Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
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.
Nat Metab ; 1(2): 222-235, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-32694784

RESUMEN

Heterogeneous populations of hypothalamic neurons orchestrate energy balance via the release of specific signatures of neuropeptides. However, how specific intracellular machinery controls peptidergic identities and function of individual hypothalamic neurons remains largely unknown. The transcription factor T-box 3 (Tbx3) is expressed in hypothalamic neurons sensing and governing energy status, whereas human TBX3 haploinsufficiency has been linked with obesity. Here, we demonstrate that loss of Tbx3 function in hypothalamic neurons causes weight gain and other metabolic disturbances by disrupting both the peptidergic identity and plasticity of Pomc/Cart and Agrp/Npy neurons. These alterations are observed after loss of Tbx3 in both immature hypothalamic neurons and terminally differentiated mouse neurons. We further establish the importance of Tbx3 for body weight regulation in Drosophila melanogaster and show that TBX3 is implicated in the differentiation of human embryonic stem cells into hypothalamic Pomc neurons. Our data indicate that Tbx3 directs the terminal specification of neurons as functional components of the melanocortin system and is required for maintaining their peptidergic identity. In summary, we report the discovery of a key mechanistic process underlying the functional heterogeneity of hypothalamic neurons governing body weight and systemic metabolism.


Asunto(s)
Hipotálamo/metabolismo , Melanocortinas/metabolismo , Neuronas/metabolismo , Proteínas de Dominio T Box/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Peso Corporal , Metabolismo Energético , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Hipotálamo/citología , Ratones , Ratones Endogámicos C57BL , Proopiomelanocortina/genética , ARN Mensajero/genética , Proteínas de Dominio T Box/genética
6.
Mol Metab ; 16: 191-202, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30093356

RESUMEN

OBJECTIVE: The metabolic role of d-serine, a non-proteinogenic NMDA receptor co-agonist, is poorly understood. Conversely, inhibition of pancreatic NMDA receptors as well as loss of the d-serine producing enzyme serine racemase have been shown to modulate insulin secretion. Thus, we aim to study the impact of chronic and acute d-serine supplementation on insulin secretion and other parameters of glucose homeostasis. METHODS: We apply MALDI FT-ICR mass spectrometry imaging, NMR based metabolomics, 16s rRNA gene sequencing of gut microbiota in combination with a detailed physiological characterization to unravel the metabolic action of d-serine in mice acutely and chronically treated with 1% d-serine in drinking water in combination with either chow or high fat diet feeding. Moreover, we identify SNPs in SRR, the enzyme converting L-to d-serine and two subunits of the NMDA receptor to associate with insulin secretion in humans, based on the analysis of 2760 non-diabetic Caucasian individuals. RESULTS: We show that chronic elevation of d-serine results in reduced high fat diet intake. In addition, d-serine leads to diet-independent hyperglycemia due to blunted insulin secretion from pancreatic beta cells. Inhibition of alpha 2-adrenergic receptors rapidly restores glycemia and glucose tolerance in d-serine supplemented mice. Moreover, we show that single nucleotide polymorphisms (SNPs) in SRR as well as in individual NMDAR subunits are associated with insulin secretion in humans. CONCLUSION: Thus, we identify a novel role of d-serine in regulating systemic glucose metabolism through modulating insulin secretion.


Asunto(s)
Secreción de Insulina/efectos de los fármacos , Serina/farmacología , Animales , Glucemia/metabolismo , Peso Corporal , Dieta Alta en Grasa , Suplementos Dietéticos , Metabolismo Energético , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Homeostasis , Hiperglucemia/metabolismo , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Serina/metabolismo
7.
J Psychiatr Res ; 102: 150-158, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29660602

RESUMEN

Astrocyte functions in white matter are less well understood than in gray matter. Our recent study of white matter in ventral prefrontal cortex (vPFC) revealed alterations in expression of myelin-related genes in major depressive disorder (MDD). Since white matter astrocytes maintain myelin, we hypothesized that morphometry of these cells will be altered in MDD in the same prefrontal white matter region in which myelin-related genes are altered. White matter adjacent to vPFC was examined in 25 MDD and 21 control subjects. Density and size of GFAP-immunoreactive (-ir) astrocyte cell bodies was measured. The area fraction of GFAP-ir astrocytes (cell bodies + processes) was also estimated. GFAP mRNA expression was determined using qRT-PCR. The density of GFAP-ir astrocytes was also measured in vPFC white matter of rats subjected to chronic unpredictable stress (CUS) and control animals. Fibrous and smooth GFAP-ir astrocytes were distinguished in human white matter. The density of both types of astrocytes was significantly decreased in MDD. Area fraction of GFAP immunoreactivity was significantly decreased in MDD, but mean soma size remained unchanged. Expression of GFAP mRNA was significantly decreased in MDD. In CUS rats there was a significant decrease in astrocyte density in prefrontal white matter. The decrease in density and area fraction of white matter astrocytes and GFAP mRNA in MDD may be linked to myelin pathology previously noted in these subjects. Astrocyte pathology may contribute to axon disturbances in axon integrity reported by neuroimaging studies in MDD and interfere with signal conduction in the white matter.


Asunto(s)
Astrocitos/patología , Depresión/patología , Corteza Prefrontal/patología , Sustancia Blanca/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Astrocitos/metabolismo , Depresión/etiología , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/complicaciones , Sustancia Blanca/metabolismo
8.
J Neuroinflammation ; 15(1): 35, 2018 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-29422055

RESUMEN

ᅟ: Astrocytosis is a reactive process involving cellular, molecular, and functional changes to facilitate neuronal survival, myelin preservation, blood brain barrier function and protective glial scar formation upon brain insult. The overall pro- or anti-inflammatory impact of reactive astrocytes appears to be driven in a context- and disease-driven manner by modulation of astrocytic Ca2+ homeostasis and activation of Ca2+/calmodulin-activated serine/threonine phosphatase calcineurin. Here, we aimed to assess whether calcineurin is dispensable for astrocytosis in the hypothalamus driven by prolonged high fat diet (HFD) feeding. Global deletion of calcineurin A beta (gene name: Ppp3cb) led to a decrease of glial fibrillary acidic protein (GFAP)-positive cells in the ventromedial hypothalamus (VMH), dorsomedial hypothalamus (DMH), and arcuate nucleus (ARC) of mice exposed chronically to HFD. The concomitant decrease in Iba1-positive microglia in the VMH further suggests a modest impact of Ppp3cb deletion on microgliosis. Pharmacological inhibition of calcineurin activity by Fk506 had no impact on IBA1-positive microglia in hypothalami of mice acutely exposed to HFD for 1 week. However, Fk506-treated mice displayed a decrease in GFAP levels in the ARC. In vivo effects could not be replicated in cell culture, where calcineurin inhibition by Fk506 had no effect on astrocytic morphology, astrocytic cell death, GFAP, and vimentin protein levels or microglia numbers in primary hypothalamic astrocytes and microglia co-cultures. Further, adenoviral overexpression of calcineurin subunit Ppp3r1 in primary glia culture did not lead to an increase in GFAP fluorescence intensity. Overall, our results point to a prominent role of calcineurin in mediating hypothalamic astrocytosis as response to acute and chronic HFD exposure. Moreover, discrepant findings in vivo and in cell culture indicate the necessity of studying astrocytes in their "natural" environment, i.e., preserving an intact hypothalamic microenvironment with neurons and non-neuronal cells in close proximity.


Asunto(s)
Calcineurina/deficiencia , Dieta Alta en Grasa/efectos adversos , Gliosis/metabolismo , Gliosis/prevención & control , Hipotálamo/metabolismo , Animales , Astrocitos/metabolismo , Supervivencia Celular/fisiología , Células Cultivadas , Gliosis/patología , Hipotálamo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
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
10.
Mol Metab ; 6(8): 897-908, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28752053

RESUMEN

OBJECTIVE: The hypothalamus of hypercaloric diet-induced obese animals is featured by a significant increase of microglial reactivity and its associated cytokine production. However, the role of dietary components, in particular fat and carbohydrate, with respect to the hypothalamic inflammatory response and the consequent impact on hypothalamic control of energy homeostasis is yet not clear. METHODS: We dissected the different effects of high-carbohydrate high-fat (HCHF) diets and low-carbohydrate high-fat (LCHF) diets on hypothalamic inflammatory responses in neurons and non-neuronal cells and tested the hypothesis that HCHF diets induce hypothalamic inflammation via advanced glycation end-products (AGEs) using mice lacking advanced glycation end-products (AGEs) receptor (RAGE) and/or the activated leukocyte cell-adhesion molecule (ALCAM). RESULTS: We found that consumption of HCHF diets, but not of LCHF diets, increases microgliosis as well as the presence of N(ε)-(Carboxymethyl)-Lysine (CML), a major AGE, in POMC and NPY neurons of the arcuate nucleus. Neuron-secreted CML binds to both RAGE and ALCAM, which are expressed on endothelial cells, microglia, and pericytes. On a HCHF diet, mice lacking the RAGE and ALCAM genes displayed less microglial reactivity and less neovasculature formation in the hypothalamic ARC, and this was associated with significant improvements of metabolic disorders induced by the HCHF diet. CONCLUSIONS: Combined overconsumption of fat and sugar, but not the overconsumption of fat per se, leads to excessive CML production in hypothalamic neurons, which, in turn, stimulates hypothalamic inflammatory responses such as microgliosis and eventually leads to neuronal dysfunction in the control of energy metabolism.


Asunto(s)
Grasas de la Dieta/metabolismo , Azúcares de la Dieta/metabolismo , Gliosis/metabolismo , Hipotálamo/metabolismo , Molécula de Adhesión Celular del Leucocito Activado/genética , Animales , Grasas de la Dieta/efectos adversos , Azúcares de la Dieta/efectos adversos , Gliosis/etiología , Productos Finales de Glicación Avanzada/metabolismo , Hipotálamo/patología , Inflamación/etiología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Receptor para Productos Finales de Glicación Avanzada/deficiencia , Receptor para Productos Finales de Glicación Avanzada/genética
11.
Neuroscience ; 359: 30-39, 2017 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-28711621

RESUMEN

Studies of major depressive disorder (MDD) in postmortem brain tissue report enhanced binding to inhibitory serotonin-1A autoreceptors in midbrain dorsal raphe and reductions in length of axons expressing the serotonin transporter (SERT) in dorsolateral prefrontal cortex. The length density of axons expressing SERT in the orbitofrontal cortex (OFC) was determined in 18 subjects with MDD and 17 age-matched control subjects. A monoclonal antibody was used to immunohistochemically label the SERT in fixed sections of OFC. The 3-dimensional length density of SERT-immunoreactive (ir) axons in layer VI of OFC was estimated. The age of subjects with MDD was negatively correlated with SERT axon length (r=-0.77, p<0.0005). The significant effect of age persisted when removing four depressed subjects with an antidepressant medication present at the time of death, or when removing nine depressed subjects that had a recent prescription for an antidepressant medication. Neither gender, tissue pH, postmortem interval, 5-HTTLPR genotype, time in fixative, nor death by suicide had a significant effect on axon length. The age-related decrease in SERT-ir axon length in MDD may reflect pathology of ascending axons passing through deep white matter hyperintensities. Greater length of axons expressing SERT in younger subjects with MDD may result in a significant deficit in serotonin availability in OFC.


Asunto(s)
Axones/metabolismo , Axones/patología , Trastorno Depresivo Mayor/metabolismo , Trastorno Depresivo Mayor/patología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad
12.
Diabetes ; 66(10): 2555-2563, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28710138

RESUMEN

Neuronal circuits in the brain help to control feeding behavior and systemic metabolism in response to afferent nutrient and hormonal signals. Although astrocytes have historically been assumed to have little relevance for such neuroendocrine control, we investigated whether lipid uptake via lipoprotein lipase (LPL) in astrocytes is required to centrally regulate energy homeostasis. Ex vivo studies with hypothalamus-derived astrocytes showed that LPL expression is upregulated by oleic acid, whereas it is decreased in response to palmitic acid or triglycerides. Likewise, astrocytic LPL deletion reduced the accumulation of lipid droplets in those glial cells. Consecutive in vivo studies showed that postnatal ablation of LPL in glial fibrillary acidic protein-expressing astrocytes induced exaggerated body weight gain and glucose intolerance in mice exposed to a high-fat diet. Intriguingly, astrocytic LPL deficiency also triggered increased ceramide content in the hypothalamus, which may contribute to hypothalamic insulin resistance. We conclude that hypothalamic LPL functions in astrocytes to ensure appropriately balanced nutrient sensing, ceramide distribution, body weight regulation, and glucose metabolism.


Asunto(s)
Astrocitos/metabolismo , Dieta Alta en Grasa/efectos adversos , Obesidad/etiología , Obesidad/metabolismo , Animales , Astrocitos/citología , Peso Corporal/fisiología , Ceramidas/metabolismo , Citometría de Flujo , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Prueba de Tolerancia a la Glucosa , Humanos , Hipotálamo/citología , Inmunohistoquímica , Hibridación in Situ , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/patología , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
Nat Commun ; 8: 15143, 2017 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-28489068

RESUMEN

Consuming a calorically dense diet stimulates microglial reactivity in the mediobasal hypothalamus (MBH) in association with decreased number of appetite-curbing pro-opiomelanocortin (POMC) neurons; whether the reduction in POMC neuronal function is secondary to the microglial activation is unclear. Here we show that in hypercaloric diet-induced obese mice, persistently activated microglia in the MBH hypersecrete TNFα that in turn stimulate mitochondrial ATP production in POMC neurons, promoting mitochondrial fusion in their neurites, and increasing POMC neuronal firing rates and excitability. Specific disruption of the gene expressions of TNFα downstream signals TNFSF11A or NDUFAB1 in the MBH of diet-induced obese mice reverses mitochondrial elongation and reduces obesity. These data imply that in a hypercaloric environment, persistent elevation of microglial reactivity and consequent TNFα secretion induces mitochondrial stress in POMC neurons that contributes to the development of obesity.


Asunto(s)
Hipotálamo Medio/metabolismo , Microglía/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Proopiomelanocortina , Estrés Fisiológico , Factor de Necrosis Tumoral alfa/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Complejo I de Transporte de Electrón/genética , Regulación de la Expresión Génica , Ratones , Dinámicas Mitocondriales , Neuritas/metabolismo , Ligando RANK/genética , Transducción de Señal
14.
Cell ; 166(4): 867-880, 2016 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-27518562

RESUMEN

We report that astrocytic insulin signaling co-regulates hypothalamic glucose sensing and systemic glucose metabolism. Postnatal ablation of insulin receptors (IRs) in glial fibrillary acidic protein (GFAP)-expressing cells affects hypothalamic astrocyte morphology, mitochondrial function, and circuit connectivity. Accordingly, astrocytic IR ablation reduces glucose-induced activation of hypothalamic pro-opio-melanocortin (POMC) neurons and impairs physiological responses to changes in glucose availability. Hypothalamus-specific knockout of astrocytic IRs, as well as postnatal ablation by targeting glutamate aspartate transporter (GLAST)-expressing cells, replicates such alterations. A normal response to altering directly CNS glucose levels in mice lacking astrocytic IRs indicates a role in glucose transport across the blood-brain barrier (BBB). This was confirmed in vivo in GFAP-IR KO mice by using positron emission tomography and glucose monitoring in cerebral spinal fluid. We conclude that insulin signaling in hypothalamic astrocytes co-controls CNS glucose sensing and systemic glucose metabolism via regulation of glucose uptake across the BBB.


Asunto(s)
Astrocitos/metabolismo , Glucosa/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Transducción de Señal , Sistema de Transporte de Aminoácidos X-AG/genética , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Animales , Barrera Hematoencefálica , Retículo Endoplásmico/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Homeostasis , Ratones , Mitocondrias/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo
15.
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
16.
EMBO Mol Med ; 7(3): 288-98, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25652173

RESUMEN

We assessed the efficacy of simultaneous agonism at the glucagon-like peptide-1 receptor (GLP-1R) and the melanocortin-4 receptor (MC4R) for the treatment of obesity and diabetes in rodents. Diet-induced obese (DIO) mice were chronically treated with either the long-acting GLP-1R agonist liraglutide, the MC4R agonist RM-493 or a combination of RM-493 and liraglutide. Co-treatment of DIO mice with RM-493 and liraglutide improves body weight loss and enhances glycemic control and cholesterol metabolism beyond what can be achieved with either mono-therapy. The superior metabolic efficacy of this combination therapy is attributed to the anorectic and glycemic actions of both drugs, along with the ability of RM-493 to increase energy expenditure. Interestingly, compared to mice treated with liraglutide alone, hypothalamic Glp-1r expression was higher in mice treated with the combination therapy after both acute and chronic treatment. Further, RM-493 enhanced hypothalamic Mc4r expression. Hence, co-dosing with MC4R and GLP-1R agonists increases expression of each receptor, indicative of minimized receptor desensitization. Together, these findings suggest potential opportunities for employing combination treatments that comprise parallel MC4R and GLP-1R agonism for the treatment of obesity and diabetes.


Asunto(s)
Diabetes Mellitus/tratamiento farmacológico , Péptido 1 Similar al Glucagón/análogos & derivados , Hipoglucemiantes/uso terapéutico , Obesidad/tratamiento farmacológico , Receptor de Melanocortina Tipo 4/agonistas , Receptores de Glucagón/agonistas , alfa-MSH/análogos & derivados , Animales , Sinergismo Farmacológico , Quimioterapia Combinada , Péptido 1 Similar al Glucagón/farmacología , Péptido 1 Similar al Glucagón/uso terapéutico , Receptor del Péptido 1 Similar al Glucagón , Hipoglucemiantes/farmacología , Liraglutida , Ratones Obesos , Resultado del Tratamiento , alfa-MSH/farmacología , alfa-MSH/uso terapéutico
17.
Glia ; 62(1): 17-25, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24166765

RESUMEN

The arcuate nucleus (ARC) of the hypothalamus plays a key role in sensing metabolic feedback and regulating energy homeostasis. Recent studies revealed activation of microglia in mice with high-fat diet (HFD)-induced obesity (DIO), suggesting a potential pathophysiological role for inflammatory processes within the hypothalamus. To further investigate the metabolic causes and molecular underpinnings of such glial activation, we analyzed the microglial activity in wild-type (WT), monogenic obese ob/ob (leptin deficient), db/db (leptin-receptor mutation), and Type-4 melanocortin receptor knockout (MC4R KO) mice on either a HFD or on standardized chow (SC) diet. Following HFD exposure, we observed a significant increase in the total number of ARC microglia, immunoreactivity of ionized calcium binding adaptor molecule 1 (iba1-ir), cluster of differentiation 68 (CD68-ir), and ramification of microglial processes. The ob/ob mice had significantly less iba1-ir and ramifications. Leptin replacement rescued these phenomena. The db/db mice had similar iba1-ir comparable with WT mice but had significantly lower CD68-ir and more ramifications than WT mice. After 2 weeks of HFD, ob/ob mice showed an increase of iba1-ir, and db/db mice showed increase of CD68-ir. Obese MC4R KO mice fed a SC diet had comparable iba1-ir and CD68-ir with WT mice but had significantly more ramifications than WT mice. Intriguingly, treatment of DIO mice with glucagon-like peptide-1 receptor agonists reduced microglial activation independent of body weight. Our results show that diet type, adipokines, and gut signals, but not body weight, affect the presence and activity levels of hypothalamic microglia in obesity.


Asunto(s)
Hormonas/farmacología , Microglía/metabolismo , Núcleo Supraóptico/citología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Citocininas/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Exenatida , Leptina/deficiencia , Leptina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Obesidad/inducido químicamente , Obesidad/fisiopatología , Péptidos/farmacología , Receptor de Melanocortina Tipo 4/deficiencia , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/metabolismo , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Transducción de Señal/efectos de los fármacos , Ponzoñas/farmacología
18.
Artículo en Inglés | MEDLINE | ID: mdl-22889863

RESUMEN

The activity of the mammalian target of rapamycin (mTOR), an ubiquitously expressed serine/threonine kinase, is central to the regulation of translation initiation and, consequently protein synthesis required for long-term potentiation and new synaptic connections. Recent studies show that activation of the mTOR signaling pathway is required for the rapid antidepressant actions of glutamate N-methyl-d-aspartate (NMDA) receptor antagonists such as ketamine. Our prior work documented the first evidence of robust deficits in the mTOR signaling pathway in the prefrontal cortex (PFC) from subjects diagnosed with major depressive disorder (MDD). The goal of this study was to determine whether alterations in mTOR signaling can be observed in rats exposed to the chronic unpredictable stress (CUS) model of depression. In the present study, we examined the effect of CUS on the expression of phosphorylated mTOR and its downstream signaling components in the frontal cortex, hippocampus, amygdala, and dorsal raphe. We also examined the effect of CUS on the expression of kinases that phosphorylate mTOR such as extracellular signal-regulated kinase (ERK1/2) and protein kinase B/Akt (Akt1). In addition, we examined the effect of stress on the phosphorylation of GluR1 an, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit. We found that eight-weeks of CUS exposure significantly decreased the phosphorylation levels of mTOR and its downstream signaling components in the amygdala. Reduced level of phospho-mTOR in the amygdala was accompanied by decreased phosphorylation of ERK-1/2, Akt-1, and GluR1. No significant changes were seen in the frontal cortex, hippocampus, or dorsal raphe. Our study demonstrates that long-term stress exposure results in brain region-specific abnormalities in signaling pathways previously linked to novel mechanisms for rapid antidepressant effects. These observations are in line with evidence showing that mTOR and its upstream and downstream signaling partners could be important targets for the development of novel antidepressants.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Transducción de Señal/fisiología , Estrés Fisiológico/fisiología , Estrés Psicológico/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar
19.
Pharmacol Rep ; 63(2): 537-43, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21602609

RESUMEN

Preclinical data indicate the antidepressant activity of zinc and the involvement of the brain-derived neurotrophic factor (BDNF) in this mechanism. The present study investigates the effect of chronic (16 days) combined treatment with zinc (15 mg/kg zinc hydroaspartate) and imipramine (5 mg/kg) in chronic unpredictable stress (CUS) on the BDNF mRNA level in the rat brain. Moreover, serum zinc concentrations were also assessed. CUS induced a significant reduction in the BDNF mRNA level in the hippocampus by 21% but had no effect in the frontal cortex. Repeated treatment with zinc induced a significant increase in the BDNF mRNA level in the hippocampus in the unstressed animals by 12% and as in the chronically stressed animals by 14%, compared to the appropriate controls. Imipramine treatment did not affect this factor. However, combined treatment of zinc and imipramine induced a 12% elevation of the BDNF mRNA level in the stressed but not in the unstressed rats. CUS induced a 19% reduction in the serum zinc concentration, whereas combined treatment of zinc and imipramine reduced this concentration by 24% in the unstressed and increased it (by 20%) in the stressed animals. These results indicate that: 1) CUS induces a reduction in the BDNF gene expression with a concomitant diminution of serum zinc concentration and 2) the CUS-induced reduction in the BDNF gene expression is antagonized by chronic treatment with zinc.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Imipramina/farmacología , Estrés Psicológico/tratamiento farmacológico , Zinc/farmacología , Animales , Antidepresivos Tricíclicos/farmacología , Depresión/tratamiento farmacológico , Modelos Animales de Enfermedad , Quimioterapia Combinada , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Zinc/sangre
20.
Pharmacol Rep ; 61(6): 1184-91, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20081255

RESUMEN

Preclinical data indicate the involvement of glutamatergic and serotonergic pathways in the antidepressant activity of zinc. The present study investigated alterations in N-methyl-D-aspartate (NMDA)/glutamatergic and serotonergic receptors (using radioligand binding) induced by chronic treatment (14-day) with zinc hydroaspartate (65 mg/kg). Moreover, the mRNA and protein levels of brain-derived neurotrophic factor (BDNF) were also assessed. Chronic zinc administration reduced the affinity of glycine to glycine/NMDA receptors in the rat frontal cortex and increased the density of 5-HT(1A) and 5-HT(2A) serotonin receptors in the hippocampus and frontal cortex, respectively. These receptor alterations may be in part due to increased BDNF mRNA and protein levels in the rat frontal cortex. These results indicate that chronic zinc treatment alters glutamatergic and serotonergic systems, which is a hallmark of clinically effective antidepressants.


Asunto(s)
Antidepresivos/farmacología , Ácido Aspártico/farmacología , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Zinc/farmacología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/metabolismo , Glicina/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , ARN Mensajero/metabolismo , Ensayo de Unión Radioligante , Ratas , Ratas Wistar , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT2A/efectos de los fármacos , Receptor de Serotonina 5-HT2A/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...