Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Cell Metab ; 35(4): 601-619.e10, 2023 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-36977414

RESUMO

Adipose tissue modulates energy homeostasis by secreting leptin, but little is known about the factors governing leptin production. We show that succinate, long perceived as a mediator of immune response and lipolysis, controls leptin expression via its receptor SUCNR1. Adipocyte-specific deletion of Sucnr1 influences metabolic health according to nutritional status. Adipocyte Sucnr1 deficiency impairs leptin response to feeding, whereas oral succinate mimics nutrient-related leptin dynamics via SUCNR1. SUCNR1 activation controls leptin expression via the circadian clock in an AMPK/JNK-C/EBPα-dependent manner. Although the anti-lipolytic role of SUCNR1 prevails in obesity, its function as a regulator of leptin signaling contributes to the metabolically favorable phenotype in adipocyte-specific Sucnr1 knockout mice under standard dietary conditions. Obesity-associated hyperleptinemia in humans is linked to SUCNR1 overexpression in adipocytes, which emerges as the major predictor of adipose tissue leptin expression. Our study establishes the succinate/SUCNR1 axis as a metabolite-sensing pathway mediating nutrient-related leptin dynamics to control whole-body homeostasis.


Assuntos
Relógios Circadianos , Leptina , Animais , Humanos , Camundongos , Adipócitos/metabolismo , Metabolismo Energético/fisiologia , Leptina/metabolismo , Camundongos Knockout , Obesidade/metabolismo , Succinatos/metabolismo
2.
Cell Rep ; 38(2): 110241, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35021074

RESUMO

Metabolic homeostasis is under circadian regulation to adapt energy requirements to light-dark cycles. Feeding cycles are regulated by photic stimuli reaching the suprachiasmatic nucleus via retinohypothalamic axons and by nutritional information involving dopaminergic neurotransmission. Previously, we reported that Pitx3-mutant Aphakia mice with altered development of the retinohypothalamic tract and the dopaminergic neurons projecting to the striatum, are resistant to locomotor and metabolic entrainment by time-restricted feeding. In their Matters Arising article, Scarpa et al. (2022) challenge this conclusion using mice from the same strain but following a different experimental paradigm involving calorie restriction. Here, we address their concerns by extending the analyses of our previous data, by identifying important differences in the experimental design between both studies and by presenting additional results on the dopaminergic deficit in the brain of Aphakia mice. This Matters Arising Response article addresses the Matters Arising article by Scarpa et al. (2022), published concurrently in Cell Reports.


Assuntos
Afacia , Núcleo Supraquiasmático , Animais , Dopamina , Metabolismo Energético , Camundongos , Fotoperíodo
3.
Diabetologia ; 65(3): 490-505, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34932133

RESUMO

AIMS/HYPOTHESIS: Second-generation antipsychotic (SGA) drugs have been associated with the development of type 2 diabetes and the metabolic syndrome in patients with schizophrenia. In this study, we aimed to investigate the effects of two different SGA drugs, olanzapine and aripiprazole, on metabolic state and islet function and plasticity. METHODS: We analysed the functional adaptation of beta cells in 12-week-old B6;129 female mice fed an olanzapine- or aripiprazole-supplemented diet (5.5-6.0 mg kg-1 day-1) for 6 months. Glucose and insulin tolerance tests, in vivo glucose-stimulated insulin secretion and indirect calorimetry were performed at the end of the study. The effects of SGAs on beta cell plasticity and islet serotonin levels were assessed by transcriptomic analysis and immunofluorescence. Insulin secretion was assessed by static incubations and Ca2+ fluxes by imaging techniques. RESULTS: Treatment of female mice with olanzapine or aripiprazole for 6 months induced weight gain (p<0.01 and p<0.05, respectively), glucose intolerance (p<0.01) and impaired insulin secretion (p<0.05) vs mice fed a control chow diet. Aripiprazole, but not olanzapine, induced serotonin production in beta cells vs controls, likely by increasing tryptophan hydroxylase 1 (TPH1) expression, and inhibited Ca2+ flux. Of note, aripiprazole increased beta cell size (p<0.05) and mass (p<0.01) vs mice fed a control chow diet, along with activation of mechanistic target of rapamycin complex 1 (mTORC1)/S6 signalling, without preventing beta cell dysfunction. CONCLUSIONS/INTERPRETATION: Both SGAs induced weight gain and beta cell dysfunction, leading to glucose intolerance; however, aripiprazole had a more potent effect in terms of metabolic alterations, which was likely a result of its ability to modulate the serotonergic system. The deleterious metabolic effects of SGAs on islet function should be considered while treating patients as these drugs may increase the risk for development of the metabolic syndrome and diabetes.


Assuntos
Antipsicóticos , Diabetes Mellitus Tipo 2 , Ilhotas Pancreáticas , Animais , Antipsicóticos/efeitos adversos , Aripiprazol/metabolismo , Aripiprazol/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Humanos , Ilhotas Pancreáticas/metabolismo , Camundongos , Olanzapina/efeitos adversos , Olanzapina/metabolismo
4.
Mol Metab ; 53: 101251, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34015524

RESUMO

OBJECTIVE: Pancreatic ß-cell dysfunction is a central feature in the pathogenesis of type 2 diabetes (T2D). Accumulating evidence indicates that ß-site APP-cleaving enzyme 2 (BACE2) inhibition exerts a beneficial effect on ß-cells in different models of T2D. Thus, targeting BACE2 may represent a potential therapeutic strategy for the treatment of this disease. Here, we aimed to investigate the effects of BACE2 suppression on glucose homeostasis in a model of diet-induced obesity. METHODS: BACE2 knock-out (BKO) and wild-type (WT) mice were fed with a high-fat diet (HFD) for 2 or 16 weeks. Body weight, food intake, respiratory exchange ratio, locomotor activity, and energy expenditure were determined. Glucose homeostasis was evaluated by glucose and insulin tolerance tests. ß-cell proliferation was assessed by Ki67-positive nuclei, and ß-cell function was determined by measuring glucose-stimulated insulin secretion. Leptin sensitivity was evaluated by quantifying food intake and body weight after an intraperitoneal leptin injection. Neuropeptide gene expression and insulin signaling in the mediobasal hypothalamus were determined by qPCR and Akt phosphorylation, respectively. RESULTS: After 16 weeks of HFD feeding, BKO mice exhibited an exacerbated body weight gain and hyperphagia, in comparison to WT littermates. Glucose tolerance was similar in both groups, whereas HFD-induced hyperinsulinemia, insulin resistance, and ß-cell expansion were more pronounced in BKO mice. In turn, leptin-induced food intake inhibition and hypothalamic insulin signaling were impaired in BKO mice, regardless of the diet, in accordance with deregulation of the expression of hypothalamic neuropeptide genes. Importantly, BKO mice already showed increased ß-cell proliferation and glucose-stimulated insulin secretion with respect to WT littermates after two weeks of HFD feeding, before the onset of obesity. CONCLUSIONS: Collectively, these results reveal that BACE2 suppression in an obesogenic setting leads to exacerbated body weight gain, hyperinsulinemia, and insulin resistance. Thus, we conclude that inhibition of BACE2 may aggravate the adverse metabolic effects associated with obesity.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Obesidade/metabolismo , Animais , Dieta/efeitos adversos , Masculino , Camundongos , Camundongos Transgênicos
5.
Sci Transl Med ; 13(587)2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33790021

RESUMO

Accelerated postnatal growth is a potentially modifiable risk factor for future obesity. To study how specific breast milk components contribute to early growth and obesity risk, we quantified one-carbon metabolism-related metabolites in human breast milk and found an inverse association between milk betaine content and infant growth. This association was replicated in an independent and geographically distinct cohort. To determine the potential role of milk betaine in modulating offspring obesity risk, we performed maternal betaine supplementation experiments in mice. Higher betaine intake during lactation increased milk betaine content in dams and led to lower adiposity and improved glucose homeostasis throughout adulthood in mouse offspring. These effects were accompanied by a transient increase in Akkermansia spp. abundance in the gut during early life and a long-lasting increase in intestinal goblet cell number. The link between breast milk betaine and Akkermansia abundance in the gut was also observed in humans, as infants exposed to higher milk betaine content during breastfeeding showed higher fecal Akkermansia muciniphila abundance. Furthermore, administration of A. muciniphila to mouse pups during the lactation period partially replicated the effects of maternal breast milk betaine, including increased intestinal goblet cell number, lower adiposity, and improved glucose homeostasis during adulthood. These data demonstrate a link between breast milk betaine content and long-term metabolic health of offspring.


Assuntos
Betaína , Leite Humano , Akkermansia , Animais , Dieta Hiperlipídica , Feminino , Lactação , Camundongos
6.
BMC Biol ; 19(1): 40, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33658023

RESUMO

BACKGROUND: Insulin secretion from the pancreatic ß-cell is finely modulated by different signals to allow an adequate control of glucose homeostasis. Incretin hormones such as glucagon-like peptide-1 (GLP-1) act as key physiological potentiators of insulin release through binding to the G protein-coupled receptor GLP-1R. Another key regulator of insulin signaling is the Ser/Thr kinase G protein-coupled receptor kinase 2 (GRK2). However, whether GRK2 affects insulin secretion or if GRK2 can control incretin actions in vivo remains to be analyzed. RESULTS: Using GRK2 hemizygous mice, isolated pancreatic islets, and model ß-cell lines, we have uncovered a relevant physiological role for GRK2 as a regulator of incretin-mediated insulin secretion in vivo. Feeding, oral glucose gavage, or administration of GLP-1R agonists in animals with reduced GRK2 levels (GRK2+/- mice) resulted in enhanced early phase insulin release without affecting late phase secretion. In contrast, intraperitoneal glucose-induced insulin release was not affected. This effect was recapitulated in isolated islets and correlated with the increased size or priming efficacy of the readily releasable pool (RRP) of insulin granules that was observed in GRK2+/- mice. Using nanoBRET in ß-cell lines, we found that stimulation of GLP-1R promoted GRK2 association to this receptor and that GRK2 protein and kinase activity were required for subsequent ß-arrestin recruitment. CONCLUSIONS: Overall, our data suggest that GRK2 is an important negative modulator of GLP-1R-mediated insulin secretion and that GRK2-interfering strategies may favor ß-cell insulin secretion specifically during the early phase, an effect that may carry interesting therapeutic applications.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/genética , Regulação da Expressão Gênica , Receptor do Peptídeo Semelhante ao Glucagon 1/genética , Secreção de Insulina/genética , Animais , Linhagem Celular , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos
7.
Mol Metab ; 45: 101162, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33422644

RESUMO

Childhood obesity is a strong risk factor for adult obesity, type 2 diabetes, and cardiovascular disease. The mechanisms that link early adiposity with late-onset chronic diseases are poorly characterised. We developed a mouse model of early adiposity through litter size reduction. Mice reared in small litters (SLs) developed obesity, insulin resistance, and hepatic steatosis during adulthood. The liver played a major role in the development of the disease. OBJECTIVE: To gain insight into the molecular mechanisms that link early development and childhood obesity with adult hepatic steatosis and insulin resistance. METHODS: We analysed the hepatic transcriptome (Affymetrix) of control and SL mice to uncover potential pathways involved in the long-term programming of disease in our model. RESULTS: The circadian rhythm was the most significantly deregulated Gene Ontology term in the liver of adult SL mice. Several core clock genes, such as period 1-3 and cryptochrome 1-2, were altered in two-week-old SL mice and remained altered throughout their life course until they reached 4-6 months of age. Defective circadian rhythm was restricted to the periphery since the expression of clock genes in the hypothalamus, the central pacemaker, was normal. The period-cryptochrome genes were primarily entrained by dietary signals. Hence, restricting food availability during the light cycle only uncoupled the central rhythm from the peripheral and completely normalised hepatic triglyceride content in adult SL mice. This effect was accompanied by better re-alignment of the hepatic period genes, suggesting that they might have played a causal role in mediating hepatic steatosis in the adult SL mice. Functional downregulation of Per2 in hepatocytes in vitro confirmed that the period genes regulated lipid-related genes in part through peroxisome proliferator-activated receptor alpha (Ppara). CONCLUSIONS: The hepatic circadian rhythm matures during early development, from birth to postnatal day 30. Hence, nutritional challenges during early life may misalign the hepatic circadian rhythm and secondarily lead to metabolic derangements. Specific time-restricted feeding interventions improve metabolic health in the context of childhood obesity by partially re-aligning the peripheral circadian rhythm.


Assuntos
Ritmo Circadiano/fisiologia , Lactação , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Adiposidade , Adulto , Animais , Ritmo Circadiano/genética , Diabetes Mellitus Tipo 2/metabolismo , Jejum , Feminino , Humanos , Hipotálamo/metabolismo , Recém-Nascido , Resistência à Insulina/fisiologia , Doenças Metabólicas/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Hepatopatia Gordurosa não Alcoólica/genética , Obesidade/metabolismo , Obesidade Infantil
8.
Front Genet ; 11: 583932, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33173540

RESUMO

Animal models are invaluable for biomedical research, especially in the context of rare diseases, which have a very low prevalence and are often complex. Concretely mouse models provide key information on rare disease mechanisms and therapeutic strategies that cannot be obtained by using only alternative methods, and greatly contribute to accelerate the development of new therapeutic options for rare diseases. Despite this, the use of experimental animals remains controversial. The combination of respectful management, ethical laws and transparency regarding animal experimentation contributes to improve society's opinion about biomedical research and positively impacts on research quality, which eventually also benefits patients. Here we present examples of current advances in preclinical research in rare diseases using mouse models, together with our perspective on future directions and challenges.

9.
Proc Natl Acad Sci U S A ; 117(48): 30335-30343, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33199621

RESUMO

Implementation of regular physical activity helps in the maintenance of a healthy metabolic profile both in humans and mice through molecular mechanisms not yet completely defined. Here, we show that high-intensity interval training (HIIT) modifies the microRNA (miRNA) profile of circulating exosomes in mice, including significant increases in miR-133a and miR-133b Importantly, treatment of sedentary mice with exosomes isolated from the plasma of trained mice improves glucose tolerance, insulin sensitivity, and decreases plasma levels of triglycerides. Moreover, exosomes isolated from the muscle of trained mice display similar changes in miRNA content, and their administration to sedentary mice reproduces the improvement of glucose tolerance. Exosomal miRNAs up-regulated by HIIT target insulin-regulated transcription factor forkhead box O1 (FoxO1) and, accordingly, expression of FoxO1 is decreased in the liver of trained and exosome-treated mice. Treatment with exosomes transfected with a miR-133b mimic or with a specific siRNA targeting FoxO1 recapitulates the metabolic effects observed in trained mice. Overall, our data suggest that circulating exosomes released by the muscle carry a specific miRNA signature that is modified by exercise and induce expression changes in the liver that impact whole-body metabolic profile.


Assuntos
Regulação para Baixo/genética , Exossomos/metabolismo , Proteína Forkhead Box O1/genética , Treinamento Intervalado de Alta Intensidade , Resistência à Insulina , Fígado/metabolismo , MicroRNAs/metabolismo , Músculos/metabolismo , Animais , Exossomos/ultraestrutura , Proteína Forkhead Box O1/metabolismo , Gluconeogênese , Glucose/metabolismo , Metabolismo dos Lipídeos , Masculino , Metabolômica , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Condicionamento Físico Animal
10.
Mov Disord ; 35(9): 1636-1648, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32666590

RESUMO

BACKGROUND: Numerous studies indicate an association between neurodegenerative and metabolic diseases. Although still a matter of debate, growing evidence from epidemiological and animal studies indicate that preexisting diabetes increases the risk to develop Parkinson's disease. However, the mechanisms of such an association are unknown. OBJECTIVES: We investigated whether diabetes alters striatal dopamine neurotransmission and assessed the vulnerability of nigrostriatal neurons to neurodegeneration. METHODS: We used streptozotocin-treated and genetically diabetic db/db mice. Expression of oxidative stress and nigrostriatal neuronal markers and levels of dopamine and its metabolites were monitored. Dopamine release and uptake were assessed using fast-scan cyclic voltammetry. 6-Hydroxydopamine was unilaterally injected into the striatum using stereotaxic surgery. Motor performance was scored using specific tests. RESULTS: Diabetes resulted in oxidative stress and decreased levels of dopamine and its metabolites in the striatum. Levels of proteins regulating dopamine release and uptake, including the dopamine transporter, the Girk2 potassium channel, the vesicular monoamine transporter 2, and the presynaptic vesicle protein synaptobrevin-2, were decreased in diabetic mice. Electrically evoked levels of extracellular dopamine in the striatum were enhanced, and altered dopamine uptake was observed. Striatal microinjections of a subthreshold dose of the neurotoxin 6-hydroxydopamine in diabetic mice, insufficient to cause motor alterations in nondiabetic animals, resulted in motor impairment, higher loss of striatal dopaminergic axons, and decreased neuronal cell bodies in the substantia nigra. CONCLUSIONS: Our results indicate that diabetes promotes striatal oxidative stress, alters dopamine neurotransmission, and increases vulnerability to neurodegenerative damage leading to motor impairment. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Assuntos
Diabetes Mellitus Experimental , Dopamina , Animais , Corpo Estriado/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Camundongos , Substância Negra/metabolismo , Transmissão Sináptica
11.
Bioinformatics ; 36(11): 3618-3619, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32108859

RESUMO

SUMMARY: Mass spectrometry imaging (MSI) can reveal biochemical information directly from a tissue section. MSI generates a large quantity of complex spectral data which is still challenging to translate into relevant biochemical information. Here, we present rMSIproc, an open-source R package that implements a full data processing workflow for MSI experiments performed using TOF or FT-based mass spectrometers. The package provides a novel strategy for spectral alignment and recalibration, which allows to process multiple datasets simultaneously. This enables to perform a confident statistical analysis with multiple datasets from one or several experiments. rMSIproc is designed to work with files larger than the computer memory capacity and the algorithms are implemented using a multi-threading strategy. rMSIproc is a powerful tool able to take full advantage of modern computer systems to completely develop the whole MSI potential. AVAILABILITY AND IMPLEMENTATION: rMSIproc is freely available at https://github.com/prafols/rMSIproc. CONTACT: pere.rafols@urv.cat. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Algoritmos , Software , Sistemas Computacionais , Espectrometria de Massas , Fluxo de Trabalho
12.
Cell Rep ; 29(11): 3678-3692.e4, 2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31825844

RESUMO

Daily adaptation of metabolic activity to light-dark cycles to maintain homeostasis is controlled by hypothalamic nuclei receiving information from the retina and from nutritional inputs that vary according to feeding cycles. We show that selective hypomorphic expression of the transcription factor gene Pitx3 prevents light-dependent entrainment of the central pacemaker in the suprachiasmatic nucleus. This translates into altered behavioral and metabolic outputs affecting locomotor activity, feeding patterns, energy expenditure, and corticosterone secretion that correlate with dysfunctional expression of clock genes in the ventromedial hypothalamus, liver, and brown adipose tissue. Metabolic entrainment by time-restricted feeding restores clock function in the liver and brown adipose tissue but not in the ventromedial hypothalamus and, remarkably, fails to synchronize energy expenditure and locomotor and hormonal outputs. Thus, our study reveals a central role of the priming of the suprachiasmatic nucleus with retinal innervation in the hypothalamic regulation of cyclic metabolic homeostasis.


Assuntos
Relógios Circadianos , Metabolismo Energético , Proteínas de Homeodomínio/genética , Núcleo Supraquiasmático/metabolismo , Fatores de Transcrição/genética , Tecido Adiposo/metabolismo , Animais , Corticosterona/metabolismo , Comportamento Alimentar , Proteínas de Homeodomínio/metabolismo , Hipotálamo/metabolismo , Fígado/metabolismo , Locomoção , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição/metabolismo
13.
Front Neurosci ; 13: 921, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31551685

RESUMO

The hypothalamus is the principal regulator of global energy balance, enclosing additionally essential neuronal centers for glucose-sensing and osmoregulation. Disturbances in these tightly regulated neuronal networks are thought to underlie the development of severe pandemic syndromes, including obesity and diabetes. In this work, we investigate in vivo the response of individual hypothalamic nuclei to the i.p. administration of glucose or vehicle solutions, using two groups of adult male C57BL6/J fasted mice and a combination of non-invasive T2 ∗-weighted and diffusion-weighted functional magnetic resonance imaging (fMRI) approaches. MRI parameters were assessed in both groups of animals before, during and in a post-stimulus phase, following the administration of glucose or vehicle solutions. Hypothalamic nuclei depicted different patterns of activation characterized by: (i) generalized glucose-induced increases of neuronal activation and perfusion-markers in the lateral hypothalamus, arcuate and dorsomedial nuclei, (ii) cellular shrinking events and decreases in microvascular blood flow in the dorsomedial, ventromedial and lateral hypothalamus, following the administration of vehicle solutions and (iii) increased neuronal activity markers and decreased microperfusion parameters in the ARC nuclei of vehicle-administered animals. Immunohistochemical studies performed after the post-stimulus phase confirmed the presence of c-Fos immunoreactive neurons in the arcuate nucleus (ARC) from both animal groups, with significantly higher numbers in the glucose-treated animals. Together, our results reveal that fMRI methods are able to detect in vivo diverse patterns of glucose or vehicle-induced effects in the different hypothalamic nuclei.

14.
Arch. argent. pediatr ; 117(4): 245-251, ago. 2019. tab, graf
Artigo em Inglês, Espanhol | BINACIS, LILACS | ID: biblio-1054928

RESUMO

Introducción. Debido a la fuerte industrialización de la Ciudad de Buenos Aires y alrededores, la población podría estar expuesta a metales. Para poder evaluar el nivel de exposición de los niños al cromo y al mercurio, es fundamental tener valores de referencia (VR) propios. El objetivo fue determinar los VR pediátricos para cromo y mercurio en la muestra aislada de orina. Población y métodos: Se incluyeron niños y niñas no expuestos a los contaminantes evaluados que concurrieron al Servicio de Bajo Riesgo y al Consultorio del Jardín Maternal del Hospital de Pediatría S.A.M.I.C. "Prof. Dr. Juan P. Garrahan". Se cuantificó cromo (UCr), mercurio (UHg) y creatinina urinarios. Se calcularon los p95 con su intervalo de confianza del 95 % [IC 95 %] según el concepto para VR de la German Human Biomonitoring Commission. Resultados: Se incluyeron 160 pacientes en el estudio. Se obtuvieron 144 muestras de niños y niñas de entre 1 y 17 años (mediana: 7 años). Se cuantificó UCr a 137 muestras y UHg a 129. La mediana y rango de cromo fue 0,54 (indetectable -3,06) µg/g de creatinina y la de mercurio fue 0,49 (indetectable -7,57) µg/g de creatinina.Conclusiones: Los VR fueron, para UCr, hasta 1,5 µg/l [1,2-2,8] y hasta 2,2 µg/g de creatinina [1,8-3,0] y para UHg, hasta 2,5 µg/l [1,8-4,8] y 3,2 µg/g de creatinina [2,5-4,7


Introduction. Due to the heavy industrialization of the Autonomous City of Buenos Aires and Greater Buenos Aires, the population may have become exposed to metals.To assess the level of exposure to chromium and mercury in children, it is critical to have local reference values (RVs). Our objective was to determine pediatric RVs for chromium and mercury in a single urine sample.Population and methods: Children who were not exposed to the studied contaminants and who attended the Department of Low Risk Conditions and the Daycare Center Office of Hospital de Pediatría S.A.M.I.C. "Prof. Dr. Juan P. Garrahan" were included. Urinary chromium (UCr), urinary mercury (UHg), and urinary creatinine were measured. The p95 and its corresponding 95 % confidence interval (CI) were estimated based on the RV concept proposed by the German Human Biomonitoring Commission.Results: The study included 160 patients. A total of 144 samples from children aged 1-17 years (median: 7 years) were collected. UCr was measured in 137 samples and UHg, in 129 samples. The median value of chromium was 0.54 µg/g of creatinine (range, undetectable to 3.06), while that of mercury was 0.49 µg/g of creatinine (range, undetectable to 7.57). Conclusions: The RVs for UCr were up to 1.5 µg/L [1.2-2.8] and up to 2.2 µg/g of creatinine [1.8-3.0], and for UHg, up to 2.5 µg/L [1.8-4.8] and 3.2 µg/g of creatinine [2.5-4.7]


Assuntos
Humanos , Masculino , Feminino , Recém-Nascido , Lactente , Pré-Escolar , Criança , Adolescente , Valores de Referência , Cromo/urina , Mercúrio/urina , População Urbana , Urina , Exposição Ambiental/análise
15.
Arch Argent Pediatr ; 117(4): 245-251, 2019 08 01.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-31339267

RESUMO

INTRODUCTION: Due to the heavy industrialization of the Autonomous City of Buenos Aires and Greater Buenos Aires, the population may have become exposed to metals. To assess the level of exposure to chromium and mercury in children, it is critical to have local reference values (RVs). Our objective was to determine pediatric RV s for chromium and mercury in a single urine sample. POPULATION AND METHODS: Children who were not exposed to the studied contaminants and who attended the Department of Low Risk Conditions and the Daycare Center Office of Hospital de Pediatría S.A.M.I.C. "Prof. Dr. Juan P. Garrahan" were included. Urinary chromium (UCr), urinary mercury (UHg), and urinary creatinine were measured. The p95 and its corresponding 95 % confidence interval (CI) were estimated based on the RV concept proposed by the German Human Biomonitoring Commission. RESULTS: The study included 160 patients. A total of 144 samples from children aged 1-17 years (median: 7 years) were collected. UCr was measured in 137 samples and UHg, in 129 samples. The median value of chromium was 0.54 µg/g of creatinine (range, undetectable to 3.06), while that of mercury was 0.49 µg/g of creatinine (range, undetectable to 7.57). CONCLUSIONS: The RVs for UCr were up to 1.5 µg/L [1.2-2.8] and up to 2.2 µg/g of creatinine [1.8-3.0], and for UHg, up to 2.5 µg/L [1.8-4.8] and 3.2 µg/g of creatinine [2.5-4.7].


Introducción. Debido a la fuerte industrialización de la Ciudad de Buenos Aires y alrededores, la población podría estar expuesta a metales. Para poder evaluar el nivel de exposición de los niños al cromo y al mercurio, es fundamental tener valores de referencia (VR) propios. El objetivo fue determinar los VR pediátricos para cromo y mercurio en la muestra aislada de orina. Población y métodos: Se incluyeron niños y niñas no expuestos a los contaminantes evaluados que concurrieron al Servicio de Bajo Riesgo y al Consultorio del Jardín Maternal del Hospital de Pediatría S. A. M. I. C. "Prof. Dr. Juan P. Garrahan". Se cuantificó cromo (UCr), mercurio (UHg) y creatinina urinarios. Se calcularon los p95 con su intervalo de confianza del 95 % [IC 95 %] según el concepto para VR de la German Human Biomonitoring Commission. Resultados: Se incluyeron 160 pacientes en el estudio. Se obtuvieron 144 muestras de niños y niñas de entre 1 y 17 años (mediana: 7 años). Se cuantificó UCr a 137 muestras y UHg a 129. La mediana y rango de cromo fue 0,54 (indetectable -3,06) µg/g de creatinina y la de mercurio fue 0,49 (indetectable -7,57) µg/g de creatinina. Conclusiones: Los VR fueron, para UCr, hasta 1,5 µg/l [1,2-2,8] y hasta 2,2 µg/g de creatinina [1,8-3,0] y para UHg, hasta 2,5 µg/l [1,8-4,8] y 3,2 µg/g de creatinina [2,5-4,7].


Assuntos
Cromo/urina , Mercúrio/urina , Adolescente , Argentina , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Valores de Referência , Saúde da População Urbana
16.
FASEB J ; : fj201700717RR, 2018 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-29812971

RESUMO

Postnatal overfeeding increases the risk of chronic diseases later in life, including obesity, insulin resistance, hepatic steatosis, and type 2 diabetes. Epigenetic mechanisms might underlie the long-lasting effects associated with early nutrition. Here we aimed to explore the molecular pathways involved in early development of insulin resistance and hepatic steatosis, and we examined the potential contribution of DNA methylation and histone modifications to long-term programming of metabolic disease. We used a well-characterized mouse model of neonatal overfeeding and early adiposity by litter size reduction. Neonatal overfeeding led to hepatic insulin resistance very early in life that persisted throughout adulthood despite normalizing food intake. Up-regulation of monoacylglycerol O-acyltransferase ( Mogat) 1 conceivably mediates hepatic steatosis and insulin resistance through increasing intracellular diacylglycerol content. Early and sustained deregulation of Mogat1 was associated with a combination of histone modifications that might favor Mogat1 expression. In sum, postnatal overfeeding causes extremely rapid derangements of hepatic insulin sensitivity that remain relatively stable until adulthood. Epigenetic mechanisms, particularly histone modifications, could contribute to such long-lasting effects. Our data suggest that targeting hepatic monoacylglycerol acyltransferase activity during early life might provide a novel strategy to improve hepatic insulin sensitivity and prevent late-onset insulin resistance and fatty liver disease.-Ramon-Krauel, M., Pentinat, T., Bloks, V. W., Cebrià, J., Ribo, S., Pérez-Wienese, R., Vilà, M., Palacios-Marin, I., Fernández-Pérez, A., Vallejo, M., Téllez, N., Rodríguez, M. À., Yanes, O., Lerin, C., Díaz, R., Plosch, T., Tietge, U. J. F., Jimenez-Chillaron, J. C. Epigenetic programming at the Mogat1 locus may link neonatal overnutrition with long-term hepatic steatosis and insulin resistance.

17.
PLoS One ; 12(3): e0173675, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28319140

RESUMO

The aim of the present work was to further explore the physiological roles of muscle-derived IL-6. Adult-floxed and conditional skeletal muscle IL-6 knock out male and female mice were used to study energy expenditure (indirect calorimetry at rest and during treadmill exercise, and body temperature cycle during the light phase) and energy intake (response to fast/refeeding). We also evaluated the responses to leptin and the activity of the insulin signalling pathway in skeletal muscle and liver by phosphorylation of Akt at Ser 473. The stress response was also studied. Results indicate a relevant role of muscle IL-6 in maintaining energy homeostasis, especially in males. Absence of muscle IL-6 in male mice results in lower core body temperature in the light phase, increased respiratory exchange ratio (RER) both at rest and during exercise, increased expression of TCA cycle marked gene, citrate synthase in muscle, reduced fat storage and decreased body weight and food consumption in response to leptin. In females, muscle IL-6 deficiency increases VO2 and CO2 levels similarly. Also in contrast to males, energy expenditure (EE) measured over 48h reveals a significant elevation in female mice with muscle IL-6 deficiency; moreover, they show a modified response to fasting-refeeding and to restraint stress. The present results contribute to the understanding of the role of muscle IL-6 in male and female mouse metabolism, not only during exercise but also in the basal state and in situations where energy balance is altered.


Assuntos
Metabolismo Energético , Interleucina-6/metabolismo , Músculo Esquelético/metabolismo , Caracteres Sexuais , Animais , Temperatura Corporal , Teste de Esforço , Jejum/metabolismo , Jejum/fisiologia , Técnicas de Inativação de Genes , Insulina/metabolismo , Interleucina-6/deficiência , Interleucina-6/genética , Masculino , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Descanso , Serina/metabolismo , Transdução de Sinais , Estresse Fisiológico/efeitos dos fármacos
18.
Sci Rep ; 7(1): 389, 2017 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-28341857

RESUMO

Oxidative stress constitutes a major cause for increased risk of congenital malformations associated to severe hyperglycaemia during pregnancy. Mutations in the gene encoding the transcription factor ALX3 cause congenital craniofacial and neural tube defects. Since oxidative stress and lack of ALX3 favour excessive embryonic apoptosis, we investigated whether ALX3-deficiency further increases the risk of embryonic damage during gestational hyperglycaemia in mice. We found that congenital malformations associated to ALX3-deficiency are enhanced in diabetic pregnancies. Increased expression of genes encoding oxidative stress-scavenging enzymes in embryos from diabetic mothers was blunted in the absence of ALX3, leading to increased oxidative stress. Levels of ALX3 increased in response to glucose, but ALX3 did not activate oxidative stress defence genes directly. Instead, ALX3 stimulated the transcription of Foxo1, a master regulator of oxidative stress-scavenging genes, by binding to a newly identified binding site located in the Foxo1 promoter. Our data identify ALX3 as an important component of the defence mechanisms against the occurrence of developmental malformations during diabetic gestations, stimulating the expression of oxidative stress-scavenging genes in a glucose-dependent manner via Foxo1 activation. Thus, ALX3 deficiency provides a novel molecular mechanism for developmental defects arising from maternal hyperglycaemia.


Assuntos
Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Glucose/metabolismo , Proteínas de Homeodomínio/metabolismo , Hiperglicemia/metabolismo , Estresse Oxidativo , Gravidez em Diabéticas/metabolismo , Animais , Desenvolvimento Embrionário , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Glucose/administração & dosagem , Proteínas de Homeodomínio/genética , Hiperglicemia/complicações , Hiperglicemia/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Gravidez em Diabéticas/genética
19.
Nature ; 539(7630): 518-523, 2016 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-27806375

RESUMO

Mammalian colour patterns are among the most recognizable characteristics found in nature and can have a profound impact on fitness. However, little is known about the mechanisms underlying the formation and subsequent evolution of these patterns. Here we show that, in the African striped mouse (Rhabdomys pumilio), periodic dorsal stripes result from underlying differences in melanocyte maturation, which give rise to spatial variation in hair colour. We identify the transcription factor ALX3 as a regulator of this process. In embryonic dorsal skin, patterned expression of Alx3 precedes pigment stripes and acts to directly repress Mitf, a master regulator of melanocyte differentiation, thereby giving rise to light-coloured hair. Moreover, Alx3 is upregulated in the light stripes of chipmunks, which have independently evolved a similar dorsal pattern. Our results show a previously undescribed mechanism for modulating spatial variation in hair colour and provide insights into how phenotypic novelty evolves.


Assuntos
Padronização Corporal , Regulação da Expressão Gênica no Desenvolvimento , Cor de Cabelo , Murinae/embriologia , Murinae/genética , Animais , Evolução Biológica , Padronização Corporal/genética , Diferenciação Celular , Cor de Cabelo/genética , Proteínas de Homeodomínio/metabolismo , Melaninas/biossíntese , Melanócitos/citologia , Melanócitos/metabolismo , Camundongos , Fator de Transcrição Associado à Microftalmia/antagonistas & inibidores , Fator de Transcrição Associado à Microftalmia/metabolismo , Murinae/fisiologia , Fenótipo , Regiões Promotoras Genéticas/genética , Sciuridae/genética , Pele/embriologia
20.
Diabetologia ; 59(4): 766-75, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26739814

RESUMO

AIMS/HYPOTHESIS: The stimulation of glucagon secretion in response to decreased glucose levels has been studied extensively. In contrast, little is known about the regulation of glucagon gene expression in response to fluctuations in glucose concentration. Paired box 6 (PAX6) is a key transcription factor that regulates the glucagon promoter by binding to the G1 and G3 elements. Here, we investigated the role of the transcription factor aristaless-like homeobox 3 (ALX3) as a glucose-dependent modulator of PAX6 activity in alpha cells. METHODS: Experiments were performed in wild-type or Alx3-deficient islets and alphaTC1 cells. We used chromatin immunoprecipitations and electrophoretic mobility shift assays for DNA binding, immunoprecipitations and pull-down assays for protein interactions, transfected cells for promoter activity, and small interfering RNA and quantitative RT-PCR for gene expression. RESULTS: Elevated glucose concentration resulted in stimulated expression of Alx3 and decreased glucagon gene expression in wild-type islets. In ALX3-deficient islets, basal glucagon levels were non-responsive to changes in glucose concentration. In basal conditions ALX3 bound to the glucagon promoter at G3, but not at G1. ALX3 could form heterodimers with PAX6 that were permissive for binding to G3 but not to G1. Thus, increasing the levels of ALX3 in response to glucose resulted in the sequestration of PAX6 by ALX3 for binding to G1, thus reducing glucagon promoter activation and glucagon gene expression. CONCLUSIONS/INTERPRETATION: Glucose-stimulated expression of ALX3 in alpha cells provides a regulatory mechanism for the downregulation of glucagon gene expression by interfering with PAX6-mediated transactivation on the glucagon G1 promoter element.


Assuntos
Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Glucose/farmacologia , Proteínas de Homeodomínio/metabolismo , Fator de Transcrição PAX6/metabolismo , Animais , Linhagem Celular , Ensaio de Desvio de Mobilidade Eletroforética , Imunoprecipitação , Técnicas In Vitro , Masculino , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...