Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Mol Metab ; 79: 101840, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38036170

RESUMEN

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.


Asunto(s)
Ácidos Grasos no Esterificados , Proopiomelanocortina , Ratones , Animales , Ácidos Grasos no Esterificados/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Ratones Obesos , Peso Corporal , Hipotálamo/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Metabolismo Energético/fisiología
2.
Am J Physiol Endocrinol Metab ; 324(2): E154-E166, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36598900

RESUMEN

Maternal obesity is an important risk factor for obesity, cardiovascular, and metabolic diseases in the offspring. Studies have shown that it leads to hypothalamic inflammation in the progeny, affecting the function of neurons regulating food intake and energy expenditure. In adult mice fed a high-fat diet, one of the hypothalamic abnormalities that contribute to the development of obesity is the damage of the blood-brain barrier (BBB) at the median eminence-arcuate nucleus (ME-ARC) interface; however, how the hypothalamic BBB is affected in the offspring of obese mothers requires further investigation. Here, we used confocal and transmission electron microscopy, transcript expression analysis, glucose tolerance testing, and a cross-fostering intervention to determine the impact of maternal obesity and breastfeeding on BBB integrity at the ME-ARC interface. The offspring of obese mothers were born smaller; conversely, at weaning, they presented larger body mass and glucose intolerance. In addition, maternal obesity-induced structural and functional damage of the offspring's ME-ARC BBB. By a cross-fostering intervention, some of the defects in barrier integrity and metabolism seen during development in an obesogenic diet were recovered. The offspring of obese dams breastfed by lean dams presented a reduction of body mass and glucose intolerance as compared to the offspring continuously exposed to an obesogenic environment during intrauterine and perinatal life; this was accompanied by partial recovery of the anatomical structure of the ME-ARC interface, and by the normalization of transcript expression of genes coding for hypothalamic neurotransmitters involved in energy balance and BBB integrity. Thus, maternal obesity promotes structural and functional damage of the hypothalamic BBB, which is, in part, reverted by lactation by lean mothers.NEW & NOTEWORTHY Maternal dietary habits directly influence offspring health. In this study, we aimed at determining the impact of maternal obesity on BBB integrity. We show that DIO offspring presented a leakier ME-BBB, accompanied by changes in the expression of transcripts encoding for endothelial and tanycytic proteins, as well as of hypothalamic neuropeptides. Breastfeeding in lean dams was sufficient to protect the offspring from ME-BBB disruption, providing a preventive strategy of nutritional intervention during early life.


Asunto(s)
Intolerancia a la Glucosa , Obesidad Materna , Humanos , Femenino , Animales , Ratones , Embarazo , Barrera Hematoencefálica/metabolismo , Eminencia Media/metabolismo , Obesidad Materna/metabolismo , Madres , Intolerancia a la Glucosa/metabolismo , Obesidad/metabolismo , Hipotálamo/metabolismo , Dieta Alta en Grasa/efectos adversos , Fenómenos Fisiologicos Nutricionales Maternos
3.
J Neuroinflammation ; 18(1): 192, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34465367

RESUMEN

BACKGROUND: Interleukin-6 (IL6) produced in the context of exercise acts in the hypothalamus reducing obesity-associated inflammation and restoring the control of food intake and energy expenditure. In the hippocampus, some of the beneficial actions of IL6 are attributed to its neurogenesis-inducing properties. However, in the hypothalamus, the putative neurogenic actions of IL6 have never been explored, and its potential to balance energy intake can be an approach to prevent or attenuate obesity. METHODS: Wild-type (WT) and IL6 knockout (KO) mice were employed to study the capacity of IL6 to induce neurogenesis. We used cell labeling with Bromodeoxyuridine (BrdU), immunofluorescence, and real-time PCR to determine the expression of markers of neurogenesis and neurotransmitters. We prepared hypothalamic neuroprogenitor cells from KO that were treated with IL6 in order to provide an ex vivo model to further characterizing the neurogenic actions of IL6 through differentiation assays. In addition, we analyzed single-cell RNA sequencing data and determined the expression of IL6 and IL6 receptor in specific cell types of the murine hypothalamus. RESULTS: IL6 expression in the hypothalamus is low and restricted to microglia and tanycytes, whereas IL6 receptor is expressed in microglia, ependymocytes, endothelial cells, and astrocytes. Exogenous IL6 reduces diet-induced obesity. In outbred mice, obesity-resistance is accompanied by increased expression of IL6 in the hypothalamus. IL6 induces neurogenesis-related gene expression in the hypothalamus and in neuroprogenitor cells, both from WT as well as from KO mice. CONCLUSION: IL6 induces neurogenesis-related gene expression in the hypothalamus of WT mice. In KO mice, the neurogenic actions of IL6 are preserved; however, the appearance of new fully differentiated proopiomelanocortin (POMC) and neuropeptide Y (NPY) neurons is either delayed or disturbed.


Asunto(s)
Hipotálamo/metabolismo , Interleucina-6/genética , Neurogénesis/genética , Neuronas/metabolismo , Obesidad/genética , Animales , Metabolismo Energético/fisiología , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/metabolismo , Hipotálamo/efectos de los fármacos , Interleucina-6/metabolismo , Interleucina-6/farmacología , Masculino , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Obesidad/metabolismo , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo
4.
Nutr Res ; 76: 52-70, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32155506

RESUMEN

Most phenolic compounds and dietary fiber reach intact to the colon. We hypothesized that grape peel powder (GPP), a rich source of these bioactive compounds, modulates inflammatory and oxidative pathways collaborating to attenuate colonic damage in experimental colitis. To determine which bioactive fraction would be responsible for this effect, the aim of this study was to evaluate the effect of dietary supplementation with whole GPP or the isolated bioactive-rich fractions from GPP (extractable polyphenols [EP], dietary fiber and fiber-bound polyphenols [NEP-F], and dietary fiber) in rats with experimental colitis. Colitis was induced by intrarectal injection of 2,4,6-trinitrobenzene sulfonic acid (TNBS) after 15 days of dietary supplementation. EP diet did not reverse the decrease in feed intake and indeed worsened colon shortening and increased spleen weight; however, these effects were not observed for the GPP group, which had polyphenols associated to the matrix besides the extractable ones. Colitis impaired the activity of colonic antioxidant enzymes and increased lipid peroxidation, protein oxidation, nitric oxide (NO) levels, and proinflammatory cytokines in serum and in the colon tissue. GPP restored the activity of antioxidant enzymes and decreased colon oxidation and NO levels. All grape peel fractions reduced the protein expression of the inhibitor of kappa kinase beta and NO levels in colon tissue, but only NEP-F reduced the expression of phosphorylated nuclear factor kappa B and myeloperoxidase activity. Results demonstrated that GPP attenuates inflammatory and oxidative response in TNBS-induced colitis by downregulating the nuclear factor kappa B pathway and upregulating antioxidant enzymes, with NEP-F being the fraction most likely associated to these protective effects.


Asunto(s)
Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Colitis/tratamiento farmacológico , Colon/efectos de los fármacos , FN-kappa B/metabolismo , Polifenoles/uso terapéutico , Vitis/química , Animales , Antiinflamatorios/farmacología , Antioxidantes/metabolismo , Antioxidantes/farmacología , Colitis/complicaciones , Colitis/metabolismo , Colitis/patología , Colon/metabolismo , Colon/patología , Citocinas/metabolismo , Fibras de la Dieta , Frutas , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Peroxidación de Lípido , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fitoterapia , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Polifenoles/farmacología , Ratas Wistar , Transducción de Señal , Superóxido Dismutasa/metabolismo , Ácido Trinitrobencenosulfónico
5.
Biomed Res Int ; 2019: 1365210, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31534953

RESUMEN

Interleukin-6 (IL-6) is a unique cytokine that can play both pro- and anti-inflammatory roles depending on the anatomical site and conditions under which it has been induced. Specific neurons of the hypothalamus provide important signals to control food intake and energy expenditure. In individuals with obesity, a microglia-dependent inflammatory response damages the neural circuits responsible for maintaining whole-body energy homeostasis, resulting in a positive energy balance. However, little is known about the role of IL-6 in the regulation of hypothalamic microglia. In this systematic review, we asked what types of conditions and stimuli could modulate microglial IL-6 expression in murine model. We searched the PubMed and Web of Science databases and analyzed 13 articles that evaluated diverse contexts and study models focused on IL-6 expression and microglia activation, including the effects of stress, hypoxia, infection, neonatal overfeeding and nicotine exposure, lipopolysaccharide stimulus, hormones, exercise protocols, and aging. The results presented in this review emphasized the role of "injury-like" stimuli, under which IL-6 acts as a proinflammatory cytokine, concomitant with marked microglial activation, which drive hypothalamic neuroinflammation. Emerging evidence indicates an important correlation of basal IL-6 levels and microglial function with the maintenance of hypothalamic homeostasis. Advances in our understanding of these different contexts will lead to the development of more specific pharmacological approaches for the management of acute and chronic conditions, like obesity and metabolic diseases, without disturbing the homeostatic functions of IL-6 and microglia in the hypothalamus.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Hipotálamo/inmunología , Interleucina-6/inmunología , Enfermedades Metabólicas/inmunología , Microglía/inmunología , Obesidad/inmunología , Animales , Humanos , Hipotálamo/patología , Enfermedades Metabólicas/patología , Ratones , Microglía/patología , Obesidad/patología
6.
J Cell Biochem ; 120(10): 18186-18192, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31144370

RESUMEN

Obesity and aging lead to abnormal transforming growth factor-ß1 (TGF-ß1) signaling in the hypothalamus, triggering the imbalance on glucose metabolism and energy homeostasis. Here, we determine the effect of acute exercise on TGF-ß1 expression in the hypothalamus of two models of obesity in mice. The bioinformatics analysis was performed to evaluate the correlation between hypothalamic Tgf-ß1 messenger RNA (mRNA) and genes related to thermogenesis in the brown adipose tissue (BAT) by using a large panel of isogenic BXD mice. Thereafter, leptin-deficient (ob/ob) mice and obese C57BL/6 mice fed on a high-fat diet (HFD) were submitted to the acute exercise protocol. Transcriptomic analysis by using BXD mouse reference population database revealed that hypothalamic Tgf-ß1 mRNA is negatively correlated with genes related to thermogenesis in brown adipose tissue of BXD mice, such as peroxisome proliferator-activated receptor gamma coactivator and is positively correlated with respiratory exchange ratio. In agreement with these results, leptin-deficient (ob/ob) and HFD-fed mice displayed high levels of Tgf-ß1 mRNA in the hypothalamus and reduction of Pgc1α mRNA in BAT. Interestingly, an acute exercise session reduced TGF-ß1 expression in the hypothalamus, increased Pgc1α mRNA in the BAT and reduced food consumption in obese mice. Our results demonstrated that acute physical exercise suppressed hypothalamic TGF-ß1 expression, increasing Pgc1α mRNA in BAT in obese mice.


Asunto(s)
Regulación hacia Abajo , Hipotálamo/metabolismo , Obesidad/genética , Condicionamiento Físico Animal/fisiología , Factor de Crecimiento Transformador beta1/genética , Tejido Adiposo Pardo/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/genética , Perfilación de la Expresión Génica/métodos , Leptina/deficiencia , Leptina/genética , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/etiología , Obesidad/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Termogénesis/genética , Factor de Crecimiento Transformador beta1/metabolismo
7.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1126-1137, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30738810

RESUMEN

In experimental obesity, the hypothalamus is affected by an inflammatory response activated by dietary saturated fats. This inflammation is triggered as early as one day after exposure to a high-fat diet, and during its progression, there is recruitment of inflammatory cells from the systemic circulation. The objective of the present study was identifying chemokines potentially involved in the development of hypothalamic diet-induced inflammation. In order to identify chemokines potentially involved in this process, we performed a real-time PCR array that determined Ackr2 as one of the transcripts undergoing differential regulation in obese-prone as compared to obese-resistant mice fed a high-fat diet for three days. ACKR2 is a decoy receptor that acts as an inhibitor of the signals generated by several CC inflammatory chemokines. Our results show that Ackr2 expression is rapidly induced after exposure to dietary fats both in obese-prone and obese-resistant mice. In immunofluorescence studies, ACKR2 was detected in hypothalamic neurons expressing POMC and NPY and also in microglia and astrocytes. The lentiviral overexpression of ACKR2 in the hypothalamus reduced diet-induced hypothalamic inflammation; however, there was no change in spontaneous caloric intake and body mass. Nevertheless, the overexpression of ACKR2 resulted in improvement of glucose tolerance, which was accompanied by reduced insulin secretion and increased whole body insulin sensitivity. Thus, ACKR2 is a decoy chemokine receptor expressed in most hypothalamic cells that is modulated by dietary intervention and acts to reduce diet-induced inflammation, leading to improved glucose tolerance due to improved insulin action.


Asunto(s)
Perfilación de la Expresión Génica , Glucosa/metabolismo , Hipotálamo/metabolismo , Inflamación/genética , Obesidad/genética , Receptores de Quimiocina/genética , Animales , Astrocitos/metabolismo , Citocinas/genética , Citocinas/metabolismo , Dieta Alta en Grasa/efectos adversos , Prueba de Tolerancia a la Glucosa , Hipotálamo/citología , Inflamación/etiología , Inflamación/metabolismo , Resistencia a la Insulina/genética , Masculino , Ratones , Neuronas/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Receptores de Quimiocina/metabolismo
8.
PLoS One ; 14(1): e0210059, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30608959

RESUMEN

BACKGROUND: The use of medicinal plants and their derivatives is increasing, and approximately one-third of all traditional herbal medicines are intended for wound treatment. Natural products used in these treatments include vegetable oils, which are rich in essential fatty acids. Once in contact with an ulcerative surface, the oil reaches the blood and lymphatic vessels, thus eliciting systemic effects. OBJECTIVE: This study evaluated the local and possible systemic effects of essential fatty acids (sunflower oil) applied topically to rat wounds. METHODS: Cutaneous punch wounds (6 mm) were produced on the dorsa of 30 rats. Saline (SS), mineral oil (MO) or essential fatty acid (EFA) solutions were applied topically. Healing was evaluated after 2, 4 and 10 days (n = 5 per group) by visual and histological/morphometric examination, second harmonic generation (SHG) microscopy, and cytokine and growth factor quantification in the scar tissue (real-time PCR) and in serum (ELISA). RESULTS: MO/EFA-treated animals had higher IGF-1, leptin, IL-6 and IFN-γ mRNA expression and lower serum IL-6 levels than the control (SS/MO) animals. SHG analysis showed no difference in collagen density between the animals treated with MO and EFA. CONCLUSION: EFA treatment induces topical (observed by local IGF-1, leptin, IL-6 and IFN-γ production) and systemic effects, lowering IL-6 levels in the serum. As the oil is widely used to shorten ulcer healing time, studies are needed to evaluate the treatment safety and possible undesired effects.


Asunto(s)
Ácidos Grasos Esenciales/sangre , Cicatrización de Heridas/fisiología , Animales , Femenino , Factor I del Crecimiento Similar a la Insulina/metabolismo , Interferón gamma/sangre , Interleucina-6/sangre , Leptina/sangre , Ratas , Ratas Sprague-Dawley
9.
J Cell Physiol ; 233(12): 9426-9436, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30063084

RESUMEN

Hypothalamic sphingosine-1-phosphate receptor 1 (S1PR1), the G protein-coupled receptor 1 of sphingosine-1-phosphate, has been described as a modulator in the control of energy homeostasis in rodents. However, this mechanism is still unclear. Here, we evaluate the role of interleukin 6 (IL-6) associated with acute physical exercise in the control of the hypothalamic S1PR1-signal transducer and activator of transcription 3 (STAT3) axis. Acute exercise session and an intracerebroventricular IL-6 injection increased S1PR1 protein content and STAT3 phosphorylation in the hypothalamus of lean and obese mice accompanied by a reduction in food consumption. Transcriptome analysis indicated a strong positive correlation between Il-6 and S1pr1 messenger RNA in several tissues of genetically diverse BXD mice strains and humans, including in the hypothalamus. Interestingly, exercise failed to stimulate the S1PR1-STAT3 axis in IL-6 knockout mice and the disruption of hypothalamic-specific IL-6 action blocked the anorexigenic effects of exercise. Taken together, our results indicate that physical exercise modulates the S1PR1 protein content in the hypothalamus, through the central action of IL-6.


Asunto(s)
Hipotálamo/metabolismo , Interleucina-6/metabolismo , Condicionamiento Físico Animal , Receptores de Lisoesfingolípidos/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Humanos , Inyecciones Intraventriculares , Interleucina-6/administración & dosificación , Interleucina-6/genética , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Lisoesfingolípidos/genética , Receptores de Esfingosina-1-Fosfato
10.
J Nutr Biochem ; 53: 9-19, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29175142

RESUMEN

The "first hit" to atherogenesis is driven by toll-like receptor 4, endoplasmic reticulum stress and ultimately metabolic dysfunction. In this study, we hypothesized that a flaxseed oil-enriched diet (FS) abolishes these inflammatory signaling pathway and restore metabolic homeostasis by activating the fatty acid receptor GPR120 in aorta of obese mice. Glucose homeostasis was assessed by GTT and ITT; lipidomics was performed using a Hybrid Ion Trap-Orbitrap Mass Spectrometer; serum lipids were measured using colorimetric assays; GPR120 and infiltrating macrophages were analyzed by immunofluorescence; protein immunoprecipitation and gene expression were evaluated by Western blot and RT-PCR, respectively. There were no differences in body weight and food intake between the groups from both strains (Swiss and LDLr-KO mice). GTT and cholesterol levels were improved by FS in both mice models. Lipidomics showed an increase in ω3 (C18:3) content, meanwhile stearic acid (C18:0) was not detected in endothelial tissue in response to FS. Moreover, FS markedly decreased pro-inflammatory (IL-1ß, TNF-α, pIκBα, pIKKß) and unfolded protein response markers (ATF6 and GRP78) in aorta. In Swiss mice, GPR120 was partially involved in the ω3-mediated anti-inflammatory actions, disrupting TLR4 pathway, but not in LDLr-KO mice. Partial replacement of dietary saturated by unsaturated ω3 fatty acids contributes to inhibition of cardiovascular risk markers, pro-inflammatory cytokines and ER stress sensors and effectors in the aorta. However, downregulation of inflammation is not mediated by arterial GPR120 activation.


Asunto(s)
Aortitis/prevención & control , Estrés del Retículo Endoplásmico/efectos de los fármacos , Aceite de Linaza/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Aortitis/metabolismo , Modelos Animales de Enfermedad , Dislipidemias/dietoterapia , Dislipidemias/fisiopatología , Chaperón BiP del Retículo Endoplásmico , Ácidos Grasos Omega-3/farmacología , Aceite de Linaza/química , Lípidos/sangre , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Obesidad/dietoterapia , Obesidad/fisiopatología , Sustancias Protectoras/farmacología , Receptores de LDL/genética
11.
PLoS One ; 12(12): e0188779, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29236720

RESUMEN

INTRODUCTION: Wounds are a common health problem. Coffee is widely consumed and its oil contains essential fatty acids. We evaluated the local (skin) and systemic effects associated with the topical use of coffee oils in rats. METHODS: Punch skin wounds (6 mm) incisions were generated on the backs of 75 rats. Saline (SS), mineral oil (MO), green coffee oil (GCO), roasted coffee oil (RCO), green coffee ground oil (GCGO) or roasted coffee ground oil (RCGO) were topically applied to the wounds. Healing was evaluated by visual and histological/morphometric optical microscopy examination; second harmonics generation (SHG) microscopy, wound tissue q-PCR (values in fold-change) and blood serum (ELISA, values in pg/mL). RESULTS: RCO treated animals presented faster wound healing (0.986 vs. 0.422), higher mRNA expression of IGF-1 (2.78 vs. 1.00, p = 0.01), IL-6 (10.72 vs. 1.00, p = 0.001) and IL-23 (4.10 vs. 1.2, p = 0.05) in early stages of wound healing; higher IL-12 (3.32 vs. 1.00, p = 0.05) in the later stages; and lower serum levels of IFN-γ (11.97 vs. 196.45, p = 0.01). GCO treatment led to higher mRNA expression of IL-6 (day 2: 7.94 vs. 1.00, p = 0.001 and day 4: 6.90 vs. 1.00, p = 0.01) and IL-23 (7.93 vs. 1.20, p = 0.001) in the early stages. The RCO treatment also produced higher serum IFN-α levels throughout the experiment (day 2: 52.53 vs. 21.20; day 4: 46.98 vs.21.56; day 10: 83.61 vs. 25.69, p = 0.05) and lower levels of IL-4 (day 4: 0.9 vs.13.36, p = 0.01), adiponectin (day 10: 8,367.47 vs. 16,526.38, p = 0.001) and IFN-γ (day 4: 43.03 vs.196.45, p = 0.05). The SHG analysis showed a higher collagen density in the RCO and GCO treatments (p = 0.05). CONCLUSION: Topical treatment with coffee oils led to systemic actions and faster wound healing in rats. Further studies should be performed are necessary to assess the safety of topical vegetal oil use for skin lesions.


Asunto(s)
Café , Modelos Biológicos , Aceites de Plantas/administración & dosificación , Piel/lesiones , Cicatrización de Heridas/efectos de los fármacos , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Aceites de Plantas/química , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
J Neuroinflammation ; 14(1): 178, 2017 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-28865476

RESUMEN

BACKGROUND: The consumption of large amounts of dietary fats can trigger an inflammatory response in the hypothalamus and contribute to the dysfunctional control of caloric intake and energy expenditure commonly present in obesity. The objective of this study was to identify chemokine-related transcripts that could be involved in the early stages of diet-induced hypothalamic inflammation. METHODS: We used immunoblot, PCR array, real-time PCR, immunofluorescence staining, glucose and insulin tolerance tests, and determination of general metabolic parameters to evaluate markers of inflammation, body mass variation, and glucose tolerance in mice fed a high-fat diet. RESULTS: Using a real-time PCR array, we identified leukemia inhibitory factor as a chemokine/cytokine undergoing a rapid increase in the hypothalamus of obesity-resistant and a rapid decrease in the hypothalamus of obesity-prone mice fed a high-fat diet for 1 day. We hypothesized that the increased hypothalamic expression of leukemia inhibitory factor could contribute to the protective phenotype of obesity-resistant mice. To test this hypothesis, we immunoneutralized hypothalamic leukemia inhibitory factor and evaluated inflammatory and metabolic parameters. The immunoneutralization of leukemia inhibitory factor in the hypothalamus of obesity-resistant mice resulted in increased body mass gain and increased adiposity. Body mass gain was mostly due to increased caloric intake and reduced spontaneous physical activity. This modification in the phenotype was accompanied by increased expression of inflammatory cytokines in the hypothalamus. In addition, the inhibition of hypothalamic leukemia inhibitory factor was accompanied by glucose intolerance and insulin resistance. CONCLUSION: Hypothalamic expression of leukemia inhibitory factor may protect mice from the development of diet-induced obesity; the inhibition of this protein in the hypothalamus transforms obesity-resistant into obesity-prone mice.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Hipotálamo/metabolismo , Factor Inhibidor de Leucemia/antagonistas & inhibidores , Factor Inhibidor de Leucemia/biosíntesis , Obesidad/metabolismo , Fenotipo , Animales , Ingestión de Energía/efectos de los fármacos , Ingestión de Energía/fisiología , Hipotálamo/efectos de los fármacos , Inmunoglobulina G/farmacología , Masculino , Ratones , Obesidad/etiología , Distribución Aleatoria
13.
Aging (Albany NY) ; 9(8): 1926-1940, 2017 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-28854149

RESUMEN

Obesity and aging are associated with hypothalamic inflammation, hyperphagia and abnormalities in the thermogenesis control. It has been demonstrated that the association between aging and obesity induces hypothalamic inflammation and metabolic disorders, at least in part, through the atypical hypothalamic transforming growth factor-ß (TGF-ß1). Physical exercise has been used to modulate several metabolic parameters. Thus, the aim of this study was to evaluate the impact of chronic exercise on TGF-ß1 expression in the hypothalamus of Middle-Aged mice submitted to a one year of high-fat diet (HFD) treatment. We observed that long-term of HFD-feeding induced hypothalamic TGF-ß1 accumulation, potentiated the hypothalamic inflammation, body weight gain and defective thermogenesis of Middle-Aged mice when compared to Middle-Aged animals fed on chow diet. As expected, chronic exercise induced negative energy balance, reduced food consumption and increasing the energy expenditure, which promotes body weight loss. Interestingly, exercise training reduced the TGF-ß1 expression and IkB-α ser32 phosphorylation in the hypothalamus of Middle-Aged obese mice. Taken together our study demonstrated that chronic exercise suppressed the TGF-ß1/IkB-α axis in the hypothalamus and improved the energy homeostasis in an animal model of obesity-associated to aging.


Asunto(s)
Terapia por Ejercicio , Hipotálamo/metabolismo , Obesidad/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factores de Edad , Animales , Regulación de la Temperatura Corporal , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Regulación hacia Abajo , Ingestión de Alimentos , Metabolismo Energético , Conducta Alimentaria , Hipotálamo/fisiopatología , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa/metabolismo , Obesidad/genética , Obesidad/fisiopatología , Obesidad/terapia , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta1/genética , Pérdida de Peso
14.
Sci Rep ; 6: 29290, 2016 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-27373214

RESUMEN

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.


Asunto(s)
Hipotálamo/metabolismo , Inflamación/metabolismo , Obesidad/metabolismo , Proopiomelanocortina/metabolismo , betaendorfina/metabolismo , Adolescente , Adulto , Animales , Dieta , Grasas de la Dieta/metabolismo , Ingestión de Energía , Humanos , Hipotálamo/inmunología , Inflamación/inmunología , Masculino , Ratones , Ratones Obesos , Obesidad/inmunología , Proopiomelanocortina/inmunología , Ratas , Ratas Wistar , Adulto Joven
15.
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
16.
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
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.
Diabetes ; 63(11): 3770-84, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24947351

RESUMEN

Hypothalamic inflammation is a common feature of experimental obesity. Dietary fats are important triggers of this process, inducing the activation of toll-like receptor-4 (TLR4) signaling and endoplasmic reticulum stress. Microglia cells, which are the cellular components of the innate immune system in the brain, are expected to play a role in the early activation of diet-induced hypothalamic inflammation. Here, we use bone marrow transplants to generate mice chimeras that express a functional TLR4 in the entire body except in bone marrow-derived cells or only in bone marrow-derived cells. We show that a functional TLR4 in bone marrow-derived cells is required for the complete expression of the diet-induced obese phenotype and for the perpetuation of inflammation in the hypothalamus. In an obesity-prone mouse strain, the chemokine CX3CL1 (fractalkine) is rapidly induced in the neurons of the hypothalamus after the introduction of a high-fat diet. The inhibition of hypothalamic fractalkine reduces diet-induced hypothalamic inflammation and the recruitment of bone marrow-derived monocytic cells to the hypothalamus; in addition, this inhibition reduces obesity and protects against diet-induced glucose intolerance. Thus, fractalkine is an important player in the early induction of diet-induced hypothalamic inflammation, and its inhibition impairs the induction of the obese and glucose intolerance phenotypes.


Asunto(s)
Quimiocina CX3CL1/metabolismo , Hipotálamo/metabolismo , Inflamación/metabolismo , Obesidad/metabolismo , Animales , Quimiocina CX3CL1/genética , Dieta Alta en Grasa/efectos adversos , Citometría de Flujo , Hipotálamo/inmunología , Immunoblotting , Inflamación/etiología , Inflamación/inmunología , Masculino , Ratones , Obesidad/etiología , Obesidad/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Receptor Toll-Like 4/metabolismo
19.
Br J Nutr ; 110(3): 447-55, 2013 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-23415177

RESUMEN

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.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina , Insulina/metabolismo , Myrtaceae , Obesidad/tratamiento farmacológico , Fitoterapia , Preparaciones de Plantas/uso terapéutico , Análisis de Varianza , Animales , Antocianinas/farmacología , Antocianinas/uso terapéutico , Fármacos Antiobesidad/farmacología , Fármacos Antiobesidad/uso terapéutico , Suplementos Dietéticos , Frutas , Intolerancia a la Glucosa , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inflamación/metabolismo , Leptina/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos , Myrtaceae/química , Obesidad/etiología , Obesidad/metabolismo , Preparaciones de Plantas/farmacología , Polvos , Receptor de Insulina/metabolismo , Transducción de Señal , Aumento de Peso/efectos de los fármacos
20.
Diabetes ; 62(1): 137-48, 2013 01.
Artículo en Inglés | MEDLINE | ID: mdl-22966070

RESUMEN

Mutation of tub gene in mice induces obesity, suggesting that tub could be an important regulator of energy balance. In the current study, we investigated whether insulin, leptin, and obesity can modulate Tub in vivo in hypothalamic nuclei, and we investigated possible consequences on energy balance, neuropeptide expression, and hepatic glucose metabolism. Food intake, metabolic characteristics, signaling proteins, and neuropeptide expression were measured in response to fasting and refeeding, intracerebroventricular insulin and leptin, and Tub antisense oligonucleotide (ASO). Tub tyrosine phosphorylation (Tub-p-tyr) is modulated by nutritional status. Tub is a substrate of insulin receptor tyrosine kinase (IRTK) and leptin receptor (LEPR)-Janus kinase 2 (JAK2) in hypothalamic nuclei. After leptin or insulin stimulation, Tub translocates to the nucleus. Inhibition of Tub expression in hypothalamus by ASO increased food intake, fasting blood glucose, and hepatic glucose output, decreased O(2) consumption, and blunted the effect of insulin or leptin on proopiomelanocortin, thyroid-releasing hormone, melanin-concentrating hormone, and orexin expression. In hypothalamus of mice administered a high-fat diet, there is a reduction in leptin and insulin-induced Tub-p-tyr and nuclear translocation, which is reversed by reducing protein tyrosine phosphatase 1B expression. These results indicate that Tub has a key role in the control of insulin and leptin effects on food intake, and the modulation of Tub may contribute to insulin and leptin resistance in DIO mice.


Asunto(s)
Hipotálamo/fisiología , Insulina/farmacología , Leptina/farmacología , Proteínas/fisiología , Transducción de Señal/fisiología , Transporte Activo de Núcleo Celular , Proteínas Adaptadoras Transductoras de Señales , Animales , Ayuno , Janus Quinasa 2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Oligonucleótidos Antisentido/farmacología , Fosfolipasa C beta/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/fisiología , Proteínas/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA