Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nat Metab ; 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38689023

RESUMO

The oxidative phosphorylation system1 in mammalian mitochondria plays a key role in transducing energy from ingested nutrients2. Mitochondrial metabolism is dynamic and can be reprogrammed to support both catabolic and anabolic reactions, depending on physiological demands or disease states. Rewiring of mitochondrial metabolism is intricately linked to metabolic diseases and promotes tumour growth3-5. Here, we demonstrate that oral treatment with an inhibitor of mitochondrial transcription (IMT)6 shifts whole-animal metabolism towards fatty acid oxidation, which, in turn, leads to rapid normalization of body weight, reversal of hepatosteatosis and restoration of normal glucose tolerance in male mice on a high-fat diet. Paradoxically, the IMT treatment causes a severe reduction of oxidative phosphorylation capacity concomitant with marked upregulation of fatty acid oxidation in the liver, as determined by proteomics and metabolomics analyses. The IMT treatment leads to a marked reduction of complex I, the main dehydrogenase feeding electrons into the ubiquinone (Q) pool, whereas the levels of electron transfer flavoprotein dehydrogenase and other dehydrogenases connected to the Q pool are increased. This rewiring of metabolism caused by reduced mtDNA expression in the liver provides a principle for drug treatment of obesity and obesity-related pathology.

2.
Physiol Rep ; 12(1): e15901, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38171546

RESUMO

Obesity is a global health problem characterized by excessive fat accumulation, driven by adipogenesis and lipid accumulation. Long non-coding RNAs (lncRNAs) have recently been implicated in regulating adipogenesis and adipose tissue function. Mouse lncRNA U90926 was previously identified as a repressor of in vitro adipogenesis in 3T3-L1 preadipocytes. Consequently, we hypothesized that, in vivo, U90926 may repress adipogenesis, and hence its deletion would increase weight gain and adiposity. We tested the hypothesis by applying U90926-deficient (U9-KO) mice to a high-throughput phenotyping pipeline. Compared with WT, U9-KO mice showed no major differences across a wide range of behavioral, neurological, and other physiological parameters. In mice fed a standard diet, we have found no differences in obesity-related phenotypes, including weight gain, fat mass, and plasma concentrations of glucose, insulin, triglycerides, and free fatty acids, in U9-KO mice compared to WT. U90926 deficiency lacked a major effect on white adipose tissue morphology and gene expression profile. Furthermore, in mice fed a high-fat diet, we found increased expression of U90926 in adipose tissue stromal vascular cell fraction, yet observed no effect of U90926 deficiency on weight gain, fat mass, adipogenesis marker expression, and immune cell infiltration into the adipose tissue. These data suggest that the U90926 lacks an essential role in obesity-related phenotypes and adipose tissue biology in vivo.


Assuntos
RNA Longo não Codificante , Camundongos , Animais , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Adipócitos/metabolismo , Obesidade/genética , Obesidade/metabolismo , Adipogenia/genética , Aumento de Peso , Dieta Hiperlipídica/efeitos adversos , Fenótipo , Camundongos Endogâmicos C57BL
3.
Mol Metab ; 79: 101840, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38036170

RESUMO

OBJECTIVE: Free fatty acid receptor-1 (FFAR1) is a medium- and long-chain fatty acid sensing G protein-coupled receptor that is highly expressed in the hypothalamus. Here, we investigated the central role of FFAR1 on energy balance. METHODS: Central FFAR1 agonism and virogenic knockdown were performed in mice. Energy balance studies, infrared thermographic analysis of brown adipose tissue (BAT) and molecular analysis of the hypothalamus, BAT, white adipose tissue (WAT) and liver were carried out. RESULTS: Pharmacological stimulation of FFAR1, using central administration of its agonist TUG-905 in diet-induced obese mice, decreases body weight and is associated with increased energy expenditure, BAT thermogenesis and browning of subcutaneous WAT (sWAT), as well as reduced AMP-activated protein kinase (AMPK) levels, reduced inflammation, and decreased endoplasmic reticulum (ER) stress in the hypothalamus. As FFAR1 is expressed in distinct hypothalamic neuronal subpopulations, we used an AAV vector expressing a shRNA to specifically knockdown Ffar1 in proopiomelanocortin (POMC) neurons of the arcuate nucleus of the hypothalamus (ARC) of obese mice. Our data showed that knockdown of Ffar1 in POMC neurons promoted hyperphagia and body weight gain. In parallel, these mice developed hepatic insulin resistance and steatosis. CONCLUSIONS: FFAR1 emerges as a new hypothalamic nutrient sensor regulating whole body energy balance. Moreover, pharmacological activation of FFAR1 could provide a therapeutic advance in the management of obesity and its associated metabolic disorders.


Assuntos
Ácidos Graxos não Esterificados , Pró-Opiomelanocortina , Camundongos , Animais , Ácidos Graxos não Esterificados/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Camundongos Obesos , Peso Corporal , Hipotálamo/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Metabolismo Energético/fisiologia
4.
Mamm Genome ; 34(2): 107-122, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37326672

RESUMO

Cardiovascular diseases cause a high mortality rate worldwide and represent a major burden for health care systems. Experimental rodent models play a central role in cardiovascular disease research by effectively simulating human cardiovascular diseases. Using mice, the International Mouse Phenotyping Consortium (IMPC) aims to target each protein-coding gene and phenotype multiple organ systems in single-gene knockout models by a global network of mouse clinics. In this review, we summarize the current advances of the IMPC in cardiac research and describe in detail the diagnostic requirements of high-throughput electrocardiography and transthoracic echocardiography capable of detecting cardiac arrhythmias and cardiomyopathies in mice. Beyond that, we are linking metabolism to the heart and describing phenotypes that emerge in a set of known genes, when knocked out in mice, such as the leptin receptor (Lepr), leptin (Lep), and Bardet-Biedl syndrome 5 (Bbs5). Furthermore, we are presenting not yet associated loss-of-function genes affecting both, metabolism and the cardiovascular system, such as the RING finger protein 10 (Rfn10), F-box protein 38 (Fbxo38), and Dipeptidyl peptidase 8 (Dpp8). These extensive high-throughput data from IMPC mice provide a promising opportunity to explore genetics causing metabolic heart disease with an important translational approach.


Assuntos
Doenças Cardiovasculares , Sistema Cardiovascular , Camundongos , Animais , Humanos , Camundongos Knockout , Doenças Cardiovasculares/genética , Técnicas de Inativação de Genes , Fenótipo
5.
Nutrients ; 13(12)2021 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-34959756

RESUMO

Besides their direct effects on peripheral metabolic tissues, thyroid hormones (TH) act on the hypothalamus to modulate energy homeostasis. However, since most of the hypothalamic actions of TH have been addressed in studies with direct central administration, the estimation of the relative contribution of the central vs. peripheral effects in physiologic conditions of peripheral release (or administration) of TH remains unclear. In this study we used two different models of peripherally induced hyperthyroidism (i.e., T4 and T3 oral administration) to assess and compare the serum and hypothalamic TH status and relate them to the metabolic effects of the treatment. Peripheral TH treatment affected feeding behavior, overall growth, core body temperature, body composition, brown adipose tissue (BAT) morphology and uncoupling protein 1 (UCP1) levels and metabolic activity, white adipose tissue (WAT) browning and liver metabolism. This resulted in an increased overall uncoupling capacity and a shift of the lipid metabolism from WAT accumulation to BAT fueling. Both peripheral treatment protocols induced significant changes in TH concentrations within the hypothalamus, with T3 eliciting a downregulation of hypothalamic AMP-activated protein kinase (AMPK), supporting the existence of a central action of peripheral TH. Altogether, these data suggest that peripherally administered TH modulate energy balance by various mechanisms; they also provide a unifying vision of the centrally mediated and the direct local metabolic effect of TH in the context of hyperthyroidism.


Assuntos
Proteínas Quinases Ativadas por AMP/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Hipertireoidismo/metabolismo , Hipotálamo/metabolismo , Hormônios Tireóideos/administração & dosagem , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Modelos Animais de Doenças , Hipertireoidismo/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL
6.
Nat Metab ; 3(10): 1415-1431, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34675439

RESUMO

Current pharmacological therapies for treating obesity are of limited efficacy. Genetic ablation or loss of function of AMP-activated protein kinase alpha 1 (AMPKα1) in steroidogenic factor 1 (SF1) neurons of the ventromedial nucleus of the hypothalamus (VMH) induces feeding-independent resistance to obesity due to sympathetic activation of brown adipose tissue (BAT) thermogenesis. Here, we show that body weight of obese mice can be reduced by intravenous injection of small extracellular vesicles (sEVs) delivering a plasmid encoding an AMPKα1 dominant negative mutant (AMPKα1-DN) targeted to VMH-SF1 neurons. The beneficial effect of SF1-AMPKα1-DN-loaded sEVs is feeding-independent and involves sympathetic nerve activation and increased UCP1-dependent thermogenesis in BAT. Our results underscore the potential of sEVs to specifically target AMPK in hypothalamic neurons and introduce a broader strategy to manipulate body weight and reduce obesity.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Tecido Adiposo Marrom/enzimologia , Vesículas Extracelulares/metabolismo , Hipotálamo/enzimologia , Obesidade/metabolismo , Animais , Metabolismo Energético , Camundongos , Termogênese , Redução de Peso
7.
Neuroscience ; 447: 191-215, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-33046217

RESUMO

In the last thirty years, obesity has reached epidemic proportions and is now regarded as a major health issue in contemporary society trending to serious economic and social burdens. The latest projections of the World Health Organization are alarming. By 2030, nearly 60% of the worldwide population could be either obese or overweight, highlighting the needs to find innovative treatments. Currently, bariatric surgery is the most effective way to efficiently lower body mass. Although great improvements in terms of recovery and patient care were made in these surgical procedures, bariatric surgery remains an option for extreme forms of obesity and seems unable to tackle obesity pandemic expansion. Throughout the last century, numerous pharmacological strategies targeting either peripheral or central components of the energy balance regulatory system were designed to reduce body mass, some of them reaching sufficient levels of efficiency and safety. Nevertheless, obesity drug therapy remains quite limited on its effectiveness to actually overcome the obesogenic environment. Thus, innovative unimolecular polypharmacology strategies, able to simultaneously target multiple actors involved in the obesity initiation and expansion, were developed during the last ten years opening a new promising avenue in the pharmacological management of obesity. In this review, we first describe the clinical features of obesity-associated conditions and then focus on the outcomes of currently approved drug therapies for obesity as well as new ones expecting to reach the clinic in the near future.

8.
Neuroscience ; 437: 215-239, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32360593

RESUMO

In the last thirty years, obesity has reached epidemic proportions and is now regarded as a major health issue in contemporary society trending to serious economic and social burdens. The latest projections of the World Health Organization are alarming. By 2030, nearly 60% of the worldwide population could be either obese or overweight, highlighting the needs to find innovative treatments. Currently, bariatric surgery is the most effective way to efficiently lower body mass. Although great improvements in terms of recovery and patient care were made in these surgical procedures, bariatric surgery remains an option for extreme forms of obesity and seems unable to tackle obesity pandemic expansion. Throughout the last century, numerous pharmacological strategies targeting either peripheral or central components of the energy balance regulatory system were designed to reduce body mass, some of them reaching sufficient levels of efficiency and safety. Nevertheless, obesity drug therapy remains quite limited on its effectiveness to actually overcome the obesogenic environment. Thus, innovative unimolecular polypharmacology strategies, able to simultaneously target multiple actors involved in the obesity initiation and expansion, were developed during the last ten years opening a new promising avenue in the pharmacological management of obesity. In this review, we first describe the clinical features of obesity-associated conditions and then focus on the outcomes of currently approved drug therapies for obesity as well as new ones expecting to reach the clinic in the near future.


Assuntos
Cirurgia Bariátrica , Preparações Farmacêuticas , Humanos , Obesidade/tratamento farmacológico
9.
Front Neurosci ; 11: 224, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28484368

RESUMO

Under physiological conditions, the brain consumes over 20% of the whole body energy supply. The blood-brain barrier (BBB) allows dynamic interactions between blood capillaries and the neuronal network in order to provide an adequate control of molecules that are transported in and out of the brain. Alterations in the BBB structure and function affecting brain accessibility to nutrients and exit of toxins are found in a number of diseases, which in turn may disturb brain function and nutrient signaling. In this review we explore the major advances obtained in the understanding of the BBB development and how its structure impacts on function. Furthermore, we focus on the particularities of the barrier permeability in the hypothalamus, its role in metabolic control and the potential impact of hypothalamic BBB abnormities in metabolic related diseases.

10.
J Neuroinflammation ; 14(1): 91, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28446241

RESUMO

BACKGROUND: The consumption of large amounts of dietary fats is one of the most important environmental factors contributing to the development of obesity and metabolic disorders. GPR120 and GPR40 are polyunsaturated fatty acid receptors that exert a number of systemic effects that are beneficial for metabolic and inflammatory diseases. Here, we evaluate the expression and potential role of hypothalamic GPR120 and GPR40 as targets for the treatment of obesity. METHODS: Male Swiss (6-weeks old), were fed with a high fat diet (HFD, 60% of kcal from fat) for 4 weeks. Next, mice underwent stereotaxic surgery to place an indwelling cannula into the right lateral ventricle. intracerebroventricular (icv)-cannulated mice were treated twice a day for 6 days with 2.0 µL saline or GPR40 and GPR120 agonists: GW9508, TUG1197, or TUG905 (2.0 µL, 1.0 mM). Food intake and body mass were measured during the treatment period. At the end of the experiment, the hypothalamus was collected for real-time PCR analysis. RESULTS: We show that both receptors are expressed in the hypothalamus; GPR120 is primarily present in microglia, whereas GPR40 is expressed in neurons. Upon intracerebroventricular treatment, GW9508, a non-specific agonist for both receptors, reduced energy efficiency and the expression of inflammatory genes in the hypothalamus. Reducing GPR120 hypothalamic expression using a lentivirus-based approach resulted in the loss of the anti-inflammatory effect of GW9508 and increased energy efficiency. Intracerebroventricular treatment with the GPR120- and GPR40-specific agonists TUG1197 and TUG905, respectively, resulted in milder effects than those produced by GW9508. CONCLUSIONS: GPR120 and GPR40 act in concert in the hypothalamus to reduce energy efficiency and regulate the inflammation associated with obesity. The combined activation of both receptors in the hypothalamus results in better metabolic outcomes than the isolated activation of either receptor alone.


Assuntos
Metabolismo Energético/fisiologia , Ácidos Graxos Insaturados/biossíntese , Homeostase/fisiologia , Hipotálamo/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Animais , Linhagem Celular , Ácidos Graxos Insaturados/genética , Expressão Gênica , Inflamação/genética , Inflamação/metabolismo , Masculino , Camundongos , Microglia/metabolismo , Obesidade/genética , Obesidade/metabolismo , Receptores Acoplados a Proteínas G/genética
11.
Front Horm Res ; 48: 84-96, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28245454

RESUMO

Hypothalamic resistance to adipostatic actions of leptin is a hallmark of obesity. Studies have revealed that hypothalamic inflammation, triggered in response to the consumption of large amounts of dietary fat, is an important mechanism in the development of leptin resistance. In this chapter, we will review the work that paved the way linking neuroinflammation of the hypothalamus and defective leptin action in obesity.


Assuntos
Hipotálamo/imunologia , Inflamação/imunologia , Leptina/metabolismo , Neuroimunomodulação/imunologia , Obesidade/metabolismo , Animais , Humanos
12.
Nutrition ; 32(7-8): 863-70, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27138107

RESUMO

OBJECTIVE: This study aimed to investigate the effect of Passiflora edulis peel flour (PEPF) intake on hypothalamic neuropeptides messenger RNA expression, insulin sensitivity, and other metabolic parameters in Sprague-Dawley rats fed a high-fat (HF) diet. METHODS: Sprague-Dawley rats were divided in 3 groups: a control group, fed on a normal fat diet; a HF group, fed on a high-fat diet (35% fat [w/w]); and a high-fat Passiflora flour (HFPF) group, fed on a HF diet containing PEPF. The rats from the HFPF group as well as the HF group were kept on an HF diet for the first 4 wk to induce metabolic conditions related to obesity. Then the HFPF group was switched to a HF diet containing PEPF for additional 6 wk. Other groups were kept on normal-fat and HF diet without addition of PEPF during the whole period of experiment. The glucose tolerance and insulin sensitivity were evaluated through the glucose tolerance test (GTT) and the insulin tolerance test (ITT). Gut hormones and adipokines were measured through an immunoassay. The hypothalamic neuropeptides expression was assessed by real-time polymerase chain reaction. RESULTS: The PEPF intake increased the hypothalamic cocaine- and amphetamine-regulated transcript expression (CART) (P < 0.05), counteracted cumulative body weight gain (P < 0.001), decreased adiposity (P < 0.05) and leptin level (P < 0.01), whereas increased adiponectin (P < 0.01), glucose-dependent insulinotropic polypeptide (P < 0.01), and glucagon-like peptide-1 (GLP-1) (P < 0.001) improved the insulin sensitivity in diet-induced obesity rats by increasing the kITT (glucose disappearance rate) (P < 0.01), which was calculated during the ITT. Other gut hormones (peptide tyrosine tyrosine, pancreatic polypeptide, and amylin) and interleukins (IL) (IL-6, tumor necrosis factor-α, IL-1ß, and monocyte chemoattractant protein-1) were not changed by the PEPF intake. CONCLUSION: Our findings provide a further understanding of how the PEPF works as a dietary component to improve glucose homeostasis and demonstrate a molecular mechanism that may increase satiety by PEPF in diet-induced obesity.


Assuntos
Dieta Hiperlipídica , Hipotálamo/metabolismo , Incretinas/metabolismo , Resistência à Insulina/fisiologia , Neuropeptídeos/metabolismo , Passiflora , Adiponectina/metabolismo , Animais , Polipeptídeo Inibidor Gástrico/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Hipotálamo/efeitos dos fármacos , Leptina/metabolismo , Masculino , Camundongos , Modelos Animais , Neuropeptídeos/efeitos dos fármacos , Reação em Cadeia da Polimerase , Ratos Sprague-Dawley
13.
Br J Nutr ; 110(3): 447-55, 2013 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-23415177

RESUMO

The peel of the native Brazilian fruit jaboticaba is rich in anthocyanins, which are known for their anti-obesity effects in animal models. The aim of the present study was to evaluate the effects of freeze-dried jaboticaba peel powder (FDJPP) on a number of metabolic parameters in a model of diet-induced obesity. Mice (n 8 per group) were initially fed on a high-fat diet (HFD, 35% w/w) for 4 weeks and then switched to a HFD supplemented with FDJPP (1, 2 or 4% w/w) for an additional 6 weeks. Energy intake, weight loss, glucose tolerance, insulin resistance and lipid profile were determined, and the results were evaluated using ANOVA and Tukey's tests. The FDJPP exerted no protective effect on HFD-induced weight gain, hyperleptinaemia and glucose intolerance. However, the supplementation was effective to reduce insulin resistance, as evidenced in the insulin tolerance test, and subsequently confirmed by improved signal transduction through the insulin receptor/insulin receptor substrate-1/Akt/forkhead box protein pathway and by the attenuation of HFD-induced inflammation in the liver, verified by lower expressions of IL-1b and IL-6 and decreased phosphorylated IkB-a protein levels in all jaboticaba-treated mice. These results suggest that FDJPP may exert a protective role against obesity-associated insulin resistance.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Resistência à Insulina , Insulina/metabolismo , Myrtaceae , Obesidade/tratamento farmacológico , Fitoterapia , Preparações de Plantas/uso terapêutico , Análise de Variância , Animais , Antocianinas/farmacologia , Antocianinas/uso terapêutico , Fármacos Antiobesidade/farmacologia , Fármacos Antiobesidade/uso terapêutico , Suplementos Nutricionais , Frutas , Intolerância à Glucose , Inflamação/tratamento farmacológico , Inflamação/etiologia , Inflamação/metabolismo , Leptina/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos , Myrtaceae/química , Obesidade/etiologia , Obesidade/metabolismo , Preparações de Plantas/farmacologia , Pós , Receptor de Insulina/metabolismo , Transdução de Sinais , Aumento de Peso/efeitos dos fármacos
14.
J Med Food ; 13(6): 1402-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20874245

RESUMO

The goal of the this study was to evaluate the mutagenic/antimutagenic effects of conventional (BRS133) and transgenic (BRS 245 RR) soybeans (CS and TS, respectively) in vivo using the bone marrow micronucleus (MN) test, histopathological analysis, chromosome aberration test (CAT), and mitotic index (MI) determination. Six-week-old male Swiss mice were fed with pelleted commercial diet mixed with CS or TS at 10% or 20%. Two experimental designs (MN and CAT) were conducted simultaneously with 10 groups each during a 15-day period. Animals were treated with pelleted commercial diet, CS (10% or 20%), or TS (10% or 20%), and on day 14 they also received cyclophosphamide (CP) (50 mg/kg i.p.). The 10% and 20% CS and TS diets did not significantly decrease the frequencies of micronucleated polychromatic erythrocytes in bone marrow induced by CP. However, the CAT indicated that the 10% and 20% CS diets significantly (P < .05) protected nucleated bone marrow cells against chemical-induced mutagenesis and also produced a significant (P < .05) decrease in the total percentage of spontaneous aberrations. Among the treatments with TS, only the 10% TS diet reduced the percentage of total aberrations induced by CP. The results also indicated that the treatment with 20% TS alone significantly (P < .05) decreased the MI, indicating cytotoxic effects related to the treatment. Taken together, our results suggest that, under the tested conditions, TS and CS have antimutagenic properties and are not toxic.


Assuntos
Dieta , Alimentos Geneticamente Modificados , Glycine max , Sementes , Animais , Peso Corporal/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Aberrações Cromossômicas/induzido quimicamente , Produtos Agrícolas , Ciclofosfamida/toxicidade , Alimentos Geneticamente Modificados/efeitos adversos , Intestino Delgado/citologia , Fígado/citologia , Masculino , Camundongos , Testes para Micronúcleos , Índice Mitótico , Mutagênicos/toxicidade , Plantas Geneticamente Modificadas , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/prevenção & controle , Sementes/efeitos adversos , Sementes/química , Glycine max/efeitos adversos , Glycine max/química , Glycine max/genética , Estômago/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA