Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Int J Obes (Lond) ; 43(5): 1120-1124, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30006581

RESUMO

Exogenous oxytocin administration in obese mice, rats, and monkeys was shown to induce sustained weight loss, mostly due to a decrease in fat mass, accompanied by an improvement of glucose metabolism. A pilot study in obese humans confirmed the weight-reducing effect of oxytocin. Knowledge about circulating oxytocin levels in human obesity might help indicating which obese subjects could potentially benefit from an oxytocin treatment. Conclusive results on this topic are missing. The aim of this study was to measure circulating oxytocin levels in lean (n = 37) and obese (n = 72) individuals across a wide range of body mass index (BMI) values (18.5-60 kg/m2) and to determine the impact of pronounced body weight loss following gastric bypass surgery in 12 morbidly obese patients. We observed that oxytocin levels were unchanged in overweight and in class I and II obese subjects and only morbidly obese patients (obesity class III, BMI > 40 kg/m2) exhibited significantly higher levels than lean individuals, with no modification 1 year after gastric bypass surgery, despite substantial body weight loss. In conclusion, morbidly obese subjects present elevated oxytocin levels which were unaltered following pronounced weight loss.


Assuntos
Derivação Gástrica/estatística & dados numéricos , Obesidade Mórbida/metabolismo , Ocitocina/metabolismo , Redução de Peso/fisiologia , Adiposidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Obesidade Mórbida/fisiopatologia , Obesidade Mórbida/cirurgia , Ocitocina/uso terapêutico , Projetos Piloto , Resultado do Tratamento
2.
Am J Respir Cell Mol Biol ; 59(4): 490-499, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29757677

RESUMO

Pulmonary vascular remodeling is an angiogenic-related process involving changes in smooth muscle cell (SMC) homeostasis, which is frequently observed in chronic obstructive pulmonary disease (COPD). MicroRNAs (miRNAs) are small, noncoding RNAs that regulate mRNA expression levels of many genes, leading to the manifestation of cell identity and specific cellular phenotypes. Here, we evaluate the miRNA expression profiles of pulmonary arteries (PAs) of patients with COPD and its relationship with the regulation of SMC phenotypic change. miRNA expression profiles from PAs of 12 patients with COPD, 9 smokers with normal lung function (SK), and 7 nonsmokers (NS) were analyzed using TaqMan Low-Density Arrays. In patients with COPD, expression levels of miR-98, miR-139-5p, miR-146b-5p, and miR-451 were upregulated, as compared with NS. In contrast, miR-197, miR-204, miR-485-3p, and miR-627 were downregulated. miRNA-197 expression correlated with both airflow obstruction and PA intimal enlargement. In an in vitro model of SMC differentiation, miR-197 expression was associated with an SMC contractile phenotype. miR-197 inhibition blocked the acquisition of contractile markers in SMCs and promoted a proliferative/migratory phenotype measured by both cell cycle analysis and wound-healing assay. Using luciferase assays, Western blot, and quantitative PCR, we confirmed that miR-197 targets the transcription factor E2F1. In PAs from patients with COPD, levels of E2F1 were increased as compared with NS. In PAs of patients with COPD, remodeling of the vessel wall is associated with downregulation of miR-197, which regulates SMC phenotype. The effect of miR-197 on PAs might be mediated, at least in part, by the key proproliferative factor, E2F1.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/genética , Artéria Pulmonar/metabolismo , Artéria Pulmonar/fisiopatologia , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Remodelação Vascular/genética , Idoso , Diferenciação Celular/genética , Proliferação de Células/genética , Fator de Transcrição E2F1/metabolismo , Feminino , Volume Expiratório Forçado , Redes Reguladoras de Genes , Humanos , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Índice de Gravidade de Doença
3.
Int J Mol Sci ; 20(1)2018 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-30587816

RESUMO

Whereas leptin administration only has a negligible effect on the treatment of obesity, it has been demonstrated that its action can be improved by co-administration of leptin and one of its sensitizers. Considering that oxytocin treatment decreases body weight in obese animals and humans, we investigated the effects of oxytocin and leptin cotreatment. First, lean and diet-induced obese (DIO) mice were treated with oxytocin for 2 weeks and we measured the acute leptin response. Second, DIO mice were treated for 2 weeks with saline, oxytocin (50 µg/day), leptin (20 or 40 µg/day) or oxytocin plus leptin. Oxytocin pre-treatment restored a normal acute leptin response, decreasing food intake and body weight gain. Chronic continuous administration of oxytocin or leptin at 40 µg/day decreased body weight in the presence (leptin) or in the absence (oxytocin) of cumulative differences in food intake. Saline or leptin treatment at 20 µg/day had no impact on body weight. Oxytocin and leptin cotreatments had no additional effects compared with single treatments. These results point to the fact that chronic oxytocin treatment improves the acute, but not the chronic leptin response, suggesting that this treatment could be used to improve the short-term satiety effect of leptin.


Assuntos
Dieta Hiperlipídica , Ingestão de Alimentos/efeitos dos fármacos , Leptina/farmacologia , Obesidade/etiologia , Ocitocina/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos
4.
FASEB J ; 30(2): 909-22, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26527067

RESUMO

Brown adipose tissue (BAT) is essential for adaptive thermogenesis and dissipation of caloric excess through the activity of uncoupling protein (UCP)-1. BAT in humans is of great interest for the treatment of obesity and related diseases. In this study, the expression of Twik-related acid-sensitive K(+) channel (TASK)-1 [a pH-sensitive potassium channel encoded by the potassium channel, 2-pore domain, subfamily K, member 3 (Kcnk3) gene] correlated highly with Ucp1 expression in obese and cold-exposed mice. In addition, Task1-null mice, compared with their controls, became overweight, mainly because of an increase in white adipose tissue mass and BAT whitening. Task1(-/-)-mouse-derived brown adipocytes, compared with wild-type mouse-derived brown adipocytes, displayed an impaired ß3-adrenergic receptor response that was characterized by a decrease in oxygen consumption, Ucp1 expression, and lipolysis. This phenotype was thought to be caused by an exacerbation of mineralocorticoid receptor (MR) signaling, given that it was mimicked by corticoids and reversed by an MR inhibitor. We concluded that the K(+) channel TASK1 controls the thermogenic activity in brown adipocytes through modulation of ß-adrenergic receptor signaling.


Assuntos
Adipócitos Marrons/metabolismo , Tecido Adiposo Marrom/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Receptores de Mineralocorticoides/metabolismo , Transdução de Sinais/fisiologia , Adipócitos Marrons/citologia , Tecido Adiposo Marrom/citologia , Animais , Feminino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Consumo de Oxigênio/fisiologia , Canais de Potássio de Domínios Poros em Tandem/genética , Receptores de Mineralocorticoides/genética , Termogênese/fisiologia
5.
J Hepatol ; 62(2): 421-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25234947

RESUMO

BACKGROUND & AIMS: PTEN is a dual lipid/protein phosphatase, downregulated in steatotic livers with obesity or HCV infection. Liver-specific PTEN knockout (LPTEN KO) mice develop steatosis, inflammation/fibrosis and hepatocellular carcinoma with aging, but surprisingly also enhanced glucose tolerance. This study aimed at understanding the mechanisms by which hepatic PTEN deficiency improves glucose tolerance, while promoting fatty liver diseases. METHODS: Control and LPTEN KO mice underwent glucose/pyruvate tolerance tests and euglycemic-hyperinsulinemic clamps. Body fat distribution was assessed by EchoMRI, CT-scan and dissection analyses. Primary/cultured hepatocytes and insulin-sensitive tissues were analysed ex vivo. RESULTS: PTEN deficiency in hepatocytes led to steatosis through increased fatty acid (FA) uptake and de novo lipogenesis. Although LPTEN KO mice exhibited hepatic steatosis, they displayed increased skeletal muscle insulin sensitivity and glucose uptake, as assessed by euglycemic-hyperinsulinemic clamps. Surprisingly, white adipose tissue (WAT) depots were also drastically reduced. Analyses of key enzymes involved in lipid metabolism further indicated that FA synthesis/esterification was decreased in WAT. In addition, Ucp1 expression and multilocular lipid droplet structures were observed in this tissue, indicating the presence of beige adipocytes. Consistent with a liver to muscle/adipocyte crosstalk, the expression of liver-derived circulating factors, known to impact on muscle insulin sensitivity and WAT homeostasis (e.g. FGF21), was modulated in LPTEN KO mice. CONCLUSIONS: Although steatosis develops in LPTEN KO mice, PTEN deficiency in hepatocytes promotes a crosstalk between liver and muscle, as well as adipose tissue, resulting in enhanced insulin sensitivity, improved glucose tolerance and decreased adiposity.


Assuntos
Adiposidade/genética , Fígado Gorduroso/genética , Regulação da Expressão Gênica , Resistência à Insulina , Lipogênese/genética , PTEN Fosfo-Hidrolase/genética , RNA/genética , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Western Blotting , Células Cultivadas , Fígado Gorduroso/diagnóstico , Fígado Gorduroso/metabolismo , Glucose/metabolismo , Teste de Tolerância a Glucose , Metabolismo dos Lipídeos , Imageamento por Ressonância Magnética , Camundongos , Camundongos Knockout , PTEN Fosfo-Hidrolase/deficiência , Fenótipo , Tomografia Computadorizada por Raios X
6.
Rev Med Suisse ; 11(456-457): 97-100, 2015 Jan 14.
Artigo em Francês | MEDLINE | ID: mdl-25799660

RESUMO

Oxytocin is a hormone known for a long time, mainly used in the field of gynecology. Apart from these well-defined effects, the role of oxytocin in controlling the stress response or behavior and the regulation of glucose/lipid metabolism seems to be very interesting, especially in obese patients. Several clinical studies are currently underway to assess the impact of oxytocin in the treatment of obesity. Taking these new data into consideration, the use of this hormone for weight loss in obese patients or as a complementary treatment in diabetic patients seems to be promising.


Assuntos
Metabolismo/efeitos dos fármacos , Obesidade/tratamento farmacológico , Ocitocina/farmacologia , Ocitocina/uso terapêutico , Animais , Humanos
7.
Biochim Biophys Acta ; 1829(11): 1175-83, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23938248

RESUMO

The atonal-related Neurogenin/NeuroD family of basic helix-loop-helix (bHLH) transcription factors comprises potent inducers of neuronal and endocrine differentiation programs in the nervous and digestive system. Atonal homolog 8 (Atoh8) displays high similarity in the bHLH domain with NeuroD proteins. Yet, available evidences indicate that Atoh8 has distinctive features including a ubiquitous expression pattern in embryonic tissues and the ability to inhibit differentiation. To gain insights into Atoh8 function, we aimed at identifying Atoh8 targets and investigated the effects of Atoh8 on global gene expression patterns in pancreatic mPAC cells, a model of bHLH-dependent endocrine differentiation. Our data reveal that Atoh8 is a weak transcriptional activator and does not exhibit proendocrine activity. Conversely, it blocks the induction of a reduced group of gene targets of the atonal-related proendocrine factor Neurogenin3. We show that Atoh8 lacks a transactivation domain and possesses intrinsic repressor activity that depends on a conserved Proline-rich domain. Atoh8 binds the ubiquitous E protein E47 and its ability to repress transcription may partly result from its ability to inhibit E47/E47 and Neurogenin3/E47 dimer activities. These results reveal distinctive transcriptional properties of Atoh8 within the atonal-related bHLH family that may be associated with the acquisition of new biological functions.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Transcrição Gênica/fisiologia , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Camundongos , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase em Tempo Real
8.
Diabetologia ; 56(11): 2446-55, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24006088

RESUMO

AIMS/HYPOTHESIS: During obesity, the increment in beta cell mass in response to the rising demand for insulin is essential to maintain normal glucose homeostasis. However, the precise cellular and molecular mechanisms involved in beta cell mass plasticity remain poorly understood. The Wnt signalling pathway has been suggested as one possible modulator of beta cell proliferation, which represents the principal process involved in beta cell mass expansion. Here, we sought to determine the mechanisms involved in beta cell mass proliferation using diet-induced obese rats. METHODS: Wistar rats aged 8 weeks old were fed a standard or cafeteria diet. Global transcriptomic analysis of pancreatic rat islets was performed using microarray analysis. Genetic loss-of-function approaches were performed in dispersed primary rat islets and the beta cell line INS1E. Gene expression was measured by real-time PCR, protein levels by immunoblot analysis, proliferation rates by ELISA and apoptosis by flow cytometry. RESULTS: Sfrp5, coding for secreted frizzled-related protein 5, is downregulated in the pancreatic islets of cafeteria-diet-fed rats as well as in the pancreatic islets of human obese patients. We demonstrate that silencing Sfrp5 increases beta cell proliferation, which correlates with activation of Wnt signalling and enhanced levels of proliferation markers. In addition, we show that expression of Sfrp5 in beta cells is modulated by IGF binding protein 3 (IGFBP3) secreted from visceral adipose tissue. CONCLUSIONS/INTERPRETATION: Together, these findings reveal an important role for SFRP5 and Wnt signalling in the regulation of beta cell proliferation in obesity.


Assuntos
Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Proteínas de Membrana/metabolismo , Obesidade/metabolismo , Animais , Proliferação de Células , Masculino , Proteínas de Membrana/genética , Obesidade/genética , Ratos , Ratos Wistar
9.
Nat Metab ; 4(4): 424-434, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35379970

RESUMO

Preparation for motherhood requires a myriad of physiological and behavioural adjustments throughout gestation to provide an adequate environment for proper embryonic development1. Cravings for highly palatable foods are highly prevalent during pregnancy2 and contribute to the maintenance and development of gestational overweight or obesity3. However, the neurobiology underlying the distinct ingestive behaviours that result from craving specific foods remain unknown. Here we show that mice, similarly to humans, experience gestational food craving-like episodes. These episodes are associated with a brain connectivity reorganization that affects key components of the dopaminergic mesolimbic circuitry, which drives motivated appetitive behaviours and facilitates the perception of rewarding stimuli. Pregnancy engages a dynamic modulation of dopaminergic signalling through neurons expressing dopamine D2 receptors in the nucleus accumbens, which directly modulate food craving-like events. Importantly, persistent maternal food craving-like behaviour has long-lasting effects on the offspring, particularly in males, leading to glucose intolerance, increased body weight and increased susceptibility to develop eating disorders and anxiety-like behaviours during adulthood. Our results reveal the cognitively motivated nature of pregnancy food cravings and advocates for moderating emotional eating during gestation to prevent deterioration of the offspring's neuropsychological and metabolic health.


Assuntos
Fissura , Ingestão de Alimentos , Animais , Fissura/fisiologia , Dopamina/metabolismo , Feminino , Preferências Alimentares/psicologia , Masculino , Camundongos , Obesidade/metabolismo , Gravidez , Aumento de Peso
10.
Cell Metab ; 34(2): 269-284.e9, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35108514

RESUMO

Obesity and type 2 diabetes are associated with cognitive dysfunction. Because the hypothalamus is implicated in energy balance control and memory disorders, we hypothesized that specific neurons in this brain region are at the interface of metabolism and cognition. Acute obesogenic diet administration in mice impaired recognition memory due to defective production of the neurosteroid precursor pregnenolone in the hypothalamus. Genetic interference with pregnenolone synthesis by Star deletion in hypothalamic POMC, but not AgRP neurons, deteriorated recognition memory independently of metabolic disturbances. Our data suggest that pregnenolone's effects on cognitive function were mediated via an autocrine mechanism on POMC neurons, influencing hippocampal long-term potentiation. The relevance of central pregnenolone on cognition was also confirmed in metabolically unhealthy patients with obesity. Our data reveal an unsuspected role for POMC neuron-derived neurosteroids in cognition. These results provide the basis for a framework to investigate new facets of POMC neuron biology with implications for cognitive disorders.


Assuntos
Diabetes Mellitus Tipo 2 , Doenças Metabólicas , Animais , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hipotálamo/metabolismo , Doenças Metabólicas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pregnenolona/metabolismo , Pró-Opiomelanocortina/metabolismo
11.
Nat Commun ; 12(1): 7031, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34857752

RESUMO

Intestinal surface changes in size and function, but what propels these alterations and what are their metabolic consequences is unknown. Here we report that the food amount is a positive determinant of the gut surface area contributing to an increased absorptive function, reversible by reducing daily food. While several upregulated intestinal energetic pathways are dispensable, the intestinal PPARα is instead necessary for the genetic and environment overeating-induced increase of the gut absorptive capacity. In presence of dietary lipids, intestinal PPARα knock-out or its pharmacological antagonism suppress intestinal crypt expansion and shorten villi in mice and in human intestinal biopsies, diminishing the postprandial triglyceride transport and nutrient uptake. Intestinal PPARα ablation limits systemic lipid absorption and restricts lipid droplet expansion and PLIN2 levels, critical for droplet formation. This improves the lipid metabolism, and reduces body adiposity and liver steatosis, suggesting an alternative target for treating obesity.


Assuntos
Fígado Gorduroso/genética , Intestinos/metabolismo , PPAR alfa/genética , Perilipina-2/genética , Adiposidade/genética , Animais , Dieta/métodos , Ingestão de Alimentos/fisiologia , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Regulação da Expressão Gênica , Humanos , Absorção Intestinal/fisiologia , Gotículas Lipídicas/metabolismo , Metabolismo dos Lipídeos/genética , Masculino , Camundongos , Camundongos Transgênicos , PPAR alfa/deficiência , PPAR alfa/metabolismo , Perilipina-2/metabolismo , Período Pós-Prandial , Transdução de Sinais , Triglicerídeos/metabolismo
12.
Cell Metab ; 33(9): 1820-1835.e9, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34343501

RESUMO

Appropriate cristae remodeling is a determinant of mitochondrial function and bioenergetics and thus represents a crucial process for cellular metabolic adaptations. Here, we show that mitochondrial cristae architecture and expression of the master cristae-remodeling protein OPA1 in proopiomelanocortin (POMC) neurons, which are key metabolic sensors implicated in energy balance control, is affected by fluctuations in nutrient availability. Genetic inactivation of OPA1 in POMC neurons causes dramatic alterations in cristae topology, mitochondrial Ca2+ handling, reduction in alpha-melanocyte stimulating hormone (α-MSH) in target areas, hyperphagia, and attenuated white adipose tissue (WAT) lipolysis resulting in obesity. Pharmacological blockade of mitochondrial Ca2+ influx restores α-MSH and the lipolytic program, while improving the metabolic defects of mutant mice. Chemogenetic manipulation of POMC neurons confirms a role in lipolysis control. Our results unveil a novel axis that connects OPA1 in POMC neurons with mitochondrial cristae, Ca2+ homeostasis, and WAT lipolysis in the regulation of energy balance.


Assuntos
Lipólise , Pró-Opiomelanocortina , Tecido Adiposo/metabolismo , Animais , GTP Fosfo-Hidrolases , Homeostase , Camundongos , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo
13.
Am J Physiol Endocrinol Metab ; 299(6): E1087-95, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20943756

RESUMO

BACE1 (ß-site amyloidogenic cleavage of precursor protein-cleaving enzyme 1) is a ß-secretase protein that plays a central role in the production of the ß-amyloid peptide in the brain and is thought to be involved in the Alzheimer's pathogenesis. In type 2 diabetes, amyloid deposition within the pancreatic islets is a pathophysiological hallmark, making crucial the study in the pancreas of BACE1 and its homologous BACE2 to understand the pathological mechanisms of this disease. The objectives of the present study were to characterize the localization of BACE proteins in human pancreas and determine their function. High levels of BACE enzymatic activity were detected in human pancreas. In normal human pancreas, BACE1 was observed in endocrine as well as in exocrine pancreas, whereas BACE2 expression was restricted to ß-cells. Intracellular analysis using immunofluorescence showed colocalization of BACE1 with insulin and BACE2 with clathrin-coated vesicles of the plasma membrane in MIN6 cells. When BACE1 and -2 were pharmacologically inhibited, BACE1 localization was not altered, whereas BACE2 content in clathrin-coated vesicles was increased. Insulin internalization rate was reduced, insulin receptor ß-subunit (IRß) expression was decreased at the plasma membrane and increased in the Golgi apparatus, and a significant reduction in insulin gene expression was detected. Similar results were obtained after specific BACE2 silencing in MIN6 cells. All these data point to a role for BACE2 in the IRß trafficking and insulin signaling. In conclusion, BACE2 is hereby presented as an important enzyme in ß-cell function.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Pâncreas/metabolismo , Receptor de Insulina/metabolismo , Adulto , Animais , Western Blotting , Linhagem Celular , Células Cultivadas , Vesículas Revestidas por Clatrina/metabolismo , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Transporte Proteico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Frações Subcelulares/metabolismo
14.
Mol Metab ; 36: 100963, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32283518

RESUMO

OBJECTIVE: Maternal unbalanced nutritional habits during embryonic development and perinatal stages perturb hypothalamic neuronal programming of the offspring, thus increasing obesity-associated diabetes risk. However, the underlying molecular mechanisms remain largely unknown. In this study we sought to determine the translatomic signatures associated with pro-opiomelanocortin (POMC) neuron malprogramming in maternal obesogenic conditions. METHODS: We used the RiboTag mouse model to specifically profile the translatome of POMC neurons during neonatal (P0) and perinatal (P21) life and its neuroanatomical, functional, and physiological consequences. RESULTS: Maternal high-fat diet (HFD) exposure did not interfere with offspring's hypothalamic POMC neuron specification, but significantly impaired their spatial distribution and axonal extension to target areas. Importantly, we established POMC neuron-specific translatome signatures accounting for aberrant neuronal development and axonal growth. These anatomical and molecular alterations caused metabolic dysfunction in early life and adulthood. CONCLUSIONS: Our study provides fundamental insights on the molecular mechanisms underlying POMC neuron malprogramming in obesogenic contexts.


Assuntos
Obesidade/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , DNA/genética , Metilação de DNA , Dieta Hiperlipídica , Feminino , Estudo de Associação Genômica Ampla , Hipotálamo/metabolismo , Masculino , Camundongos , Neurogênese/genética , Neurônios/metabolismo , Obesidade/metabolismo , Gravidez/genética , Gravidez/metabolismo , Pró-Opiomelanocortina/fisiologia
15.
Antioxid Redox Signal ; 32(9): 618-635, 2020 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-31931619

RESUMO

Aims: Nicotinamide adenine dinucleotide phosphate oxidases (NOX-es) produce reactive oxygen species and modulate ß-cell insulin secretion. Islets of type 2 diabetic subjects present elevated expression of NOX5. Here, we sought to characterize regulation of NOX5 expression in human islets in vitro and to uncover the relevance of NOX5 in islet function in vivo using a novel mouse model expressing NOX5 in doxycycline-inducible, ß-cell-specific manner (RIP/rtTA/NOX5 mice). Results:In situ hybridization and immunohistochemistry employed on pancreatic sections demonstrated NOX5 messenger ribonucleic acid (mRNA) and protein expressions in human islets. In cultures of dispersed islets, NOX5 protein was observed in somatostatin-positive (δ) cells in basal (2.8 mM glucose) conditions. Small interfering ribonucleic acid (siRNA)-mediated knockdown of NOX5 in human islets cultured in basal glucose concentrations resulted in diminished glucose-induced insulin secretion (GIIS) in vitro. However, when islets were preincubated in high (16.7 mM) glucose media for 12 h, NOX5 appeared also in insulin-positive (ß) cells. In vivo, mice with ß-cell NOX5 expression developed aggravated impairment of GIIS compared with control mice when challenged with 14 weeks of high-fat diet. Similarly, in vitro palmitate preincubation resulted in more severe reduction of insulin release in islets of RIP/rtTA/NOX5 mice compared with their control littermates. Decreased insulin secretion was most distinct in response to theophylline stimulation, suggesting impaired cyclic adenosine monophosphate (cAMP)-mediated signaling due to increased phosphodiesterase activation. Innovation and Conclusions: Our data provide the first insight into the complex regulation and function of NOX5 in islets implying an important role for NOX5 in δ-cell-mediated intraislet crosstalk in physiological circumstances but also identifying it as an aggravating factor in ß-cell failure in diabetic conditions.


Assuntos
Ilhotas Pancreáticas/metabolismo , NADPH Oxidase 5/genética , Animais , Células Cultivadas , Dieta Hiperlipídica/efeitos adversos , Feminino , Humanos , Secreção de Insulina/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , NADPH Oxidase 5/metabolismo
16.
BMC Genomics ; 10: 406, 2009 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-19715561

RESUMO

BACKGROUND: Sodium tungstate is known to be an effective anti-diabetic agent, able to increase beta cell mass in animal models of diabetes, although the molecular mechanisms of this treatment and the genes that control pancreas plasticity are yet to be identified. Using a transcriptomics approach, the aim of the study is to unravel the molecular mechanisms which participate in the recovery of exocrine and endocrine function of streptozotocin (STZ) diabetic rats treated with tungstate, determining the hyperglycemia contribution and the direct effect of tungstate. RESULTS: Streptozotocin (STZ)-diabetic rats were treated orally with tungstate for five weeks. Treated (STZ)-diabetic rats showed a partial recovery of exocrine and endocrine function, with lower glycemia, increased insulinemia and amylasemia, and increased beta cell mass achieved by reducing beta cell apoptosis and raising beta cell proliferation. The microarray analysis of the pancreases led to the identification of three groups of differentially expressed genes: genes altered due to diabetes, genes restored by the treatment, and genes specifically induced by tungstate in the diabetic animals. The results were corroborated by quantitative PCR. A detailed description of the pathways involved in the pancreatic effects of tungstate is provided in this paper. Hyperglycemia contribution was studied in STZ-diabetic rats treated with phloridzin, and the direct effect of tungstate was determined in INS-1E cells treated with tungstate or serum from untreated or treated STZ-rats, observing that tungstate action in the pancreas takes places via hyperglycemia-independent pathways and via a combination of tungstate direct and indirect (through the serum profile modification) effects. Finally, the MAPK pathway was evaluated, observing that it has a key role in the tungstate-induced increase of beta cell proliferation as tungstate activates the mitogen-activated protein kinase (MAPK) pathway directly by increasing p42/p44 phosphorylation and indirectly by decreasing the expression of raf kinase inhibitor protein (Rkip), a negative modulator of the pathway. CONCLUSION: In conclusion, tungstate improves pancreatic function through a combination of hyperglycemia-independent pathways and through its own direct and indirect effects, whereas the MAPK pathway has a key role in the tungstate-induced increase of beta cell proliferation.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Perfilação da Expressão Gênica , Pâncreas/metabolismo , Compostos de Tungstênio/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/tratamento farmacológico , Hiperglicemia/metabolismo , Hipoglicemiantes/farmacologia , Células Secretoras de Insulina/metabolismo , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Pâncreas/efeitos dos fármacos , Florizina/farmacologia , Ratos , Ratos Wistar , Estreptozocina
17.
Nat Commun ; 10(1): 3545, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31391467

RESUMO

Tens of millions suffer from insulin deficiency (ID); a defect leading to severe metabolic imbalance and death. The only means for management of ID is insulin therapy; yet, this approach is sub-optimal and causes life-threatening hypoglycemia. Hence, ID represents a great medical and societal challenge. Here we report that S100A9, also known as Calgranulin B or Myeloid-Related Protein 14 (MRP14), is a leptin-induced circulating cue exerting beneficial anti-diabetic action. In murine models of ID, enhanced expression of S100A9 alone (i.e. without administered insulin and/or leptin) slightly improves hyperglycemia, and normalizes key metabolic defects (e.g. hyperketonemia, hypertriglyceridemia, and increased hepatic fatty acid oxidation; FAO), and extends lifespan by at least a factor of two. Mechanistically, we report that Toll-Like Receptor 4 (TLR4) is required, at least in part, for the metabolic-improving and pro-survival effects of S100A9. Thus, our data identify the S100A9/TLR4 axis as a putative target for ID care.


Assuntos
Calgranulina B/metabolismo , Diabetes Mellitus Experimental/metabolismo , Hiperglicemia/metabolismo , Longevidade/fisiologia , Receptor 4 Toll-Like/metabolismo , Animais , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/etiologia , Toxina Diftérica/toxicidade , Ácidos Graxos/metabolismo , Humanos , Hiperglicemia/sangue , Hiperglicemia/etiologia , Insulina/deficiência , Leptina/administração & dosagem , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Oxirredução , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Estreptozocina/toxicidade , Receptor 4 Toll-Like/genética
18.
Metabolism ; 87: 13-17, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29936173

RESUMO

BACKGROUND: Oleoylethanolamide (OEA) is an endocannabinoid that controls food intake, energy expenditure and locomotor activity. Its anorexigenic effect appears to be mediated by PPARα, but the tissue where the presence of this receptor is required for OEA to inhibit feeding is unknown as yet. Previous studies point to a possible role of proximal enterocytes and neurons of the nodose ganglion. MATERIALS AND METHODS: Acute intraperitoneal OEA effects on food intake, energy expenditure, respiratory exchange ratio (RER) and locomotor activity were studied in control mice (PPARα-loxP) and intestinal (Villin-Cre;PPARα-loxP) or nodose ganglion (Phox2B-Cre;PPARα-loxP) specific PPARα knockout mice placed in calorimetric cages. RESULTS: OEA administration to both intestinal and nodose ganglion PPARα knockout mice decreased food intake, RER (leading to increased lipid oxidation) and locomotor activity as in control mice. However, while OEA injection acutely decreased energy expenditure in controls, this effect was not observed in mice devoid of PPARα in the intestine. CONCLUSION: These results indicate that the OEA effect on food intake is independent from the presence of PPARα in the intestine and the nodose ganglion, while the impact of OEA on energy expenditure requires the presence of PPARα in the intestine.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Endocanabinoides/farmacologia , Metabolismo Energético/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Gânglio Nodoso/metabolismo , Ácidos Oleicos/farmacologia , PPAR alfa/metabolismo , Animais , Mucosa Intestinal/efeitos dos fármacos , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Gânglio Nodoso/efeitos dos fármacos , PPAR alfa/efeitos dos fármacos , PPAR alfa/genética
20.
Nat Commun ; 8(1): 1820, 2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29180649

RESUMO

Obesity-induced inflammation engenders insulin resistance and type 2 diabetes mellitus (T2DM) but the inflammatory effectors linking obesity to insulin resistance are incompletely understood. Here, we show that hepatic expression of Protein Tyrosine Phosphatase Receptor Gamma (PTPR-γ) is stimulated by inflammation in obese/T2DM mice and positively correlates with indices of inflammation and insulin resistance in humans. NF-κB binds to the promoter of Ptprg and is required for inflammation-induced PTPR-γ expression. PTPR-γ loss-of-function lowers glycemia and insulinemia by enhancing insulin-stimulated suppression of endogenous glucose production. These phenotypes are rescued by re-expression of Ptprg only in liver of mice lacking Ptprg globally. Hepatic PTPR-γ overexpression that mimics levels found in obesity is sufficient to cause severe hepatic and systemic insulin resistance. We propose hepatic PTPR-γ as a link between obesity-induced inflammation and insulin resistance and as potential target for treatment of T2DM.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Resistência à Insulina/fisiologia , Fígado/metabolismo , Obesidade/metabolismo , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Adulto , Idoso , Animais , Glicemia , Linhagem Celular , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Células Hep G2 , Humanos , Inflamação/metabolismo , Insulina/sangue , Interleucina-6/metabolismo , Metabolismo dos Lipídeos , Lipopolissacarídeos/efeitos adversos , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Pessoa de Meia-Idade , Modelos Animais , NF-kappa B/metabolismo , Obesidade/sangue , Obesidade/complicações , Proteínas Tirosina Fosfatases/metabolismo , RNA Mensageiro/biossíntese , Proteínas Tirosina Fosfatases Semelhantes a Receptores/genética , Sirtuína 1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA