Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Endocrinol (Oxf) ; 99(6): 517-524, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37820012

RESUMO

OBJECTIVE: Worldwide, adults and children are at risk of adrenal insufficiency largely due to infectious diseases and adrenal suppression from use of anti-inflammatory glucocorticoids. Home waking salivary cortisone is an accurate screening test for adrenal insufficiency, it has potential to reduce costs, and patients prefer it to the adrenocorticotropin (ACTH) (synacthen) stimulation test. We carried out a service evaluation of home waking salivary cortisone in clinical care to identify implementation barriers. DESIGN, PATIENTS AND MEASUREMENTS: Service evaluation in a centre where 212 patients referred for adrenal insufficiency had a waking salivary cortisone. Problems encountered during testing were recorded and patient feedback, via focus groups, collected. RESULTS: From all patients providing a waking salivary cortisone 55% had a normal test, 23% adrenal suppression, and 22% an equivocal result requiring a clinical centre ACTH stimulation test. The median (interquartile range [IQR]) for the time of the saliva sample was 07:40 (07:00-08:40). The median (IQR) days between collection and (i) delivery to local laboratory was 1 (0.25-2) day; (ii) reporting by local laboratory was 13 (11-18) days. Patients considered the test is "easy to do" and preferred it to the inpatient ACTH stimulation test. The principal challenge to clinical implementation was results reporting to clinicians due to delays at the local laboratory. CONCLUSIONS: This service evaluation provides real-world evidence that home waking salivary cortisone is an effective, practical screening test for adrenal insufficiency. It identified key barriers to testing implementation that need to be addressed when introducing the test to a health service.


Assuntos
Insuficiência Adrenal , Cortisona , Adulto , Criança , Humanos , Hidrocortisona , Saliva , Insuficiência Adrenal/diagnóstico , Hormônio Adrenocorticotrópico
2.
NEJM Evid ; 2(2): EVIDoa2200182, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38320034

RESUMO

At-Home Cortisone Screen for Adrenal InsufficiencyThis prospective study tested the use of home waking salivary cortisone levels to predict adrenal insufficiency. Salivary cortisone had diagnostic accuracy similar to that of a standard adrenocorticotropin stimulation test and was preferred by patients.


Assuntos
Insuficiência Adrenal , Cortisona , Humanos , Hidrocortisona , Estudos Prospectivos , Saliva , Insuficiência Adrenal/diagnóstico
3.
Sci Rep ; 9(1): 3656, 2019 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-30842494

RESUMO

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.


Assuntos
Cromossomos de Mamíferos/genética , Metabolômica/métodos , Hepatopatia Gordurosa não Alcoólica/genética , Octopamina/administração & dosagem , Polimorfismo de Nucleotídeo Único , Animais , Dieta Hiperlipídica/efeitos adversos , Masculino , Camundongos Endogâmicos BALB C , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo , Octopamina/farmacologia , Espectroscopia de Prótons por Ressonância Magnética , Locos de Características Quantitativas , Biologia de Sistemas , Resultado do Tratamento
4.
Cell Rep ; 20(1): 136-148, 2017 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-28683308

RESUMO

The influence of the gut microbiome on metabolic and behavioral traits is widely accepted, though the microbiome-derived metabolites involved remain unclear. We carried out untargeted urine 1H-NMR spectroscopy-based metabolic phenotyping in an isogenic C57BL/6J mouse population (n = 50) and show that microbial-host co-metabolites are prodromal (i.e., early) markers predicting future divergence in metabolic (obesity and glucose homeostasis) and behavioral (anxiety and activity) outcomes with 94%-100% accuracy. Some of these metabolites also modulate disease phenotypes, best illustrated by trimethylamine-N-oxide (TMAO), a product of microbial-host co-metabolism predicting future obesity, impaired glucose tolerance (IGT), and behavior while reducing endoplasmic reticulum stress and lipogenesis in 3T3-L1 adipocytes. Chronic in vivo TMAO treatment limits IGT in HFD-fed mice and isolated pancreatic islets by increasing insulin secretion. We highlight the prodromal potential of microbial metabolites to predict disease outcomes and their potential in shaping mammalian phenotypic heterogeneity.


Assuntos
Ansiedade/microbiologia , Microbioma Gastrointestinal , Intolerância à Glucose/microbiologia , Metaboloma , Obesidade/microbiologia , Fenótipo , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Ansiedade/metabolismo , Biomarcadores/metabolismo , Glicemia/metabolismo , Linhagem Celular , Estresse do Retículo Endoplasmático , Intolerância à Glucose/metabolismo , Interações Hospedeiro-Patógeno , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Lipogênese , Masculino , Metilaminas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Oxidantes/farmacologia
5.
PLoS One ; 8(12): e82825, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24324835

RESUMO

Nutritional factors play important roles in the etiology of obesity, type 2 diabetes mellitus and their complications through genotype x environment interactions. We have characterised molecular adaptation to high fat diet (HFD) feeding in inbred mouse strains widely used in genetic and physiological studies. We carried out physiological tests, plasma lipid assays, obesity measures, liver histology, hepatic lipid measurements and liver genome-wide gene transcription profiling in C57BL/6J and BALB/c mice fed either a control or a high fat diet. The two strains showed marked susceptibility (C57BL/6J) and relative resistance (BALB/c) to HFD-induced insulin resistance and non alcoholic fatty liver disease (NAFLD). Global gene set enrichment analysis (GSEA) of transcriptome data identified consistent patterns of expression of key genes (Srebf1, Stard4, Pnpla2, Ccnd1) and molecular pathways in the two strains, which may underlie homeostatic adaptations to dietary fat. Differential regulation of pathways, including the proteasome, the ubiquitin mediated proteolysis and PPAR signalling in fat fed C57BL/6J and BALB/c suggests that altered expression of underlying diet-responsive genes may be involved in contrasting nutrigenomic predisposition and resistance to insulin resistance and NAFLD in these models. Collectively, these data, which further demonstrate the impact of gene x environment interactions on gene expression regulations, contribute to improved knowledge of natural and pathogenic adaptive genomic regulations and molecular mechanisms associated with genetically determined susceptibility and resistance to metabolic diseases.


Assuntos
Dieta Hiperlipídica , Fígado Gorduroso/etiologia , Fígado Gorduroso/metabolismo , Nutrigenômica , Obesidade/complicações , Obesidade/etiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Adaptação Fisiológica , Animais , Suscetibilidade a Doenças , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Glucose/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Hepatopatia Gordurosa não Alcoólica , Reprodutibilidade dos Testes , Transdução de Sinais , Transcriptoma
6.
J Proteome Res ; 10(4): 1675-89, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21322573

RESUMO

Maintaining homeostasis in higher organisms involves a complex interplay of multiple ubiquitous and organ-specific molecular mechanisms that can be characterized using functional genomics technologies such as transcriptomics, proteomics, and metabonomics and dissected out through genetic investigations in healthy and diseased individuals. We characterized the genomic, metabolic, and physiological divergence of several inbred rat strains--Brown Norway, Lewis, Wistar Kyoto, Fisher (F344)--frequently used as healthy controls in genetic studies of the cardiometabolic syndrome. Hierarchical clustering of (1)H NMR-based metabolic profiles (n = 20 for urine, n = 16 for plasma) identified metabolic phenotype (metabotype) divergence patterns similar to the phylogenetic variability based on single nucleotide polymorphisms. However, the observed urinary metabotype variation exceeded that explainable by genetic polymorphisms. To understand further this natural variation, we used an integrative, knowledge-based network biology metabolic pathway analysis approach, coined Metabolite-Set Enrichment Analysis (MSEA). MSEA reveals that homeostasis and physiological plasticity can be achieved despite widespread divergences in glucose, lipid, amino acid, and energy metabolism in the host, together with different gut microbiota contributions suggestive of strain-specific transgenomic interactions. This work illustrates the concept of natural metabolomic variation, leading to physiologically stable albeit diverse strategies within the range of normality, all of which are highly relevant to animal model physiology, genetical genomics, and patient stratification in personalized healthcare.


Assuntos
Redes e Vias Metabólicas/fisiologia , Metaboloma , Metabolômica/métodos , Ratos/metabolismo , Ratos/fisiologia , Animais , Análise por Conglomerados , Humanos , Masculino , Ressonância Magnética Nuclear Biomolecular , Fenótipo , Ratos Endogâmicos
7.
Diabetes ; 59(5): 1192-201, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20150285

RESUMO

OBJECTIVE: The aim of the study was to elucidate the cellular mechanism underlying the suppression of glucose-induced insulin secretion in mice fed a high-fat diet (HFD) for 15 weeks. RESEARCH DESIGN AND METHODS: C57BL6J mice were fed a HFD or a normal diet (ND) for 3 or 15 weeks. Plasma insulin and glucose levels in vivo were assessed by intraperitoneal glucose tolerance test. Insulin secretion in vitro was studied using static incubations and a perfused pancreas preparation. Membrane currents, electrical activity, and exocytosis were examined by patch-clamp technique measurements. Intracellular calcium concentration ([Ca(2+)](i)) was measured by microfluorimetry. Total internal reflection fluorescence microscope (TIRFM) was used for optical imaging of exocytosis and submembrane depolarization-evoked [Ca(2+)](i). The functional data were complemented by analyses of histology and gene transcription. RESULTS: After 15 weeks, but not 3 weeks, mice on HFD exhibited hyperglycemia and hypoinsulinemia. Pancreatic islet content and beta-cell area increased 2- and 1.5-fold, respectively. These changes correlated with a 20-50% reduction of glucose-induced insulin secretion (normalized to insulin content). The latter effect was not associated with impaired electrical activity or [Ca(2+)](i) signaling. Single-cell capacitance and TIRFM measurements of exocytosis revealed a selective suppression (>70%) of exocytosis elicited by short (50 ms) depolarization, whereas the responses to longer depolarizations were (500 ms) less affected. The loss of rapid exocytosis correlated with dispersion of Ca(2+) entry in HFD beta-cells. No changes in gene transcription of key exocytotic protein were observed. CONCLUSIONS: HFD results in reduced insulin secretion by causing the functional dissociation of voltage-gated Ca(2+) entry from exocytosis. These observations suggest a novel explanation to the well-established link between obesity and diabetes.


Assuntos
Canais de Cálcio/metabolismo , Diabetes Mellitus Tipo 2/induzido quimicamente , Diabetes Mellitus Tipo 2/fisiopatologia , Gorduras na Dieta/efeitos adversos , Vesículas Secretórias/metabolismo , Animais , Diabetes Mellitus Tipo 2/metabolismo , Exocitose/fisiologia , Citometria de Fluxo , Intolerância à Glucose/metabolismo , Imuno-Histoquímica , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência
8.
PLoS One ; 3(2): e1668, 2008 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-18301746

RESUMO

Insulin resistance plays a central role in type 2 diabetes and obesity, which develop as a consequence of genetic and environmental factors. Dietary changes including high fat diet (HFD) feeding promotes insulin resistance in rodent models which present useful systems for studying interactions between genetic background and environmental influences contributing to disease susceptibility and progression. We applied a combination of classical physiological, biochemical and hormonal studies and plasma (1)H NMR spectroscopy-based metabonomics to characterize the phenotypic and metabotypic consequences of HFD (40%) feeding in inbred mouse strains (C57BL/6, 129S6, BALB/c, DBA/2, C3H) frequently used in genetic studies. We showed the wide range of phenotypic and metabonomic adaptations to HFD across the five strains and the increased nutrigenomic predisposition of 129S6 and C57BL/6 to insulin resistance and obesity relative to the other strains. In contrast mice of the BALB/c and DBA/2 strains showed relative resistance to HFD-induced glucose intolerance and obesity. Hierarchical metabonomic clustering derived from (1)H NMR spectral data of the strains provided a phylometabonomic classification of strain-specific metabolic features and differential responses to HFD which closely match SNP-based phylogenetic relationships between strains. Our results support the concept of genomic clustering of functionally related genes and provide important information for defining biological markers predicting spontaneous susceptibility to insulin resistance and pathological adaptations to fat feeding.


Assuntos
Adaptação Fisiológica , Gorduras na Dieta/administração & dosagem , Metabolismo , Filogenia , Animais , Gorduras na Dieta/metabolismo , Resistência à Insulina , Espectroscopia de Ressonância Magnética/métodos , Camundongos , Camundongos Endogâmicos , Obesidade , Fenótipo , Especificidade da Espécie
9.
Nat Genet ; 39(5): 666-72, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17435758

RESUMO

Characterizing the relationships between genomic and phenotypic variation is essential to understanding disease etiology. Information-dense data sets derived from pathophysiological, proteomic and transcriptomic profiling have been applied to map quantitative trait loci (QTLs). Metabolic traits, already used in QTL studies in plants, are essential phenotypes in mammalian genetics to define disease biomarkers. Using a complex mammalian system, here we show chromosomal mapping of untargeted plasma metabolic fingerprints derived from NMR spectroscopic analysis in a cross between diabetic and control rats. We propose candidate metabolites for the most significant QTLs. Metabolite profiling in congenic strains provided evidence of QTL replication. Linkage to a gut microbial metabolite (benzoate) can be explained by deletion of a uridine diphosphate glucuronosyltransferase. Mapping metabotypic QTLs provides a practical approach to understanding genome-phenotype relationships in mammals and may uncover deeper biological complexity, as extended genome (microbiome) perturbations that affect disease processes through transgenomic effects may influence QTL detection.


Assuntos
Diabetes Mellitus/genética , Ligação Genética , Genoma/genética , Metabolismo/genética , Fenótipo , Locos de Características Quantitativas , Animais , Sequência de Bases , Benzoatos/química , Biomarcadores/análise , Glucuronosiltransferase/genética , Escore Lod , Dados de Sequência Molecular , Estrutura Molecular , Ressonância Magnética Nuclear Biomolecular , Ratos , Análise de Sequência de DNA
10.
Proc Natl Acad Sci U S A ; 103(33): 12511-6, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16895997

RESUMO

Here, we study the intricate relationship between gut microbiota and host cometabolic phenotypes associated with dietary-induced impaired glucose homeostasis and nonalcoholic fatty liver disease (NAFLD) in a mouse strain (129S6) known to be susceptible to these disease traits, using plasma and urine metabotyping, achieved by (1)H NMR spectroscopy. Multivariate statistical modeling of the spectra shows that the genetic predisposition of the 129S6 mouse to impaired glucose homeostasis and NAFLD is associated with disruptions of choline metabolism, i.e., low circulating levels of plasma phosphatidylcholine and high urinary excretion of methylamines (dimethylamine, trimethylamine, and trimethylamine-N-oxide), coprocessed by symbiotic gut microbiota and mammalian enzyme systems. Conversion of choline into methylamines by microbiota in strain 129S6 on a high-fat diet reduces the bioavailability of choline and mimics the effect of choline-deficient diets, causing NAFLD. These data also indicate that gut microbiota may play an active role in the development of insulin resistance.


Assuntos
Fígado Gorduroso/fisiopatologia , Trato Gastrointestinal/microbiologia , Resistência à Insulina/fisiologia , Animais , Peso Corporal , Gorduras na Dieta , Glucose/metabolismo , Homeostase , Insulina/metabolismo , Lipídeos/sangue , Fígado/anatomia & histologia , Fígado/metabolismo , Masculino , Metilaminas/sangue , Metilaminas/urina , Camundongos , Camundongos Endogâmicos BALB C , Análise Multivariada , Ressonância Magnética Nuclear Biomolecular , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...