Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomolecules ; 13(10)2023 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-37892238

RESUMO

CETP activity reduces plasma HDL-cholesterol concentrations, a correlate of an increased risk of atherosclerotic events. However, our recent findings suggest that CETP expression in macrophages promotes an intracellular antioxidant state, reduces free cholesterol accumulation and phagocytosis, and attenuates pro-inflammatory gene expression. To determine whether CETP expression in macrophages affects atherosclerosis development, we transplanted bone marrow from transgenic mice expressing simian CETP or non-expressing littermates into hypercholesterolemic LDL-receptor-deficient mice. The CETP expression did not change the lipid-stained lesion areas but decreased the macrophage content (CD68), neutrophil accumulation (LY6G), and TNF-α aorta content of young male transplanted mice and decreased LY6G, TNF-α, iNOS, and nitrotyrosine (3-NT) in aged female transplanted mice. These findings suggest that CETP expression in bone-marrow-derived cells reduces the inflammatory features of atherosclerosis. These novel mechanistic observations may help to explain the failure of CETP inhibitors in reducing atherosclerotic events in humans.


Assuntos
Aterosclerose , Medula Óssea , Humanos , Camundongos , Animais , Masculino , Feminino , Idoso , Medula Óssea/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas de Transferência de Ésteres de Colesterol/genética , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Aterosclerose/metabolismo , Colesterol/metabolismo , Camundongos Transgênicos , Camundongos Endogâmicos C57BL
2.
Am J Reprod Immunol ; 89(3): e13673, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36585861

RESUMO

BACKGROUND: Recurrent Pregnancy Loss (RPL) and Recurrent Implantation Failure (RIF) are highly heterogeneous condition and many of the mechanisms involved still require elucidation. The aim was to analyze the lipidomic profile in plasma of women with RPL and RIF before and after receiving the Lipid Emulsion Therapy (LET) containing 10% fish oil (SMOFlipid® 20%). METHODS: This study included twenty-six women with RPL or RIF from immunological or inflammatory causes, with elevated natural killer cell levels and divided into a Pregnancy Loss or a Live Birth group according to the outcome. The women received intravenous LET and sample collecting was done before the first, third and fifth dose of LET in the pregnant women. Ultra-performance liquid chromatography quadrupole time of flight mass spectrometry (UPLC-QTOF MS) and multivariate statistical methods were performed to evaluate the profile of phospholipids present in the women's plasma. RESULTS: An increase of phosphatidylcholines (PC) 40:8 and 36:5 levels with predominance of n6 polyunsaturated fatty acids (PUFA) was observed in plasma lipids of the Pregnancy Loss Group compared to Live Birth Group. We also observed an increase in the relative abundance of n3 PUFA-PC species (42:10 and 36:6) and LysoPC 15:0 with the long term use of LET. CONCLUSION: The greater availability of n3 PUFA in plasma of the pregnant women stemming from LET use can be considered advantageous regarding the alteration of the phospholipid profile and its postulated anti-inflammatory and immunomodulatory role.


Assuntos
Aborto Habitual , Ácidos Graxos Ômega-3 , Humanos , Feminino , Gravidez , Fosfolipídeos , Aborto Habitual/terapia , Aborto Habitual/etiologia , Ácidos Graxos Ômega-3/uso terapêutico , Emulsões Gordurosas Intravenosas , Cromatografia Líquida
3.
Neurosci Lett ; 792: 136955, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36347339

RESUMO

GPR139 is an orphan G-protein-coupled receptor that is expressed in restricted areas of the nervous system, including the hypothalamus. In this study, we hypothesized that GPR139 could be involved in the regulation of energy balance and metabolism. In the first part of the study, we confirmed that GPR139 is expressed in the hypothalamus and particularly in proopiomelanocortin and agouti-related peptide neurons of the mediobasal hypothalamus. Using a lentivirus with a short-hairpin RNA, we inhibited the expression of GPR139 bilaterally in the mediobasal hypothalamus of mice. The intervention promoted a 40% reduction in the hypothalamic expression of GPR139, which was accompanied by an increase in body mass, a reduction in fasting blood glucose levels, and an increase in insulin levels. In the hypothalamus, inhibition of GPR139 was accompanied by a reduction in the expression of orexin. As previous studies using a pharmacological antagonist of orexin showed a beneficial impact on type 2 diabetes and glucose metabolism, we propose that the inhibition of hypothalamic GPR139 could be acting indirectly through the orexin system to control systemic glucose and insulin. In conclusion, this study advances the characterization of GPR139 in the hypothalamus, demonstrating its involvement in the regulation of body mass, blood insulin, and glycemia.


Assuntos
Diabetes Mellitus Tipo 2 , Insulina , Camundongos , Animais , Orexinas/metabolismo , Insulina/metabolismo , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Hipotálamo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas do Tecido Nervoso/metabolismo
4.
Clin Nutr ESPEN ; 50: 326-329, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35871943

RESUMO

BACKGROUND & AIMS: Obesity courses with metabolic and inflammatory changes that include, among others, higher expression of the renin-angiotensin-aldosterone system. The pathophysiology of the new coronavirus suggests an affinity for angiotensin-2 converting enzyme receptors, cytokine storm, and systemic hypercoagulability. Thus, obesity could contribute to the worse evolution of individuals with COVID-19. Here we evaluated the clinical outcome and age of SARS-CoV-2 infection in patients with higher BMI compared with normal BMI at the São Francisco de Assis University Hospital (HUSF), in Bragança Paulista, SP. METHODS: Retrospective observational study with a review of medical records from June of 2020 to May of 2021 of patients positive for SARS-CoV-2 from HUSF. Demographic, anthropometric, and metabolic data were collected for correlation analysis. The study was approved by the Ethical Committee under CAAE: 34121820.3.0000.5514. RESULTS: 360 medical records were analyzed, of which 125 were included. The mean age of patients with obesity was significantly lower than overweight and normal weight, both in the overall mean (p-value 0.002-66 versus 56 and 56) and in the mean age of mortality (p-value 0.003-59 versus 61 and 76). The mean plasma calcium in the last sample collected during hospitalization of patients with obesity was significantly higher than that of overweight and normal weight (p-value < 0.001-7.8 versus 8.1 and 8.6). The mean hemoglobin in the first admission sample was also significantly higher in patients with obesity compared to the other groups (p-value 0.041-12.5 versus 12.9 and 13.6). On the other hand, the plasma concentration of urea in the first sample of hospitalization of patients with normal weight was higher than in patients with overweight and obesity (p-value 0.036-90.4 versus 64.8 and 57.1). CONCLUSION: Our findings suggest that age is not a determining factor for the death outcome in patients with obesity. However, obesity contributes to metabolic changes and mortality in SARS-CoV-2 infected patients.


Assuntos
COVID-19 , SARS-CoV-2 , Cálcio , Humanos , Obesidade , Estudos Observacionais como Assunto , Sobrepeso , Ureia
5.
Neurosci Lett ; 781: 136660, 2022 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-35489647

RESUMO

Currently, up to 35% off all drugs approved for the treatment of human diseases belong to the G-protein-coupled receptor (GPCR) family. Out of the almost 800 existing GPCRs, 25% have no known endogenous ligands and are regarded as orphan receptors; many of these are currently under investigation as potential pharmacological targets. Here, we hypothesised that orphan GPCRs expressed in the hypothalamus could be targets for the treatment of obesity and other metabolic diseases. Using bioinformatic tools, we identified 78 class A orphan GPCRs that are expressed in the hypothalamus of mice. Initially, we selected two candidates and determined their responsivities to nutritional interventions: GPR162, the GPCR with highest expression in the hypothalamus, and GPR68, a GPCR with intermediate expression in the hypothalamus and that has never been explored for its potential involvement in metabolic regulation. GPR162 expression was not modified by fasting/feeding or by the consumption of a high-fat diet, and was therefore not subsequently evaluated. Conversely, GPR68 expression increased in response to the consumption of a high-fat diet and reduced under fasting conditions. Using immunofluorescence, GPR68 was identified in both proopiomelanocortin-expressing and agouti-related peptide-expressing neurons in the hypothalamic arcuate nucleus. Acute inhibition of GPR68 with an allosteric modulator promoted an increase in the expression of the orexigenic agouti-related peptide and neuropeptide Y, whereas 4- and 12-h inhibition of GPR68 resulted in increased caloric intake. Thus, GPR68 has emerged as an orphan GPCR that is expressed in the hypothalamus and is involved in the regulation of feeding.


Assuntos
Núcleo Arqueado do Hipotálamo , Hipotálamo , Receptores Acoplados a Proteínas G , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Hipotálamo/metabolismo , Camundongos , Neuropeptídeo Y/metabolismo , Pró-Opiomelanocortina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
6.
J Cell Mol Med ; 25(5): 2290-2296, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33544456

RESUMO

Altered immune and/or inflammatory response plays an important role in cases of recurrent pregnancy loss (RPL) and repeated implantation failure (RIF). Exacerbation of the maternal immune response through increased NK cell activity and inflammatory cytokines can cause embryo rejection leading to abortion or embryo implantation failure. Immunosuppressors or immunomodulators can help or prevent this condition. Currently, lipid emulsion therapy (LET) has emerged as a treatment for RPL and RIF in women with abnormal NK cell activity, by decreasing the exacerbated immune response of the maternal uterus and providing a more receptive environment for the embryo. However, the mechanisms by which the intralipid acts to reduce NK cell activity are still unclear. In this review, we focus on the studies that conducted LET to treat patients with RPL and RIF with abnormal NK cell activity. We find that although some authors recommend LET as an effective intervention, more studies are necessary to confirm its effectiveness in restoring NK cell activity to normal levels and to comprehend the underlying mechanisms of the lipids action in ameliorating the maternal environment and improving the pregnancy rate.


Assuntos
Aborto Habitual/terapia , Lipídeos/uso terapêutico , Aborto Habitual/diagnóstico , Aborto Habitual/etiologia , Citocinas , Gerenciamento Clínico , Suscetibilidade a Doenças , Implantação do Embrião , Emulsões , Feminino , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Lipídeos/administração & dosagem , Ativação Linfocitária/genética , Gravidez , Resultado do Tratamento
7.
Proc Natl Acad Sci U S A ; 117(38): 23932-23941, 2020 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-32900951

RESUMO

DICER is a key enzyme in microRNA (miRNA) biogenesis. Here we show that aerobic exercise training up-regulates DICER in adipose tissue of mice and humans. This can be mimicked by infusion of serum from exercised mice into sedentary mice and depends on AMPK-mediated signaling in both muscle and adipocytes. Adipocyte DICER is required for whole-body metabolic adaptations to aerobic exercise training, in part, by allowing controlled substrate utilization in adipose tissue, which, in turn, supports skeletal muscle function. Exercise training increases overall miRNA expression in adipose tissue, and up-regulation of miR-203-3p limits glycolysis in adipose under conditions of metabolic stress. We propose that exercise training-induced DICER-miR-203-3p up-regulation in adipocytes is a key adaptive response that coordinates signals from working muscle to promote whole-body metabolic adaptations.


Assuntos
Tecido Adiposo/metabolismo , RNA Helicases DEAD-box/metabolismo , Exercício Físico/fisiologia , Ribonuclease III/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Adaptação Fisiológica/fisiologia , Adipócitos/metabolismo , Animais , Células Cultivadas , RNA Helicases DEAD-box/deficiência , RNA Helicases DEAD-box/genética , Feminino , Glicólise , Humanos , Masculino , Camundongos , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , Condicionamento Físico Animal , Ribonuclease III/deficiência , Ribonuclease III/genética
9.
Neuroendocrinology ; 110(1-2): 92-104, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31104058

RESUMO

In outbred mice, susceptibility or resistance to diet-induced obesity is associated with rapid changes in hypothalamic proopiomelanocortin (POMC) levels. Here, we evaluated 3 hypotheses that potentially explain the development of the different obesity phenotypes in outbred Swiss mice. First, rapid and differential changes in the gut microbiota in obesity-prone (OP) and obesity-resistant (OR) mice fed on a high-fat diet (HFD) might cause differential efficiencies in fatty acid harvesting leading to changes in systemic fatty acid concentrations that in turn affect POMC expression and processing. Second, independently of the gut microbiota, OP mice might have increased blood fatty acid levels after the introduction of a HFD, which could affect POMC expression and processing. Third, fatty acids might act directly in the hypothalamus to differentially regulate POMC expression and/or processing in OP and OR mice. We evaluated OP and OR male Swiss mice using 16S rRNA sequencing for the determination of gut microbiota; gas chromatography for blood lipid determination; and immunoblot and real-time polymerase chain reaction for protein and transcript determination and indirect calorimetry. Some experiments were performed with human pluripotent stem cells differentiated into hypothalamic neurons. We did not find evidence supporting the first 2 hypotheses. However, we found that in OP but not in OR mice, palmitate induces a rapid increase in hypothalamic POMC, which is followed by increased expression of proprotein convertase subtilisin/kexin type 1 PC1/3. Lentiviral inhibition of hypothalamic PC1/3 increased caloric intake and body mass in both OP and OR mice. In human stem cell-derived hypothalamic cells, we found that palmitate potently suppressed the production of POMC-derived peptides. Palmitate directly regulates PC1/3 in OP mice and likely has a functional impact on POMC processing.


Assuntos
Microbioma Gastrointestinal , Hipotálamo/metabolismo , Inflamação/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo , Palmitatos/farmacologia , Pró-Opiomelanocortina/metabolismo , Animais , Dieta Hiperlipídica , Modelos Animais de Doenças , Humanos , Ácido Linoleico/farmacologia , Masculino , Camundongos , Obesidade/sangue , Obesidade/etiologia , Células-Tronco Pluripotentes , RNA Ribossômico 16S
10.
J Chem Neuroanat ; 103: 101713, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31726089

RESUMO

The arcuate and the paraventricular and lateral hypothalamic nuclei, related to hunger and satiety control, are generally compromised by excess fatty acids. In this situation, fatty acids cause inflammation via TLR4 (toll like receptor 4) and the nuclei become less responsive to the hormones leptin and insulin, contributing to the development of obesity. In this work, these nuclei were analyzed in animals fed with high-fat diet and submitted to swimming without and with load for two months. For this, frontal sections of the hypothalamus were immunolabelled with GFAP (glial fibrillary acidic protein), synaptophysin, IL-6 (interleukin 6) and TLR4. Also, proteins extracted from the hypothalamus were analyzed using Western blotting (GFAP and synaptophysin), fluorometric analysis for caspases 3 and 7, and CBA (cytometric bead array) for Th1, Th2, and Th17 profiles. The high-fat diet significantly caused overweight and, in the hypothalamus, decreased synapses and increased astrocytic reactivity. The swimming with load, especially 80 % of the maximum load, reduced those consequences. The high-fat diet increased TLR4 in the arcuate nucleus and the swimming exercise with 80 % of the maximum load showed a tendency of reducing this expression. Swimming did not significantly influence the inflammatory or anti-inflammatory cytokines in the hypothalamus or in plasma. The high-fat diet in sedentary animals increased the expression of caspases 3 and 7 and swimming practice reduced this increment to levels compatible with animals fed on a normal diet. The set of results conclude that the impact of swimming on the damage caused in the hypothalamus by a high-fat diet is positive. The different aspects analyzed in here point to better cellular viability and conservation of the synapses in the hypothalamic nuclei of overweight animals that practiced swimming with a load.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Hipotálamo/metabolismo , Neurônios/metabolismo , Sobrepeso/metabolismo , Natação/fisiologia , Animais , Caspases/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Interleucina-6/metabolismo , Masculino , Camundongos , Sobrepeso/etiologia , Sinaptofisina/metabolismo , Receptor 4 Toll-Like/metabolismo
11.
Int J Obes (Lond) ; 43(12): 2361-2369, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31548571

RESUMO

The characterization of the hypothalamic neuronal network, that controls food intake and energy expenditure, has provided great advances in the understanding of the pathophysiology of obesity. Most of the advances in this field were obtained thanks to the development of a number of genetic and nongenetic animal models that, at least in part, overtook the anatomical constraints that impair the study of the human hypothalamus. Despite the undisputed differences between human and rodent physiology, most seminal studies undertaken in rodents that have unveiled details of the neural regulation of energy homeostasis were eventually confirmed in humans; thus, placing experimental studies in the forefront of obesity research. During the last 15 years, researchers have provided extensive experimental proof that supports the existence of hypothalamic dysfunction, which leads to a progressive whole-body positive energy balance, and thus, to obesity. Here, we review the experimental work that unveiled the mechanisms behind hypothalamic dysfunction in obesity.


Assuntos
Hipotálamo , Neurônios/fisiologia , Obesidade/fisiopatologia , Animais , Modelos Animais de Doenças , Humanos , Hipotálamo/citologia , Hipotálamo/fisiopatologia , Inflamação/fisiopatologia , Camundongos
12.
EBioMedicine ; 39: 448-460, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30522931

RESUMO

BACKGROUND: The Iroquois homeobox 3 (Irx3) gene has been identified as a functional long-range target of obesity-associated variants within the fat mass and obesity-associated protein (FTO) gene. It is highly expressed in the hypothalamus, and both whole-body knockout and hypothalamic restricted abrogation of its expression results in a lean phenotype, which is mostly explained by the resulting increased energy expenditure in the brown adipose tissue. Because of its potential implication in the pathogenesis of obesity, we evaluated the hypothalamic cell distribution of Irx3 and the outcomes of inhibiting its expression in a rodent model of diet-induced obesity. METHODS: Bioinformatics tools were used to evaluate the correlations between hypothalamic Irx3 and neurotransmitters, markers of thermogenesis and obesity related phenotypes. Droplet-sequencing analysis in >20,000 hypothalamic cells was used to explore the types of hypothalamic cells expressing Irx3. Lentivirus was used to inhibit hypothalamic Irx3 and the resulting phenotype was studied. FINDINGS: IRX3 is expressed predominantly in POMC neurons. Its expression is inhibited during prolonged fasting, as well as when mice are fed a high-fat diet. The partial inhibition of hypothalamic Irx3 using a lentivirus resulted in increased diet-induced body mass gain and adiposity due to increased caloric intake and reduced energy expenditure. INTERPRETATION: Contrary to the results obtained when lean mice are submitted to complete inhibition of Irx3, partial inhibition of hypothalamic Irx3 in obese mice causes an exacerbation of the obese phenotype. These data suggest that at least some of the Irx3 functions in the hypothalamus are regulated according to a hormetic pattern, and modulation of its expression can be a novel approach to modifying the body's energy-handling regulation. FUND: Sao Paulo Research Foundation grants 2013/07607-8 (LAV) and 2017/02983-2 (JDJ); NIH grants R01DK083567 (YBK).


Assuntos
Dieta Hiperlipídica/efeitos adversos , Regulação para Baixo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Hipotálamo/metabolismo , Obesidade/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Peso Corporal , Linhagem Celular , Biologia Computacional/métodos , Modelos Animais de Doenças , Ingestão de Energia , Metabolismo Energético , Jejum/metabolismo , Humanos , Masculino , Camundongos , Obesidade/induzido quimicamente , Obesidade/metabolismo , Fenótipo , Análise de Sequência de RNA
13.
Lipids Health Dis ; 17(1): 90, 2018 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-29678181

RESUMO

BACKGROUND: Peroxisome proliferator-activated receptor γ coactivator- 1alpha (PGC-1α) plays an important role in whole body metabolism and, particularly in glucose homeostasis. Its expression is highly regulated and, small variations in tissue levels can have a major impact in a number of physiological and pathological conditions. Recent studies have shown that the ubiquitin/proteasome system plays a role in the control of PGC-1α degradation. METHODS: Here we evaluated the interaction of PGC-1α with the protein A20, which plays a dual-role in the control of the ubiquitin/proteasome system acting as a deubiquitinase and as an E3 ligase. We employed immunoprecipitation, quantitative real-time PCR and immunofluorescence staining to evaluate PGC-1α, A20, PPARγ and ubiquitin in the adipose tissue of humans and mice. RESULTS: In distinct sites of the adipose tissue, A20 binds to PGC-1α. At least in the subcutaneous fat of humans and mice the levels of PGC-1α decrease during obesity, while its physical association with A20 increases. The inhibition of A20 leads to a reduction of PGC-1α and PPARγ expression, suggesting that A20 acts as a protective factor against PGC-1α disposal. CONCLUSION: We provide evidence that mechanisms regulating PGC-1α ubiquitination are potentially involved in the control of the function of this transcriptional co-activator.


Assuntos
Tecido Adiposo/metabolismo , Obesidade/genética , PPAR gama/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/genética , Tecido Adiposo/patologia , Adulto , Animais , Estudos de Casos e Controles , Metabolismo Energético/genética , Feminino , Regulação da Expressão Gênica , Glucose/metabolismo , Homeostase/genética , Humanos , Masculino , Camundongos , Obesidade/metabolismo , Obesidade/patologia , PPAR gama/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Transdução de Sinais , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
14.
Mol Cell Endocrinol ; 460: 238-245, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-28760600

RESUMO

Hypothalamic dysfunction is a common feature of experimental obesity. Studies have identified at least three mechanisms involved in the development of hypothalamic neuronal defects in diet-induced obesity: i, inflammation; ii, endoplasmic reticulum stress; and iii, mitochondrial abnormalities. However, which of these mechanisms is activated earliest in response to the consumption of large portions of dietary fats is currently unknown. Here, we used immunoblot, real-time PCR, mitochondrial respiration assays and transmission electron microscopy to evaluate markers of inflammation, endoplasmic reticulum stress and mitochondrial abnormalities in the hypothalamus of Swiss mice fed a high-fat diet for up to seven days. In the present study we show that the expression of the inflammatory chemokine fractalkine was the earliest event detected. Its hypothalamic expression increased as early as 3 h after the introduction of a high-fat diet and was followed by the increase of cytokines. GPR78, an endoplasmic reticulum chaperone, was increased 6 h after the introduction of a high-fat diet, however the actual triggering of endoplasmic reticulum stress was only detected three days later, when IRE-1α was increased. Mitofusin-2, a protein involved in mitochondrial fusion and tethering of mitochondria to the endoplasmic reticulum, underwent a transient reduction 24 h after the introduction of a high-fat diet and then increased after seven days. There were no changes in hypothalamic mitochondrial respiration during the experimental period, however there were reductions in mitochondria/endoplasmic reticulum contact sites, beginning three days after the introduction of a high-fat diet. The inhibition of TNF-α with infliximab resulted in the normalization of mitofusin-2 levels 24 h after the introduction of the diet. Thus, inflammation is the earliest mechanism activated in the hypothalamus after the introduction of a high-fat diet and may play a mechanistic role in the development of mitochondrial abnormalities in diet-induced obesity.


Assuntos
Hipotálamo/patologia , Inflamação/patologia , Mitocôndrias/patologia , Obesidade/patologia , Animais , Biomarcadores/metabolismo , Dieta Hiperlipídica , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático , GTP Fosfo-Hidrolases/metabolismo , Hipotálamo/ultraestrutura , Camundongos , Mitocôndrias/ultraestrutura , Testes de Neutralização , Fator de Necrose Tumoral alfa/metabolismo
15.
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
16.
Nutrients ; 9(4)2017 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-28346369

RESUMO

Recent studies show that the metabolic effects of fructose may vary depending on the phase of its consumption along with the light/dark cycle. Here, we investigated the metabolic outcomes of fructose consumption by rats during either the light (LPF) or the dark (DPF) phases of the light/dark cycle. This experimental approach was combined with other interventions, including restriction of chow availability to the dark phase, melatonin administration or intracerebroventricular inhibition of adenosine monophosphate-activated protein kinase (AMPK) with Compound C. LPF, but not DPF rats, exhibited increased hypothalamic AMPK phosphorylation, glucose intolerance, reduced urinary 6-sulfatoxymelatonin (6-S-Mel) (a metabolite of melatonin) and increased corticosterone levels. LPF, but not DPF rats, also exhibited increased chow ingestion during the light phase. The mentioned changes were blunted by Compound C. LPF rats subjected to dark phase-restricted feeding still exhibited increased hypothalamic AMPK phosphorylation but failed to develop the endocrine and metabolic changes. Moreover, melatonin administration to LPF rats reduced corticosterone and prevented glucose intolerance. Altogether, the present data suggests that consumption of fructose during the light phase results in out-of-phase feeding due to increased hypothalamic AMPK phosphorylation. This shift in spontaneous chow ingestion is responsible for the reduction of 6-S-Mel and glucose intolerance.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Ritmo Circadiano , Frutose/efeitos adversos , Hipotálamo/efeitos dos fármacos , Melatonina/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Corticosterona/sangue , Relação Dose-Resposta a Droga , Intolerância à Glucose , Hipotálamo/metabolismo , Masculino , Melatonina/administração & dosagem , Melatonina/análogos & derivados , Melatonina/urina , Fosforilação , Ratos , Ratos Sprague-Dawley
17.
J Neuroinflammation ; 14(1): 5, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-28086928

RESUMO

BACKGROUND: Diet-induced hypothalamic inflammation is an important mechanism leading to dysfunction of neurons involved in controlling body mass. Studies have shown that polyunsaturated fats can reduce hypothalamic inflammation. Here, we evaluated the presence and function of RvD2, a resolvin produced from docosahexaenoic acid, in the hypothalamus of mice. METHODS: Male Swiss mice were fed either chow or a high-fat diet. RvD2 receptor and synthetic enzymes were evaluated by real-time PCR and immunofluorescence. RvD2 was determined by mass spectrometry. Dietary and pharmacological approaches were used to modulate the RvD2 system in the hypothalamus, and metabolic phenotype consequences were determined. RESULTS: All enzymes involved in the synthesis of RvD2 were detected in the hypothalamus and were modulated in response to the consumption of dietary saturated fats, leading to a reduction of hypothalamic RvD2. GPR18, the receptor for RvD2, which was detected in POMC and NPY neurons, was also modulated by dietary fats. The substitution of saturated by polyunsaturated fats in the diet resulted in increased hypothalamic RvD2, which was accompanied by reduced body mass and improved glucose tolerance. The intracerebroventricular treatment with docosahexaenoic acid resulted in increased expression of the RvD2 synthetic enzymes, increased expression of anti-inflammatory cytokines and improved metabolic phenotype. Finally, intracerebroventricular treatment with RvD2 resulted in reduced adiposity, improved glucose tolerance and increased hypothalamic expression of anti-inflammatory cytokines. CONCLUSIONS: Thus, RvD2 is produced in the hypothalamus, and its receptor and synthetic enzymes are modulated by dietary fats. The improved metabolic outcomes of RvD2 make this substance an attractive approach to treat obesity.


Assuntos
Ácidos Docosa-Hexaenoicos/uso terapêutico , Encefalite/tratamento farmacológico , Encefalite/etiologia , Hipotálamo/metabolismo , Obesidade/complicações , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Proteínas de Ligação ao Cálcio/metabolismo , Citocinas/genética , Citocinas/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/química , Ácidos Docosa-Hexaenoicos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Teste de Tolerância a Glucose , Hipotálamo/patologia , Masculino , Camundongos , Proteínas dos Microfilamentos/metabolismo , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Obesidade/induzido quimicamente , Consumo de Oxigênio/fisiologia , Pró-Opiomelanocortina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
18.
J Neurosci ; 36(48): 12106-12116, 2016 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-27903721

RESUMO

Considerable clinical and epidemiological evidence links Alzheimer's disease (AD) and depression. However, the molecular mechanisms underlying this connection are largely unknown. We reported recently that soluble Aß oligomers (AßOs), toxins that accumulate in AD brains and are thought to instigate synapse damage and memory loss, induce depressive-like behavior in mice. Here, we report that the mechanism underlying this action involves AßO-induced microglial activation, aberrant TNF-α signaling, and decreased brain serotonin levels. Inactivation or ablation of microglia blocked the increase in brain TNF-α and abolished depressive-like behavior induced by AßOs. Significantly, we identified serotonin as a negative regulator of microglial activation. Finally, AßOs failed to induce depressive-like behavior in Toll-like receptor 4-deficient mice and in mice harboring a nonfunctional TLR4 variant in myeloid cells. Results establish that AßOs trigger depressive-like behavior via a double impact on brain serotonin levels and microglial activation, unveiling a cross talk between brain innate immunity and serotonergic signaling as a key player in mood alterations in AD. SIGNIFICANCE STATEMENT: Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the main cause of dementia in the world. Brain accumulation of amyloid-ß oligomers (AßOs) is a major feature in the pathogenesis of AD. Although clinical and epidemiological data suggest a strong connection between AD and depression, the underlying mechanisms linking these two disorders remain largely unknown. Here, we report that aberrant activation of the brain innate immunity and decreased serotonergic tonus in the brain are key players in AßO-induced depressive-like behavior in mice. Our findings may open up new possibilities for the development of effective therapeutics for AD and depression aimed at modulating microglial function.


Assuntos
Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/imunologia , Encéfalo/imunologia , Depressão/imunologia , Imunidade Inata/imunologia , Receptor Cross-Talk/imunologia , Serotonina/imunologia , Animais , Comportamento Animal , Depressão/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Microglia/imunologia , Fator de Necrose Tumoral alfa/imunologia
19.
Sci Rep ; 6: 29290, 2016 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-27373214

RESUMO

Obesity is the result of a long-term positive energy balance in which caloric intake overrides energy expenditure. This anabolic state results from the defective activity of hypothalamic neurons involved in the sensing and response to adiposity. However, it is currently unknown what the earliest obesity-linked hypothalamic defect is and how it orchestrates the energy imbalance present in obesity. Using an outbred model of diet-induced obesity we show that defective regulation of hypothalamic POMC is the earliest marker distinguishing obesity-prone from obesity-resistant mice. The early inhibition of hypothalamic POMC was sufficient to transform obesity-resistant in obesity-prone mice. In addition, the post-prandial change in the blood level of ß-endorphin, a POMC-derived peptide, correlates with body mass gain in rodents and humans. Taken together, these results suggest that defective regulation of POMC expression, which leads to a change of ß-endorphin levels, is the earliest hypothalamic defect leading to obesity.


Assuntos
Hipotálamo/metabolismo , Inflamação/metabolismo , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo , beta-Endorfina/metabolismo , Adolescente , Adulto , Animais , Dieta , Gorduras na Dieta/metabolismo , Ingestão de Energia , Humanos , Hipotálamo/imunologia , Inflamação/imunologia , Masculino , Camundongos , Camundongos Obesos , Obesidade/imunologia , Pró-Opiomelanocortina/imunologia , Ratos , Ratos Wistar , Adulto Jovem
20.
Aging (Albany NY) ; 9(1): 142-155, 2016 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-28039439

RESUMO

Recently, we demonstrated that the hypothalamic S1PR1/STAT3 axis plays a critical role in the control of food consumption and energy expenditure in rodents. Here, we found that reduction of hypothalamic S1PR1 expression occurs in an age-dependent manner, and was associated with defective thermogenic signaling and weight gain. To address the physiological relevance of these findings, we investigated the effects of chronic and acute exercise on the hypothalamic S1PR1/STAT3 axis. Chronic exercise increased S1PR1 expression and STAT3 phosphorylation in the hypothalamus, restoring the anorexigenic and thermogenic signals in middle-aged mice. Acutely, exercise increased sphingosine-1-phosphate (S1P) levels in the cerebrospinal fluid (CSF) of young rats, whereas the administration of CSF from exercised young rats into the hypothalamus of middle-aged rats at rest was sufficient to reduce the food intake. Finally, the intracerebroventricular (ICV) administration of S1PR1 activators, including the bioactive lipid molecule S1P, and pharmacological S1PR1 activator, SEW2871, induced a potent STAT3 phosphorylation and anorexigenic response in middle-aged rats. Overall, these results suggest that hypothalamic S1PR1 is important for the maintenance of energy balance and provide new insights into the mechanism by which exercise controls the anorexigenic and thermogenic signals in the central nervous system during the aging process.


Assuntos
Metabolismo Energético/fisiologia , Hipotálamo/metabolismo , Lisofosfolipídeos/metabolismo , Condicionamento Físico Animal/fisiologia , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais/fisiologia , Esfingosina/análogos & derivados , Absorciometria de Fóton , Tecido Adiposo Marrom/diagnóstico por imagem , Envelhecimento/fisiologia , Animais , Homeostase/fisiologia , Interleucina-6/sangue , Masculino , Camundongos , Consumo de Oxigênio/fisiologia , Ratos , Ratos Wistar , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Proteína Desacopladora 1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...