Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell Metab ; 35(3): 438-455.e7, 2023 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-36889283

RESUMEN

Until menopause, women have a lower propensity to develop metabolic diseases than men, suggestive of a protective role for sex hormones. Although a functional synergy between central actions of estrogens and leptin has been demonstrated to protect against metabolic disturbances, the underlying cellular and molecular mechanisms mediating this crosstalk have remained elusive. By using a series of embryonic, adult-onset, and tissue/cell-specific loss-of-function mouse models, we document an unprecedented role of hypothalamic Cbp/P300-interacting transactivator with Glu/Asp-rich carboxy-terminal domain 1 (Cited1) in mediating estradiol (E2)-dependent leptin actions that control feeding specifically in pro-opiomelanocortin (Pomc) neurons. We reveal that within arcuate Pomc neurons, Cited1 drives leptin's anorectic effects by acting as a co-factor converging E2 and leptin signaling via direct Cited1-ERα-Stat3 interactions. Together, these results provide new insights on how melanocortin neurons integrate endocrine inputs from gonadal and adipose axes via Cited1, thereby contributing to the sexual dimorphism in diet-induced obesity.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Leptina , Ratones , Animales , Femenino , Leptina/metabolismo , Estradiol/farmacología , Proopiomelanocortina/metabolismo , Hipotálamo/metabolismo , Obesidad/metabolismo
2.
Glia ; 70(11): 2062-2078, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35802021

RESUMEN

Hypothalamic astrocytes are particularly affected by energy-dense food consumption. How the anatomical location of these glial cells and their spatial molecular distribution in the arcuate nucleus of the hypothalamus (ARC) determine the cellular response to a high caloric diet remains unclear. In this study, we investigated their distinctive molecular responses following exposure to a high-fat high-sugar (HFHS) diet, specifically in the ARC. Using RNA sequencing and proteomics, we showed that astrocytes have a distinct transcriptomic and proteomic profile dependent on their anatomical location, with a major proteomic reprogramming in hypothalamic astrocytes. By ARC single-cell sequencing, we observed that a HFHS diet dictates time- and cell- specific transcriptomic responses, revealing that astrocytes have the most distinct regulatory pattern compared to other cell types. Lastly, we topographically and molecularly characterized astrocytes expressing glial fibrillary acidic protein and/or aldehyde dehydrogenase 1 family member L1 in the ARC, of which the abundance was significantly increased, as well as the alteration in their spatial and molecular profiles, with a HFHS diet. Together, our results provide a detailed multi-omics view on the spatial and temporal changes of astrocytes particularly in the ARC during different time points of adaptation to a high calorie diet.


Asunto(s)
Astrocitos , Proteómica , Núcleo Arqueado del Hipotálamo/metabolismo , Astrocitos/metabolismo , Dieta Alta en Grasa/efectos adversos , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipotálamo/metabolismo
3.
Cell Metab ; 33(6): 1155-1170.e10, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33951475

RESUMEN

Pathologies of the micro- and macrovascular systems are a hallmark of the metabolic syndrome, which can lead to chronically elevated blood pressure. However, the underlying pathomechanisms involved still need to be clarified. Here, we report that an obesity-associated increase in serum leptin triggers the select expansion of the micro-angioarchitecture in pre-autonomic brain centers that regulate hemodynamic homeostasis. By using a series of cell- and region-specific loss- and gain-of-function models, we show that this pathophysiological process depends on hypothalamic astroglial hypoxia-inducible factor 1α-vascular endothelial growth factor (HIF1α-VEGF) signaling downstream of leptin signaling. Importantly, several distinct models of HIF1α-VEGF pathway disruption in astrocytes are protected not only from obesity-induced hypothalamic angiopathy but also from sympathetic hyperactivity or arterial hypertension. These results suggest that hyperleptinemia promotes obesity-induced hypertension via a HIF1α-VEGF signaling cascade in hypothalamic astrocytes while establishing a novel mechanistic link that connects hypothalamic micro-angioarchitecture with control over systemic blood pressure.


Asunto(s)
Astrocitos/metabolismo , Hipertensión/metabolismo , Hipotálamo/metabolismo , Leptina/fisiología , Obesidad/metabolismo , Animales , Astrocitos/patología , Femenino , Hipotálamo/patología , Masculino , Ratones , Ratones Endogámicos C57BL
4.
Cell Metab ; 33(4): 833-844.e5, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33571454

RESUMEN

Uncertainty exists as to whether the glucose-dependent insulinotropic polypeptide receptor (GIPR) should be activated or inhibited for the treatment of obesity. Gipr was recently demonstrated in hypothalamic feeding centers, but the physiological relevance of CNS Gipr remains unknown. Here we show that HFD-fed CNS-Gipr KO mice and humanized (h)GIPR knockin mice with CNS-hGIPR deletion show decreased body weight and improved glucose metabolism. In DIO mice, acute central and peripheral administration of acyl-GIP increases cFos neuronal activity in hypothalamic feeding centers, and this coincides with decreased body weight and food intake and improved glucose handling. Chronic central and peripheral administration of acyl-GIP lowers body weight and food intake in wild-type mice, but shows blunted/absent efficacy in CNS-Gipr KO mice. Also, the superior metabolic effect of GLP-1/GIP co-agonism relative to GLP-1 is extinguished in CNS-Gipr KO mice. Our data hence establish a key role of CNS Gipr for control of energy metabolism.


Asunto(s)
Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Polipéptido Inhibidor Gástrico/farmacología , Receptores de la Hormona Gastrointestinal/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Sistema Nervioso Central/metabolismo , Dieta Alta en Grasa , Polipéptido Inhibidor Gástrico/química , Péptido 1 Similar al Glucagón/farmacología , Humanos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/metabolismo , Obesidad/patología , Obesidad/prevención & control , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de la Hormona Gastrointestinal/deficiencia , Receptores de la Hormona Gastrointestinal/genética
5.
J Clin Invest ; 130(11): 6093-6108, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32780722

RESUMEN

Recent genome-wide association studies (GWAS) identified DUSP8, encoding a dual-specificity phosphatase targeting mitogen-activated protein kinases, as a type 2 diabetes (T2D) risk gene. Here, we reveal that Dusp8 is a gatekeeper in the hypothalamic control of glucose homeostasis in mice and humans. Male, but not female, Dusp8 loss-of-function mice, either with global or corticotropin-releasing hormone neuron-specific deletion, had impaired systemic glucose tolerance and insulin sensitivity when exposed to high-fat diet (HFD). Mechanistically, we found impaired hypothalamic-pituitary-adrenal axis feedback, blunted sympathetic responsiveness, and chronically elevated corticosterone levels driven by hypothalamic hyperactivation of Jnk signaling. Accordingly, global Jnk1 ablation, AAV-mediated Dusp8 overexpression in the mediobasal hypothalamus, or metyrapone-induced chemical adrenalectomy rescued the impaired glucose homeostasis of obese male Dusp8-KO mice, respectively. The sex-specific role of murine Dusp8 in governing hypothalamic Jnk signaling, insulin sensitivity, and systemic glucose tolerance was consistent with functional MRI data in human volunteers that revealed an association of the DUSP8 rs2334499 risk variant with hypothalamic insulin resistance in men. Further, expression of DUSP8 was increased in the infundibular nucleus of T2D humans. In summary, our findings suggest the GWAS-identified gene Dusp8 as a novel hypothalamic factor that plays a functional role in the etiology of T2D.


Asunto(s)
Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Tipo 2/enzimología , Fosfatasas de Especificidad Dual/metabolismo , Hipotálamo/enzimología , Resistencia a la Insulina , MAP Quinasa Quinasa 4/metabolismo , Transducción de Señal , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , Fosfatasas de Especificidad Dual/genética , MAP Quinasa Quinasa 4/genética , Ratones , Ratones Noqueados
6.
J Lipid Res ; 59(9): 1649-1659, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29991652

RESUMEN

Meta-inflammation of hypothalamic areas governing energy homeostasis has recently emerged as a process of potential pathophysiological relevance for the development of obesity and its metabolic sequelae. The current model suggests that diet-induced neuronal injury triggers microgliosis and astrocytosis, conditions which ultimately may induce functional impairment of hypothalamic circuits governing feeding behavior, systemic metabolism, and body weight. Epidemiological data indicate that low circulating HDL levels, besides conveying cardiovascular risk, also correlate strongly with obesity. We simulated that condition by using a genetic loss of function mouse model (apoA-I-/-) with markedly reduced HDL levels to investigate whether HDL may directly modulate hypothalamic inflammation. Astrogliosis was significantly enhanced in the hypothalami of apoA-I-/- compared with apoA-I+/+ mice and was associated with compromised mitochondrial function. apoA-I-/- mice exhibited key components of metabolic disease, like increased fat mass, fasting glucose levels, hepatic triglyceride content, and hepatic glucose output compared with apoA-I+/+ controls. Administration of reconstituted HDL (CSL-111) normalized hypothalamic inflammation and mitochondrial function markers in apoA-I-/- mice. Treatment of primary astrocytes with apoA-I resulted in enhanced mitochondrial activity, implying that circulating HDL levels are likely important for astrocyte function. HDL-based therapies may consequently avert reactive gliosis in hypothalamic astrocytes by improving mitochondrial bioenergetics and thereby offering potential treatment and prevention for obesity and metabolic disease.


Asunto(s)
Apolipoproteína A-I/metabolismo , Gliosis/metabolismo , Gliosis/patología , Hipotálamo/patología , Lipoproteínas HDL/sangre , Adenosina Trifosfato/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Biomarcadores/metabolismo , Gliosis/sangre , Glucólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/patología , Fosforilación Oxidativa , Fenotipo
7.
Obes Facts ; 11(2): 93-108, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29631267

RESUMEN

OBJECTIVE: We aimed to determine the effect of human SH2B1 variants on leptin and insulin signaling, major regulators of energy homeostasis, on the RNA level. METHODS: We analyzed the expression of infrequent alleles of seven SH2B1 variants (Arg67Cys, Lys150Arg, Thr175Ala, Thr343Met, Thr484Ala, Ser616Pro and Pro689Leu) in response to insulin or leptin cell stimulation. Two of these were identified in own mutation screens, the others were predicted to be deleterious or to serve as controls. The variants were analyzed in a homologous system of mouse hypothalamic cells. Changes in expression of downstream genes were measured. Student's t-test for independent samples was applied and effect sizes using Cohen's d were calculated. RESULTS: In 34 of 54 analyzed genes involved in leptin (JAK/STAT or AKT) signaling, variants nominally changed expression. The expression of three genes was considerably increased (p values ≤ 0.001: Gbp2b (67Cys; d = 25.11), Irf9 (689Leu; d = 44.65) and Isg15 (150Arg; d = 20.35)). Of 32 analyzed genes in the insulin signaling pathway, the expression of 10 genes nominally changed (p ≤ 0.05), three resulted in p values ≤ 0.01 ( Cap1 (150Arg; d = 7.48), Mapk1 (343Met; d = ­6.80) and Sorbs1 (689Leu; d = 7.82)). CONCLUSION: The increased expression of genes in leptin (JAK/STAT or AKT) signaling implies that the main mode of action for human SH2B1 mutations might affect leptin signaling rather than insulin signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Hipotálamo/efectos de los fármacos , Insulina/farmacología , Leptina/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adolescente , Adulto , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Masculino , Ratones , Ratones Noqueados , Mutación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Adulto Joven
8.
Cell Metab ; 26(4): 620-632.e6, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-28943448

RESUMEN

Chronic inflammation has been proposed to contribute to the pathogenesis of diet-induced obesity. However, scarce therapeutic options are available to treat obesity and the associated immunometabolic complications. Glucocorticoids are routinely employed for the management of inflammatory diseases, but their pleiotropic nature leads to detrimental metabolic side effects. We developed a glucagon-like peptide-1 (GLP-1)-dexamethasone co-agonist in which GLP-1 selectively delivers dexamethasone to GLP-1 receptor-expressing cells. GLP-1-dexamethasone lowers body weight up to 25% in obese mice by targeting the hypothalamic control of feeding and by increasing energy expenditure. This strategy reverses hypothalamic and systemic inflammation while improving glucose tolerance and insulin sensitivity. The selective preference for GLP-1 receptor bypasses deleterious effects of dexamethasone on glucose handling, bone integrity, and hypothalamus-pituitary-adrenal axis activity. Thus, GLP-1-directed glucocorticoid pharmacology represents a safe and efficacious therapy option for diet-induced immunometabolic derangements and the resulting obesity.


Asunto(s)
Dexametasona/uso terapéutico , Péptido 1 Similar al Glucagón/uso terapéutico , Glucocorticoides/uso terapéutico , Incretinas/uso terapéutico , Inflamación/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Animales , Peso Corporal/efectos de los fármacos , Dexametasona/análogos & derivados , Metabolismo Energético/efectos de los fármacos , Péptido 1 Similar al Glucagón/análogos & derivados , Glucocorticoides/química , Glucosa/metabolismo , Células HEK293 , Humanos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Incretinas/química , Inflamación/complicaciones , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/complicaciones , Obesidad/metabolismo
9.
Sci Signal ; 9(425): ra43, 2016 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-27117253

RESUMEN

PEN is an abundant peptide in the brain that has been implicated in the regulation of feeding. We identified a receptor for PEN in mouse hypothalamus and Neuro2A cells. PEN bound to and activated GPR83, a G protein (heterotrimeric guanine nucleotide)-binding protein)-coupled receptor (GPCR). Reduction of GPR83 expression in mouse brain and Neuro2A cells reduced PEN binding and signaling, consistent with GPR83 functioning as the major receptor for PEN. In some brain regions, GPR83 colocalized with GPR171, a GPCR that binds the neuropeptide bigLEN, another neuropeptide that is involved in feeding and is generated from the same precursor protein as is PEN. Coexpression of these two receptors in cell lines altered the signaling properties of each receptor, suggesting a functional interaction. Our data established PEN as a neuropeptide that binds GPR83 and suggested that these two ligand-receptor systems-PEN-GPR83 and bigLEN-GPR171-may be functionally coupled in the regulation of feeding.


Asunto(s)
Hipotálamo/metabolismo , Neuropéptido Y/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Regulación del Apetito/fisiología , Western Blotting , Células CHO , Membrana Celular/metabolismo , Células Cultivadas , Cricetulus , Células HEK293 , Humanos , Masculino , Ratones , Fosforilación , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/genética
10.
Nat Commun ; 7: 10782, 2016 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-26923837

RESUMEN

Hypothalamic leptin signalling has a key role in food intake and energy-balance control and is often impaired in obese individuals. Here we identify histone deacetylase 5 (HDAC5) as a regulator of leptin signalling and organismal energy balance. Global HDAC5 KO mice have increased food intake and greater diet-induced obesity when fed high-fat diet. Pharmacological and genetic inhibition of HDAC5 activity in the mediobasal hypothalamus increases food intake and modulates pathways implicated in leptin signalling. We show HDAC5 directly regulates STAT3 localization and transcriptional activity via reciprocal STAT3 deacetylation at Lys685 and phosphorylation at Tyr705. In vivo, leptin sensitivity is substantially impaired in HDAC5 loss-of-function mice. Hypothalamic HDAC5 overexpression improves leptin action and partially protects against HFD-induced leptin resistance and obesity. Overall, our data suggest that hypothalamic HDAC5 activity is a regulator of leptin signalling that adapts food intake and body weight to our dietary environment.


Asunto(s)
Hipotálamo/metabolismo , Leptina/metabolismo , Animales , Glucemia , Línea Celular , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Prueba de Tolerancia a la Glucosa , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Infusiones Intraventriculares , Resistencia a la Insulina , Captura por Microdisección con Láser , Leptina/genética , Masculino , Hormonas Estimuladoras de los Melanocitos/farmacología , Ratones , Ratones Endogámicos , Ratones Noqueados , Neuronas/fisiología , Ratas , Ratas Wistar
11.
Diabetes ; 65(2): 406-20, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26512025

RESUMEN

Ghrelin, the natural ligand of the growth hormone secretagogue receptor type 1a (GHS-R1a), is mainly secreted from the stomach and regulates food intake and energy homeostasis. p27 regulates cell cycle progression in many cell types. Here, we report that rats affected by the multiple endocrine neoplasia syndrome MENX, caused by a p27 mutation, develop pancreatic islet hyperplasia containing elevated numbers of ghrelin-producing ε-cells. The metabolic phenotype of MENX-affected rats featured high endogenous acylated and unacylated plasma ghrelin levels. Supporting increased ghrelin action, MENX rats show increased food intake, enhanced body fat mass, and elevated plasma levels of triglycerides and cholesterol. Ghrelin effect on food intake was confirmed by treating MENX rats with a GHS-R1a antagonist. At 7.5 months, MENX-affected rats show decreased mRNA levels of hypothalamic GHS-R1a, neuropeptide Y (NPY), and agouti-related protein (AgRP), suggesting that prolonged hyperghrelinemia may lead to decreased ghrelin efficacy. In line with ghrelin's proposed role in glucose metabolism, we find decreased glucose-stimulated insulin secretion in MENX rats, while insulin sensitivity is improved. In summary, we provide a novel nontransgenic rat model with high endogenous ghrelin plasma levels and, interestingly, improved glucose tolerance. This model might aid in identifying new therapeutic approaches for obesity and obesity-related diseases, including type 2 diabetes.


Asunto(s)
Ghrelina/sangre , Resistencia a la Insulina/fisiología , Neoplasia Endocrina Múltiple/sangre , Obesidad/sangre , Proteína Relacionada con Agouti/metabolismo , Animales , Regulación del Apetito/efectos de los fármacos , Regulación del Apetito/fisiología , Glucemia/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Hipotálamo/metabolismo , Neoplasia Endocrina Múltiple/complicaciones , Neoplasia Endocrina Múltiple/genética , Mutación , Neuropéptido Y/metabolismo , Obesidad/complicaciones , ARN Mensajero/metabolismo , Ratas , Receptores de Ghrelina/antagonistas & inhibidores , Receptores de Ghrelina/metabolismo , Receptores de Ghrelina/uso terapéutico
12.
Diabetes ; 63(4): 1422-7, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24379349

RESUMEN

We recently reported restoration of leptin responsiveness in diet-induced obese (DIO) mice using a pharmacologically optimized, polyethylene-glycolated (PEG)-leptin analog in combination with exendin-4 or FGF21. However, the return of leptin action required discontinuation of high-fat diet (HFD) exposure. Here we assess whether a single peptide possessing balanced coagonism at the glucagon-like peptide 1 (GLP-1) and glucagon receptors can restore leptin responsiveness in DIO mice maintained on a HFD. DIO mice were treated with PEG-GLP-1/glucagon (30 nmol/kg every fourth day) to induce an ∼15% body weight loss, upon which they were randomized to continue PEG-GLP-1/glucagon therapy or reassigned to receive supplemental daily PEG-leptin (185 nmol/kg/day). The addition of PEG-leptin to PEG-GLP-1/glucagon resulted in an ∼18% greater weight loss as compared with PEG-GLP-1/glucagon alone and was accompanied by further decreases in food intake and improved glucose and lipid metabolism. The beneficial effect of PEG-leptin supplementation occurred after an initial body weight loss similar to what we previously reported following reduced dietary fat along with PEG-leptin and exendin-4 or FGF21 cotreatment. In summary, we report that GLP-1/glucagon coagonism restores leptin responsiveness in mice maintained on a HFD, thus emphasizing the translational value of this polypharmacotherapy for the treatment of obesity and diabetes.


Asunto(s)
Péptido 1 Similar al Glucagón/agonistas , Péptido 1 Similar al Glucagón/uso terapéutico , Leptina/agonistas , Obesidad/tratamiento farmacológico , Receptores de Glucagón/agonistas , Animales , Dieta Alta en Grasa , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Glucagón/agonistas , Glucagón/uso terapéutico , Leptina/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Polietilenglicoles/uso terapéutico , Pérdida de Peso
13.
Diabetes ; 61(11): 2734-42, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22787140

RESUMEN

Although obesity rates are rapidly rising, caloric restriction remains one of the few safe therapies. Here we tested the hypothesis that obesity-associated disorders are caused by increased adipose tissue as opposed to excess dietary lipids. Fat mass (FM) of lean C57B6 mice fed a high-fat diet (HFD; FMC mice) was "clamped" to match the FM of mice maintained on a low-fat diet (standard diet [SD] mice). FMC mice displayed improved glucose and insulin tolerance as compared with ad libitum HFD mice (P < 0.001) or SD mice (P < 0.05). These improvements were associated with fewer signs of inflammation, consistent with the less-impaired metabolism. In follow-up studies, diet-induced obese mice were food restricted for 5 weeks to achieve FM levels identical with those of age-matched SD mice. Previously, obese mice exhibited improved glucose and insulin tolerance but showed markedly increased fasting-induced hyperphagia (P < 0.001). When mice were given ad libitum access to the HFD, the hyperphagia of these mice led to accelerated body weight gain as compared with otherwise matched controls without a history of obesity. These results suggest that although caloric restriction on a HFD provides metabolic benefits, maintaining those benefits may require lifelong continuation, at least in individuals with a history of obesity.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Restricción Calórica/efectos adversos , Metabolismo Energético , Adiposidad , Animales , Dieta Alta en Grasa/efectos adversos , Dieta Reductora/efectos adversos , Regulación de la Expresión Génica , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/inmunología , Intolerancia a la Glucosa/metabolismo , Hiperfagia/etiología , Hipotálamo/metabolismo , Mediadores de Inflamación/metabolismo , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/dietoterapia , Obesidad/inmunología , Obesidad/metabolismo , Obesidad/prevención & control , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos , Distribución Aleatoria , Prevención Secundaria , Aumento de Peso
14.
Physiol Behav ; 106(4): 485-90, 2012 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-22483785

RESUMEN

Hypothalamic inflammation is a potentially important process in the pathogenesis of high-fat diet-induced metabolic disorders that has recently received significant attention. Microglia are macrophage-like cells of the central nervous system which are activated by pro-inflammatory signals causing local production of specific interleukins and cytokines, and these in turn may further promote systemic metabolic disease. Whether or how this microglial activation can be averted or reversed is unknown. Since running exercise improves systemic metabolic health and has been found to promote neuronal survival as well as the recovery of brain functions after injury, we hypothesized that regular treadmill running may blunt the effect of western diet on hypothalamic inflammation. Using low-density lipoprotein receptor deficient (l dlr-/-) mice to better reflect human lipid metabolism, we first confirmed that microglial activation in the hypothalamus is severely increased upon exposure to a high-fat, or "western", diet. Moderate, but regular, treadmill running exercise markedly decreased hypothalamic inflammation in these mice. Furthermore, the observed decline in microglial activation was associated with an improvement of glucose tolerance. Our findings support the hypothesis that hypothalamic inflammation can be reversed by exercise and suggest that interventions to avert or reverse neuronal damage may offer relevant potential in obesity treatment and prevention.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Hipotálamo/patología , Inflamación/patología , Inflamación/prevención & control , Condicionamiento Físico Animal/fisiología , Animales , Peso Corporal/fisiología , Calorimetría Indirecta , Prueba de Tolerancia a la Glucosa , Homeostasis/fisiología , Inmunohistoquímica , Activación de Macrófagos/fisiología , Masculino , Ratones , Ratones Noqueados , Microglía/fisiología , Resistencia Física/fisiología , Receptores de LDL/genética , Carrera/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA