Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Cell Host Microbe ; 31(6): 1038-1053.e10, 2023 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-37279756

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Humanos , Animales , Ratones , Homeostasis , Purinas/metabolismo , Bacterias/genética , Bacterias/metabolismo , Ácido Úrico/metabolismo
2.
Elife ; 112022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-35072627

RESUMEN

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.


Asunto(s)
Colina/análogos & derivados , Ritmo Circadiano/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Obesidad/metabolismo , Animales , Colina/administración & dosificación , Colina/metabolismo , Dieta Alta en Grasa , Inhibidores Enzimáticos/farmacología , Leptina/deficiencia , Liasas/efectos de los fármacos , Masculino , Metilaminas/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/microbiología
3.
Eur Heart J ; 42(9): 919-933, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33532862

RESUMEN

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.


Asunto(s)
Enfermedad de la Arteria Coronaria , Infarto del Miocardio , Enfermedad de la Arteria Coronaria/genética , Células Endoteliales , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Humanos , Japón , Infarto del Miocardio/genética , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo
4.
Mol Syst Biol ; 17(1): e9684, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33417276

RESUMEN

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.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Hígado Graso/genética , Glucosa/efectos adversos , Resistencia a la Insulina/genética , MAP Quinasa Quinasa 6/genética , Proteínas Nucleares/genética , Animales , Modelos Animales de Enfermedad , Hígado Graso/inducido químicamente , Hígado Graso/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Variación Genética , Lipidómica , Masculino , Ratones , Fosfatidilcolinas/metabolismo , Triglicéridos/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 41(1): 220-233, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33086870

RESUMEN

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.


Asunto(s)
Apoptosis , Aterosclerosis/metabolismo , Proliferación Celular , Células Endoteliales/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Aorta/metabolismo , Aorta/patología , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Cadherinas/genética , Cadherinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/patología , Femenino , Factor Estimulante de Colonias de Macrófagos/deficiencia , Factor Estimulante de Colonias de Macrófagos/genética , Macrófagos/patología , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transducción de Señal
6.
Cell Mol Gastroenterol Hepatol ; 11(1): 199-220, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32866618

RESUMEN

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.


Asunto(s)
Tetracloruro de Carbono/toxicidad , Redes Reguladoras de Genes/efectos de los fármacos , Predisposición Genética a la Enfermedad , Cirrosis Hepática/inducido químicamente , Hígado/patología , Animales , Tetracloruro de Carbono/administración & dosificación , Modelos Animales de Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Inyecciones Intraperitoneales , Hígado/efectos de los fármacos , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Masculino , Ratones , Sitios de Carácter Cuantitativo
7.
Cell Metab ; 29(4): 932-949.e4, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30639359

RESUMEN

We studied sex differences in over 50 cardio-metabolic traits in a panel of 100 diverse inbred strains of mice. The results clearly showed that the effects of sex on both clinical phenotypes and gene expression depend on the genetic background. In support of this, genetic loci associated with the traits frequently showed sex specificity. For example, Lyplal1, a gene implicated in human obesity, was shown to underlie a sex-specific locus for diet-induced obesity. Global gene expression analyses of tissues across the panel implicated adipose tissue "beiging" and mitochondrial functions in the sex differences. Isolated mitochondria showed gene-by-sex interactions in oxidative functions, such that some strains (C57BL/6J) showed similar function between sexes, whereas others (DBA/2J and A/J) showed increased function in females. Reduced adipose mitochondrial function in males as compared to females was associated with increased susceptibility to obesity and insulin resistance. Gonadectomy studies indicated that gonadal hormones acting in a tissue-specific manner were responsible in part for the sex differences.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Mitocondrias/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Enfermedades Cardiovasculares/patología , Femenino , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos , Obesidad/metabolismo , Obesidad/patología , Fenotipo , Análisis de Componente Principal , Caracteres Sexuales
8.
Artículo en Inglés | MEDLINE | ID: mdl-31920990

RESUMEN

Responses to a high fat, high sucrose (HFHS) diet vary greatly among inbred strains of mice. We sought to examine the epigenetic (DNA methylation) changes underlying these differences as well as variation in weight loss when switched to a low-fat chow diet. We surveyed DNA methylation from livers of 45 inbred mouse strains fed a HFHS diet for 8 weeks using reduced-representation bisulfite sequencing (RRBS). We observed a total of 1,045,665 CpGs of which 83 candidate sites were significantly associated with HFHS diet. Many of these CpGs correlated strongly with gene expression or clinical traits such as body fat percentage and plasma glucose. Five inbred strains were then studied in the context of weight loss to test for evidence of epigenetic "memory." The mice were first fed a HFHS diet for 6 weeks followed by a low-fat chow diet for 4 weeks. Four of the five strains returned to initial levels of body fat while one strain, A/J, retained almost 50% of the fat gained. A total of 36 of the HFHS diet responsive CpGs exhibited evidence of persistent epigenetic modifications following weight normalization, including CpGs near the genes Scd1 and Cdk1. Our study identifies DNA methylation changes in response to a HFHS diet challenge that revert more slowly than overall body fat percentage in weight loss and provides evidence for epigenetic mediated "memory."

9.
Nat Microbiol ; 3(12): 1461-1471, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30397344

RESUMEN

Humans with metabolic and inflammatory diseases frequently harbour lower levels of butyrate-producing bacteria in their gut. However, it is not known whether variation in the levels of these organisms is causally linked with disease development and whether diet modifies the impact of these bacteria on health. Here we show that a prominent gut-associated butyrate-producing bacterial genus (Roseburia) is inversely correlated with atherosclerotic lesion development in a genetically diverse mouse population. We use germ-free apolipoprotein E-deficient mice colonized with synthetic microbial communities that differ in their capacity to generate butyrate to demonstrate that Roseburia intestinalis interacts with dietary plant polysaccharides to: impact gene expression in the intestine, directing metabolism away from glycolysis and toward fatty acid utilization; lower systemic inflammation; and ameliorate atherosclerosis. Furthermore, intestinal administration of butyrate reduces endotoxaemia and atherosclerosis development. Together, our results illustrate how modifiable diet-by-microbiota interactions impact cardiovascular disease, and suggest that interventions aimed at increasing the representation of butyrate-producing bacteria may provide protection against atherosclerosis.


Asunto(s)
Aterosclerosis , Clostridiales/metabolismo , Dieta , Microbioma Gastrointestinal , Intestinos/microbiología , Animales , Apolipoproteínas E/genética , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/patología , Butiratos/metabolismo , Butiratos/farmacología , Enfermedades Cardiovasculares , Clostridiales/genética , Colon/metabolismo , Colon/microbiología , Carbohidratos de la Dieta/metabolismo , Modelos Animales de Enfermedad , Endotoxemia , Metabolismo Energético , Ácidos Grasos/metabolismo , Heces/microbiología , Expresión Génica , Vida Libre de Gérmenes , Masculino , Metaboloma , Ratones , Ratones Noqueados , ARN Ribosómico 16S/genética
10.
Biol Sex Differ ; 9(1): 46, 2018 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-30343673

RESUMEN

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) encompasses benign steatosis and more severe conditions such as non-alcoholic steatohepatitis (NASH), cirrhosis, and liver cancer. This chronic liver disease has a poorly understood etiology and demonstrates sexual dimorphisms. We aim to examine the molecular mechanisms underlying sexual dimorphisms in NAFLD pathogenesis through a comprehensive multi-omics study. We integrated genomics (DNA variations), transcriptomics of liver and adipose tissue, and phenotypic data of NAFLD derived from female mice of ~ 100 strains included in the hybrid mouse diversity panel (HMDP) and compared the NAFLD molecular pathways and gene networks between sexes. RESULTS: We identified both shared and sex-specific biological processes for NAFLD. Adaptive immunity, branched chain amino acid metabolism, oxidative phosphorylation, and cell cycle/apoptosis were shared between sexes. Among the sex-specific pathways were vitamins and cofactors metabolism and ion channel transport for females, and phospholipid, lysophospholipid, and phosphatidylinositol metabolism and insulin signaling for males. Additionally, numerous lipid and insulin-related pathways and inflammatory processes in the adipose and liver tissue appeared to show more prominent association with NAFLD in male HMDP. Using data-driven network modeling, we identified plausible sex-specific and tissue-specific regulatory genes as well as those that are shared between sexes. These key regulators orchestrate the NAFLD pathways in a sex- and tissue-specific manner. Gonadectomy experiments support that sex hormones may partially underlie the sexually dimorphic genes and pathways involved in NAFLD. CONCLUSIONS: Our multi-omics integrative study reveals sex- and tissue-specific genes, processes, and networks underlying sexual dimorphism in NAFLD and may facilitate sex-specific precision medicine.


Asunto(s)
Tejido Adiposo/metabolismo , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Caracteres Sexuales , Animales , Castración , Femenino , Genómica , Metabolismo de los Lípidos , Masculino , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Transducción de Señal , Transcriptoma
11.
Curr Opin Lipidol ; 29(5): 404-410, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30156571

RESUMEN

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.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Enfermedades Metabólicas/fisiopatología , Caracteres Sexuales , Animales , Enfermedades Cardiovasculares/metabolismo , Humanos , Enfermedades Metabólicas/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 38(9): 2016-2027, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30026271

RESUMEN

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.


Asunto(s)
Apoptosis , Aterosclerosis/fisiopatología , Proteínas de Homeodominio/fisiología , Macrófagos/fisiología , Factores de Transcripción/fisiología , Dedos de Zinc/fisiología , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Expresión Génica , Proteínas de Homeodominio/genética , Macrófagos/citología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Factores de Transcripción/genética , Dedos de Zinc/genética
13.
Cell Metab ; 27(5): 1138-1155.e6, 2018 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-29719227

RESUMEN

Inter-tissue communication via secreted proteins has been established as a vital mechanism for proper physiologic homeostasis. Here, we report a bioinformatics framework using a mouse reference population, the Hybrid Mouse Diversity Panel (HMDP), which integrates global multi-tissue expression data and publicly available resources to identify and functionally annotate novel circuits of tissue-tissue communication. We validate this method by showing that we can identify known as well as novel endocrine factors responsible for communication between tissues. We further show the utility of this approach by identification and mechanistic characterization of two new endocrine factors. Adipose-derived Lipocalin-5 is shown to enhance skeletal muscle mitochondrial function, and liver-secreted Notum promotes browning of white adipose tissue, also known as "beiging." We demonstrate the general applicability of the method by providing in vivo evidence for three additional novel molecules mediating tissue-tissue interactions.


Asunto(s)
Sistema Endocrino/metabolismo , Homeostasis , Lipocalinas/metabolismo , Proteómica/métodos , Tejido Adiposo/metabolismo , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Músculo Esquelético/metabolismo
14.
J Lipid Res ; 59(7): 1164-1174, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29739864

RESUMEN

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.


Asunto(s)
Ceramidas/metabolismo , Dieta , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Caracteres Sexuales , Animales , Ceramidas/biosíntesis , Femenino , Hígado/efectos de los fármacos , Masculino , Ratones , Testosterona/farmacología
15.
Genome Biol ; 18(1): 70, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28407784

RESUMEN

BACKGROUND: The gut microbiome is a complex and metabolically active community that directly influences host phenotypes. In this study, we profile gut microbiota using 16S rRNA gene sequencing in 531 well-phenotyped Finnish men from the Metabolic Syndrome In Men (METSIM) study. RESULTS: We investigate gut microbiota relationships with a variety of factors that have an impact on the development of metabolic and cardiovascular traits. We identify novel associations between gut microbiota and fasting serum levels of a number of metabolites, including fatty acids, amino acids, lipids, and glucose. In particular, we detect associations with fasting plasma trimethylamine N-oxide (TMAO) levels, a gut microbiota-dependent metabolite associated with coronary artery disease and stroke. We further investigate the gut microbiota composition and microbiota-metabolite relationships in subjects with different body mass index and individuals with normal or altered oral glucose tolerance. Finally, we perform microbiota co-occurrence network analysis, which shows that certain metabolites strongly correlate with microbial community structure and that some of these correlations are specific for the pre-diabetic state. CONCLUSIONS: Our study identifies novel relationships between the composition of the gut microbiota and circulating metabolites and provides a resource for future studies to understand host-gut microbiota relationships.


Asunto(s)
Bacterias/clasificación , Bacterias/metabolismo , Microbioma Gastrointestinal , Síndrome Metabólico/sangre , Síndrome Metabólico/microbiología , Anciano , Aminoácidos/sangre , Glucemia/metabolismo , Índice de Masa Corporal , Estudios de Cohortes , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/microbiología , Ácidos Grasos/sangre , Prueba de Tolerancia a la Glucosa , Humanos , Lípidos/sangre , Masculino , Metilaminas , Microbiota/fisiología , Persona de Mediana Edad , Fenotipo
16.
J Lipid Res ; 58(1): 178-187, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27811231

RESUMEN

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.


Asunto(s)
Hígado Graso/genética , Lipoproteínas HDL/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/genética , Triglicéridos/genética , Animales , Dieta Alta en Grasa , Hígado Graso/sangre , Hígado Graso/patología , Femenino , Estudio de Asociación del Genoma Completo , Hormonas/genética , Hormonas/metabolismo , Hiperlipidemias/sangre , Hiperlipidemias/genética , Hiperlipidemias/patología , Resistencia a la Insulina/genética , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/sangre , Obesidad/patología , Polimorfismo de Nucleótido Simple/genética , Caracteres Sexuales
17.
Gut Microbes ; 7(4): 313-322, 2016 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-27355107

RESUMEN

We previously reported quantitation of gut microbiota in a panel of 89 different inbred strains of mice, and we now examine the question of sex differences in microbiota composition. When the total population of 689 mice was examined together, several taxa exhibited significant differences in abundance between sexes but a larger number of differences were observed at the single strain level, suggesting that sex differences can be obscured by host genetics and environmental factors. We also examined a subset of mice on chow and high fat diets and observed sex-by-diet interactions. We further investigated the sex differences using gonadectomized and hormone treated mice from 3 different inbred strains. Principal coordinate analysis with unweighted UniFrac distances revealed very clear effects of gonadectomy and hormone replacement on microbiota composition in all 3 strains. Moreover, bile acid analyses showed gender-specific differences as well as effects of gonodectomy, providing one possible mechanism mediating sex differences in microbiota composition.


Asunto(s)
Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Hormonas/metabolismo , Ratones/microbiología , Animales , Ácidos y Sales Biliares/metabolismo , Conducta Alimentaria , Femenino , Masculino , Ratones/fisiología , Factores Sexuales
18.
J Lipid Res ; 57(6): 925-42, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27099397

RESUMEN

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.


Asunto(s)
Enfermedades Cardiovasculares/genética , Modelos Animales de Enfermedad , Enfermedades Metabólicas/genética , Transcriptoma/genética , Animales , Aterosclerosis/genética , Enfermedades Cardiovasculares/patología , Estudio de Asociación del Genoma Completo , Insuficiencia Cardíaca/genética , Humanos , Hibridación Genética , Resistencia a la Insulina/genética , Enfermedades Metabólicas/patología , Ratones , Microbiota/genética , Obesidad/genética , Osteoporosis/genética , Sitios de Carácter Cuantitativo/genética
19.
Cell ; 165(1): 111-124, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-26972052

RESUMEN

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.


Asunto(s)
Plaquetas/metabolismo , Microbioma Gastrointestinal , Metilaminas/metabolismo , Trombosis/metabolismo , Animales , Calcio/metabolismo , Traumatismos de las Arterias Carótidas/patología , Ciego/microbiología , Cloruros , Colina/metabolismo , Dieta , Femenino , Compuestos Férricos , Vida Libre de Gérmenes , Humanos , Metilaminas/sangre , Ratones , Ratones Endogámicos C57BL , Trombosis/patología
20.
Genome Res ; 25(10): 1558-69, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26260972

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal/genética , Animales , Dieta , Dieta Alta en Grasa , Ambiente , Femenino , Estudio de Asociación del Genoma Completo , Herencia , Masculino , Ratones , Ratones Endogámicos , Obesidad/microbiología , ARN Ribosómico 16S , Sacarosa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...