Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomolecules ; 13(10)2023 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-37892238

RESUMEN

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.


Asunto(s)
Aterosclerosis , Médula Ósea , Humanos , Ratones , Animales , Masculino , Femenino , Anciano , Médula Ósea/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas de Transferencia de Ésteres de Colesterol/genética , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Aterosclerosis/metabolismo , Colesterol/metabolismo , Ratones Transgénicos , Ratones Endogámicos C57BL
2.
Neurosci Lett ; 792: 136955, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36347339

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 2 , Insulina , Ratones , Animales , Orexinas/metabolismo , Insulina/metabolismo , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Hipotálamo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas del Tejido Nervioso/metabolismo
3.
Neurosci Lett ; 781: 136660, 2022 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-35489647

RESUMEN

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.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Hipotálamo , Receptores Acoplados a Proteínas G , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Hipotálamo/metabolismo , Ratones , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
5.
Neuroendocrinology ; 110(1-2): 92-104, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31104058

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Hipotálamo/metabolismo , Inflamación/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Palmitatos/farmacología , Proopiomelanocortina/metabolismo , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Humanos , Ácido Linoleico/farmacología , Masculino , Ratones , Obesidad/sangre , Obesidad/etiología , Células Madre Pluripotentes , ARN Ribosómico 16S
6.
Int J Obes (Lond) ; 43(12): 2361-2369, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31548571

RESUMEN

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.


Asunto(s)
Hipotálamo , Neuronas/fisiología , Obesidad/fisiopatología , Animales , Modelos Animales de Enfermedad , Humanos , Hipotálamo/citología , Hipotálamo/fisiopatología , Inflamación/fisiopatología , Ratones
7.
EBioMedicine ; 39: 448-460, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30522931

RESUMEN

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).


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Regulación hacia Abajo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Hipotálamo/metabolismo , Obesidad/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Peso Corporal , Línea Celular , Biología Computacional/métodos , Modelos Animales de Enfermedad , Ingestión de Energía , Metabolismo Energético , Ayuno/metabolismo , Humanos , Masculino , Ratones , Obesidad/inducido químicamente , Obesidad/metabolismo , Fenotipo , Análisis de Secuencia de ARN
8.
Lipids Health Dis ; 17(1): 90, 2018 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-29678181

RESUMEN

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.


Asunto(s)
Tejido Adiposo/metabolismo , Obesidad/genética , PPAR gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Tejido Adiposo/patología , Adulto , Animales , Estudios de Casos y Controles , Metabolismo Energético/genética , Femenino , Regulación de la Expresión Génica , Glucosa/metabolismo , Homeostasis/genética , Humanos , Masculino , Ratones , Obesidad/metabolismo , Obesidad/patología , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Transducción de Señal , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
9.
Mol Cell Endocrinol ; 460: 238-245, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28760600

RESUMEN

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.


Asunto(s)
Hipotálamo/patología , Inflamación/patología , Mitocondrias/patología , Obesidad/patología , Animales , Biomarcadores/metabolismo , Dieta Alta en Grasa , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , GTP Fosfohidrolasas/metabolismo , Hipotálamo/ultraestructura , Ratones , Mitocondrias/ultraestructura , Pruebas de Neutralización , Factor de Necrosis Tumoral alfa/metabolismo
10.
J Neuroinflammation ; 14(1): 91, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28446241

RESUMEN

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.


Asunto(s)
Metabolismo Energético/fisiología , Ácidos Grasos Insaturados/biosíntesis , Homeostasis/fisiología , Hipotálamo/metabolismo , Receptores Acoplados a Proteínas G/biosíntesis , Animales , Línea Celular , Ácidos Grasos Insaturados/genética , Expresión Génica , Inflamación/genética , Inflamación/metabolismo , Masculino , Ratones , Microglía/metabolismo , Obesidad/genética , Obesidad/metabolismo , Receptores Acoplados a Proteínas G/genética
11.
Nutrients ; 9(4)2017 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-28346369

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Ritmo Circadiano , Fructosa/efectos adversos , Hipotálamo/efectos de los fármacos , Melatonina/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Animales , Corticosterona/sangre , Relación Dosis-Respuesta a Droga , Intolerancia a la Glucosa , Hipotálamo/metabolismo , Masculino , Melatonina/administración & dosificación , Melatonina/análogos & derivados , Melatonina/orina , Fosforilación , Ratas , Ratas Sprague-Dawley
12.
J Neurosci ; 36(48): 12106-12116, 2016 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-27903721

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Encéfalo/inmunología , Depresión/inmunología , Inmunidad Innata/inmunología , Receptor Cross-Talk/inmunología , Serotonina/inmunología , Animales , Conducta Animal , Depresión/etiología , Masculino , Ratones , Ratones Endogámicos C3H , Microglía/inmunología , Factor de Necrosis Tumoral alfa/inmunología
13.
Aging (Albany NY) ; 9(1): 142-155, 2016 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-28039439

RESUMEN

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.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Lisofosfolípidos/metabolismo , Condicionamiento Físico Animal/fisiología , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal/fisiología , Esfingosina/análogos & derivados , Absorciometría de Fotón , Tejido Adiposo Pardo/diagnóstico por imagen , Envejecimiento/fisiología , Animales , Homeostasis/fisiología , Interleucina-6/sangre , Masculino , Ratones , Consumo de Oxígeno/fisiología , Ratas , Ratas Wistar , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Proteína Desacopladora 1/metabolismo
14.
Diabetes ; 65(3): 673-86, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26512023

RESUMEN

Apoptosis of hypothalamic neurons is believed to play an important role in the development and perpetuation of obesity. Similar to the hippocampus, the hypothalamus presents constitutive and stimulated neurogenesis, suggesting that obesity-associated hypothalamic dysfunction can be repaired. Here, we explored the hypothesis that n-3 polyunsaturated fatty acids (PUFAs) induce hypothalamic neurogenesis. Both in the diet and injected directly into the hypothalamus, PUFAs were capable of increasing hypothalamic neurogenesis to levels similar or superior to the effect of brain-derived neurotrophic factor (BDNF). Most of the neurogenic activity induced by PUFAs resulted in increased numbers of proopiomelanocortin but not NPY neurons and was accompanied by increased expression of BDNF and G-protein-coupled receptor 40 (GPR40). The inhibition of GPR40 was capable of reducing the neurogenic effect of a PUFA, while the inhibition of BDNF resulted in the reduction of global hypothalamic cell. Thus, PUFAs emerge as a potential dietary approach to correct obesity-associated hypothalamic neuronal loss.


Asunto(s)
Glucemia/efectos de los fármacos , Ácidos Grasos Omega-3/farmacología , Hipotálamo/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Animales , Glucemia/metabolismo , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Prueba de Tolerancia a la Glucosa , Hipotálamo/citología , Hipotálamo/metabolismo , Ratones , Neuronas/metabolismo , Neuropéptido Y , Proopiomelanocortina/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
PLoS One ; 10(3): e0119850, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25786112

RESUMEN

Autophagy is an important process that regulates cellular homeostasis by degrading dysfunctional proteins, organelles and lipids. In this study, the hypothesis that obesity could lead to impairment in hypothalamic autophagy in mice was evaluated by examining the hypothalamic distribution and content of autophagic proteins in animal with obesity induced by 8 or 16 weeks high fat diet to induce obesity and in response to intracerebroventricular injections of palmitic acid. The results showed that chronic exposure to a high fat diet leads to an increased expression of inflammatory markers and downregulation of autophagic proteins. In obese mice, autophagic induction leads to the downregulation of proteins, such as JNK and Bax, which are involved in the stress pathways. In neuron cell-line, palmitate has a direct effect on autophagy even without inflammatory activity. Understanding the cellular and molecular bases of overnutrition is essential for identifying new diagnostic and therapeutic targets for obesity.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales , Autofagia/fisiología , Ácidos Grasos/metabolismo , Hipotálamo/fisiología , Obesidad/fisiopatología , Análisis de Varianza , Animales , Línea Celular , Técnica del Anticuerpo Fluorescente , Prueba de Tolerancia a la Glucosa , Hipotálamo/metabolismo , Immunoblotting , MAP Quinasa Quinasa 4/metabolismo , Masculino , Ratones , Ratones Obesos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína X Asociada a bcl-2/metabolismo
16.
Endocrinology ; 156(1): 103-13, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25375037

RESUMEN

The anomalous activation of toll-like receptor 4 (TLR4) by dietary fats is one of the most important mechanisms linking obesity to insulin resistance. TLR4 is expressed in most tissues of the body, but its activity in the cells of the immune system is expected to underlie its most important roles of inducing inflammation and insulin resistance. Here we explore the hypothesis that TLR4 expression in bone marrow-derived cells mediates most of the actions of this receptor as an inducer of insulin resistance. Wild type and TLR4-mutant mice were used in bone marrow transplant experiments producing chimeras that harbored the functional receptor in all cells of the body except bone marrow-derived cells or only in bone marrow-derived cells. Transplanted mice were fed chow or a high-fat diet, and glucose homeostasis was evaluated by glucose and insulin tolerance tests. Insulin signal transduction and the expression of markers of inflammation were evaluated in the liver and white adipose tissue. In addition, we performed liver histology and evaluated the expression of gluconeogenic enzymes. The expression of TLR4 in bone marrow-derived cells only, but not in non-bone marrow-derived tissues only, was a determining factor in the induction of diet-induced insulin resistance, which was accompanied by an increased expression of inflammatory markers in both white adipose tissue and liver as well as increased liver steatosis and increased expression of gluconeogenic enzymes. TLR4 expressed in bone marrow-derived cells is an important mediator of obesity-associated insulin resistance in mice.


Asunto(s)
Grasas de la Dieta/efectos adversos , Regulación de la Expresión Génica/fisiología , Resistencia a la Insulina/fisiología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Receptor Toll-Like 4/metabolismo , Tejido Adiposo/metabolismo , Animales , Trasplante de Médula Ósea , Grasas de la Dieta/administración & dosificación , Inflamación/genética , Inflamación/metabolismo , Resistencia a la Insulina/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Obesidad/etiología , Obesidad/metabolismo , Proteínas Proto-Oncogénicas c-akt , Receptor Toll-Like 4/genética
17.
Nat Commun ; 5: 4859, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25255053

RESUMEN

Sphingosine 1-phosphate receptor 1 (S1PR1) is a G-protein-coupled receptor for sphingosine-1-phosphate (S1P) that has a role in many physiological and pathophysiological processes. Here we show that the S1P/S1PR1 signalling pathway in hypothalamic neurons regulates energy homeostasis in rodents. We demonstrate that S1PR1 protein is highly enriched in hypothalamic POMC neurons of rats. Intracerebroventricular injections of the bioactive lipid, S1P, reduce food consumption and increase rat energy expenditure through persistent activation of STAT3 and the melanocortin system. Similarly, the selective disruption of hypothalamic S1PR1 increases food intake and reduces the respiratory exchange ratio. We further show that STAT3 controls S1PR1 expression in neurons via a positive feedback mechanism. Interestingly, several models of obesity and cancer anorexia display an imbalance of hypothalamic S1P/S1PR1/STAT3 axis, whereas pharmacological intervention ameliorates these phenotypes. Taken together, our data demonstrate that the neuronal S1P/S1PR1/STAT3 signalling axis plays a critical role in the control of energy homeostasis in rats.


Asunto(s)
Metabolismo Energético , Hipotálamo/metabolismo , Lisofosfolípidos/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Ratas , Ratas Wistar , Receptores de Lisoesfingolípidos/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato
18.
Endocrinology ; 155(8): 2831-44, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24892821

RESUMEN

In both human and experimental obesity, inflammatory damage to the hypothalamus plays an important role in the loss of the coordinated control of food intake and energy expenditure. Upon prolonged maintenance of increased body mass, the brain changes the defended set point of adiposity, and returning to normal weight becomes extremely difficult. Here we show that in prolonged but not in short-term obesity, the ubiquitin/proteasome system in the hypothalamus fails to maintain an adequate rate of protein recycling, leading to the accumulation of ubiquitinated proteins. This is accompanied by an increased colocalization of ubiquitin and p62 in the arcuate nucleus and reduced expression of autophagy markers in the hypothalamus. Genetic protection from obesity is accompanied by the normal regulation of the ubiquitin/proteasome system in the hypothalamus, whereas the inhibition of proteasome or p62 results in the acceleration of body mass gain in mice exposed for a short period to a high-fat diet. Thus, the defective regulation of the ubiquitin/proteasome system in the hypothalamus may be an important mechanism involved in the progression and autoperpetuation of obesity.


Asunto(s)
Hipotálamo/metabolismo , Obesidad/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Animales , Autofagia , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Microglía/metabolismo , Neuronas/metabolismo , Fenotipo , Factor de Transcripción TFIIH , Factores de Transcripción/metabolismo , Aumento de Peso , Pérdida de Peso
19.
Am J Physiol Endocrinol Metab ; 305(2): E230-42, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23695212

RESUMEN

Melatonin can contribute to glucose homeostasis either by decreasing gluconeogenesis or by counteracting insulin resistance in distinct models of obesity. However, the precise mechanism through which melatonin controls glucose homeostasis is not completely understood. Male Wistar rats were administered an intracerebroventricular (icv) injection of melatonin and one of following: an icv injection of a phosphatidylinositol 3-kinase (PI3K) inhibitor, an icv injection of a melatonin receptor (MT) antagonist, or an intraperitoneal (ip) injection of a muscarinic receptor antagonist. Anesthetized rats were subjected to pyruvate tolerance test to estimate in vivo glucose clearance after pyruvate load and in situ liver perfusion to assess hepatic gluconeogenesis. The hypothalamus was removed to determine Akt phosphorylation. Melatonin injections in the central nervous system suppressed hepatic gluconeogenesis and increased hypothalamic Akt phosphorylation. These effects of melatonin were suppressed either by icv injections of PI3K inhibitors and MT antagonists and by ip injection of a muscarinic receptor antagonist. We conclude that melatonin activates hypothalamus-liver communication that may contribute to circadian adjustments of gluconeogenesis. These data further suggest a physiopathological relationship between the circadian disruptions in metabolism and reduced levels of melatonin found in type 2 diabetes patients.


Asunto(s)
Antioxidantes/farmacología , Gluconeogénesis/efectos de los fármacos , Hipotálamo/metabolismo , Hígado/metabolismo , Melatonina/farmacología , Proteína Oncogénica v-akt/metabolismo , Receptor de Melatonina MT1/efectos de los fármacos , Receptor de Melatonina MT2/efectos de los fármacos , Animales , Western Blotting , Técnica del Anticuerpo Fluorescente , Prueba de Tolerancia a la Glucosa , Hipotálamo/efectos de los fármacos , Inyecciones Intraventriculares , Hígado/efectos de los fármacos , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Ácido Pirúvico/metabolismo , Ratas , Ratas Wistar , Receptores Muscarínicos/efectos de los fármacos
20.
PLoS One ; 8(4): e62669, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23626844

RESUMEN

BACKGROUND: Hypothalamic AMPK acts as a cell energy sensor and can modulate food intake, glucose homeostasis, and fatty acid biosynthesis. Intrahypothalamic fatty acid injection is known to suppress liver glucose production, mainly by activation of hypothalamic ATP-sensitive potassium (K(ATP)) channels. Since all models employed seem to involve malonyl-CoA biosynthesis, we hypothesized that acetyl-CoA carboxylase can modulate the counter-regulatory response independent of nutrient availability. METHODOLOGY/PRINCIPAL FINDINGS: In this study employing immunoblot, real-time PCR, ELISA, and biochemical measurements, we showed that reduction of the hypothalamic expression of acetyl-CoA carboxylase by antisense oligonucleotide after intraventricular injection increased food intake and NPY mRNA, and diminished the expression of CART, CRH, and TRH mRNA. Additionally, as in fasted rats, in antisense oligonucleotide-treated rats, serum glucagon and ketone bodies increased, while the levels of serum insulin and hepatic glycogen diminished. The reduction of hypothalamic acetyl-CoA carboxylase also increased PEPCK expression, AMPK phosphorylation, and glucose production in the liver. Interestingly, these effects were observed without modification of hypothalamic AMPK phosphorylation. CONCLUSION/SIGNIFICANCE: Hypothalamic ACC inhibition can activate hepatic counter-regulatory response independent of hypothalamic AMPK activation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/genética , Hipotálamo/metabolismo , Hígado/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Animales , Peso Corporal , Dieta , Regulación de la Expresión Génica , Gluconeogénesis/fisiología , Hormonas/sangre , Masculino , Oligonucleótidos/administración & dosificación , Fosforilación , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...