Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 113(14): E2073-82, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-27001850

RESUMEN

Previous studies implicate the hypothalamic ventromedial nucleus (VMN) in glycemic control. Here, we report that selective inhibition of the subset of VMN neurons that express the transcription factor steroidogenic-factor 1 (VMN(SF1) neurons) blocks recovery from insulin-induced hypoglycemia whereas, conversely, activation of VMN(SF1) neurons causes diabetes-range hyperglycemia. Moreover, this hyperglycemic response is reproduced by selective activation of VMN(SF1) fibers projecting to the anterior bed nucleus of the stria terminalis (aBNST), but not to other brain areas innervated by VMN(SF1) neurons. We also report that neurons in the lateral parabrachial nucleus (LPBN), a brain area that is also implicated in the response to hypoglycemia, make synaptic connections with the specific subset of glucoregulatory VMN(SF1) neurons that project to the aBNST. These results collectively establish a physiological role in glucose homeostasis for VMN(SF1) neurons and suggest that these neurons are part of an ascending glucoregulatory LPBN→VMN(SF1)→aBNST neurocircuit.


Asunto(s)
Glucemia/metabolismo , Neuronas Aferentes/fisiología , Núcleo Hipotalámico Ventromedial/fisiología , Animales , Insulina/administración & dosificación , Ratones , Núcleo Hipotalámico Ventromedial/citología
2.
Hum Mol Genet ; 24(14): 3908-17, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25859008

RESUMEN

The neurodegenerative disorder spinocerebellar ataxia type 7 (SCA7) is caused by a polyglutamine (polyQ) expansion in the ataxin-7 protein, categorizing SCA7 as one member of a large class of heritable neurodegenerative proteinopathies. Cleavage of ataxin-7 by the protease caspase-7 has been demonstrated in vitro, and the accumulation of proteolytic cleavage products in SCA7 patients and mouse models has been identified as an early pathological change. However, it remains unknown whether a causal relationship exists between ataxin-7 proteolysis and in vivo SCA7 disease progression. To determine whether caspase cleavage is a critical event in SCA7 disease pathogenesis, we generated transgenic mice expressing polyQ-expanded ataxin-7 with a second-site mutation (D266N) to prevent caspase-7 proteolysis. When we compared SCA7-D266N mice with SCA7 mice lacking the D266N mutation, we found that SCA7-D266N mice exhibited improved motor performance, reduced neurodegeneration and substantial lifespan extension. Our findings indicate that proteolysis at the D266 caspase-7 cleavage site is an important mediator of ataxin-7 neurotoxicity, suggesting that inhibition of caspase-7 cleavage of polyQ-ataxin-7 may be a promising therapeutic strategy for this untreatable disorder.


Asunto(s)
Ataxina-7/metabolismo , Enfermedades Neurodegenerativas/genética , Péptidos/metabolismo , Regiones Promotoras Genéticas , Proteolisis , Degeneración Retiniana/genética , Animales , Ácido Aspártico/metabolismo , Ataxina-7/genética , Caspasa 7/genética , Caspasa 7/metabolismo , Modelos Animales de Enfermedad , Terapia Genética , Humanos , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas/terapia , Fenotipo , Células de Purkinje/metabolismo , Degeneración Retiniana/terapia
4.
Nature ; 493(7433): 480, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23344350
5.
Am J Physiol Endocrinol Metab ; 304(7): E734-46, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23384771

RESUMEN

Recent advances in human brown adipose tissue (BAT) imaging technology have renewed interest in the identification of BAT activators for the treatment of obesity and diabetes. In uncontrolled diabetes (uDM), activation of BAT is implicated in glucose lowering mediated by intracerebroventricular (icv) administration of leptin, which normalizes blood glucose levels in streptozotocin (STZ)-induced diabetic rats. The potent effect of icv leptin to increase BAT glucose uptake in STZ-diabetes is accompanied by the return of reduced plasma thyroxine (T4) levels and BAT uncoupling protein-1 (Ucp1) mRNA levels to nondiabetic controls. We therefore sought to determine whether activation of thyroid hormone receptors is sufficient in and of itself to lower blood glucose levels in STZ-diabetes and whether this effect involves activation of BAT. We found that, although systemic administration of the thyroid hormone (TR)ß-selective agonist GC-1 increases energy expenditure and induces further weight loss in STZ-diabetic rats, it neither increased BAT glucose uptake nor attenuated diabetic hyperglycemia. Even when GC-1 was administered in combination with a ß(3)-adrenergic receptor agonist to mimic sympathetic nervous system activation, glucose uptake was not increased in STZ-diabetic rats, nor was blood glucose lowered, yet this intervention potently activated BAT. Similar results were observed in animals treated with active thyroid hormone (T3) instead of GC-1. Taken together, our data suggest that neither returning normal plasma thyroid hormone levels nor BAT activation has any impact on diabetic hyperglycemia, and that in BAT, increases of Ucp1 gene expression and glucose uptake are readily dissociated from one another in this setting.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Diabetes Mellitus Experimental/metabolismo , Glucosa/metabolismo , Hiperglucemia/metabolismo , Sistema Nervioso Simpático/metabolismo , Termogénesis/fisiología , Receptores beta de Hormona Tiroidea/metabolismo , Acetatos/farmacología , Tejido Adiposo Pardo/efectos de los fármacos , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Animales , Composición Corporal/efectos de los fármacos , Diabetes Mellitus Experimental/complicaciones , Ingestión de Alimentos/efectos de los fármacos , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/etiología , Canales Iónicos/efectos de los fármacos , Canales Iónicos/metabolismo , Masculino , Proteínas Mitocondriales/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Fenoles/farmacología , Ratas , Ratas Wistar , Receptores Adrenérgicos beta 3/metabolismo , Estreptozocina , Sistema Nervioso Simpático/efectos de los fármacos , Termogénesis/efectos de los fármacos , Receptores beta de Hormona Tiroidea/agonistas , Triyodotironina/farmacología , Proteína Desacopladora 1
6.
J Neurosci ; 29(48): 15134-44, 2009 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-19955365

RESUMEN

Polyglutamine (polyQ) expansion within the ataxin-7 protein, a member of the STAGA [SPT3-TAF(II)31-GCN5L acetylase] and TFTC (GCN5 and TRRAP) chromatin remodeling complexes, causes the neurodegenerative disease spinocerebellar ataxia type 7 (SCA7). Proteolytic processing of ataxin-7 by caspase-7 generates N-terminal toxic polyQ-containing fragments that accumulate with disease progression and play an important role in SCA7 pathogenesis. To elucidate the basis for the toxicity of these fragments, we evaluated which posttranslational modifications of the N-terminal fragment of ataxin-7 modulate turnover and toxicity. Here, we show that mutating lysine 257 (K257), an amino acid adjacent to the caspase-7 cleavage site of ataxin-7 regulates turnover of the truncation product in a repeat-dependent manner. Modification of ataxin-7 K257 by acetylation promotes accumulation of the fragment, while unmodified ataxin-7 is degraded. The degradation of the caspase-7 cleavage product is mediated by macroautophagy in cell culture and primary neuron models of SCA7. Consistent with this, the fragment colocalizes with autophagic vesicle markers, and enhanced fragment accumulation increases in these lysosomal structures. We suggest that the levels of fragment accumulation within the cell is a key event in SCA7 neurodegeneration, and enhancing clearance of polyQ-containing fragments may be an effective target to reduce neurotoxicity in SCA7.


Asunto(s)
Autofagia/genética , Caspasa 7/metabolismo , Mucoproteínas/genética , Proteínas del Tejido Nervioso/metabolismo , Péptidos/genética , Procesamiento Proteico-Postraduccional/genética , Acetilación , Animales , Animales Recién Nacidos , Ataxina-7 , Caspasa 7/genética , Células Cultivadas , Cerebelo/citología , Modelos Animales de Enfermedad , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Neuronas/fisiología , Priones/genética , Priones/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Interferencia de ARN/fisiología , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo , Ataxias Espinocerebelosas/patología , Transfección/métodos
7.
Nat Neurosci ; 9(10): 1302-11, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16936724

RESUMEN

Non-neuronal cells may be pivotal in neurodegenerative disease, but the mechanistic basis of this effect remains ill-defined. In the polyglutamine disease spinocerebellar ataxia type 7 (SCA7), Purkinje cells undergo non-cell-autonomous degeneration in transgenic mice. We considered the possibility that glial dysfunction leads to Purkinje cell degeneration, and generated mice that express ataxin-7 in Bergmann glia of the cerebellum with the Gfa2 promoter. Bergmann glia-specific expression of mutant ataxin-7 was sufficient to produce ataxia and neurodegeneration. Expression of the Bergmann glia-specific glutamate transporter GLAST was reduced in Gfa2-SCA7 mice and was associated with impaired glutamate transport in cultured Bergmann glia, cerebellar slices and cerebellar synaptosomes. Ultrastructural analysis of Purkinje cells revealed findings of dark cell degeneration consistent with excitotoxic injury. Our studies indicate that impairment of glutamate transport secondary to glial dysfunction contributes to SCA7 neurodegeneration, and suggest a similar role for glial dysfunction in other polyglutamine diseases and SCAs.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/metabolismo , Expresión Génica/fisiología , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuroglía/metabolismo , Factores de Edad , Anciano , Animales , Animales Recién Nacidos , Ataxina-7 , Conducta Animal , Western Blotting/métodos , Encéfalo/patología , Células Cultivadas , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Ácido Glutámico/metabolismo , Humanos , Inmunohistoquímica/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión/métodos , Proteínas del Tejido Nervioso/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Neuroglía/ultraestructura , Transfección/métodos
8.
Front Nutr ; 6: 66, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31139631

RESUMEN

Background: The energy content of whole, fresh fruit derives primarily from simple sugars, which are currently under heightened scrutiny for their potential contribution to obesity and chronic disease risk. Yet fruit also has a relatively low energy density, moderate palatability/reward value, and high fiber content, which together may limit energy intake. Although reasoned arguments can be made that fruit is fattening or slimming, the question is best resolved empirically. Methods: Methods were preregistered with PROSPERO (CRD42018111830). The primary outcome is the impact of whole, fresh fruit consumption on measures of adiposity including body weight in randomized controlled trials (RCTs). Secondary outcomes are the impact of whole, fresh fruit consumption on energy intake in RCTs, and the association between whole, fresh fruit consumption and changes in measures of adiposity in prospective observational studies. CENTRAL and PubMed databases were searched through October 2018. Cochrane risk of bias tool was used to assess risk of bias in RCTs, and the GRADE method was used to judge and convey the certainty of conclusions. Reporting follows PRISMA guidelines. Results: RCTs, and particularly those of higher quality, suggest that increasing whole, fresh fruit consumption promotes weight maintenance or modest weight loss over periods of 3-24 weeks (moderate certainty), with limited evidence suggesting that a high intake of fruit favors weight loss among people with overweight or obesity. Consistent with this, single-meal RCTs suggest that consuming whole, fresh fruit tends to decrease energy intake, particularly when consumed prior to a meal or when displacing more energy-dense foods (moderate certainty). Prospective observational studies suggest that habitually higher fruit intake is not associated with weight change, or is associated with modest protection against weight gain, over five or more years. Conclusions: Current evidence suggests that whole, fresh fruit consumption is unlikely to contribute to excess energy intake and adiposity, but rather has little effect on these outcomes or constrains them modestly. Single-meal RCTs, RCTs lasting 3-24 weeks, and long-term observational studies are relatively consistent in supporting this conclusion. Whole, fresh fruit probably does not contribute to obesity and may have a place in the prevention and management of excess adiposity.

9.
Diabetes ; 66(4): 920-934, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28073831

RESUMEN

Effectors of the phosphoinositide 3-kinase (PI3K) signal transduction pathway contribute to the hypothalamic regulation of energy and glucose homeostasis in divergent ways. Here we show that central nervous system (CNS) action of the PI3K signaling intermediate atypical protein kinase C (aPKC) constrains food intake, weight gain, and glucose intolerance in both rats and mice. Pharmacological inhibition of CNS aPKC activity acutely increases food intake and worsens glucose tolerance in chow-fed rodents and causes excess weight gain during high-fat diet (HFD) feeding. Similarly, selective deletion of the aPKC isoform Pkc-λ in proopiomelanocortin (POMC) neurons disrupts leptin action, reduces melanocortin content in the paraventricular nucleus, and markedly increases susceptibility to obesity, glucose intolerance, and insulin resistance specifically in HFD-fed male mice. These data implicate aPKC as a novel regulator of energy and glucose homeostasis downstream of the leptin-PI3K pathway in POMC neurons.


Asunto(s)
Ingestión de Alimentos/genética , Intolerancia a la Glucosa/genética , Glucosa/metabolismo , Isoenzimas/genética , Neuronas/metabolismo , Obesidad/genética , Proteína Quinasa C/genética , Aumento de Peso/genética , Animales , Dieta Alta en Grasa , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Intolerancia a la Glucosa/metabolismo , Hipotálamo/metabolismo , Resistencia a la Insulina , Leptina/metabolismo , Masculino , Melanocortinas/metabolismo , Ratones , Obesidad/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proopiomelanocortina/efectos de los fármacos , Proopiomelanocortina/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Ratas , Transducción de Señal , Aumento de Peso/efectos de los fármacos
10.
Adv Nutr ; 6(6): 660-4, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26567191

RESUMEN

Linoleic acid (LA) is a bioactive fatty acid with diverse effects on human physiology and pathophysiology. LA is a major dietary fatty acid, and also one of the most abundant fatty acids in adipose tissue, where its concentration reflects dietary intake. Over the last half century in the United States, dietary LA intake has greatly increased as dietary fat sources have shifted toward polyunsaturated seed oils such as soybean oil. We have conducted a systematic literature review of studies reporting the concentration of LA in subcutaneous adipose tissue of US cohorts. Our results indicate that adipose tissue LA has increased by 136% over the last half century and that this increase is highly correlated with an increase in dietary LA intake over the same period of time.


Asunto(s)
Tejido Adiposo/química , Ácido Linoleico/análisis , Adulto , Dieta/tendencias , Grasas Insaturadas en la Dieta/administración & dosificación , Grasas Insaturadas en la Dieta/análisis , Humanos , Ácido Linoleico/administración & dosificación , Grasa Subcutánea/química , Estados Unidos
11.
Obesity (Silver Spring) ; 23(11): 2142-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26530930

RESUMEN

OBJECTIVE: To use quantitative magnetic resonance imaging (MRI) to test whether mediobasal hypothalamic (MBH) gliosis is associated with obesity and insulin resistance in humans. METHODS: Sixty-seven participants underwent a fasting blood draw and MRI. Cases with radiologic evidence of MBH gliosis (N = 22) were identified as the upper tertile of left MBH T2 relaxation time and were compared to controls (N = 23) from the lowest tertile. In a separate postmortem study, brain slices (N = 10) through the MBH were imaged by MRI and stained for glial fibrillary acidic protein (GFAP). RESULTS: In all participants, longer T2 relaxation time in the left MBH was associated with higher BMI (P = 0.01). Compared with controls, cases had longer T2 relaxation times in the right MBH (P < 0.05), as well as higher BMI (P < 0.05), fasting insulin concentrations (P < 0.01), and HOMA-IR values (P < 0.01), adjusted for sex and age. Elevations in insulin and HOMA-IR were also independent of BMI. In the postmortem study, GFAP staining intensity was positively associated with MBH T2 relaxation time (P < 0.05), validating an MRI-based method for the detection of MBH gliosis in humans. CONCLUSIONS: These findings link hypothalamic gliosis to insulin resistance in humans and suggest that the link is independent of the level of adiposity.


Asunto(s)
Gliosis/diagnóstico , Hipotálamo/patología , Resistencia a la Insulina , Obesidad/diagnóstico , Adiposidad/fisiología , Adolescente , Adulto , Estudios de Casos y Controles , Ayuno/metabolismo , Femenino , Humanos , Insulina/metabolismo , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Adulto Joven
12.
Endocrinology ; 155(8): 2858-67, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24914942

RESUMEN

Gliosis, the activation of astrocyte and microglial cell populations, is a hallmark of central nervous system injury and is detectable using either immunohistochemistry or in vivo magnetic resonance imaging (MRI). Obesity in rodents and humans is associated with gliosis of the arcuate nucleus, a key hypothalamic region for the regulation of energy homeostasis and adiposity, but whether this response is permanent or reversible is unknown. Here we combine terminal immunohistochemistry analysis with serial, noninvasive MRI to characterize the progression and reversibility of hypothalamic gliosis in high-fat diet (HFD)-fed mice. The effects of HFD feeding for 16 weeks to increase body weight and adiposity relative to chow were nearly normalized after the return to chow feeding for an additional 4 weeks in the diet-reversal group. Mice maintained on the HFD for the full 20-week study period experienced continued weight gain associated with the expected increases of astrocyte and microglial activation in the arcuate nucleus, but these changes were not observed in the diet-reversal group. The proopiomelanocortin neuron number did not differ between groups. Although MRI demonstrated a positive correlation between body weight, adiposity, and the gliosis-associated T2 signal in the mediobasal hypothalamus, it did not detect the reversal of gliosis among the HFD-fed mice after the return to chow diet. We conclude that hypothalamic gliosis associated with 16-week HFD feeding is largely reversible in rodents, consistent with the reversal of the HFD-induced obesity phenotype, and extend published evidence regarding the utility of MRI as a tool for studying obesity-associated hypothalamic gliosis in vivo.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Dieta Alta en Grasa/efectos adversos , Gliosis/etiología , Enfermedades Hipotalámicas/etiología , Enfermedades Hipotalámicas/metabolismo , Obesidad/complicaciones , Obesidad/metabolismo , Adiposidad , Animales , Ingestión de Alimentos , Enfermedades Hipotalámicas/prevención & control , Inmunohistoquímica , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/dietoterapia , Pérdida de Peso
13.
Cell Rep ; 8(2): 477-86, 2014 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-25043177

RESUMEN

We have analyzed the yeast replicative lifespan of a large number of open reading frame (ORF) deletions. Here, we report that strains lacking genes SGF73, SGF11, and UBP8 encoding SAGA/SLIK complex histone deubiquitinase module (DUBm) components are exceptionally long lived. Strains lacking other SAGA/SALSA components, including the acetyltransferase encoded by GCN5, are not long lived; however, these genes are required for the lifespan extension observed in DUBm deletions. Moreover, the SIR2-encoded histone deacetylase is required, and we document both a genetic and physical interaction between DUBm and Sir2. A series of studies assessing Sir2-dependent functions lead us to propose that DUBm strains are exceptionally long lived because they promote multiple prolongevity events, including reduced rDNA recombination and altered silencing of telomere-proximal genes. Given that ataxin-7, the human Sgf73 ortholog, causes the neurodegenerative disease spinocerebellar ataxia type 7, our findings indicate that the genetic and epigenetic interactions between DUBm and SIR2 will be relevant to neurodegeneration and aging.


Asunto(s)
Replicación del ADN , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/metabolismo , Sirtuina 2/metabolismo , Transactivadores/metabolismo , Proliferación Celular , Endopeptidasas/genética , Endopeptidasas/metabolismo , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Unión Proteica , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/fisiología , Proteínas de Saccharomyces cerevisiae/genética , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/genética , Sirtuina 2/genética , Transactivadores/genética
15.
Mol Metab ; 2(2): 116-22, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24199157

RESUMEN

The mediobasal hypothalamus (MBH) plays a central role in the regulation of food intake and energy balance. Although the excitatory neurotransmitter glutamate is implicated in energy balance regulation by the MBH, the hypothesis that feeding elicits local glutamate release remains untested. To test this hypothesis, we employed a glutamate biosensor that measures glutamate concentrations at 1-s intervals in conscious, freely behaving rats. Results indicate that feeding is associated with an increase of MBH glutamate concentration that occurs within 1-2 s of oral contact with a food pellet, and the glutamate response to a palatable high-fat pellet is greatly exaggerated relative to chow. In contrast, glutamate responses were not observed during water ingestion or other observed behaviors. These findings indicate that feeding is associated with rapid release of glutamate in the MBH, that this release is exaggerated with an obesogenic food, and that this response is likely stimulated by orosensory factors.

16.
Endocrinology ; 154(9): 3067-76, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23782941

RESUMEN

In rodent models of type 1 diabetes, leptin administration into brain ventricles normalizes blood glucose at doses that have no effect when given peripherally. The ventromedial nucleus of the hypothalamus (VMN) is a potential target for leptin's antidiabetic effects because leptin-sensitive neurons in this brain area are implicated in glucose homeostasis. To test this hypothesis, we injected leptin directly into the bilateral VMN of rats with streptozotocin-induced uncontrolled diabetes mellitus. This intervention completely normalized both hyperglycemia and the elevated rates of hepatic glucose production and plasma glucagon levels but had no effect on tissue glucose uptake in the skeletal muscle or brown adipose tissue as measured using tracer dilution techniques during a basal clamp. To determine whether VMN leptin signaling is required for leptin-mediated normalization of diabetic hyperglycemia, we studied mice in which the leptin receptor gene was deleted in VMN steroidogenic factor 1 neurons using cre-loxP technology. Our findings indicate leptin action within these neurons is not required for the correction of diabetic hyperglycemia by central leptin infusion. We conclude that leptin signaling in the VMN is sufficient to mediate leptin's antidiabetic action but may not be necessary for this effect. Leptin action within a distributed neuronal network may mediate its effects on glucose homeostasis.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Hiperglucemia/prevención & control , Leptina/metabolismo , Neuronas/metabolismo , Receptores de Leptina/metabolismo , Transducción de Señal , Núcleo Hipotalámico Ventromedial/metabolismo , Animales , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/patología , Glucagón/sangre , Glucagón/metabolismo , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/metabolismo , Gluconeogénesis/efectos de los fármacos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/metabolismo , Hipoglucemiantes/uso terapéutico , Infusiones Intraventriculares , Inyecciones Intraventriculares , Leptina/administración & dosificación , Leptina/genética , Leptina/uso terapéutico , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Ratas , Ratas Wistar , Receptores de Leptina/agonistas , Receptores de Leptina/genética , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Transducción de Señal/efectos de los fármacos , Núcleo Hipotalámico Ventromedial/efectos de los fármacos , Núcleo Hipotalámico Ventromedial/patología
17.
Brain Res ; 1512: 97-105, 2013 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-23548599

RESUMEN

Astrocytes respond to multiple forms of central nervous system (CNS) injury by entering a reactive state characterized by morphological changes and a specific pattern of altered protein expression. Termed astrogliosis, this response has been shown to strongly influence the injury response and functional recovery of CNS tissues. This pattern of CNS inflammation and injury associated with astrogliosis has recently been found to occur in the energy homeostasis centers of the hypothalamus during diet-induced obesity (DIO) in rodent models, but the characterization of the astrocyte response remains incomplete. Here, we report that astrocytes in the mediobasal hypothalamus respond robustly and rapidly to purified high-fat diet (HFD) feeding by cleaving caspase-3, a protease whose cleavage is often associated with apoptosis. Although obesity develops in HFD-fed rats by day 14, caspase-3 cleavage occurs by day 3, prior to the development of obesity, suggesting the possibility that it could play a causal role in the hypothalamic neuropathology and fat gain observed in DIO. Caspase-3 cleavage is not associated with an increase in the rate of apoptosis, as determined by TUNEL staining, suggesting it plays a non-apoptotic role analogous to the response to excitotoxic neuron injury. Our results indicate that astrocytes in the mediobasal hypothalamus respond rapidly and robustly to HFD feeding, activating caspase-3 in the absence of apoptosis, a process that has the potential to influence the course of DIO.


Asunto(s)
Astrocitos/metabolismo , Caspasa 3/metabolismo , Dieta Alta en Grasa/efectos adversos , Hipotálamo/patología , Obesidad/inducido químicamente , Obesidad/patología , Análisis de Varianza , Animales , Apoptosis/fisiología , Composición Corporal/fisiología , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Etiquetado Corte-Fin in Situ , Masculino , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Wistar
18.
Diabetes ; 62(5): 1512-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23274899

RESUMEN

Recent evidence suggests that central leptin administration fully normalizes hyperglycemia in a rodent model of uncontrolled insulin-deficient diabetes by reducing hepatic glucose production (HGP) and by increasing glucose uptake. The current studies were undertaken to determine whether brain-derived neurotrophic factor (BDNF) action in the brain lowers blood glucose in uncontrolled insulin-deficient diabetes and to investigate the mechanisms mediating this effect. Adult male rats implanted with cannulas to either the lateral cerebral ventricle or the ventromedial hypothalamic nucleus (VMN) received either vehicle or streptozotocin to induce uncontrolled insulin-deficient diabetes. Three days later, animals received daily intracerebroventricular or intra-VMN injections of either BDNF or its vehicle. We found that repeated daily intracerebroventricular administration of BDNF attenuated diabetic hyperglycemia independent of changes in food intake. Instead, using tracer dilution techniques during a basal clamp, we found that BDNF lowered blood glucose levels by potently suppressing HGP, without affecting tissue glucose uptake, an effect associated with normalization of both plasma glucagon levels and hepatic expression of gluconeogenic genes. Moreover, BDNF microinjection directly into the VMN also lowered fasting blood glucose levels in uncontrolled insulin-deficient diabetes, but this effect was modest compared with intracerebroventricular administration. We conclude that central nervous system BDNF attenuates diabetic hyperglycemia via an insulin-independent mechanism. This action of BDNF likely involves the VMN and is associated with inhibition of glucagon secretion and a decrease in the rate of HGP.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Gluconeogénesis , Hiperglucemia/prevención & control , Hígado/metabolismo , Neuronas/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Transporte Biológico/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/administración & dosificación , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Conducta Alimentaria/efectos de los fármacos , Glucagón/sangre , Glucagón/metabolismo , Gluconeogénesis/efectos de los fármacos , Glucosa/metabolismo , Hipoglucemiantes/uso terapéutico , Inyecciones Intraventriculares , Insulina/uso terapéutico , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/metabolismo , Hígado/efectos de los fármacos , Masculino , Neuronas/efectos de los fármacos , Ratas , Ratas Wistar , Núcleo Hipotalámico Ventromedial/efectos de los fármacos
19.
J Clin Endocrinol Metab ; 97(3): 745-55, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22238401

RESUMEN

CONTEXT: Obesity has emerged as one of the leading medical challenges of the 21st century. The resistance of this disorder to effective, long-term treatment can be traced to the fact that body fat stores are subject to homeostatic regulation in obese individuals, just as in lean individuals. Because the growing obesity epidemic is linked to a substantial increase in daily energy intake, a key priority is to delineate how mechanisms governing food intake and body fat content are altered in an obesogenic environment. EVIDENCE ACQUISITION: We considered all relevant published research and cited references that represented the highest quality evidence available. Where space permitted, primary references were cited. EVIDENCE SYNTHESIS: The increase of energy intake that has fueled the U.S. obesity epidemic is linked to greater availability of highly rewarding/palatable and energy-dense food. Obesity occurs in genetically susceptible individuals and involves the biological defense of an elevated body fat mass, which may result in part from interactions between brain reward and homeostatic circuits. Inflammatory signaling, accumulation of lipid metabolites, or other mechanisms that impair hypothalamic neurons may also contribute to the development of obesity and offer a plausible mechanism to explain the biological defense of elevated body fat mass. CONCLUSIONS: Despite steady research progress, mechanisms underlying the resistance to fat loss once obesity is established remain incompletely understood. Breakthroughs in this area may be required for the development of effective new obesity prevention and treatment strategies.


Asunto(s)
Tejido Adiposo/fisiología , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Obesidad/etiología , Índice de Masa Corporal , Homeostasis/fisiología , Humanos , Obesidad/fisiopatología
20.
J Clin Invest ; 122(1): 153-62, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22201683

RESUMEN

Rodent models of obesity induced by consuming high-fat diet (HFD) are characterized by inflammation both in peripheral tissues and in hypothalamic areas critical for energy homeostasis. Here we report that unlike inflammation in peripheral tissues, which develops as a consequence of obesity, hypothalamic inflammatory signaling was evident in both rats and mice within 1 to 3 days of HFD onset, prior to substantial weight gain. Furthermore, both reactive gliosis and markers suggestive of neuron injury were evident in the hypothalamic arcuate nucleus of rats and mice within the first week of HFD feeding. Although these responses temporarily subsided, suggesting that neuroprotective mechanisms may initially limit the damage, with continued HFD feeding, inflammation and gliosis returned permanently to the mediobasal hypothalamus. Consistent with these data in rodents, we found evidence of increased gliosis in the mediobasal hypothalamus of obese humans, as assessed by MRI. These findings collectively suggest that, in both humans and rodent models, obesity is associated with neuronal injury in a brain area crucial for body weight control.


Asunto(s)
Hipotálamo/patología , Obesidad/patología , Adolescente , Adulto , Animales , Secuencia de Bases , Citocinas/genética , Dieta Alta en Grasa/efectos adversos , Femenino , Gliosis/etiología , Gliosis/patología , Humanos , Hipotálamo/lesiones , Hipotálamo/metabolismo , Inflamación/etiología , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , FN-kappa B/metabolismo , Neuronas/patología , Obesidad/genética , Obesidad/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Long-Evans , Transducción de Señal , Factores de Tiempo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA