where many of the corresponding genes identified have already been implicated in a number of human diseases. The utility of these data for elucidating the causes of common human diseases is demonstrated by integrating them with genotypic and expression data from other human and mouse populations. This provides much-needed functional support for the candidate susceptibility genes being identified at a growing number of genetic loci that have been identified as key drivers of disease from genome-wide association studies of disease. By using an integrative genomics approach, we highlight how the gene RPS26 and not ERBB3 is supported by our data as the most likely susceptibility gene for a novel type 1 diabetes locus recently identified in a large-scale, genome-wide association study. We also identify SORT1 and CELSR2 as candidate susceptibility genes for a locus recently associated with coronary artery disease and plasma low-density lipoprotein cholesterol levels in the process.


Assuntos
Perfilação da Expressão Gênica , Predisposição Genética para Doença/genética , Fígado/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Transcrição Gênica/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Criança , Pré-Escolar , LDL-Colesterol/sangue , LDL-Colesterol/genética , Doença da Artéria Coronariana/genética , Diabetes Mellitus Tipo 1/genética , Feminino , Genes MHC da Classe II/genética , Genoma Humano , Genótipo , Humanos , Lactente , Masculino , Camundongos , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Locos de Características Quantitativas/genética , RNA Mensageiro/análise , RNA Mensageiro/genética
19.
Cell Mol Gastroenterol Hepatol ; 11(1): 199-220, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32866618

RESUMO

BACKGROUND & AIMS: Liver fibrosis is a multifactorial trait that develops in response to chronic liver injury. Our aim was to characterize the genetic architecture of carbon tetrachloride (CCl4)-induced liver fibrosis using the Hybrid Mouse Diversity Panel, a panel of more than 100 genetically distinct mouse strains optimized for genome-wide association studies and systems genetics. METHODS: Chronic liver injury was induced by CCl4 injections twice weekly for 6 weeks. Four hundred thirty-seven mice received CCl4 and 256 received vehicle, after which animals were euthanized for liver histology and gene expression. Using automated digital image analysis, we quantified fibrosis as the collagen proportionate area of the whole section, excluding normal collagen. RESULTS: We discovered broad variation in fibrosis among the Hybrid Mouse Diversity Panel strains, demonstrating a significant genetic influence. Genome-wide association analyses revealed significant and suggestive loci underlying susceptibility to fibrosis, some of which overlapped with loci identified in mouse crosses and human population studies. Liver global gene expression was assessed by RNA sequencing across the strains, and candidate genes were identified using differential expression and expression quantitative trait locus analyses. Gene set enrichment analyses identified the underlying pathways, of which stellate cell involvement was prominent, and coexpression network modeling identified modules associated with fibrosis. CONCLUSIONS: Our results provide a rich resource for the design of experiments to understand mechanisms underlying fibrosis and for rational strain selection when testing antifibrotic drugs.


Assuntos
Tetracloreto de Carbono/toxicidade , Redes Reguladoras de Genes/efeitos dos fármacos , Predisposição Genética para Doença , Cirrose Hepática/induzido quimicamente , Fígado/patologia , Animais , Tetracloreto de Carbono/administração & dosagem , Modelos Animais de Doenças , Estudo de Associação Genômica Ampla , Humanos , Injeções Intraperitoneais , Fígado/efeitos dos fármacos , Cirrose Hepática/genética , Cirrose Hepática/patologia , Masculino , Camundongos , Locos de Características Quantitativas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA