Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Mol Neurosci ; 56(4): 829-839, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25687329

RESUMEN

To test the hypothesis that astrocytic leptin signaling induces an overall potentiation of the neuronal response to leptin, we generated a new line of astrocyte-specific leptin receptor knockout (ALKO-Δ1) mice in which no leptin receptor is expressed in astrocytes. Corresponding to cell-specific Cre recombinase expression in hypothalamic astrocytes but not neurons, this new strain of ALKO mice had attenuated pSTAT3 signaling in the arcuate nucleus of the hypothalamus 30 min after intracerebroventricular delivery of leptin. In response to high-fat diet for 2 months, the ALKO mice showed a greater increase of percent fat and blood leptin concentration. This coincided with a mild reactive gliosis in the hypothalamus. Overall, the absence of leptin receptors in astrocytes attenuated hypothalamic pSTAT3 signaling, induced a mild reactive morphology, and promoted the development of diet-induced obesity. We conclude that leptin signaling in astrocytes is essential for the homeostasis of neuroendocrine regulation in obesity.


Asunto(s)
Astrocitos/metabolismo , Leptina/metabolismo , Receptores de Leptina/metabolismo , Transducción de Señal , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/metabolismo , Femenino , Gliosis , Leptina/sangre , Masculino , Ratones , Receptores de Leptina/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
2.
J Neurosci ; 34(44): 14697-706, 2014 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-25355222

RESUMEN

The blood-brain barrier (BBB) is a large regulatory and exchange interface between the brain and peripheral circulation. We propose that changes of the BBB contribute to many pathophysiological processes in the brain of subjects with chronic sleep restriction (CSR). To achieve CSR that mimics a common pattern of human sleep loss, we quantified a new procedure of sleep disruption in mice by a week of consecutive sleep recording. We then tested the hypothesis that CSR compromises microvascular function. CSR not only diminished endothelial and inducible nitric oxide synthase, endothelin1, and glucose transporter expression in cerebral microvessels of the BBB, but it also decreased 2-deoxy-glucose uptake by the brain. The expression of several tight junction proteins also was decreased, whereas the level of cyclooxygenase-2 increased. This coincided with an increase of paracellular permeability of the BBB to the small tracers sodium fluorescein and biotin. CSR for 6 d was sufficient to impair BBB structure and function, although the increase of paracellular permeability returned to baseline after 24 h of recovery sleep. This merits attention not only in neuroscience research but also in public health policy and clinical practice.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Encéfalo/fisiopatología , Privación de Sueño/fisiopatología , Sueño/fisiología , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Regulación hacia Abajo , Células Endoteliales/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Permeabilidad , Privación de Sueño/metabolismo
3.
Brain Behav Immun ; 40: 61-73, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24576482

RESUMEN

Leptin, a pleiotropic adipokine, crosses the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB) from the periphery and facilitates experimental autoimmune encephalomyelitis (EAE). EAE induces dynamic changes of leptin receptors in enriched brain and spinal cord microvessels, leading to further questions about the potential roles of endothelial leptin signaling in EAE progression. In endothelial leptin receptor specific knockout (ELKO) mice, there were lower EAE behavioral scores in the early phase of the disorder, better preserved BSCB function shown by reduced uptake of sodium fluorescein and leukocyte infiltration into the spinal cord. Flow cytometry showed that the ELKO mutation decreased the number of CD3 and CD45 cells in the spinal cord, although immune cell profiles in peripheral organs were unchanged. Not only were CD4(+) and CD8(+) T lymphocytes reduced, there were also lower numbers of CD11b(+)Gr1(+) granulocytes in the spinal cord of ELKO mice. In enriched microvessels from the spinal cord of the ELKO mice, the decreased expression of mRNAs for a few tight junction proteins was less pronounced in ELKO than WT mice, as was the elevation of mRNA for CCL5, CXCL9, IFN-γ, and TNF-α. Altogether, ELKO mice show reduced inflammation at the level of the BSCB, less leukocyte infiltration, and better preserved tight junction protein expression and BBB function than WT mice after EAE. Although leptin concentrations were high in ELKO mice and microvascular leptin receptors show an initial elevation before inhibition during the course of EAE, removal of leptin signaling helped to reduce disease burden. We conclude that endothelial leptin signaling exacerbates BBB dysfunction to worsen EAE.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Leptina/sangre , Leucocitos/inmunología , Receptores de Leptina/metabolismo , Médula Espinal/inmunología , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Citocinas/inmunología , Células Endoteliales/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Receptores de Leptina/genética , Índice de Severidad de la Enfermedad , Transducción de Señal , Médula Espinal/irrigación sanguínea , Uniones Estrechas/inmunología
4.
J Cereb Blood Flow Metab ; 34(1): 43-51, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24064496

RESUMEN

The blood-brain barrier (BBB) is a regulatory interface between the central nervous system and the rest of the body. However, BBB changes in obesity and metabolic syndrome have not been fully elucidated. We hypothesized that obesity reduces energy metabolism in the cerebral microvessels composing the BBB, reflected by downregulation of protein expression and function. We performed comparative proteomic analyses in enriched microvessels from the cerebral cortex of mice 2 months after ingestion of a high-fat diet or regular rodent chow. In mice with diet-induced obesity (DIO), there was downregulation of 47 proteins in the cerebral microvessels, including cytoskeletal proteins, chaperons, enzymes, transport-related proteins, and regulators for transcriptional and translational activities. Only two proteins, involved in messenger RNA (mRNA) transport and processing, were upregulated. The changes of these proteins were further validated by quantitative polymerase chain reaction (qPCR), western blotting, and immunofluorescent staining of freshly isolated microvessels, in samples obtained from different batches of mice. The predominant downregulation suggests that DIO suppresses metabolic activity of BBB microvessels. The finding of a hypometabolic state of the BBB in mice at the chronic stage of DIO is unexpected and unprecedented; it may provide novel mechanistic insight into how obesity influences CNS function via regulatory changes of the BBB.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Dieta Alta en Grasa/efectos adversos , Obesidad/metabolismo , Biosíntesis de Proteínas , Animales , Western Blotting , Modelos Animales de Enfermedad , Regulación hacia Abajo , Electroforesis en Gel Bidimensional , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microvasos/metabolismo , Microvasos/patología , Obesidad/etiología , Obesidad/patología , Proteómica , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación hacia Arriba
5.
Fluids Barriers CNS ; 11(1): 27, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25601899

RESUMEN

BACKGROUND: We have recently shown that mice with experimental autoimmune encephalomyelitis (EAE) have increased sleep fragmentation (SF) and reduced sleep efficiency, and that the extent of SF correlates with the severity of disease. It is not yet clear whether and how sleep promotes recovery from autoimmune attacks. We hypothesized that SF promotes leukocyte infiltration across the blood-spinal cord barrier, impairs immune regulation, and thus worsens EAE. METHODS: Three groups of C57 mice were studied: Resting EAE; SF EAE with the mice subjected to the SF maneuver 12 h /day during zeitgeber time (ZT) 0-12 h; and naïve controls with neither EAE nor SF. Besides monitoring of the incidence and severity of EAE, the immune profiles of leukocytes in the spinal cord as well as those in the spleen were determined. RESULTS: When analyzed 16 days after EAE induction, at which time the SF was terminated, the SF group had a greater number of CD4(+) T cells and a higher percent of CD4(+) cells among all leukocytes in the spinal cord than the resting EAE group. When allowed to recover to 28 days after EAE induction, the SF mice had lower EAE scores than the resting EAE group. EAE induced splenomegaly and an increase of Gr1(+)CD11b(+) myeloid-derived suppressor cells in the splenocytes. However, SF treatment had no additional effect on either peripheral splenocytes or granulocytes that reached the spinal cord. CONCLUSION: The SF maneuver facilitated the migration of encephalopathic lymphocytes into the spinal cord. Paradoxically, these mice had a better EAE score after cessation of SF compared with mice without SF.

6.
Fluids Barriers CNS ; 10(1): 32, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24176017

RESUMEN

BACKGROUND: Fibroblast growth factor (FGF)-19, an endocrine FGF protein mainly produced by the ileum, stimulates metabolic activity and alleviates obesity. FGF19 modulates metabolism after either intravenous or intracerebroventricular injection, and its receptor FGFR4 is present in the hypothalamus. This led to the question whether blood-borne FGF19 crosses the blood-brain barrier (BBB) to exert its metabolic effects. METHODS: We determined the pharmacokinetics of FGF19 permeation from blood to brain in comparison with its distribution in peripheral organs. Multiple-time regression analysis after intravenous bolus injection, in-situ brain perfusion, and HPLC assays were performed. RESULTS: FGF19 was relatively stable in blood and in the brain compartment. Significant influx was seen in the presence of excess unlabeled FGF19 in blood. This coincided with a slower decline of 125I-FGF19 in blood which suggested there was decreased clearance or peripheral tissue uptake. In support of an altered pattern of peripheral processing of 125I-FGF19 by excess unlabeled FGF19, the high influx to liver was significantly attenuated, whereas the minimal renal uptake was linearly accelerated. In the present setting, we did not detect a saturable transport of FGF19 across the BBB, as the entry rate of 125I-FGF19 was not altered by excess unlabeled FGF19 or its mouse homologue FGF15 during in-situ brain perfusion. CONCLUSION: FGF19 remained stable in the blood and brain compartments for up to 10 min. Its influx to the brain was non-linear, non-saturable, and affected by its blood concentration and distribution in peripheral organs. Liver showed a robust and specific uptake of FGF19 that could be inhibited by the presence of excess unlabeled FGF19, whereas kidney clearance was dose-dependent.

7.
J Mol Neurosci ; 51(3): 1038-45, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23955775

RESUMEN

Excessive obesity correlates with hypersomnolence and impaired cognitive function, presumably induced by metabolic factors and cytokines. Production of the adipokine leptin correlates with the amount of adiposity, and leptin has been shown to promote sleep. To determine whether leptin plays a major role in the hypersomnolence of obesity, we measured sleep architecture in pan-leptin receptor knockout (POKO) mice that do not respond to leptin because of the production of a mutant, non-signaling receptor. The obese POKO mice had more non-rapid eye movement (NREM) sleep and less waking time than their littermate controls. This was mainly seen during the light span, although increased bouts of rapid eye movement sleep were also seen in the dark span. The increase of NREM sleep correlated with the extent of obesity. The POKO mice also had decreased locomotor activity and more immobility in the open field test, but there was no increase of forced immobility nor reduction of sucrose intake as would be seen in depression. The increased NREM sleep and reduced locomotor activity in the POKO mice suggest that it was obesity, rather than leptin signaling, that played a predominant role in altering sleep architecture and activity.


Asunto(s)
Trastornos de Somnolencia Excesiva/genética , Actividad Motora , Receptores de Leptina/genética , Animales , Trastornos de Somnolencia Excesiva/etiología , Trastornos de Somnolencia Excesiva/fisiopatología , Ratones , Ratones Noqueados , Obesidad/complicaciones , Obesidad/genética , Sueño REM , Vigilia
8.
PLoS One ; 8(8): e69356, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23950892

RESUMEN

Interleukin (IL)-15 is a ubiquitously expressed cytokine that in the basal state is mainly localized intracellularly, including the nucleus. Unexpectedly, tumor necrosis factor-α (TNF) time-dependently induced nuclear export of IL-15Rα and IL15. This process was inhibited by leptomycine B (LMB), a specific inhibitor of nuclear export receptor chromosomal region maintenance 1 (CRM1). In the presence of TNF, LMB co-treatment led to accumulation of both IL-15Rα and IL-15 in the nucleus of HeLa cells, suggesting that CRM1 facilitates nuclear export and that TNF enhances CRM1 activity. Once in the cytoplasm, IL-15 showed partial co-localization with late endosomes but very little with other organelles tested 4 h after TNF treatment. IL-15Rα showed co-localization with both early and late endosomes, and to a lesser extent with endoplasmic reticulum and Golgi. This indicates different kinetics and possibly different trafficking routes of IL-15 from its specific receptor. The TNF-induced secretion of IL-15 was attenuated by pretreatment of cells by brefeldin A that inhibits ER-to-Golgi transport, or by use of domain negative ADP-ribosylation factor 6 (ARF6) that interferes with exocytotic sorting. We conclude that TNF abolishes nuclear localization of IL-15 and IL-15Rα by acting on CRM1, and it facilitates exocytosis of IL-15 with the involvement of ARF6.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Interleucina-15/metabolismo , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/genética , Transporte Activo de Núcleo Celular/efectos de los fármacos , Western Blotting , Brefeldino A/farmacología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Retículo Endoplásmico/metabolismo , Endosomas/metabolismo , Ácidos Grasos Insaturados/farmacología , Aparato de Golgi/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Inmunohistoquímica , Subunidad alfa del Receptor de Interleucina-15/genética , Subunidad alfa del Receptor de Interleucina-15/metabolismo , Carioferinas/antagonistas & inhibidores , Microscopía Fluorescente , Unión Proteica , Transporte de Proteínas/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Factores de Tiempo , Proteína Exportina 1
9.
Brain Behav Immun ; 34: 98-107, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23916894

RESUMEN

Leptin is commonly thought to play a detrimental role in exacerbating experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis. Paradoxically, we show here that astrocytic leptin signaling has beneficial effects in reducing disease severity. In the astrocyte specific leptin receptor knockout (ALKO) mouse in which leptin signaling is absent in astrocytes, there were higher EAE scores (more locomotor deficits) than in the wildtype counterparts. The difference mainly occurred at a late stage of EAE when wildtype mice showed signs of recovery whereas ALKO mice continued to deteriorate. The more severe symptoms in ALKO mice coincided with more infiltrating cells in the spinal cord and perivascular brain parenchyma, more demyelination, more infiltrating CD4 cells, and a lower percent of neutrophils in the spinal cord 28 days after EAE induction. Cultured astrocytes from wildtype mice showed increased adenosine release in response to interleukin-6 and the hippocampus of wildtype mice had increased adenosine production 28 days after EAE induction, but the ALKO mutation abolished the increase in both conditions. This indicates a role of astrocytic leptin in normal gliotransmitter release and astrocyte functions. The worsening of EAE in the ALKO mice in the late stage suggests that astrocytic leptin signaling helps to clear infiltrating leukocytes and reduce autoimmune destruction of the CNS.


Asunto(s)
Astrocitos/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Receptores de Leptina/genética , Adenosina/análisis , Adenosina/metabolismo , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Hipocampo/patología , Leucocitos/metabolismo , Ratones , Ratones Noqueados , Receptores de Leptina/metabolismo , Médula Espinal/patología
10.
J Appl Physiol (1985) ; 115(7): 995-1003, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23869060

RESUMEN

Leptin, a hormone mainly produced by fat cells, shows cell-specific effects to regulate feeding and metabolic activities. We propose that an important feature of metabolic dysregulation resulting in obesity is the loss of the circadian rhythm of biopotentials. This was tested in the pan-leptin receptor knockout (POKO) mice newly generated in our laboratory. In the POKO mice, leptin no longer induced pSTAT-3 signaling after intracerebroventricular injection. Three basic phenotypes were observed: the heterozygotes had similar weight and adiposity as the wild-type (WT) mice (>60% of the mice); the homozygotes were either fatter (∼30%), or rarely leaner (<5%) than the WT mice. By early adulthood, the POKO mice had higher average body weight and adiposity than their respective same-sex WT littermate controls, and this was consistent among different batches. The homozygote fat POKO showed significant reduction of midline estimating statistic of rhythm of circadian parameters, and shifts of ultradian rhythms. The blunted circadian rhythm of these extremely obese POKO mice was also seen in their physical inactivity, longer feeding bouts, and higher food intake. The extent of obesity correlated with the blunted circadian amplitude, accumulative metabolic and locomotor activities, and the severity of hyperphagia. This contrasts with the heterozygote POKO mice which showed little obesity and metabolic disturbance, and only subtle changes of the circadian rhythm of metabolic activity without alterations in feeding behavior. The results provide a novel aspect of leptin resistance, almost manifesting as an "all or none" phenomenon.


Asunto(s)
Ritmo Circadiano/fisiología , Conducta Alimentaria/fisiología , Leptina/metabolismo , Transducción de Señal/fisiología , Adiposidad/fisiología , Animales , Peso Corporal/fisiología , Grasas de la Dieta/metabolismo , Ingestión de Alimentos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/metabolismo , Obesidad/fisiopatología , Receptores de Leptina/metabolismo , Delgadez/metabolismo , Delgadez/fisiopatología
11.
J Mol Neurosci ; 51(2): 364-70, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23504255

RESUMEN

We have shown that mice with experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis, have upregulated leptin receptor expression in reactive astrocytes of the hippocampus, a region involved in sickness behavior. Leptin can exacerbate EAE when its serum concentration is high. Although leptin receptors in astrocytes modulate leptin transport across cultured endothelial cell monolayers, it is not known how leptin transport in EAE mice is regulated. Here, we determined brain and cervical spinal cord uptake of leptin in early and recovery stages of EAE, after either intravenous delivery or in situ brain perfusion of (125)I-leptin and the vascular marker (131)I-albumin. While increased vascular space and general blood-brain barrier (BBB) permeability after EAE were expected, the specific saturable transport system for leptin crossing the BBB also persisted. Moreover, there was upregulation of leptin transport in hippocampus and cervical spinal cord in the early stage of EAE, shown by higher leptin uptake in these regions and by competitive inhibition with coadministered excess unlabeled leptin. We conclude that EAE induced a time- and region-specific increase of leptin transport. The results provide a link between circulating leptin and enhanced leptin signaling that may play a crucial role in disease progression.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar , Encefalomielitis Autoinmune Experimental/metabolismo , Leptina/metabolismo , Animales , Femenino , Hipocampo/metabolismo , Leptina/farmacocinética , Ratones , Médula Espinal/metabolismo
12.
J Cell Physiol ; 228(7): 1610-6, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23359322

RESUMEN

Hyperleptinemia is usually associated with obesity and leptin resistance. Endothelial cell leptin receptor knockout (ELKO) mice without a signaling membrane-bound leptin receptor in endothelia, however, have profound hyperleptinemia without signs of leptin resistance. Leptin mRNA in adipose tissue was unchanged. To test the hypothesis that the ELKO mutation results in delayed degradation and slowed excretion, we determined the kinetics of leptin transfer in groups of ELKO and wildtype mice after intravenous bolus injection of (125) I-leptin and the reference substance (131) I-albumin. The degradation pattern of (125) I-leptin in serum and brain homogenates at different time points between 10 and 60 min was measured by HPLC and acid precipitation. Although ELKO mice had reduced uptake of (125) I-leptin uptake by the brain and several peripheral organs, leptin was more stable in blood and tissue. There was no change in the rate of renal excretion. ELISA showed that serum soluble leptin receptor, known to antagonize leptin transport, had a 400-fold increase, probably contributing to the hyperleptinemia and reduced tissue uptake. Thus, the ELKO mutation unexpectedly increased the stability of leptin but suppressed its tissue uptake. These changes probably contribute to the known partial resistance of the ELKO mice to diet-induced obesity.


Asunto(s)
Leptina/sangre , Receptores de Leptina/deficiencia , Tejido Adiposo/metabolismo , Animales , Transporte Biológico Activo , Encéfalo/metabolismo , Células Endoteliales/metabolismo , Riñón/metabolismo , Leptina/metabolismo , Ratones , Ratones Noqueados , Obesidad/etiología , Obesidad/metabolismo , Estabilidad Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Albúmina Sérica/metabolismo , Distribución Tisular
13.
J Appl Physiol (1985) ; 114(6): 734-41, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23329815

RESUMEN

To determine how astrocytic leptin signaling regulates the physiological response of mice to diet-induced obesity (DIO), we performed metabolic analyses and hypothalamic leptin signaling assays on astrocytic leptin-receptor knockout (ALKO) mice in which astrocytes lack functional leptin receptor (ObR) signaling. ALKO mice and wild-type (WT) littermate controls were studied at different stages of DIO with measurement of body wt, percent fat, metabolic activity, and biochemical parameters. When fed regular chow, the ALKO mice had similar body wt, percent fat, food intake, heat dissipation, respiratory exchange ratio, and activity as their WT littermates. There was no change in blood concentrations of triglyceride, soluble leptin receptor (sObR), mRNA for leptin and uncoupling protein 1 (UCP1) in adipose tissue, and insulin sensitivity. Unexpectedly, in response to a high-fat diet the ALKO mice had attenuated hyperleptinemia and sObR, a lower level of leptin mRNA in subcutaneous fat, and a paradoxical increase in UCP1 mRNA. Thus, ALKO mice did not show the worsening of obesity that occurs with normal WT mice and the neuronal ObR mutation that results in morbid obesity. The findings are consistent with a competing, counterregulatory model between neuronal and astrocytic leptin signaling.


Asunto(s)
Astrocitos/metabolismo , Dieta Alta en Grasa , Hipotálamo/metabolismo , Leptina/metabolismo , Obesidad/prevención & control , Receptores de Leptina/deficiencia , Adiposidad , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Peso Corporal , Modelos Animales de Enfermedad , Metabolismo Energético , Genotipo , Insulina/sangre , Canales Iónicos/genética , Canales Iónicos/metabolismo , Leptina/sangre , Leptina/genética , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Actividad Motora , Obesidad/sangre , Obesidad/genética , Fenotipo , ARN Mensajero/sangre , Receptores de Leptina/sangre , Receptores de Leptina/genética , Transducción de Señal , Grasa Subcutánea/metabolismo , Factores de Tiempo , Triglicéridos/sangre , Proteína Desacopladora 1
14.
J Mol Neurosci ; 49(3): 446-56, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22684620

RESUMEN

The detrimental role of leptin in experimental autoimmune encephalomyelitis (EAE) is opposite to its neuroprotective role in other neuropathologies. We hypothesize that a shifted cellular distribution of leptin receptors underlies the differential effects of leptin. A robust increase of ObR immunoreactivity was seen along glial fibrillary acidic protein (GFAP)(+) intermediate filaments in reactive astrocytes in the hippocampus and hypothalamus of mice with EAE. Although astrocyte-specific GFAP mRNA and protein were both increased, ObRa mRNA was elevated only after resolution of EAE symptoms, and ObRb mRNA was even decreased at the peak time of symptoms of EAE. A cell type-specific action of leptin may underlie its differential effects.


Asunto(s)
Astrocitos/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Hipocampo/metabolismo , Hipotálamo/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Receptores de Leptina/biosíntesis , Animales , Convalecencia , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Regulación de la Expresión Génica , Gliosis/etiología , Gliosis/genética , Gliosis/metabolismo , Gliosis/patología , Hipocampo/patología , Hipotálamo/patología , Filamentos Intermedios/metabolismo , Leptina/fisiología , Ratones , Ratones Endogámicos , Proteína Proteolipídica de la Mielina/inmunología , Proteína Proteolipídica de la Mielina/toxicidad , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Especificidad de Órganos , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Procesamiento Proteico-Postraduccional , ARN Mensajero/biosíntesis , Receptores de Leptina/genética , Transcripción Genética , Regulación hacia Arriba
15.
J Mol Neurosci ; 49(3): 523-30, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23180096

RESUMEN

Both proconvulsive and anticonvulsive roles of leptin have been reported, suggesting cell-specific actions of leptin in different models of seizure and epilepsy. The goal of our study was to determine the regulation and function of astrocytic leptin receptors in a mouse model of epilepsy and glutamate-induced cytotoxicity. We show that in pilocarpine-challenged mice developing epilepsy with recurrent seizures after a latent period of 2 weeks, hippocampal leptin receptor (ObR) immunofluorescence was increased at 6 weeks. This was more pronounced in astrocytes than in neurons. In cultured astrocytes, glutamate increased ObRa and ObRb expression, whereas leptin pretreatment attenuated glial cytotoxicity by excess glutamate, reflected by better preserved adenosine triphosphate production. The protective role of astrocytic leptin signaling is further supported by the higher lethality of the astrocyte-specific leptin receptor knockout mice in the initial phase of seizure production. Thus, leptin signaling in astrocytes plays a protective role against seizure, and the effects are at least partially mediated by attenuation of glutamate toxicity. Astrocytic leptin signaling, therefore, may be a novel therapeutic target.


Asunto(s)
Astrocitos/fisiología , Epilepsia/prevención & control , Ácido Glutámico/toxicidad , Leptina/fisiología , Neurotoxinas/toxicidad , Animales , Astrocitoma/patología , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Convulsivantes/toxicidad , Epilepsia/inducido químicamente , Epilepsia/patología , Epilepsia/fisiopatología , Femenino , Regulación de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/biosíntesis , Proteína Ácida Fibrilar de la Glía/genética , Gliosis/etiología , Gliosis/patología , Hipocampo/metabolismo , Hipocampo/patología , Leptina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Pilocarpina/toxicidad , Receptores de Leptina/biosíntesis , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Receptores de Leptina/fisiología , Recurrencia , Transducción de Señal , Regulación hacia Arriba
16.
Neurosci Biobehav Rev ; 37(2): 184-92, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23201098

RESUMEN

Interleukin (IL)-15 is a ubiquitously expressed cytokine existing in both intracellular and secretory forms. Here we review the expression, regulation, and functions of IL15 and its receptors in the brain. IL15 receptors show robust upregulation after neuroinflammation, suggesting a major role of IL15 signaling in cerebral function. Involvement of the IL15 system in neuropsychiatric behavior is reflected by the effects of IL15, IL15Rα, and IL2Rγ deletions on neurobehavior and neurotransmitters, the effects of IL15 treatment on neuronal activity, and the potential role of IL15 in neuroplasticity/neurogenesis. The results show that IL15 modulates GABA and serotonin transmission. This may underlie deficits in mood (depressive-like behavior and decreased normal anxiety) and memory, as well as activity level, sleep, and thermoregulation. Although IL15 has only a low level of permeation across the blood-brain barrier, peripheral IL15 is able to activate multiple signaling pathways in neurons widely distributed in CNS regions. The effects of IL15 in "preventing" neuropsychiatric symptoms in normal mice implicate a potential therapeutic role of this polypeptide cytokine.


Asunto(s)
Afecto/fisiología , Encéfalo/metabolismo , Encéfalo/fisiología , Inflamación/metabolismo , Interleucina-15/fisiología , Memoria/fisiología , Neurogénesis/fisiología , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiología , Humanos , Interleucina-15/metabolismo , Subunidad alfa del Receptor de Interleucina-15/metabolismo , Subunidad alfa del Receptor de Interleucina-15/fisiología , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Regulación hacia Arriba
17.
Cell Physiol Biochem ; 30(5): 1109-19, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23018453

RESUMEN

BACKGROUND/AIMS: Acute phase C-reactive protein (CRP), elevated in obesity and inflammation, is a major binding protein for leptin. It is thought that CRP contributes to leptin resistance by preventing leptin from crossing the blood-brain barrier (BBB). Here we determined how CRP interacts with the BBB and whether it deters leptin from reaching CNS targets. METHODS: BBB permeability, compartmental distribution, tracer stability, and expression of tight junction protein and inflammatory marker were determined. RESULTS: CRP was stable in blood, but did not permeate the BBB in trace amounts. However, it increased paracellular permeability at a higher dose. Agouti viable (A(vy)) mice with adult-onset obesity show higher CRP entry into the brain. CRP did not permeate hCMEC/D3 cells nor change zona occludin-1 or cyclooxygenase-2 expression. An intermediate dose of CRP had no effect on leptin transport across the BBB after co-treatment. Thus, acute interactions between CRP and leptin at the BBB level were negligible and did not explain the leptin resistance seen in obesity. CONCLUSIONS: The interactions of CRP and the BBB are a two-phase process, with increased paracellular permeability at a high dose that enables its entry into the CNS and serves to induce reactive gliosis and impair CNS function.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Proteína C-Reactiva/metabolismo , Permeabilidad Capilar , Sistema Nervioso Central/metabolismo , Inflamación/metabolismo , Obesidad/metabolismo , Animales , Leptina/metabolismo , Ratones , Ratones Endogámicos C57BL
18.
Ann N Y Acad Sci ; 1264: 64-71, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22530983

RESUMEN

Leptin, an adipocyte-derived cytokine, crosses the blood-brain barrier to act on many regions of the central nervous system (CNS). It participates in the regulation of energy balance, inflammatory processes, immune regulation, synaptic formation, memory condensation, and neurotrophic activities. This review focuses on the newly identified actions of leptin on astrocytes. We first summarize the distribution of leptin receptors in the brain, with a focus on the hypothalamus, where the leptin receptor is known to mediate essential feeding suppression activities, and on the hippocampus, where leptin facilitates memory, reduces neurodegeneration, and plays a dual role in seizures. We will then discuss regulation of the nonneuronal leptin system in obesity. Its relationship with neuronal leptin signaling is illustrated by in vitro assays in primary astrocyte culture and by in vivo studies on mice after pretreatment with a glial metabolic inhibitor or after cell-specific deletion of intracellular signaling leptin receptors. Overall, the glial leptin system shows robust regulation and plays an essential role in obesity. Strategies to manipulate this nonneuronal leptin signaling may have major clinical impact.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Leptina/metabolismo , Receptores de Leptina/metabolismo , Animales , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Metabolismo Energético , Hipocampo/metabolismo , Humanos , Hipotálamo/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad
19.
J Mol Neurosci ; 47(2): 267-77, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22392152

RESUMEN

Reactive gliosis, a sign of neuroinflammation, has been observed in mice with adult-onset obesity as well as CNS injury. The hypothesis that obesity-derived metabolic factors exacerbate reactive gliosis in response to mechanical injury was tested here on cultured primary glial cells subjected to a well-established model of scratch wound injury. Cells treated with serum from mice with diet-induced obesity (DIO) showed higher immunoreactivity of CD11b (marker for microglia) and GFAP (marker for astrocytes), with morphological changes at both the injury border and areas away from the injury. The effect of DIO serum was greater than that of scratch injury alone. Leptin was almost as effective as DIO serum in inducing microgliosis and astrogliosis in a dose-response manner. By contrast, C-reactive protein (CRP) mainly induced microgliosis in noninjured cells; injury-induced factors appeared to attenuate this effect. The effect of CRP also differed from the effect of the antibiotic minocycline. Minocycline attenuated the microgliosis and to a lesser extent astrogliosis, particularly in CRP-treated cells, thus serving as a negative control. We conclude that blood-borne proinflammatory metabolic factors in obesity increase reactive gliosis and probably exacerbate CNS injury.


Asunto(s)
Astrocitos/metabolismo , Gliosis/metabolismo , Microglía/metabolismo , Inflamación Neurogénica/metabolismo , Obesidad/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Gliosis/sangre , Gliosis/fisiopatología , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/patología , Inflamación Neurogénica/sangre , Inflamación Neurogénica/patología , Obesidad/sangre , Cultivo Primario de Células
20.
J Appl Physiol (1985) ; 112(8): 1410-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22323652

RESUMEN

Leptin, a polypeptide hormone produced mainly by adipocytes, has diverse effects in both the brain and peripheral organs, including suppression of feeding. Other than mediating leptin transport across the blood-brain barrier, the role of the endothelial leptin receptor remains unclear. We recently generated a mutant mouse strain lacking endothelial leptin receptor signaling, and showed that there is an increased uptake of leptin by brain parenchyma after its delivery by in situ brain perfusion. Here, we tested the hypothesis that endothelial leptin receptor mutation confers partial resistance to diet-induced obesity. These ELKO mice had similar body weight and percent fat as their wild-type littermates when fed with rodent chow, but blood concentrations of leptin were significantly elevated. In response to a high-fat diet, wild-type mice had a greater gain of body weight and fat than ELKO mice. As shown by metabolic chamber measurement, the ELKO mice had higher oxygen consumption, carbon dioxide production, and heat dissipation, although food intake was similar to that of the wild-type mice and locomotor activity was even reduced. This indicates that the partial resistance to diet-induced obesity was mediated by higher metabolic activity in the ELKO mice. Since neuronal leptin receptor knockout mice show obesity and diabetes, the results suggest that endothelial leptin signaling shows opposite effects from that of neuronal leptin signaling, with a facilitatory role in diet-induced obesity.


Asunto(s)
Grasas de la Dieta/efectos adversos , Endotelio Vascular/metabolismo , Mutación/genética , Obesidad/etiología , Obesidad/prevención & control , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Adiposidad/efectos de los fármacos , Adiposidad/fisiología , Animales , Peso Corporal/fisiología , Dióxido de Carbono/metabolismo , Ritmo Circadiano/fisiología , Grasas de la Dieta/farmacología , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Masculino , Ratones , Ratones Noqueados , Obesidad/metabolismo , Consumo de Oxígeno/fisiología , Receptores de Leptina/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...