Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gut ; 70(11): 2105-2114, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33975870

RESUMEN

OBJECTIVE: Gut microbial products are involved in regulation of host metabolism. In human and experimental studies, we explored the potential role of hippurate, a hepatic phase 2 conjugation product of microbial benzoate, as a marker and mediator of metabolic health. DESIGN: In 271 middle-aged non-diabetic Danish individuals, who were stratified on habitual dietary intake, we applied 1H-nuclear magnetic resonance (NMR) spectroscopy of urine samples and shotgun-sequencing-based metagenomics of the gut microbiome to explore links between the urine level of hippurate, measures of the gut microbiome, dietary fat and markers of metabolic health. In mechanistic experiments with chronic subcutaneous infusion of hippurate to high-fat-diet-fed obese mice, we tested for causality between hippurate and metabolic phenotypes. RESULTS: In the human study, we showed that urine hippurate positively associates with microbial gene richness and functional modules for microbial benzoate biosynthetic pathways, one of which is less prevalent in the Bacteroides 2 enterotype compared with Ruminococcaceae or Prevotella enterotypes. Through dietary stratification, we identify a subset of study participants consuming a diet rich in saturated fat in which urine hippurate concentration, independently of gene richness, accounts for links with metabolic health. In the high-fat-fed mice experiments, we demonstrate causality through chronic infusion of hippurate (20 nmol/day) resulting in improved glucose tolerance and enhanced insulin secretion. CONCLUSION: Our human and experimental studies show that a high urine hippurate concentration is a general marker of metabolic health, and in the context of obesity induced by high-fat diets, hippurate contributes to metabolic improvements, highlighting its potential as a mediator of metabolic health.


Asunto(s)
Biomarcadores/metabolismo , Microbioma Gastrointestinal , Hipuratos/metabolismo , Animales , Biodiversidad , Dinamarca , Femenino , Humanos , Espectroscopía de Resonancia Magnética , Masculino , Metaboloma , Metagenómica , Ratones , Persona de Mediana Edad , Fenotipo
2.
Diabetologia ; 63(6): 1223-1235, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32173762

RESUMEN

AIMS/HYPOTHESIS: Drug and surgical-based therapies in type 2 diabetes are associated with altered gut microbiota architecture. Here we investigated the role of the gut microbiome in improved glucose homeostasis following bariatric surgery. METHODS: We carried out gut microbiome analyses in gastrectomised (by vertical sleeve gastrectomy [VSG]) rats of the Goto-Kakizaki (GK) non-obese model of spontaneously occurring type 2 diabetes, followed by physiological studies in the GK rat. RESULTS: VSG in the GK rat led to permanent improvement of glucose tolerance associated with minor changes in the gut microbiome, mostly characterised by significant enrichment of caecal Prevotella copri. Gut microbiota enrichment with P. copri in GK rats through permissive antibiotic treatment, inoculation of gut microbiota isolated from gastrectomised GK rats, and direct inoculation of P. copri, resulted in significant improvement of glucose tolerance, independent of changes in body weight. Plasma bile acids were increased in GK rats following inoculation with P. copri and P. copri-enriched microbiota from VSG-treated rats; the inoculated GK rats then showed increased liver glycogen and upregulated expression of Fxr (also known as Nr1h4), Srebf1c, Chrebp (also known as Mlxipl) and Il10 and downregulated expression of Cyp7a1. CONCLUSIONS: Our data underline the impact of intestinal P. copri on improved glucose homeostasis through enhanced bile acid metabolism and farnesoid X receptor (FXR) signalling, which may represent a promising opportunity for novel type 2 diabetes therapeutics.


Asunto(s)
Diabetes Mellitus Tipo 2/microbiología , Microbioma Gastrointestinal/fisiología , Prevotella/fisiología , Animales , Glucemia/metabolismo , Peso Corporal/fisiología , Masculino , Ratas , Transducción de Señal/fisiología
3.
Cell Mol Life Sci ; 75(21): 3977-3990, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30101405

RESUMEN

Evidence from the literature keeps highlighting the impact of mutualistic bacterial communities of the gut microbiota on human health. The gut microbita is a complex ecosystem of symbiotic bacteria which contributes to mammalian host biology by processing, otherwise, indigestible nutrients, supplying essential metabolites, and contributing to modulate its immune system. Advances in sequencing technologies have enabled structural analysis of the human gut microbiota and allowed detection of changes in gut bacterial composition in several common diseases, including cardiometabolic disorders. Biological signals sent by the gut microbiota to the host, including microbial metabolites and pro-inflammatory molecules, mediate microbiome-host genome cross-talk. This rapidly expanding line of research can identify disease-causing and disease-predictive microbial metabolite biomarkers, which can be translated into novel biodiagnostic tests, dietary supplements, and nutritional interventions for personalized therapeutic developments in common diseases. Here, we review results from the most significant studies dealing with the association of products from the gut microbial metabolism with cardiometabolic disorders. We underline the importance of these postbiotic biomarkers in the diagnosis and treatment of human disorders.


Asunto(s)
Biomarcadores/metabolismo , Enfermedades Cardiovasculares/genética , Microbioma Gastrointestinal/genética , Enfermedades Metabólicas/genética , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/microbiología , Enfermedades Cardiovasculares/patología , Humanos , Enfermedades Metabólicas/microbiología , Enfermedades Metabólicas/patología
4.
Lipids Health Dis ; 18(1): 38, 2019 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-30711004

RESUMEN

BACKGROUND: Lipoproteins are major players in the development and progression of atherosclerotic plaques leading to coronary stenosis and myocardial infarction. Epidemiological, genetic and experimental observations have implicated the association of sphingolipids and intermediates of sphingolipid synthesis in atherosclerosis. We aimed to investigate relationships between quantitative changes in serum sphingolipids, the regulation of the metabolism of lipoproteins (LDL, HDL), and endophenotypes of coronary artery disease (CAD). METHODS: We carried out untargeted liquid chromatography - mass spectrometry (UPLC-MS) lipidomics of serum samples of subjects belonging to a cross-sectional study and recruited on the basis of absence or presence of angiographically-defined CAD, and extensively characterized for clinical and biochemical phenotypes. RESULTS: Among the 2998 spectral features detected in the serum samples, 1328 metabolic features were significantly correlated with at least one of the clinical or biochemical phenotypes measured in the cohort. We found evidence of significant associations between 34 metabolite signals, corresponding to a set of sphingomyelins, and serum HDL cholesterol. Many of these metabolite associations were also observed with serum LDL and total cholesterol levels but not as much with serum triglycerides. CONCLUSION: Among patients with CAD, sphingolipids in the form of sphingomyelins are directly correlated with serum levels of lipoproteins and total cholesterol. Results from this study support the fundamental role of sphingolipids in modulating lipid serum levels, highlighting the importance to identify novel targets in the sphingolipid metabolic pathway for anti-atherogenic therapies.


Asunto(s)
Colesterol/sangre , Esfingomielinas/sangre , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de la Arteria Coronaria/sangre , Femenino , Cromatografía de Gases y Espectrometría de Masas , Humanos , Lipoproteínas HDL/sangre , Lipoproteínas LDL/sangre , Masculino , Espectrometría de Masas , Metabolómica/instrumentación , Metabolómica/métodos , Persona de Mediana Edad , Adulto Joven
5.
Sci Rep ; 13(1): 13926, 2023 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-37626071

RESUMEN

Gut-microbiota derived metabolites are important regulators of host biology and metabolism. To understand the impacts of the microbial metabolite 4-cresol sulfate (4-CS) on four chronic diseases [type 2 diabetes mellitus, metabolic syndrome (MetS), non-alcoholic fatty liver disease, and chronic kidney disease (CKD)], we conducted association analyses of plasma 4-CS quantified by liquid chromatography coupled to mass spectrometry (LC-MS) in 3641 participants of the Nagahama study. Our results validated the elevation of 4-CS in CKD and identified a reducing trend in MetS. To delineate the holistic effects of 4-CS, we performed a phenome-wide association analysis (PheWAS) with 937 intermediate biological and behavioral traits. We detected associations between 4-CS and 39 phenotypes related to blood pressure regulation, hepatic and renal functions, hematology, sleep quality, intraocular pressure, ion regulation, ketone and fatty acid metabolisms, disease history and dietary habits. Among them, 19 PheWAS significant traits, including fatty acids and 14 blood pressure indices, were correlated with MetS, suggesting that 4-CS is a potential biomarker for MetS. Consistent associations of this gut microbial-derived metabolite on multiple endophenotypes underlying distinct etiopathogenesis support its role in the overall host health, with prospects of probiotic-based therapeutic solutions in chronic diseases.


Asunto(s)
Diabetes Mellitus Tipo 2 , Humanos , Ésteres del Ácido Sulfúrico , Fenómica , Endofenotipos
6.
Biochimie ; 194: 35-42, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34965461

RESUMEN

The gut microbiota contributes to mammalian host biology by supplying metabolites from nutrients and pro-inflammatory molecules. We have recently shown that urinary hippurate is associated with reduced risk of obesity, increased gut microbiome diversity and gene richness, and functional modules for microbial production of its precursor benzoate. Obese mice infused with hippurate exhibit profound alterations of glucose homeostasis. Here, we tested the biological effects of chronic administration of benzoate on cardiometabolic phenotypes in lean and obese mice. Benzoate induced glucose intolerance, enhanced insulin secretion, increased adiposity and stimulated liver inflammation in lean mice fed control diet. In contrast, in condition of obesity and diabetes induced by high fat diet feeding, benzoate infusion resulted in reduction of glucose intolerance, stimulation of both glucose-induced insulin secretion and ß-cell proliferation, and reduction of liver triglyceride and collagen accumulation. These results combined with those previously obtained in mice treated with hippurate underline the importance of the benzoate-hippurate pathway in cardiometabolic diseases and pave the way to diagnostic and therapeutic solutions.


Asunto(s)
Benzoatos , Diabetes Mellitus , Animales , Benzoatos/farmacología , Diabetes Mellitus/tratamiento farmacológico , Dieta Alta en Grasa/efectos adversos , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/metabolismo
7.
Metabolites ; 12(7)2022 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-35888720

RESUMEN

Analysis of the genetic control of small metabolites provides powerful information on the regulation of the endpoints of genome expression. We carried out untargeted liquid chromatography−high-resolution mass spectrometry in 273 individuals characterized for pathophysiological elements of the cardiometabolic syndrome. We quantified 3013 serum lipidomic features, which we used in both genome-wide association studies (GWAS), using a panel of over 2.5 M imputed single-nucleotide polymorphisms (SNPs), and metabolome-wide association studies (MWAS) with phenotypes. Genetic analyses showed that 926 SNPs at 551 genetic loci significantly (q-value < 10−8) regulate the abundance of 74 lipidomic features in the group, with evidence of monogenic control for only 22 of these. In addition to this strong polygenic control of serum lipids, our results underscore instances of pleiotropy, when a single genetic locus controls the abundance of several distinct lipid features. Using the LIPID MAPS database, we assigned putative lipids, predominantly fatty acyls and sterol lipids, to 77% of the lipidome signals mapped to the genome. We identified significant correlations between lipids and clinical and biochemical phenotypes. These results demonstrate the power of untargeted lipidomic profiling for high-density quantitative molecular phenotyping in human-genetic studies and illustrate the complex genetic control of lipid metabolism.

8.
Rev Prat ; 71(7): 747-751, 2021 Sep.
Artículo en Francés | MEDLINE | ID: mdl-34792911

RESUMEN

PESTICIDES: WHAT IS KNOWN OR SUSPECTED ABOUT THEIR HARMFUL EFFECTS ON HEALTH Pesticides are molecules of various kinds used to combat harmful organisms (insects, weeds, fungi…). Their non-specificity means that they also have an impact on human health at many levels (cancers, Parkinson's disease, developmental problems in children), as shown by the Inserm expertise on "pesticides and health". Numerous other health events could be associated with their exposure. This highlights the importance of developing complementary research between epidemiology and mechanistic toxicology, of reinforcing studies encompassing the deregulation of ecosystems with those of human health, and of producing new study methodologies enabling the toxicology of molecules recently placed on the market or soon to be placed on the market to be predicted.


PESTICIDES: CE QUI EST CONNU OU SUSPECTÉ DE LEUR ACTION DÉLÉTÈRE SUR LA SANTÉ Les pesticides sont des molécules de nature variée permettant de lutter contre des organismes nuisibles (insectes, adventices, champignons…). Leur non-spécificité fait qu'ils impactent aussi la santé humaine à de multiples niveaux (cancers, maladie de Parkinson, problèmes de développement chez les enfants), comme le montre l'expertise de l'INSERM « pesticides et santé ¼. De nombreux autres événements de santé pourraient être associés à leur exposition. Ce qui permet de souligner l'importance de développer des recherches complémentaires entre épidémiologie et toxicologie mécanistique, renforcer les études englobant les dérégulations des écosystèmes à celles de la santé humaine, et produire de nouvelles méthodologies capables de prédire la toxicologie des molécules récemment mises sur le marché ou qui le seront prochainement.


Asunto(s)
Neoplasias , Enfermedad de Parkinson , Plaguicidas , Niño , Ecosistema , Humanos , Plaguicidas/toxicidad
9.
Cells ; 11(1)2021 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-35011671

RESUMEN

Environmental factors including diet, sedentary lifestyle and exposure to pollutants largely influence human health throughout life. Cellular and molecular events triggered by an exposure to environmental pollutants are extremely variable and depend on the age, the chronicity and the doses of exposure. Only a fraction of all relevant mechanisms involved in the onset and progression of pathologies in response to toxicants has probably been identified. Mitochondria are central hubs of metabolic and cell signaling responsible for a large variety of biochemical processes, including oxidative stress, metabolite production, energy transduction, hormone synthesis, and apoptosis. Growing evidence highlights mitochondrial dysfunction as a major hallmark of environmental insults. Here, we present mitochondria as crucial organelles for healthy metabolic homeostasis and whose dysfunction induces critical adverse effects. Then, we review the multiple mechanisms of action of pollutants causing mitochondrial toxicity in link with chronic diseases. We propose the Aryl hydrocarbon Receptor (AhR) as a model of "exposome receptor", whose activation by environmental pollutants leads to various toxic events through mitochondrial dysfunction. Finally, we provide some remarks related to mitotoxicity and risk assessment.


Asunto(s)
Contaminantes Ambientales/efectos adversos , Mitocondrias/patología , Xenobióticos/uso terapéutico , Apoptosis , Humanos , Xenobióticos/farmacología
10.
PLoS One ; 15(11): e0242019, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33237940

RESUMEN

Aortic valve stenosis (AVS) is a prevalent condition among the elderly population that eventually requires aortic valve replacement. The lack of reliable biomarkers for AVS poses a challenge for its early diagnosis and the application of preventive measures. Untargeted gas chromatography mass spectrometry (GC-MS) metabolomics was applied in 46 AVS cases and 46 controls to identify plasma and urine metabolites underlying AVS risk. Multivariate data analyses were performed on pre-processed data (e.g. spectral peak alignment), in order to detect changes in metabolite levels in AVS patients and to evaluate their performance in group separation and sensitivity of AVS prediction, followed by regression analyses to test for their association with AVS. Through untargeted analysis of 190 urine and 130 plasma features that could be detected and quantified in the GC-MS spectra, we identified contrasting levels of 22 urine and 21 plasma features between AVS patients and control subjects. Following metabolite assignment, we observed significant changes in the concentration of known metabolites in urine (n = 14) and plasma (n = 15) that distinguish the metabolomic profiles of AVS patients from healthy controls. Associations with AVS were replicated in both plasma and urine for about half of these metabolites. Among these, 2-Oxovaleric acid, elaidic acid, myristic acid, palmitic acid, estrone, myo-inositol showed contrasting trends of regulation in the two biofluids. Only trans-Aconitic acid and 2,4-Di-tert-butylphenol showed consistent patterns of regulation in both plasma and urine. These results illustrate the power of metabolomics in identifying potential disease-associated biomarkers and provide a foundation for further studies towards early diagnostic applications in severe heart conditions that may prevent surgery in the elderly.


Asunto(s)
Estenosis de la Válvula Aórtica/metabolismo , Válvula Aórtica/metabolismo , Biomarcadores/sangre , Biomarcadores/orina , Plasma/metabolismo , Orina/química , Estenosis de la Válvula Aórtica/sangre , Estenosis de la Válvula Aórtica/orina , Estudios de Casos y Controles , Ecocardiografía/métodos , Femenino , Cromatografía de Gases y Espectrometría de Masas/métodos , Prótesis Valvulares Cardíacas , Humanos , Masculino , Metabolómica/métodos , Persona de Mediana Edad , Análisis Multivariante , Análisis de Regresión , Urinálisis/métodos
11.
Cell Rep ; 30(7): 2306-2320.e5, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32075738

RESUMEN

Exposure to natural metabolites contributes to the risk of cardiometabolic diseases (CMDs). Through metabolome profiling, we identify the inverse correlation between serum concentrations of 4-cresol and type 2 diabetes. The chronic administration of non-toxic doses of 4-cresol in complementary preclinical models of CMD reduces adiposity, glucose intolerance, and liver triglycerides, enhances insulin secretion in vivo, stimulates islet density and size, and pancreatic ß-cell proliferation, and increases vascularization, suggesting activated islet enlargement. In vivo insulin sensitivity is not affected by 4-cresol. The incubation of mouse isolated islets with 4-cresol results in enhanced insulin secretion, insulin content, and ß-cell proliferation of a magnitude similar to that induced by GLP-1. In both CMD models and isolated islets, 4-cresol is associated with the downregulated expression of the kinase DYRK1A, which may mediate its biological effects. Our findings identify 4-cresol as an effective regulator of ß-cell function, which opens up perspectives for therapeutic applications in syndromes of insulin deficiency.


Asunto(s)
Linfocitos B/metabolismo , Glucemia/metabolismo , Cresoles/uso terapéutico , Diabetes Mellitus Tipo 2/genética , Secreción de Insulina/efectos de los fármacos , Metabolómica/métodos , Obesidad/metabolismo , Animales , Proliferación Celular , Cresoles/farmacología , Homeostasis , Humanos , Ratones , Ratas
12.
Dis Model Mech ; 12(7)2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31213483

RESUMEN

To define renal molecular mechanisms that are affected by permanent hyperglycaemia and might promote phenotypes relevant to diabetic nephropathy, we carried out linkage analysis of genome-wide gene transcription in the kidneys of F2 offspring from the Goto-Kakizaki (GK) rat model of type 2 diabetes and normoglycaemic Brown Norway (BN) rats. We mapped 2526 statistically significant expression quantitative trait loci (eQTLs) in the cross. More than 40% of eQTLs mapped in the close vicinity of the linked transcripts, underlying possible cis-regulatory mechanisms of gene expression. We identified eQTL hotspots on chromosomes 5 and 9 regulating the expression of 80-165 genes, sex or cross direction effects, and enriched metabolic and immunological processes by segregating GK alleles. Comparative analysis with adipose tissue eQTLs in the same cross showed that 496 eQTLs, in addition to the top enriched biological pathways, are conserved in the two tissues. Extensive similarities in eQTLs mapped in the GK rat and in the spontaneously hypertensive rat (SHR) suggest a common aetiology of disease phenotypes common to the two strains, including insulin resistance, which is a prominent pathophysiological feature in both GK rats and SHRs. Our data shed light on shared and tissue-specific molecular mechanisms that might underlie aetiological aspects of insulin resistance in the context of spontaneously occurring hyperglycaemia and hypertension.


Asunto(s)
Tejido Adiposo/metabolismo , Modelos Animales de Enfermedad , Resistencia a la Insulina/genética , Riñón/metabolismo , Transcriptoma , Animales , Mapeo Cromosómico , Diabetes Mellitus Tipo 2/genética , Predisposición Genética a la Enfermedad , Sitios de Carácter Cuantitativo , Ratas , Ratas Endogámicas BN , Ratas Endogámicas SHR
13.
Sci Rep ; 9(1): 3656, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30842494

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is often associated with obesity and type 2 diabetes. To disentangle etiological relationships between these conditions and identify genetically-determined metabolites involved in NAFLD processes, we mapped 1H nuclear magnetic resonance (NMR) metabolomic and disease-related phenotypes in a mouse F2 cross derived from strains showing resistance (BALB/c) and increased susceptibility (129S6) to these diseases. Quantitative trait locus (QTL) analysis based on single nucleotide polymorphism (SNP) genotypes identified diet responsive QTLs in F2 mice fed control or high fat diet (HFD). In HFD fed F2 mice we mapped on chromosome 18 a QTL regulating liver micro- and macrovesicular steatosis and inflammation, independently from glucose intolerance and adiposity, which was linked to chromosome 4. Linkage analysis of liver metabolomic profiling data identified a QTL for octopamine, which co-localised with the QTL for liver histopathology in the cross. Functional relationship between these two QTLs was validated in vivo in mice chronically treated with octopamine, which exhibited reduction in liver histopathology and metabolic benefits, underlining its role as a mechanistic biomarker of fatty liver with potential therapeutic applications.


Asunto(s)
Cromosomas de los Mamíferos/genética , Metabolómica/métodos , Enfermedad del Hígado Graso no Alcohólico/genética , Octopamina/administración & dosificación , Polimorfismo de Nucleótido Simple , Animales , Dieta Alta en Grasa/efectos adversos , Masculino , Ratones Endogámicos BALB C , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Octopamina/farmacología , Espectroscopía de Protones por Resonancia Magnética , Sitios de Carácter Cuantitativo , Biología de Sistemas , Resultado del Tratamiento
14.
Microbiome ; 6(1): 73, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29678198

RESUMEN

BACKGROUND: The dietary methylamines choline, carnitine, and phosphatidylcholine are used by the gut microbiota to produce a range of metabolites, including trimethylamine (TMA). However, little is known about the use of trimethylamine N-oxide (TMAO) by this consortium of microbes. RESULTS: A feeding study using deuterated TMAO in C57BL6/J mice demonstrated microbial conversion of TMAO to TMA, with uptake of TMA into the bloodstream and its conversion to TMAO. Microbial activity necessary to convert TMAO to TMA was suppressed in antibiotic-treated mice, with deuterated TMAO being taken up directly into the bloodstream. In batch-culture fermentation systems inoculated with human faeces, growth of Enterobacteriaceae was stimulated in the presence of TMAO. Human-derived faecal and caecal bacteria (n = 66 isolates) were screened on solid and liquid media for their ability to use TMAO, with metabolites in spent media analysed by 1H-NMR. As with the in vitro fermentation experiments, TMAO stimulated the growth of Enterobacteriaceae; these bacteria produced most TMA from TMAO. Caecal/small intestinal isolates of Escherichia coli produced more TMA from TMAO than their faecal counterparts. Lactic acid bacteria produced increased amounts of lactate when grown in the presence of TMAO but did not produce large amounts of TMA. Clostridia (sensu stricto), bifidobacteria, and coriobacteria were significantly correlated with TMA production in the mixed fermentation system but did not produce notable quantities of TMA from TMAO in pure culture. CONCLUSIONS: Reduction of TMAO by the gut microbiota (predominantly Enterobacteriaceae) to TMA followed by host uptake of TMA into the bloodstream from the intestine and its conversion back to TMAO by host hepatic enzymes is an example of metabolic retroconversion. TMAO influences microbial metabolism depending on isolation source and taxon of gut bacterium. Correlation of metabolomic and abundance data from mixed microbiota fermentation systems did not give a true picture of which members of the gut microbiota were responsible for converting TMAO to TMA; only by supplementing the study with pure culture work and additional metabolomics was it possible to increase our understanding of TMAO bioconversions by the human gut microbiota.


Asunto(s)
Microbioma Gastrointestinal , Metaboloma , Metabolómica , Metilaminas/metabolismo , Adulto , Animales , Bacterias , Cromatografía Líquida de Alta Presión , Femenino , Fermentación , Humanos , Hibridación Fluorescente in Situ , Espectroscopía de Resonancia Magnética , Masculino , Metabolómica/métodos , Metilaminas/sangre , Ratones , Espectrometría de Masas en Tándem
15.
Diabetes ; 64(9): 3314-20, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25979074

RESUMEN

Hepatocyte nuclear factor-1α (HNF1α) is a transcription factor expressed in tissues of endoderm origin. Mutations in HNF1A are associated with maturity-onset diabetes of the young 3 (MODY3). Mice deficient for Hnf1α are hyperglycemic, with their pancreatic ß-cells being defective in glucose-sensing insulin secretion. The specific mechanisms involved in this defect are unclear. Gut hormones control glucose homeostasis. Our objective was to explore whether changes in these hormones play a role in glucose homeostasis in the absence of Hnf1α. An increase in ghrelin gene transcript and a decrease in glucose-dependent insulinotropic polypeptide (GIP) gene transcripts were observed in the gut of Hnf1α-null mice. These changes correlated with an increase of ghrelin and a decrease of GIP-labeled cells. Ghrelin serological levels were significantly induced in Hnf1α-null mice. Paradoxically, GIP levels were also induced in these mice. Treatment of Hnf1α-null mice with a ghrelin antagonist led to a recovery of the diabetic symptoms. We conclude that upregulation of ghrelin in the absence of Hnf1α impairs insulin secretion and can be reversed by pharmacological inhibition of ghrelin/GHS-R interaction. These observations open up on future strategies to counteract ghrelin action in a program that could become beneficial in controlling non-insulin-dependent diabetes.


Asunto(s)
Glucemia/metabolismo , Polipéptido Inhibidor Gástrico/genética , Mucosa Gástrica/metabolismo , Ghrelina/genética , Factor Nuclear 1-alfa del Hepatocito/genética , Yeyuno/metabolismo , ARN Mensajero/metabolismo , Animales , Glucemia/efectos de los fármacos , Polipéptido Inhibidor Gástrico/metabolismo , Ghrelina/metabolismo , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Homeostasis , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Noqueados , Oligopéptidos/farmacología , Receptores de Ghrelina/antagonistas & inhibidores , Regulación hacia Arriba
16.
PLoS One ; 5(8): e12378, 2010 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-20808783

RESUMEN

BACKGROUND AND AIMS: Although Hnf1alpha is crucial for pancreas and liver functions, it is believed to play a limited functional role for intestinal epithelial functions. The aim of this study was to assess the consequences of abrogating Hnf1alpha on the maintenance of adult small intestinal epithelial functions. METHODOLOGY/PRINCIPAL FINDINGS: An Hnf1alpha knockout mouse model was used. Assessment of histological abnormalities, crypt epithelial cell proliferation, epithelial barrier, glucose transport and signalling pathways were measured in these animals. Changes in global gene expression were also analyzed. Mice lacking Hnf1alpha displayed increased crypt proliferation and intestinalomegaly as well as a disturbance of intestinal epithelial cell lineages production during adult life. This phenotype was associated with a decrease of the mucosal barrier function and lumen-to-blood glucose delivery. The mammalian target of rapamycin (mTOR) signalling pathway was found to be overly activated in the small intestine of adult Hnf1alpha mutant mice. The intestinal epithelium of Hnf1alpha null mice displayed a reduction of the enteroendocrine cell population. An impact was also observed on proper Paneth cell differentiation with abnormalities in the granule exocytosis pathway. CONCLUSIONS/SIGNIFICANCE: Together, these results unravel a functional role for Hnf1alpha in regulating adult intestinal growth and sustaining the functions of intestinal epithelial cell lineages.


Asunto(s)
Diferenciación Celular , Factor Nuclear 1-alfa del Hepatocito/deficiencia , Factor Nuclear 1-alfa del Hepatocito/genética , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Animales , Transporte Biológico/genética , Proliferación Celular , Enterocitos/citología , Enterocitos/metabolismo , Células Enteroendocrinas/citología , Células Enteroendocrinas/metabolismo , Eliminación de Gen , Técnicas de Inactivación de Genes , Glucosa/metabolismo , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Homeostasis/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Células de Paneth/citología , Células de Paneth/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA