Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Transl Psychiatry ; 12(1): 226, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35654778

RESUMEN

Objective measures, such as activity monitoring, can potentially complement clinical assessment for psychiatric patients. Alterations in rest-activity patterns are commonly encountered in patients with major depressive disorder. The aim of this study was to investigate whether features of activity patterns correlate with severity of depression symptoms (evaluated by Montgomery-Åsberg Rating Scale (MADRS) for depression). We used actigraphy recordings collected during ongoing major depressive episodes from patients not undergoing any antidepressant treatment. The recordings were acquired from two independent studies using different actigraphy systems. Data was quality-controlled and pre-processed for feature extraction following uniform procedures. We trained multiple regression models to predict MADRS score from features of activity patterns using brute-force and semi-supervised machine learning algorithms. The models were filtered based on the precision and the accuracy of fitting on training dataset before undergoing external validation on an independent dataset. The features enriched in the models surviving external validation point to high depressive symptom severity being associated with less complex activity patterns and stronger coupling to external circadian entrainers. Our results bring proof-of-concept evidence that activity patterns correlate with severity of depressive symptoms and suggest that actigraphy recordings may be a useful tool for individual evaluation of patients with major depressive disorder.


Asunto(s)
Trastorno Depresivo Mayor , Antidepresivos/uso terapéutico , Trastorno Depresivo Mayor/tratamiento farmacológico , Humanos , Escalas de Valoración Psiquiátrica
2.
Toxicol Appl Pharmacol ; 354: 94-100, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29499248

RESUMEN

Methylmercury (MeHg) is a widespread environmental contaminant with established developmental neurotoxic effects. Computational models have identified glucocorticoid receptor (GR) signaling to be a key mediator behind the birth defects induced by Hg, but the mechanisms were not elucidated. Using molecular dynamics simulations, we found that MeHg can bind to the GR protein at Cys736 (located close to the ligand binding site) and distort the conformation of the ligand binging site. To assess the functional consequences of MeHg interaction with GR, we used a human cell line expressing a luciferase reporter system (HeLa AZ-GR). We found that 100 nM MeHg does not have any significant effect on GR activity alone, but the transactivation of gene expression by GR upon Dex (a synthetic GR agonist) administration was reduced in cells pre-treated with MeHg. Similar effects were found in transgenic zebrafish larvae expressing a GR reporter system (SR4G). Next we asked whether the effects of developmental exposure to MeHg are mediated by the effects on GR. Using a mutant zebrafish line carrying a loss-of-function mutation in the GR (grS357) we could show that the effects of developmental exposure to 2.5 nM MeHg are mitigated in absence of functional GR signaling. Taken together, our data indicate that inhibition of GR signaling may have a role in the developmental neurotoxic effects of MeHg.


Asunto(s)
Intoxicación del Sistema Nervioso por Mercurio/etiología , Compuestos de Metilmercurio/toxicidad , Sistema Nervioso/efectos de los fármacos , Receptores de Glucocorticoides/efectos de los fármacos , Animales , Animales Modificados Genéticamente , Sitios de Unión , Desarrollo Embrionario/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Ligandos , Intoxicación del Sistema Nervioso por Mercurio/embriología , Intoxicación del Sistema Nervioso por Mercurio/genética , Intoxicación del Sistema Nervioso por Mercurio/metabolismo , Compuestos de Metilmercurio/química , Compuestos de Metilmercurio/metabolismo , Simulación de Dinámica Molecular , Sistema Nervioso/embriología , Sistema Nervioso/metabolismo , Unión Proteica , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Medición de Riesgo , Transducción de Señal/efectos de los fármacos , Pruebas de Toxicidad , Toxicología/métodos , Pez Cebra
3.
Neurotoxicol Teratol ; 66: 94-101, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29221727

RESUMEN

The constant interplay between environment (including both exogenous and endogenous factors) and epigenome (defined as the combination of chromatin, its covalent modifications and noncoding RNAs) triggers epigenetic events that, by modulating gene expression, capture information about changes in the environment. In this mini review, we will focus on the neurodevelopmental implications of exposure to adverse prenatal milieu with emphasis on mechanistic and functional aspects. Several neurotoxic insults have been shown to affect epigenetics with negative consequences on the development of the nervous system; among them are methylmercury, lead, arsenic and cadmium, as well as excess of glucocorticoids. Further investigations on the individual susceptibility to epigenetic changes are needed to propose and validate such modifications as possible biomarkers for early identification of neurological/neurodevelopmental disorders and for predicting/monitoring response to treatment.


Asunto(s)
Contaminantes Ambientales/toxicidad , Epigénesis Genética/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glucocorticoides/toxicidad , Síndromes de Neurotoxicidad/genética , Efectos Tardíos de la Exposición Prenatal/genética , Femenino , Humanos , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Receptores de Glucocorticoides/genética
4.
Transl Psychiatry ; 5: e603, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26171984

RESUMEN

Growing evidence links adverse prenatal conditions to mood disorders. We investigated the long-term behavioral alterations induced by prenatal exposure to excess glucocorticoids (dexamethasone--DEX). At 12 months, but not earlier, DEX-exposed mice displayed depression-like behavior and impaired hippocampal neurogenesis, not reversible by the antidepressant fluoxetine (FLX). Concomitantly, we observed arrhythmic glucocorticoid secretion and absent circadian oscillations in hippocampal clock gene expression. Analysis of spontaneous activity showed progressive alterations in circadian entrainment preceding depression. Circadian oscillations in clock gene expression (measured by means of quantitative PCR) were also attenuated in skin fibroblasts before the appearance of depression. Interestingly, circadian entrainment is not altered in a model of depression (induced by methylmercury prenatal exposure) that responds to FLX. Altogether, our results suggest that alterations in circadian entrainment of spontaneous activity, and possibly clock gene expression in fibroblasts, may predict the onset of depression and the response to FLX in patients.


Asunto(s)
Antidepresivos de Segunda Generación/uso terapéutico , Ritmo Circadiano/fisiología , Depresión/fisiopatología , Fluoxetina/uso terapéutico , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Corticosterona/metabolismo , Depresión/tratamiento farmacológico , Depresión/psicología , Dexametasona/farmacología , Femenino , Fibroblastos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/psicología
5.
Cell Death Dis ; 6: e1793, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26086966

RESUMEN

Developmental exposure to excess glucocorticoids (GCs) has harmful neurodevelopmental effects, which include persistent alterations in the differentiation potential of embryonic neural stem cells (NSCs). The mechanisms, however, are largely unknown. Here, we investigated the effects of dexamethasone (Dex, a synthetic GC analog) by MeDIP-like genome-wide analysis of differentially methylated DNA regions (DMRs) in NSCs isolated from embryonic rat cortices. We found that Dex-induced genome-wide DNA hypomethylation in the NSCs in vitro. Similarly, in utero exposure to Dex resulted in global DNA hypomethylation in the cerebral cortex of 3-day-old mouse pups. Dex-exposed NSCs displayed stable changes in the expression of the DNA methyltransferase Dnmt3a, and Dkk1, an essential factor for neuronal differentiation. These alterations were dependent on Tet3 upregulation. In conclusion, we propose that GCs elicit strong and persistent effects on DNA methylation in NSCs with Tet3 playing an essential role in the regulation of Dnmt3a and Dkk1. Noteworthy is the occurrence of similar changes in Dnmt3a and Dkk1 gene expression after exposure to excess GC in vivo.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Proteínas de Unión al ADN/metabolismo , Dexametasona/farmacología , Glucocorticoides/farmacología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , ADN Metiltransferasa 3A , Dioxigenasas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
7.
J Intern Med ; 273(5): 490-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23600401

RESUMEN

Amongst environmental chemical contaminants, methylmercury (MeHg) remains a major concern because of its detrimental effects on developing organisms, which appear to be particularly susceptible to its toxicity. Here, we investigated the effects of low MeHg levels on the development of the nervous system using both in vitro and in vivo experimental models. In neural stem cells (NSCs), MeHg decreased proliferation and neuronal differentiation and induced cellular senescence associated with impairment in mitochondrial function and a concomitant decrease in global DNA methylation. Interestingly, the effects were heritable and could be observed in daughter NSCs never directly exposed to MeHg. By chronically exposing pregnant/lactating mice to MeHg, we found persistent behavioural changes in the male offspring, which exhibited depression-like behaviour that could be reversed by chronic treatment with the antidepressant fluoxetine. The behavioural alterations were associated with a decreased number of proliferating cells and lower expression of brain-derived neurotrophic factor (Bdnf) mRNA in the hippocampal dentate gyrus. MeHg exposure also induced long-lasting DNA hypermethylation, increased histone H3-K27 tri-methylation and decreased H3 acetylation at the Bdnf promoter IV, indicating that epigenetic mechanisms play a critical role in mediating the long-lasting effects of perinatal exposure to MeHg. Fluoxetine treatment restored the Bdnf mRNA expression levels, as well as the number of proliferating cells in the granule cell layer of the dentate gyrus, which further supports the hypothesis that links depression to impaired neurogenesis. Altogether, our findings have shown that low concentrations of MeHg induce long-lasting effects in NSCs that can potentially predispose individuals to depression, which we have reported earlier to occur in experimental animals exposed to MeHg during prenatal and early postnatal development.


Asunto(s)
Compuestos de Metilmercurio/toxicidad , Células-Madre Neurales/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Efectos Tardíos de la Exposición Prenatal , Animales , Antidepresivos de Segunda Generación/uso terapéutico , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Depresión/inducido químicamente , Depresión/tratamiento farmacológico , Modelos Animales de Enfermedad , Medicina Basada en la Evidencia , Femenino , Fluoxetina/uso terapéutico , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Resultado del Tratamiento
8.
Cell Death Dis ; 1: e92, 2010 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-21368868

RESUMEN

Alterations in intrauterine programming occurring during critical periods of development have adverse consequences for whole-organ systems or individual tissue functions in later life. In this paper, we show that rat embryonic neural stem cells (NSCs) exposed to the synthetic glucocorticoid dexamethasone (Dex) undergo heritable alterations, possibly through epigenetic mechanisms. Exposure to Dex results in decreased NSC proliferation, with no effects on survival or differentiation, and changes in the expression of genes associated with cellular senescence and mitochondrial functions. Dex upregulates cell cycle-related genes p16 and p21 in a glucocorticoid receptor(GR)-dependent manner. The senescence-associated markers high mobility group (Hmg) A1 and heterochromatin protein 1 (HP1) are also upregulated in Dex-exposed NSCs, whereas Bmi1 (polycomb ring finger oncogene) and mitochondrial genes Nd3 (NADH dehydrogenase 3) and Cytb (cytochrome b) are downregulated. The concomitant decrease in global DNA methylation and DNA methyltransferases (Dnmts) suggests the occurrence of epigenetic changes. All these features are retained in daughter NSCs (never directly exposed to Dex) and are associated with a higher susceptibility to oxidative stress, as shown by the increased occurrence of apoptotic cell death on exposure to the redox-cycling reactive oxygen species (ROS) generator 2,3-dimethoxy-1-naphthoquinone (DMNQ). Our study provides novel evidence for programming effects induced by glucocorticoids (GCs) on NSCs and supports the idea that fetal exposure to endogenous or exogenous GCs is likely to result in long-term consequences that may predispose to neurodevelopmental and/or neurodegenerative disorders.


Asunto(s)
Senescencia Celular , Dexametasona/farmacología , Glucocorticoides/farmacología , Células-Madre Neurales/metabolismo , Animales , Proliferación Celular , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citocromos b/genética , Citocromos b/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Epigénesis Genética , Mitocondrias/metabolismo , NADH Deshidrogenasa/genética , NADH Deshidrogenasa/metabolismo , Naftoquinonas/farmacología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 1 , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Receptores de Glucocorticoides/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
9.
Genes Immun ; 11(1): 11-20, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19847194

RESUMEN

Alternative splicing of pre-mRNA increases proteomic diversity, a crucial mechanism in defining tissue identity. We demonstrate differentially spliced interleukin (IL)-7 in distinct anatomic areas in the adult, in developing human brains and in normal human neuronal progenitor (NHNP) cells. IL-7c (c, the canonical form spanning all six exons) or its variants IL-7 delta 5, delta 4 or delta 4/5 were cloned and expressed as recombinant proteins. IL-7 and splice variants were able to shift the differentiation of NHNP cells as compared with the diluent control (P<0.01) defined by anti-beta (III)-tubulin and glial fibrillary acidic protein expression, with different degrees (IL-7c>delta 4/5>IL-7 delta 5); IL-7 delta 4 exhibited a significantly weaker potency. Differentiation was confirmed by transcriptome analysis of IL-7c-stimulated neural NHNP cells, resulting in 58 differentially expressed genes; some of these are involved in neural differentiation, for example, the developmentally regulated transcription factor krüppel-like factor 12, musashi 2, a translational regulator of cell fate or the sonic hedgehog receptor patch 1. This suggests that IL-7 influences neural development at a molecular level by participating in human brain architecture through glia cell formation: a paradigm that alternative splicing in cytokines, for example, for IL-7, has a physiological role in human organ development and progenitor cell differentiation.


Asunto(s)
Empalme Alternativo/fisiología , Encéfalo/metabolismo , Diferenciación Celular/fisiología , Interleucina-7/biosíntesis , Precursores del ARN/metabolismo , Células Madre/metabolismo , Adulto , Encéfalo/citología , Encéfalo/embriología , Humanos , Neuroglía/citología , Neuroglía/metabolismo , Células Madre/citología
10.
Proc Natl Acad Sci U S A ; 105(7): 2717-22, 2008 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-18272487

RESUMEN

Galanin is a neuropeptide with a wide range of effects in the nervous and endocrine systems, mediated through three G protein-coupled receptor subtypes (GalR1-3). Interestingly, galanin and its receptors are also expressed in certain tumors. Here we studied the effects of galanin in rat pheochromocytoma (PC12) cells stably transfected with GFP-tagged GalR2. Galanin at 100 nM inhibited cell proliferation in both nontransfected and transfected cells. Conversly, both galanin and the GalR2(R3)-agonist AR-M1896 induced caspase-dependent apoptotic cell death only in GalR2-transfected cells. Western-blot analyses of downstream mediators of the G(q/11)-type G protein showed down-regulation of pAkt and pBad in galanin-exposed transfected cells. Also, the specific PI3 kinase inhibitor LY-294002 increased the level of pBad and decreased activation of caspases. In addition, p21(cip1) levels were up-regulated in galanin-exposed PC12 cells and down-regulated in galanin-exposed GalR2-transfected cells. In agreement, FACS analyses of galanin exposed cells showed occurrence of cell cycle arrest in PC12 cells and cell death in transfected cells. Finally, as shown with real-time PCR, galanin and its receptors were expressed at very high levels in human pheochromocytoma tissues as compared with normal adrenal medulla. These findings point to GalR2 as a possible target for therapeuthic interventions in pheochromocytoma.


Asunto(s)
Apoptosis/efectos de los fármacos , Galanina/farmacología , Receptor de Galanina Tipo 2/metabolismo , Animales , Caspasas/metabolismo , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Galanina/genética , Regulación de la Expresión Génica , Humanos , Células PC12 , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptor de Galanina Tipo 1/genética , Receptor de Galanina Tipo 2/genética , Transducción de Señal/efectos de los fármacos
11.
Food Chem Toxicol ; 46(2): 808-11, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17980472

RESUMEN

Me-Hg and PCB153 are known neurotoxic contaminants which tend to accumulate in food, particularly in fish. Aim of this study was to perform asynchronous and combined exposure to Me-Hg and PCB153 in a neuronal rat cell line (PC12) to better characterise the antagonism observed at some combination concentrations. PC12 cells were treated with three concentrations of Me-Hg (0.1-0.5-1.0 microM) and PCB153 at one concentration (175 microM) in single and combined asynchronous exposures, using viability (MTT assay) as end-point. At all concentrations used, a statistically significant antagonistic effect was observed when Me-Hg preceded PCB153 exposure, while effect was additive when PCB153 preceded Me-Hg exposure. The antagonism is particularly evident at low concentrations of Me-Hg (0.1 microM). In conclusion, combined asynchronous exposure showed that whereas Me-Hg can modulate PCB153 toxicity, the opposite seems not to be true. Therefore, the use of asynchronous exposure could be a promising approach to study the mechanisms of toxicity of binary mixtures.


Asunto(s)
Compuestos de Metilmercurio/toxicidad , Células PC12/efectos de los fármacos , Bifenilos Policlorados/toxicidad , Animales , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Antagonismo de Drogas , Combinación de Medicamentos , Contaminación de Alimentos , Compuestos de Metilmercurio/administración & dosificación , Células PC12/metabolismo , Ratas
12.
Brain Res ; 1098(1): 1-8, 2006 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-16777078

RESUMEN

The neurotoxic effects of carbon monoxide (CO) are well known. Brain hypoxia due to the binding of CO to hemoglobin is a recognized cause of CO neurotoxicity, while the direct effect of CO on intracellular targets remains poorly understood. In the present study, we have investigated the pathways leading to neural cell death induced by in vitro exposure to CO using a gas exposure chamber that we have developed. Mouse hippocampal neurons (HT22) and human glial cells (D384) were exposed to concentrations of CO ranging from 300 to 1000 ppm in the presence of 20% oxygen. Cytotoxicity was observed after 48 h exposure to 1000 ppm, corresponding to approximately 1 microM CO in the cultured medium, as measured by gas chromatography. CO induced cell death with characteristic features of apoptosis. Exposed cells exhibited loss of mitochondrial membrane potential, release of cytochrome c into the cytosol, nuclei with chromatin condensation, and exposure of phosphatidyl serine on the external leaflet of the plasma membrane. CO also triggered activation of caspase and calpain proteases. Pre-incubation with either the pancaspase inhibitor Z-VAD-fmk (20 microM) or the calpain inhibitor E64d (25 microM) reduced by 50% the occurrence of apoptosis. When pre-incubating the cells with the two inhibitors together there was an additional reduction in the number of cells with apoptotic nuclei. These data suggest that CO causes apoptosis via activation of parallel proteolytic pathways involving both caspases and calpains. Furthermore, pre-treatment with the antioxidant MnTBAP (100 microM) significantly reduced the number of apoptotic nuclei, pointing to a critical role of oxidative stress in CO toxicity.


Asunto(s)
Apoptosis/efectos de los fármacos , Intoxicación por Monóxido de Carbono/patología , Hipoxia Encefálica/patología , Neuronas/patología , Animales , Anexina A5/metabolismo , Calpaína/antagonistas & inhibidores , Calpaína/metabolismo , Proteínas Portadoras/metabolismo , Inhibidores de Caspasas , Caspasas/metabolismo , Línea Celular , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Núcleo Celular/ultraestructura , Medios de Cultivo , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Hipocampo/patología , Humanos , Immunoblotting , Inmunohistoquímica , Potenciales de la Membrana/efectos de los fármacos , Ratones , Proteínas de Microfilamentos/metabolismo , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Fosfatidilserinas/farmacología , Propidio , Transducción de Señal/efectos de los fármacos , Azul de Tripano
13.
Food Chem Toxicol ; 44(9): 1505-12, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16757078

RESUMEN

The study of interactions for those substances which tend to accumulate in food and affect the nervous system appears to be a fundamental point to characterize the combined exposure in vitro. In this study we included two food contaminants which are known neurotoxicants: methyl-mercury (Me-Hg) and the ortho-substituted PCB 153. PC12 cells were treated with Me-Hg (range 1e-7, 2e-6 M) and PCB153 (range 1e-5, 4e-4 M) in single and combined synchronous experiments and a mathematical model was set up according to the Loewe additivity criterion to evaluate the level of interaction between toxicants, using viability as end-point. At some concentrations (Me-Hg 5e-7 M and PCB153 1e-4 and 2e-4 M; Me-Hg 1e-6M and PCB153 5e-5 M; Me-Hg 1e-7 M and PCB153 4e-4 M), a statistically significant antagonist effect was observed. No interaction was observed for other combinations. The analysis of other toxicological parameters known to be modified in single exposure experiments (TBARS and intra-cellular dopamine) confirmed the viability results. The results of our work represent a starting point to generate novel information on the interactions between PCB153 and Me-Hg in vitro, as well as a new relevant experimental and mathematical approach useful to investigate the effects of different toxicant mixtures.


Asunto(s)
Contaminación de Alimentos , Compuestos de Metilmercurio/toxicidad , Células PC12/efectos de los fármacos , Bifenilos Policlorados/toxicidad , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Antagonismo de Drogas , Combinación de Medicamentos , Contaminantes Ambientales/toxicidad , Formazáns/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Modelos Biológicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Síndromes de Neurotoxicidad/etiología , Células PC12/metabolismo , Ratas , Sales de Tetrazolio/metabolismo , Tiobarbitúricos/metabolismo
14.
Neuroscience ; 140(1): 293-304, 2006 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-16564642

RESUMEN

Neuropeptide Y has been implicated in pain modulation and is substantially up-regulated in dorsal root ganglia after peripheral nerve injury. To identify the role of neuropeptide Y after axotomy, we investigated the behavioral and neurochemical phenotype of neuropeptide Y Y1 receptor knockout mice with focus on dorsal root ganglion neurons and spinal cord. Using a specific antibody Y1 receptor immunoreactivity was found in dorsal root ganglia and in dorsal horn neurons of wild-type, but not knockout mice. The Y1 receptor knockout mice exhibited a pronounced mechanical hypersensitivity. After sciatic nerve axotomy, the deletion of Y1 receptor protected knockout mice from the axotomy-induced loss of dorsal root ganglion neurons seen in wild-type mice. Lower levels of calcitonin gene-related peptide and substance P were identified by immunohistochemistry in dorsal root ganglia and dorsal horn of knockout mice, and the axotomy-induced down-regulation of both calcitonin gene-related peptide and substance P was accentuated in Y1 receptor knockout. However, the transcript levels for calcitonin gene-related peptide and substance P were significantly higher in knockout than in wild-type dorsal root ganglia ipsilateral to the axotomy, while more calcitonin gene-related peptide- and substance P-like immunoreactivity accumulated proximal and distal to a crush of the sciatic nerve. These results indicate that the deletion of the Y1 receptor causes increased release and compensatory increased synthesis of calcitonin gene-related peptide and substance P in dorsal root ganglion neurons. Together, these findings suggest that, after peripheral nerve injury, neuropeptide Y, via its Y1 receptor receptor, plays a key role in cell survival as well as in transport and synthesis of the excitatory dorsal horn messengers calcitonin gene-related peptide and substance P and thus may contribute to pain hypersensitivity.


Asunto(s)
Ganglios Espinales/citología , Expresión Génica/genética , Neuronas/metabolismo , Neuropéptidos/metabolismo , Umbral del Dolor/fisiología , Receptores de Neuropéptido Y/deficiencia , Animales , Axotomía/métodos , Conducta Animal , Transporte Biológico/genética , Péptido Relacionado con Gen de Calcitonina/genética , Péptido Relacionado con Gen de Calcitonina/metabolismo , Recuento de Células/métodos , Lateralidad Funcional , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Dimensión del Dolor/métodos , Umbral del Dolor/efectos de los fármacos , Células del Asta Posterior/metabolismo , Sustancia P/genética , Sustancia P/metabolismo
15.
Cell Death Differ ; 12(8): 1134-40, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15861186

RESUMEN

Apoptotic cell death is an essential process in the development of the central nervous system and in the pathogenesis of its degenerative diseases. Efflux of K(+) and Cl(-) ions leads to the shrinkage of the apoptotic cell and facilitates the activation of caspases. Here, we present electrophysiological and immunocytochemical evidences for the activation of a voltage-dependent anion channel (VDAC) in the plasma membrane of neurons undergoing apoptosis. Anti-VDAC antibodies blocked the channel and inhibited the apoptotic process. In nonapoptotic cells, plasma membrane VDAC1 protein can function as a NADH (-ferricyanide) reductase. Opening of VDAC channels in apoptotic cells was associated with an increase in this activity, which was partly blocked by VDAC antibodies. Hence, it appears that there might be a dual role for this protein in the plasma membrane: (1) maintenance of redox homeostasis in normal cells and (2) promotion of anion efflux in apoptotic cells.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Neuronas/metabolismo , Porinas/fisiología , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Membrana Celular/metabolismo , Células Cultivadas , Canales de Cloruro/fisiología , Electrofisiología , Activación Enzimática , Hipocampo/citología , Hipocampo/fisiología , Humanos , Immunoblotting , Inmunoquímica , Ratones , NADH NADPH Oxidorreductasas/metabolismo , Neuroblastoma , Neuronas/citología , Neuronas/enzimología , Técnicas de Placa-Clamp , Porinas/antagonistas & inhibidores , Porinas/metabolismo , Canales de Potasio/fisiología , Canal Aniónico 1 Dependiente del Voltaje , Canales Aniónicos Dependientes del Voltaje
16.
Toxicol In Vitro ; 19(1): 11-20, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15582351

RESUMEN

Styrene-7,8-oxide (SO) is the main metabolite of styrene, a neurotoxic volatile organic compound used industrially. Here we report the novel observation that several markers of oxidative stress were affected in SK-N-MC cells exposed for 16 h to concentrations of SO that induce apoptotic cell death. The production of Thiobarbituric Acid Reactive Substances (TBARS), rose from 69.1 +/- 15.7 nmol/g protein (control) to 119.3 +/- 39.2 and 102.0 +/- 17.3 nmol/g protein after exposure to 0.3 and 1 mM SO, respectively. Carbonyl group levels were significantly enhanced by SO at both concentrations. The lower dose also decreased sulphydryl groups. SO caused a marked oxidative DNA damage, as shown by a fivefold increase in 8-hydroxy-2(')-deoxyguanosine (8-OHdG). In addition, SO exposure resulted in alterations of scavenging abilities, given the reduction of both glutathione (GSH) and glutathione-S-transferase (GST) activity. Induction of expression of the oxidative stress response gene heme-oxygenase-1 (HO-1) and an increased HO-1 activity were also observed. These data provide compelling evidence that oxidative stress significantly contributes to SO toxicity in neuronal cells.


Asunto(s)
Carcinógenos/toxicidad , Desoxiguanosina/análogos & derivados , Compuestos Epoxi/toxicidad , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , 8-Hidroxi-2'-Desoxicoguanosina , Biomarcadores/metabolismo , Línea Celular Tumoral , Daño del ADN , Desoxiguanosina/metabolismo , Relación Dosis-Respuesta a Droga , Depuradores de Radicales Libres/metabolismo , Glutatión/metabolismo , Glutatión Transferasa/metabolismo , Humanos , Neuroblastoma , Neuronas/metabolismo , Neuronas/patología , Oxidación-Reducción/efectos de los fármacos , Proteínas/efectos de los fármacos , Proteínas/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
17.
Toxicology ; 201(1-3): 125-32, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15297027

RESUMEN

Styrene 7,8-oxide (SO) is the main metabolite of styrene, a neurotoxic compound used industrially. Neurons exposed to SO undergo apoptosis with characteristic features including chromatin rearrangements and caspase activation. We report that the execution phase of apoptosis induced by SO (0.3 mM) in SK-N-MC neurons is triggered by translocation of apoptogenic factors (e.g., cytochrome c) into the cytosol. In addition, mitochondria exhibit lower Ca2+ capacity and loss of mitochondrial membrane potential (DeltaPsi). Lipid peroxidation, measured as thiobarbituric acid reactive substances (TBARS), is increased after 12 h. Pre-treatment with the antioxidant MnTBAP (100 microM) prevents the decrease of Ca2+ capacity, cytochrome c release, activation of caspases, exposure of phosphatidylserine and cell death. Hence, the neurotoxic effects of SO are related to mitochondrial damage and oxidative stress.


Asunto(s)
Apoptosis/efectos de los fármacos , Calcio/metabolismo , Carcinógenos/toxicidad , Citocromos c/metabolismo , Compuestos Epoxi/toxicidad , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Humanos , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/metabolismo , Neuronas/metabolismo , Especies Reactivas de Oxígeno/farmacología , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Células Tumorales Cultivadas
18.
Toxicol Lett ; 149(1-3): 59-66, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15093249

RESUMEN

Neural stem cells (NSC) undergo apoptotic cell death as an essential component of neural development. Here, we present the results of our studies on the mechanisms by which NSC undergo cell death in response to neurotoxic insults. As experimental models we used primary culture of adult NSC from the subventricular zone of the rat brain, and the neural stem cell line C17.2 initially derived from developing mouse cerebellum. NSC undergo apoptosis in response to staurosporine (0.25 microM) as well as agents inducing oxidative stress such as 2,3-dimethoxy-1,4-naphthoquinone (DMNQ). Exposed cells demonstrate an apoptotic morphology, positive TUNEL staining and phosphatidyl serine exposure as labeled with Annexin V. Using an antibody specific for cytochrome c, we found that cells exposed to staurosporine or DMNQ exhibited diffuse fluorescence throughout the cytosol, implying a release of cytochrome c from the mitochondria. In addition to positive immunoreactivity against the active fragment (p17) of caspase-3, the administration of the pan-caspase inhibitor, zVAD-fmk (40 microM), prevents apoptosis. Both NSC and C17.2 express the Fas receptor, and procaspase-8, but exposure to agonistic Fas mAb (250 ng/ml) fails to induce apoptosis. Pretreatment with cycloheximide or actinomycin D does not influence the cell response to Fas mAb, suggesting that the endogenous inhibitor of caspase-8 FLICE-inhibitory protein (FLIP) is not responsible for the inhibition of the Fas pathway. Thus, it appears that the Fas dependent cell death pathway is not operative in these cells, while the mitochondrial pathway is active and caspase-3 serves as an executioner caspase in the apoptotic machinery. It is known that Fas not only induces apoptosis, but can also deliver growth stimulatory signals through activation of the extracellular-signal regulated kinase (ERK) pathway. The Fas-induced ERK phosphorylation that we detect in C17.2 cells suggests that in NSC Fas may function as a mediator of growth rather than death.


Asunto(s)
Neuronas/fisiología , Células Madre/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Muerte Celular/fisiología , Humanos , Ratones , Mitocondrias/fisiología , Proteínas Quinasas Activadas por Mitógenos/biosíntesis , Fosforilación , Ratas , Regulación hacia Arriba/efectos de los fármacos , Receptor fas/fisiología
19.
Neurochem Int ; 42(7): 525-33, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12590934

RESUMEN

2,5-Hexanedione is a neurotoxic metabolite of hexane. The mechanisms of its neurotoxicity remain unclear. We assessed whether chronic exposure to 2,5-hexanedione affects the glutamate-nitric oxide-cGMP pathway in primary cultures of cerebellar neurons and/or in the cerebellum of rats. Chronic exposure of cultured cerebellar neurons to 2,5-hexanedione (200 microM) reduced by approximately 50% NMDA-induced formation of cGMP. Activation of soluble guanylate cyclase by nitric oxide was reduced by 46%. This treatment reduced the content of neuronal nitric oxide synthase and soluble guanylate cyclase in neurons by 23 and 20%, respectively. In the cerebellum of rats chronically exposed to 2,5-hexanedione (in the drinking water) NMDA-induced formation of cGMP was reduced by 55% as determined by in vivo brain microdialysis. Activation of soluble guanylate cyclase by nitric oxide was reduced by 65%. The content of neuronal nitric oxide synthase and of soluble guanylate cyclase was reduced by 25 and 21%, respectively, in the cerebellum of these rats. The effects are the same in both systems, indicating that cultured neurons are a good model to study the mechanisms of neurotoxicity of 2,5-hexanedione. These results indicate that chronic exposure to 2,5-hexanedione affects the glutamate-nitric oxide-cGMP pathway at different steps both in cultured neurons and in cerebellum of the animal in vivo. The alteration of this pathway may contribute to the neurotoxic effects of 2,5-hexanedione.


Asunto(s)
Cerebelo/metabolismo , GMP Cíclico/metabolismo , Ácido Glutámico/metabolismo , Hexanonas/farmacología , Neuronas/metabolismo , Neurotoxinas/farmacología , Óxido Nítrico/metabolismo , Animales , Células Cultivadas , Microdiálisis , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...