Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Endocrinol (Lausanne) ; 15: 1397081, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38887268

RESUMEN

Introduction: Unlike white adipose tissue depots, bone marrow adipose tissue (BMAT) expands during caloric restriction (CR). Although mechanisms for BMAT expansion remain unclear, prior research suggested an intermediary role for increased circulating glucocorticoids. Methods: In this study, we utilized a recently described mouse model (BMAd-Cre) to exclusively target bone marrow adipocytes (BMAds) for elimination of the glucocorticoid receptor (GR) (i.e. Nr3c1) whilst maintaining GR expression in other adipose depots. Results: Mice lacking GR in BMAds (BMAd-Nr3c1 -/-) and control mice (BMAd-Nr3c1 +/+) were fed ad libitum or placed on a 30% CR diet for six weeks. On a normal chow diet, tibiae of female BMAd-Nr3c1-/- mice had slightly elevated proximal trabecular metaphyseal bone volume fraction and thickness. Both control and BMAd-Nr3c1-/- mice had increased circulating glucocorticoids and elevated numbers of BMAds in the proximal tibia following CR. However, no significant differences in trabecular and cortical bone were observed, and quantification with osmium tetroxide and µCT revealed no difference in BMAT accumulation between control or BMAd-Nr3c1 -/- mice. Differences in BMAd size were not observed between BMAd-Nr3c1-/- and control mice. Interestingly, BMAd-Nr3c1-/- mice had decreased circulating white blood cell counts 4 h into the light cycle. Discussion: In conclusion, our data suggest that eliminating GR from BMAd has minor effects on bone and hematopoiesis, and does not impair BMAT accumulation during CR.


Asunto(s)
Adipocitos , Adiposidad , Médula Ósea , Restricción Calórica , Hematopoyesis , Receptores de Glucocorticoides , Animales , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/deficiencia , Ratones , Adipocitos/metabolismo , Adiposidad/fisiología , Femenino , Médula Ósea/metabolismo , Ratones Noqueados , Huesos/metabolismo , Ratones Endogámicos C57BL , Tejido Adiposo/metabolismo , Masculino , Errores Innatos del Metabolismo
2.
Mol Metab ; 83: 101916, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38492843

RESUMEN

OBJECTIVE: Exposure of adipocytes to 'cool' temperatures often found in the periphery of the body induces expression of Stearoyl-CoA Desaturase-1 (Scd1), an enzyme that converts saturated fatty acids to monounsaturated fatty acids. The goal of this study is to further investigate the roles of Scd in adipocytes. METHOD: In this study, we employed Scd1 knockout cells and mouse models, along with pharmacological Scd1 inhibition to dissect the enzyme's function in adipocyte physiology. RESULTS: Our study reveals that production of monounsaturated lipids by Scd1 is necessary for fusion of autophagosomes to lysosomes and that with a Scd1-deficiency, autophagosomes accumulate. In addition, Scd1-deficiency impairs lysosomal and autolysosomal acidification resulting in vacuole accumulation and eventual cell death. Blocking autophagosome formation or supplementation with monounsaturated fatty acids maintains vitality of Scd1-deficient adipocytes. CONCLUSION: This study demonstrates the indispensable role of Scd1 in adipocyte survival, with its inhibition in vivo triggering autophagy-dependent cell death and its depletion in vivo leading to the loss of bone marrow adipocytes.


Asunto(s)
Adipocitos , Autofagia , Ácidos Grasos Monoinsaturados , Ratones Noqueados , Estearoil-CoA Desaturasa , Estearoil-CoA Desaturasa/metabolismo , Estearoil-CoA Desaturasa/genética , Animales , Ratones , Adipocitos/metabolismo , Ácidos Grasos Monoinsaturados/metabolismo , Ácidos Grasos Monoinsaturados/farmacología , Ratones Endogámicos C57BL , Lisosomas/metabolismo , Supervivencia Celular , Células 3T3-L1 , Masculino , Metabolismo de los Lípidos , Autofagosomas/metabolismo
3.
Redox Biol ; 71: 103074, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38367511

RESUMEN

Brain iron accumulation constitutes a pathognomonic indicator in several neurodegenerative disorders. Metal accumulation associated with dopaminergic neuronal death has been documented in Parkinson's disease. Through the use of in vivo and in vitro models, we demonstrated that lipid dysregulation manifests as a neuronal and glial response during iron overload. In this study, we show that cholesterol content and triacylglycerol (TAG) hydrolysis were strongly elevated in mice midbrain. Lipid cacostasis was concomitant with the loss of dopaminergic neurons, astrogliosis and elevated expression of α-synuclein. Exacerbated lipid peroxidation and markers of ferroptosis were evident in the midbrain from mice challenged with iron overload. An imbalance in the activity of lipolytic and acylation enzymes was identified, favoring neutral lipid hydrolysis, and consequently reducing TAG and cholesteryl ester levels. Notably, these observed alterations were accompanied by motor impairment in iron-treated mice. In addition, neuronal and glial cultures along with their secretomes were used to gain further insight into the mechanism underlying TAG hydrolysis and cholesterol accumulation as cellular responses to iron accumulation. We demonstrated that TAG hydrolysis in neurons is triggered by astrocyte secretomes. Moreover, we found that the ferroptosis inhibitor, ferrostatin-1, effectively prevents cholesterol accumulation both in neurons and astrocytes. Taken together, these results indicate that lipid disturbances occur in iron-overloaded mice as a consequence of iron-induced oxidative stress and depend on neuron-glia crosstalk. Our findings suggest that developing therapies aimed at restoring lipid homeostasis may lead to specific treatment for neurodegeneration associated with ferroptosis and brain iron accumulation.


Asunto(s)
Ferroptosis , Sobrecarga de Hierro , Trastornos Motores , Ratones , Animales , Metabolismo de los Lípidos , Trastornos Motores/metabolismo , Hierro/metabolismo , Peroxidación de Lípido , Neuronas Dopaminérgicas/metabolismo , Colesterol/metabolismo , Lípidos
4.
bioRxiv ; 2023 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-37961537

RESUMEN

Exposure of adipocytes to 'cool' temperatures often found in the periphery of the body induces expression of Stearoyl-CoA Desaturase-1 (SCD1), an enzyme that converts saturated fatty acids to monounsaturated fatty acids. In this study, we employed Scd1 knockout cells and mouse models, along with pharmacological SCD1 inhibition, to investigate further the roles of SCD1 in adipocytes. Our study reveals that production of monounsaturated lipids by SCD1 is necessary for fusion of autophagosomes to lysosomes and that with a SCD1-deficiency, autophagosomes accumulate. In addition, SCD1-deficiency impairs lysosomal and autolysosomal acidification resulting in vacuole accumulation and eventual cell death. Blocking autophagosome formation or supplementation with monounsaturated fatty acids maintains vitality of SCD1-deficient adipocytes. Taken together, our results demonstrate that in vitro inhibition of SCD1 in adipocytes leads to autophagy-dependent cell death, and in vivo depletion leads to loss of bone marrow adipocytes.

5.
Chem Biol Interact ; 343: 109491, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33945810

RESUMEN

Inhibition of adipocyte differentiation can be used as a strategy for preventing adipose tissue expansion and, consequently, for obesity management. Since reactive oxygen species (ROS) have emerged as key modulators of adipogenesis, the effect of menadione (a synthetic form of vitamin K known to induce the increase of intracellular ROS) on 3T3-L1 preadipocyte differentiation was studied. Menadione (15 µM) increased ROS and lipid peroxidation, generating mild oxidative stress without affecting cell viability. Menadione drastically inhibited adipogenesis, accompanied by decreased intracellular lipid accumulation and diminished expression of the lipo/adipogenic markers peroxisome proliferator-activated receptor (PPAR)γ, fatty acid synthase (FAS), CCAAT/enhancer-binding protein (C/EBP) α, fatty acid binding protein (FABP) 4, and perilipin. Menadione treatment also increased lipolysis, as indicated by augmented glycerol release and reinforced by the increased expression of hormone-sensitive lipase (HSL). Additionally, menadione increased the inhibitory phosphorylation of acetyl-CoA-carboxylase (ACC), which results in the inhibition of fatty acid synthesis. As a consequence, triglyceride content was decreased. Menadione also inhibited the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Further, treatment with increased concentration of insulin, a potent physiological activator of the PI3K/Akt pathway, rescued the normal level of expression of PPARγ, the master regulator of adipogenesis, and overcame the restraining effect of menadione on the differentiation capacity of 3T3-L1 preadipocytes. Our study reveals novel antiadipogenic action for menadione, which is, at least in part, mediated by the PI3K/Akt pathway signaling and raises its potential as a therapeutic agent in the treatment or prevention of adiposity.


Asunto(s)
Adipogénesis/efectos de los fármacos , Vitamina K 3/farmacología , Células 3T3-L1 , Animales , Diferenciación Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Ratones , Estrés Oxidativo/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Triglicéridos/metabolismo
6.
Biochim Biophys Acta Mol Cell Res ; 1867(4): 118643, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31917282

RESUMEN

Long non-coding RNAs transcribed from telomeres, known as TERRA (telomeric repeat-containing RNA), are associated with telomere and genome stability. TERRA abundance responds to different cell stresses; however, no studies have focused on oxidative stress, condition that damages biomolecules and is involved in aging and disease. Since telomeres are prone to oxidative damage leading to their dysfunction, our objective was to characterize TERRAs and the mechanisms that control their expression. TERRA increased in cells exposed to H2O2 and reverted by antioxidant treatment. TERRAs are also induced in brown adipose tissue of mice exposed to cold, which raises mitochondrial ROS. In cells exposed to H2O2, ChIP showed that chromatin landscape was modified favoring telomere transcription. TERRAs interacted with HP1α/γ, proteins that were found recruited to subtelomeres. Since HP1γ interacts with the transcriptional machinery, TERRAs may stimulate their own expression by recruiting HP1γ to subtelomeres. TERRA induction reverted within 2 h after removal of H2O2 from culture medium, suggesting they have protective functions. This was supported by rapid TERRA induction following a second H2O2 challenge. PKA inhibitors H89 and PKI blocked TERRA increase by H2O2 or IBMX+Forskolin treatment, suggesting PKA signaling regulates TERRA induction. Treatment of cells with drugs that disturb cytoskeleton integrity or growing cells on surfaces of different stiffness known to generate differential cytoskeleton tension also modified TERRA levels and sensitized cells to lower H2O2 concentrations. In summary, we show that TERRAs are induced in response to oxidative stress and are regulated by PKA as well as by changes in cytoskeleton dynamics.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citoesqueleto/metabolismo , Estrés Oxidativo , ARN Largo no Codificante/genética , Transducción de Señal , Tejido Adiposo Pardo/metabolismo , Animales , Homólogo de la Proteína Chromobox 5 , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Células HEK293 , Histonas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , ARN Largo no Codificante/metabolismo
8.
Front Neurosci ; 13: 513, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31178685

RESUMEN

Obesity is increasing at unprecedented levels globally, and the overall impact of obesity on the various organ systems of the body is only beginning to be fully appreciated. Because of the myriad of direct and indirect effects of obesity causing dysfunction of multiple tissues and organs, it is likely that there will be heterogeneity in the presentation of obesity effects in any given population. Taken together, these realities make it increasingly difficult to understand the complex interplay between obesity effects on different organs, including the brain. The focus of this review is to provide a comprehensive view of metabolic disturbances present in obesity, their direct and indirect effects on the different organ systems of the body, and to discuss the interaction of these effects in the context of brain aging and the development of neurodegenerative diseases.

9.
Toxicol In Vitro ; 60: 400-411, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31247335

RESUMEN

Neuronal exposure to 6-hydroxydopamine (6-OHDA), a hydroxylated analog of dopamine, constitutes a very useful strategy for studying the molecular events associated with neuronal death in Parkinson's disease. 6-OHDA increases oxidant levels and impairs mitochondrial respiratory chain, thus promoting neuronal injury and death. Despite the extensive use of 6-OHDA in animal models, the exact molecular events triggered by this neurotoxicant at the neuronal level have not been yet fully understood. Human IMR-32 neuroblastoma cells exposed to increasing concentrations of 6-OHDA displayed high levels of reactive oxygen species and increased plasma membrane permeability with concomitant cell viability diminution. As part of the neuronal response to 6-OHDA exposure, the nuclear translocation of nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) p65 subunit was observed. NFκB nuclear localization was also accompanied by an increase of IκB phosphorylation as well as a rise in cyclooxygenase-2 (COX-2) and the prostaglandin receptor, EP4, mRNA levels. Even though the canonical pathways participating in the modulation of NFκB have been extensively described, here we tested the hypothesis that 6-OHDA-induced injury can activate lipid signaling and, in turn, modulate the transcriptional response. 6-OHDA challenge triggered the activation of lipid signaling pathways and increased phosphatidic acid (PA), diacylglycerol and free fatty acid levels in human neuroblastoma cells. The inhibition of PA production was able to prevent the decrease in cell viability triggered by 6-OHDA, the nuclear translocation of NFκB p65 subunit and the rise in COX-2 mRNA expression. Our results indicate that the onset of the inflammatory process triggered by 6-OHDA involves the activation of PA signaling that, in turn, governs NFκB subcellular localization and COX-2 expression.


Asunto(s)
Adrenérgicos/toxicidad , FN-kappa B/metabolismo , Oxidopamina/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 2/genética , Dopamina/metabolismo , Humanos , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad , Subtipo EP4 de Receptores de Prostaglandina E/genética
10.
Front Cell Neurosci ; 13: 175, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31118888

RESUMEN

Since its discovery, the study of the biological role of α-synuclein and its pathological implications has been the subject of increasing interest. The propensity to adopt different conformational states governing its aggregation and fibrillation makes this small 14-kDa cytosolic protein one of the main etiologic factors associated with degenerative disorders known as synucleinopathies. The structure, function, and toxicity of α-synuclein and the possibility of different therapeutic approaches to target the protein have been extensively investigated and reviewed. One intriguing characteristic of α-synuclein is the different ways in which it interacts with lipids. Though in-depth studies have been carried out in this field, the information they have produced is puzzling and the precise role of lipids in α-synuclein biology and pathology and vice versa is still largely unknown. Here we provide an overview and discussion of the main findings relating to α-synuclein/lipid interaction and its involvement in the modulation of lipid metabolism and signaling.

11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(6): 639-650, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29571767

RESUMEN

We have previously shown that phospholipase D (PLD) pathways have a role in neuronal degeneration; in particular, we found that PLD activation is associated with synaptic injury induced by oxidative stress. In the present study, we investigated the effect of α-synuclein (α-syn) overexpression on PLD signaling. Wild Type (WT) α-syn was found to trigger the inhibition of PLD1 expression as well as a decrease in ERK1/2 phosphorylation and expression levels. Moreover, ERK1/2 subcellular localization was shown to be modulated by WT α-syn in a PLD1-dependent manner. Indeed, PLD1 inhibition was found to alter the neurofilament network and F-actin distribution regardless of the presence of WT α-syn. In line with this, neuroblastoma cells expressing WT α-syn exhibited a degenerative-like phenotype characterized by a marked reduction in neurofilament light subunit (NFL) expression and the rearrangement of the F-actin organization, compared with either the untransfected or the empty vector-transfected cells. The gain of function of PLD1 through the overexpression of its active form had the effect of restoring NFL expression in WT α-syn neurons. Taken together, our findings reveal an unforeseen role for α-syn in PLD regulation: PLD1 downregulation may constitute an early mechanism in the initial stages of WT α-syn-triggered neurodegeneration.


Asunto(s)
Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Enfermedad de Parkinson/metabolismo , Fosfolipasa D/biosíntesis , alfa-Sinucleína/metabolismo , Línea Celular Tumoral , Mutación con Ganancia de Función , Humanos , Filamentos Intermedios/genética , Filamentos Intermedios/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Fosfolipasa D/genética , alfa-Sinucleína/genética
12.
Oxid Med Cell Longev ; 2018: 2850341, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29581821

RESUMEN

Iron overload is a hallmark of many neurodegenerative processes such as Alzheimer's, Parkinson's, and Huntington's diseases. Unbound iron accumulated as a consequence of brain aging is highly reactive with water and oxygen and produces reactive oxygen species (ROS) or free radicals. ROS are toxic compounds able to damage cell membranes, DNA, and mitochondria. Which are the mechanisms involved in neuronal iron homeostasis and in neuronal response to iron-induced oxidative stress constitutes a cutting-edge topic in metalloneurobiology. Increasing our knowledge about the underlying mechanisms that operate in iron accumulation and their consequences would shed light on the comprehension of the molecular events that participate in the pathophysiology of the abovementioned neurodegenerative diseases. In this review, current evidences about iron accumulation in the brain, the signaling mechanisms triggered by metal overload, as well as the interaction between amyloid ß (Aß) and iron, will be summarized.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Hierro/metabolismo , Humanos , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Estrés Oxidativo
13.
Sci Rep ; 7(1): 3697, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28623340

RESUMEN

Glucokinase (GK), the hexokinase involved in glucosensing in pancreatic ß-cells, is also expressed in arcuate nucleus (AN) neurons and hypothalamic tanycytes, the cells that surround the basal third ventricle (3V). Several lines of evidence suggest that tanycytes may be involved in the regulation of energy homeostasis. Tanycytes have extended cell processes that contact the feeding-regulating neurons in the AN, particularly, agouti-related protein (AgRP), neuropeptide Y (NPY), cocaine- and amphetamine-regulated transcript (CART) and proopiomelanocortin (POMC) neurons. In this study, we developed an adenovirus expressing GK shRNA to inhibit GK expression in vivo. When injected into the 3V of rats, this adenovirus preferentially transduced tanycytes. qRT-PCR and Western blot assays confirmed GK mRNA and protein levels were lower in GK knockdown animals compared to the controls. In response to an intracerebroventricular glucose injection, the mRNA levels of anorexigenic POMC and CART and orexigenic AgRP and NPY neuropeptides were altered in GK knockdown animals. Similarly, food intake, meal duration, frequency of eating events and the cumulative eating time were increased, whereas the intervals between meals were decreased in GK knockdown rats, suggesting a decrease in satiety. Thus, GK expression in the ventricular cells appears to play an important role in feeding behavior.


Asunto(s)
Adenoviridae/fisiología , Conducta Alimentaria , Glucoquinasa/metabolismo , Hipotálamo/metabolismo , Hipotálamo/fisiopatología , Infecciones por Adenoviridae , Animales , Encefalitis/etiología , Encefalitis/metabolismo , Encefalitis/patología , Expresión Génica , Regulación de la Expresión Génica , Genes Reporteros , Hipotálamo/patología , Hipotálamo/virología , Masculino , Neuropéptidos/genética , Neuropéptidos/metabolismo , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
14.
Mol Neurobiol ; 54(5): 3236-3252, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-27080543

RESUMEN

We have previously demonstrated that oligomeric amyloid ß peptide (oAß) together with iron overload generates synaptic injury and activation of several signaling cascades. In this work, we characterized hippocampal neuronal response to oAß. HT22 neurons exposed to 500 nM oAß showed neither increased lipid peroxidation nor altered mitochondrial function. In addition, biophysical studies showed that oAß did not perturb the lipid order of the membrane. Interestingly, although no neuronal damage could be demonstrated, oAß was found to trigger bifurcated phosphoinositide-dependent signaling in the neuron, on one hand, the phosphorylation of insulin receptor, the phosphatidylinositol 3-kinase (PI3K)-dependent activation of Akt, its translocation to the nucleus and the concomitant phosphorylation, inactivation, and nuclear exclusion of the transcription factor Forkhead Box O3a (FoxO3a), and on the other, phosphoinositide-phospholipase C (PI-PLC)-dependent extracellular signal-regulated kinase 1/2 (ERK1/2) activation. Pharmacological manipulation of the signaling cascades was used in order to better characterize the role of oAß-activated signals, and mitochondrial function was determined as a measure of neuronal viability. The inhibition of PI3K, PI-PLC, and general phosphoinositide metabolism impaired neuronal mitochondrial function. Furthermore, increased oAß-induced cell death was observed in the presence of phosphoinositide metabolism inhibition. Our results allow us to conclude that oAß triggers the activation of phosphoinositide-dependent signaling, which results in the subsequent activation of neuroprotective mechanisms that could be involved in the determination of neuronal fate.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Fosfatidilinositoles/metabolismo , Multimerización de Proteína , Transducción de Señal , Animales , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína Forkhead Box O3/metabolismo , Humanos , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Agregado de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptor de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos
15.
J Biol Chem ; 288(27): 19773-84, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23687303

RESUMEN

The PI3K/Akt pathway is a key component in synaptic plasticity and neuronal survival. The aim of this work was to investigate the participation of the PI3K/Akt pathway and its outcome on different molecular targets such as glycogen synthase kinase 3ß (GSK3ß) and Forkhead box-O (FoxO) transcription factors during mild oxidative stress triggered by iron overload. The exposure of mouse hippocampal neurons (HT22) to different concentrations of Fe(2+) (25-200 µm) for 24 h led us to define a mild oxidative injury status (50 µm Fe(2+)) in which cell morphology showed changes typical of neuronal damage with increased lipid peroxidation and cellular oxidant levels but no alteration of cellular viability. There was a simultaneous increase in both Akt and GSK3ß phosphorylation. Levels of phospho-FoxO3a (inactive form) increased in the cytosolic fraction of cells treated with iron in a PI3K-dependent manner. Moreover, PI3K and Akt translocated to the nucleus in response to oxidative stress. Iron-overloaded cells harboring a constitutively active form of Akt showed decreased oxidants levels. Indeed, GSH synthesis under oxidative stress conditions was regulated by activated Akt. Our results show that activation of the PI3K/Akt pathway during iron-induced neurotoxicity regulates multiple targets such as GSK3ß, FoxO transcriptional activity, and glutathione metabolism, thus modulating the neuronal response to oxidative stress.


Asunto(s)
Hipocampo/enzimología , Hierro/farmacología , Neuronas/enzimología , Estrés Oxidativo/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Animales , Línea Celular , Núcleo Celular/enzimología , Núcleo Celular/patología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/metabolismo , Humanos , Hierro/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/genética , Ratones , Neuronas/patología , Estrés Oxidativo/genética , Fosfatidilinositol 3-Quinasas/genética , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/genética
16.
J Neurosci Res ; 89(9): 1471-7, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21608013

RESUMEN

Amino acid analogs promote translational errors that result in aberrant protein synthesis and have been used to understand the effects of protein misfolding in a variety of physiological and pathological settings. TDP-43 is a protein that is linked to protein aggregation and toxicity in a variety of neurodegenerative diseases. This study exposed primary rat neurons and astrocyte cultures to established amino acid analogs (canavanine and azetidine-2-carboxylic acid) and showed that both cell types undergo a dose-dependent increase in toxicity, with neurons exhibiting a greater degree of toxicity compared with astrocytes. Neurons and astrocytes exhibited similar increases in ubiquitinated and oxidized protein following analog treatment. Analog treatment increased heat shock protein (Hsp) levels in both neurons and astrocytes. In neurons, and to a lesser extent astrocytes, the levels of TDP-43 increased in response to analog treatment. Taken together, these data indicate that neurons exhibit preferential toxicity and alterations in TDP-43 in response to increased protein misfolding compared with astrocytes.


Asunto(s)
Astrocitos/efectos de los fármacos , Ácido Azetidinocarboxílico/toxicidad , Canavanina/toxicidad , Proteínas de Unión al ADN/metabolismo , Neuronas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Aminoácidos/agonistas , Aminoácidos/toxicidad , Animales , Astrocitos/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Proteínas de Choque Térmico/efectos de los fármacos , Proteínas de Choque Térmico/metabolismo , Neuronas/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
17.
PLoS One ; 6(1): e16411, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21297988

RESUMEN

Metabolic interaction via lactate between glial cells and neurons has been proposed as one of the mechanisms involved in hypothalamic glucosensing. We have postulated that hypothalamic glial cells, also known as tanycytes, produce lactate by glycolytic metabolism of glucose. Transfer of lactate to neighboring neurons stimulates ATP synthesis and thus contributes to their activation. Because destruction of third ventricle (III-V) tanycytes is sufficient to alter blood glucose levels and food intake in rats, it is hypothesized that tanycytes are involved in the hypothalamic glucose sensing mechanism. Here, we demonstrate the presence and function of monocarboxylate transporters (MCTs) in tanycytes. Specifically, MCT1 and MCT4 expression as well as their distribution were analyzed in Sprague Dawley rat brain, and we demonstrate that both transporters are expressed in tanycytes. Using primary tanycyte cultures, kinetic analyses and sensitivity to inhibitors were undertaken to confirm that MCT1 and MCT4 were functional for lactate influx. Additionally, physiological concentrations of glucose induced lactate efflux in cultured tanycytes, which was inhibited by classical MCT inhibitors. Because the expression of both MCT1 and MCT4 has been linked to lactate efflux, we propose that tanycytes participate in glucose sensing based on a metabolic interaction with neurons of the arcuate nucleus, which are stimulated by lactate released from MCT1 and MCT4-expressing tanycytes.


Asunto(s)
Epéndimo/metabolismo , Glucosa/metabolismo , Lactatos/metabolismo , Transportadores de Ácidos Monocarboxílicos/análisis , Proteínas Musculares/análisis , Neuroglía/metabolismo , Neuronas/metabolismo , Simportadores/análisis , Animales , Células Cultivadas , Epéndimo/química , Epéndimo/citología , Hipotálamo/química , Hipotálamo/citología , Metabolismo , Ratas , Ratas Sprague-Dawley
18.
Antiviral Res ; 88(3): 334-42, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20970459

RESUMEN

It is well established that HIV antiretroviral drugs, particularly protease inhibitors, frequently elicit a metabolic syndrome that may include hyperlipidemia, lipodystrophy, and insulin resistance. Metabolic dysfunction in non-HIV-infected subjects has been repeatedly associated with cognitive impairment in epidemiological and experimental studies, but it is not yet understood if antiretroviral therapy-induced metabolic syndrome might contribute to HIV-associated neurologic decline. To determine if protease inhibitor-induced metabolic dysfunction in mice is accompanied by adverse neurologic effects, C57BL/6 mice were given combined lopinavir/ritonavir (50/12.5-200/50 mg/kg) daily for 3 weeks. Data show that lopinavir/ritonavir administration caused significant metabolic derangement, including alterations in body weight and fat mass, as well as dose-dependent patterns of hyperlipidemia, hypoadiponectinemia, hypoleptinemia, and hyperinsulinemia. Evaluation of neurologic function revealed that even the lowest dose of lopinavir/ritonavir caused significant cognitive impairment assessed in multi-unit T-maze, but did not affect motor functions assessed as rotarod performance. Collectively, our results indicate that repeated lopinavir/ritonavir administration produces cognitive as well as metabolic impairments, and suggest that the development of selective aspects of metabolic syndrome in HIV patients could contribute to HIV-associated neurocognitive disorders.


Asunto(s)
Inhibidores de la Proteasa del VIH , Pirimidinonas/efectos adversos , Ritonavir/efectos adversos , Animales , Cognición/efectos de los fármacos , Esquema de Medicación , Combinación de Medicamentos , VIH/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , Inhibidores de la Proteasa del VIH/efectos adversos , Inhibidores de la Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/uso terapéutico , Lopinavir , Masculino , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Pirimidinonas/administración & dosificación , Pirimidinonas/metabolismo , Ritonavir/administración & dosificación , Ritonavir/metabolismo , Pérdida de Peso/efectos de los fármacos
19.
Free Radic Biol Med ; 49(8): 1290-7, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20678570

RESUMEN

Maintaining protein homeostasis is vital to cell viability, with numerous studies demonstrating a role for proteasome inhibition occurring during the aging of a variety of tissues and, presumably, contributing to the disruption of cellular homeostasis during aging. In this study we sought to elucidate the differences between neurons and astrocytes in regard to basal levels of protein synthesis, proteasome-mediated protein degradation, and sensitivity to cytotoxicity after proteasome inhibitor treatment. In these studies we demonstrate that neurons have an increased vulnerability, compared to astrocyte cultures, to proteasome-inhibitor-induced cytotoxicity. No significant difference was observed between these two cell types in regard to the basal rates of protein synthesis, or basal rates of protein degradation, in the pool of short-lived proteins. After proteasome inhibitor treatment neuronal crude lysates were observed to undergo greater increases in the levels of ubiquitinated and oxidized proteins and selectively exhibited increased levels of newly synthesized proteins accumulating within the insoluble protein pool, compared to astrocytes. Together, these data suggest a role for increased oxidized proteins and sequestration of newly synthesized proteins in the insoluble protein pool, as potential mediators of the selective neurotoxicity after proteasome inhibitor treatment. The implications for neurons exhibiting increased sensitivity to acute proteasome inhibitor exposure, and the corresponding changes in protein homeostasis observed after proteasome inhibition, are discussed in the context of both aging and age-related disorders of the nervous system.


Asunto(s)
Astrocitos/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Leupeptinas/farmacología , Neuronas/metabolismo , Estrés Oxidativo , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Extractos Celulares , Células Cultivadas , Inhibidores de Cisteína Proteinasa/efectos adversos , Inhibidores de Cisteína Proteinasa/uso terapéutico , Humanos , Leupeptinas/efectos adversos , Leupeptinas/uso terapéutico , Neuronas/efectos de los fármacos , Neuronas/patología , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/prevención & control , Oxidación-Reducción/efectos de los fármacos , Inhibidores de Proteasoma , Ratas , Ratas Sprague-Dawley , Solubilidad/efectos de los fármacos , Ubiquitinación/efectos de los fármacos
20.
J Neurochem ; 114(6): 1581-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20557430

RESUMEN

Long term consumption of a high fat diet (HFD) contributes to increased morbidity and mortality. Yet the specific effects of HFD consumption on brain aging are poorly understood. In the present study 20-month old male C57Bl/6 mice were fed either 'western diet' (41% fat), very high fat lard diet (60% fat), or corresponding control diets for 16 weeks and then assessed for changes in metabolism and brain homeostasis. Although both HFDs increased adiposity and fasting blood glucose, only the high fat lard diet increased age-related oxidative damage (protein carbonyls) and impaired retention in the behavioral test. This selective increase in oxidative damage and cognitive decline was also associated with a decline in NF-E2-related factor 2 (Nrf2) levels and Nrf2 activity, suggesting a potential role for decreased antioxidant response. Taken together, these data suggest that while adiposity and insulin resistance following HFD consumption are linked to increased morbidity, the relationship between these factors and brain homeostasis during aging is not a linear relationship. More specifically, these data implicate impaired Nrf2 signaling and increased cerebral oxidative stress as mechanisms underlying HFD-induced declines in cognitive performance in the aged brain.


Asunto(s)
Envejecimiento/metabolismo , Trastornos del Conocimiento/metabolismo , Grasas de la Dieta/administración & dosificación , Hipocampo/metabolismo , Factor 2 Relacionado con NF-E2/fisiología , Estrés Oxidativo , Adiposidad , Envejecimiento/psicología , Animales , Glucemia/metabolismo , Peso Corporal , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/psicología , Insulina/sangre , Leptina/sangre , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Carbonilación Proteica , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...