Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Biomed Sci ; 23: 6, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26786850

RESUMEN

BACKGROUND: Developing brain is a major target for alcohol's actions and neurological/functional abnormalities include microencephaly, reduced frontal cortex, mental retardation and attention-deficits. Previous studies have shown that ethanol altered the lateral ventricular neuroepithelial cell proliferation. However, the effect of ethanol on subventricular basal progenitors which generate majority of the cortical layers is not known. METHODS: We utilized spontaneously immortalized rat brain neuroblasts obtained from cultures of 18-day-old fetal rat cerebral cortices using in vitro ethanol exposures and an in utero binge model. In the in vitro acute model, cells were exposed to 86 mM ethanol for 8, 12 and 24 h. The second in vitro model comprised of chronic intermittent ethanol (CIE) exposure which consisted of 14 h of ethanol treatment followed by 10 h of withdrawal with three repetitions. RESULTS: E18 neuroblasts expressing Tbr2 representing immature basal progenitors displayed significant reduction of proliferation in response to ethanol in both the models. The decreased proliferation was accompanied by absence of apoptosis or autophagy as illustrated by FACS analysis and expression of apoptotic and autophagic markers. The BrdU incorporation assay indicated that ethanol enhanced the accumulation of cells at G1 with reduced cell number in S phase. In addition, the ethanol-inhibited basal neuroblasts proliferation was connected to decrease in cyclin D1 and Rb phosphorylation indicating cell cycle arrest. Further, in utero ethanol exposure in pregnant rats during E15-E18 significantly decreased Tbr2 and cyclin D1 positive cell number in cerebral cortex of embryos as assessed by cell sorting analysis by flow cytometry. CONCLUSIONS: Altogether, the current findings demonstrate that ethanol impacts the expansion of basal progenitors by inducing cytostasis that might explain the anomalies of cortico-cerebral development associated with fetal alcohol syndrome.


Asunto(s)
Trastornos del Sistema Nervioso Inducidos por Alcohol/metabolismo , Etanol/toxicidad , Trastornos del Espectro Alcohólico Fetal/metabolismo , Lóbulo Frontal/metabolismo , Fase G1/efectos de los fármacos , Células-Madre Neurales/metabolismo , Fase S/efectos de los fármacos , Trastornos del Sistema Nervioso Inducidos por Alcohol/patología , Animales , Ciclina D1/metabolismo , Femenino , Trastornos del Espectro Alcohólico Fetal/patología , Lóbulo Frontal/patología , Células-Madre Neurales/patología , Embarazo , Ratas , Proteínas de Dominio T Box/metabolismo
2.
J Biol Chem ; 287(52): 43533-42, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23118224

RESUMEN

The brain is one of the major targets of chronic alcohol abuse. Yet the fundamental mechanisms underlying alcohol-mediated brain damage remain unclear. The products of alcohol metabolism cause DNA damage, which in conditions of DNA repair dysfunction leads to genomic instability and neural death. We propose that one-carbon metabolism (OCM) impairment associated with long term chronic ethanol intake is a key factor in ethanol-induced neurotoxicity, because OCM provides cells with DNA precursors for DNA repair and methyl groups for DNA methylation, both critical for genomic stability. Using histological (immunohistochemistry and stereological counting) and biochemical assays, we show that 3-week chronic exposure of adult mice to 5% ethanol (Lieber-Decarli diet) results in increased DNA damage, reduced DNA repair, and neuronal death in the brain. These were concomitant with compromised OCM, as evidenced by elevated homocysteine, a marker of OCM dysfunction. We conclude that OCM dysfunction plays a causal role in alcohol-induced genomic instability in the brain because OCM status determines the alcohol effect on DNA damage/repair and genomic stability. Short ethanol exposure, which did not disturb OCM, also did not affect the response to DNA damage, whereas additional OCM disturbance induced by deficiency in a key OCM enzyme, methylenetetrahydrofolate reductase (MTHFR) in Mthfr(+/-) mice, exaggerated the ethanol effect on DNA repair. Thus, the impact of long term ethanol exposure on DNA repair and genomic stability in the brain results from OCM dysfunction, and MTHFR mutations such as Mthfr 677C→T, common in human population, may exaggerate the adverse effects of ethanol on the brain.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Depresores del Sistema Nervioso Central/efectos adversos , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Etanol/efectos adversos , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Adulto , Consumo de Bebidas Alcohólicas/metabolismo , Animales , Carbono/metabolismo , Depresores del Sistema Nervioso Central/farmacología , Reparación del ADN/genética , Etanol/farmacología , Inestabilidad Genómica/efectos de los fármacos , Inestabilidad Genómica/genética , Homocisteína/genética , Homocisteína/metabolismo , Humanos , Masculino , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Ratones , Ratones Mutantes , Mutación
3.
Alcohol Clin Exp Res ; 37(1): 96-109, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22757755

RESUMEN

BACKGROUND: Prenatal exposure to ethanol (EtOH) elicits a range of neuro-developmental abnormalities, microcephaly to behavioral deficits. Impaired protein synthesis has been connected to pathogenesis of EtOH-induced brain damage and abnormal neuron development. However, mechanisms underlying these impairments of protein synthesis are not known. In this study, we illustrate the effects of EtOH on programmed cell death protein 4 (PDCD4), a tumor and translation repressor. METHODS: Primary cortical neurons (PCNs) were treated with 2.5 and 4 mg/ml EtOH for different time points (4 to 24 hours), and PDCD4 expression was detected by Western blotting. Protein synthesis was determined using [(35) S] methionine incorporation assay. Methyl cap pull-down assay was performed to establish the effect of EtOH on association of eukaryotic initiation factor 4A (eIF4A) with capped mRNA. Luciferase assay was performed to determine the in vivo translation. A 2-day acute 5-dose binge model with EtOH (4 g/kg body wt, 25% v/v) was performed in Sprague-Dawley rats at 12-hour intervals and analyzed for PDCD4, eIF4A, and eIF4A-methyl cap association. RESULTS: EtOH increased PDCD4 expression in a time- and dose-dependent manner in PCNs, which inhibited the association of eIF4A with methyl cap. EtOH and ectopic PDCD4 expression suppressed in vivo translation in PCNs and RNAi targeting of PDCD4 blocked the inhibitory effect of EtOH on protein synthesis. In utero exposure of pregnant rats to EtOH resulted in a significant increase in PDCD4 in fetal cerebral cortex along with the inhibition of methyl cap-associated eIF4A, compared with isocaloric controls. Increased PDCD4 also occurred in pooled fractions of remaining brain regions. CONCLUSIONS: Our data, for the first time, illustrate that PDCD4 mediates inhibitory effects of EtOH on protein synthesis in PCNs and developing brain.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Corteza Cerebral/efectos de los fármacos , Etanol/farmacología , Neuronas/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Animales , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Núcleo Celular/metabolismo , Depresores del Sistema Nervioso Central/farmacología , Corteza Cerebral/citología , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Citoplasma/metabolismo , Factor 4A Eucariótico de Iniciación/metabolismo , Femenino , Trastornos del Espectro Alcohólico Fetal/etiología , Neuronas/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
4.
Alcohol Clin Exp Res (Hoboken) ; 47(8): 1530-1543, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37364904

RESUMEN

BACKGROUND: Prenatal ethanol exposure hinders oxidative stress-mediated neuroblast/neural progenitor cell proliferation by inhibiting G1-S transition, a process vital to neocortical development. We previously showed that ethanol elicits this redox imbalance by repressing cystathionine γ-lyase (CSE), the rate-limiting enzyme in the transsulfuration pathway in fetal brain and cultured cerebral cortical neurons. However, the mechanism by which ethanol impacts the CSE pathway in proliferating neuroblasts is not known. We conducted experiments to define the effects of ethanol on CSE regulation and the molecular signaling events that control this vital pathway. This enabled us to develop an intervention to prevent the ethanol-associated cytostasis. METHODS: Spontaneously immortalized undifferentiated E18 rat neuroblasts from brain cerebral cortex were exposed to ethanol to mimic an acute consumption pattern in humans. We performed loss- and gain-of-function studies to evaluate whether NFATc4 is a transcriptional regulator of CSE. The neuroprotective effects of chlorogenic acid (CGA) against the effects of ethanol were assessed using ROS and GSH/GSSG assays as measures of oxidative stress, transcriptional activation of NFATc4, and expression of NFATc4 and CSE by qRT-PCR and immunoblotting. RESULTS: Ethanol treatment of E18-neuroblast cells elicited oxidative stress and significantly reduced CSE expression with a concomitant decrease in NFATc4 transcriptional activation and expression. In parallel, inhibition of the calcineurin/NFAT pathway by FK506 exaggerated ethanol-induced CSE loss. In contrast, NFATc4 overexpression prevented loss of ethanol-induced CSE. CGA increased and activated NFATc4, amplified CSE expression, rescued ethanol-induced oxidative stress, and averted the cytostasis of neuroblasts by rescuing cyclin D1 expression. CONCLUSIONS: These findings demonstrate that ethanol can perturb CSE-dependent redox homeostasis by impairing the NFATc4 signaling pathway in neuroblasts. Notably, ethanol-associated impairments were rescued by genetic or pharmacological activation of NFATc4. Furthermore, we found a potential role for CGA in mitigating the ethanol-related neuroblast toxicity with a compelling connection to the NFATc4/CSE pathway.

5.
Mol Pharmacol ; 80(6): 988-99, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21873460

RESUMEN

Ethanol (ETOH) can cause apoptotic death of neurons by depleting GSH with an associated increase in oxidative stress. The current study illustrates a means to overcome this ETOH-induced neurotoxicity by enhancing GSH through boosting Nrf2, a transcription factor that controls GSH homeostasis. ETOH treatment caused a significant increase in Nrf2 protein, transcript expression, Nrf2-DNA binding activity, and expression of its transcriptional target, NQO1, in primary cortical neuron (PCNs). However, this increase in Nrf2 did not maintain GSH levels in response to ETOH, and apoptotic death still occurred. To elucidate this phenomenon, we silenced Nrf2 in neurons and found that ETOH-induced GSH depletion and the increase in superoxide levels were exacerbated. Furthermore, Nrf2 knockdown resulted in significantly increased (P < 0.05) caspase 3 activity and apoptosis. Adenovirus-mediated overexpression of Nrf2 prevented ETOH-induced depletion of GSH from the medium and high GSH subpopulations and prevented ETOH-related apoptotic death. These studies illustrate the importance of Nrf2-dependent maintenance of GSH homeostasis in cerebral cortical neurons in the defense against oxidative stress and apoptotic death elicited by ETOH exposure.


Asunto(s)
Apoptosis/fisiología , Corteza Cerebral/metabolismo , Etanol/toxicidad , Regulación de la Expresión Génica , Factor 2 Relacionado con NF-E2/biosíntesis , Neuronas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Técnicas de Cocultivo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glutatión/deficiencia , Factor 2 Relacionado con NF-E2/genética , Neuronas/efectos de los fármacos , Embarazo , Ratas , Ratas Sprague-Dawley
6.
Biomolecules ; 9(11)2019 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-31671572

RESUMEN

NRF2 is a redox-sensitive transcription factor that depending on the duration or magnitude of the stress, either translocates to the nucleus (beneficial) or is degraded in the cytosol (harmful). However, the role of NRF2-based mechanism(s) under ethanol (E)-induced developmental toxicity in the placental context remains unknown. Here, we used a rat prenatal model of maternal alcohol stress consisting of intermittent ethanol vapor (IEV) daily from GD11 to GD20 with a 6 h ON/18 h OFF in a vapor chamber and in vitro placental model consisting of HTR-8 trophoblasts exposed to 86 mM of E for either 24 h or 48 h. The role of NRF2 was evaluated through the NRF2-transactivation reporter assay, qRT-PCR, and Western blotting for NRF2 and cell growth-promoting protein, and cell proliferation assay. In utero and in vitro E decreased the nuclear NRF2 content and diminished its transactivation ability along with dysregulation of the proliferation indices, PCNA, CYCLIN-D1, and p21. This was associated with a ~50% reduction in cell proliferation in vitro in trophoblasts. Interestingly, this was found to be partially rescued by ectopic Nrf2 overexpression. These results indicate that ethanol-induced dysregulation of NRF2 coordinately regulates PCNA/CYCLIN-D1/p21 involving growth network, at least partially to set a stage for placental perturbations.


Asunto(s)
Etanol/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Transducción de Señal/efectos de los fármacos , Trofoblastos/citología , Trofoblastos/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Factor 2 Relacionado con NF-E2/genética , Embarazo , Activación Transcripcional/efectos de los fármacos , Trofoblastos/metabolismo
7.
J Neurosci Res ; 86(5): 1064-76, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18058941

RESUMEN

Ethanol ingestion during pregnancy elicits damage to the developing brain, some of which appears to result from enhanced apoptotic death of neurons. A consistent characteristic of this phenomenon is a highly differing sensitivity to ethanol within specific neuron populations. One possible explanation for this "selective vulnerability" could be cellular variations in glutathione (GSH) homeostasis. Prior studies have illustrated that ethanol elicits apoptotic death of neurons in the developing brain, that oxidative stress may be an underlying mechanism, and that GSH can be neuroprotective. In the present study, both multiphoton microscopy and flow cytometry demonstrate a striking heterogeneity in GSH content within cortical neuron populations. Ethanol differentially elicits apoptotic death and oxidative stress in these neurons. When neuron GSH content is reduced by treatment with butathione sulfoxamine, the ethanol-mediated enhancement of reactive oxygen species is exacerbated. Sorting of cells into high- and low-GSH populations further exemplifies ethanol-mediated oxidative stress whereby apoptotic indices are preferentially elevated in the low-GSH population. Western blot analysis of the low-GSH subpopulations shows higher ethanol-mediated expression of active caspase 3 and 24-kDa PARP-1 fragments compared with the high-GSH subpopulation. In addition, neuronal content of 4-hydroxynonenal adducts is higher in low-GSH neurons in response to ethanol. These studies suggest that GSH content is an important predictor of neuronal sensitivity to ethanol-mediated oxidative stress and subsequent cell death. The data support the proposition that the differences in proapoptotic responses to ethanol within specific neuron populations reflect a heterogeneity of neuron GSH content.


Asunto(s)
Trastornos del Sistema Nervioso Inducidos por Alcohol/metabolismo , Apoptosis/fisiología , Corteza Cerebral/metabolismo , Etanol/toxicidad , Glutatión/metabolismo , Neuronas/metabolismo , Trastornos del Sistema Nervioso Inducidos por Alcohol/fisiopatología , Aldehídos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/fisiopatología , Resistencia a Medicamentos/fisiología , Femenino , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Neuronas/clasificación , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Embarazo , Ratas
8.
Alcohol Clin Exp Res ; 32(11): 1884-92, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18717656

RESUMEN

BACKGROUND: Prior studies by many laboratories have illustrated that ethanol can elicit a cascade of caspase-dependent apoptotic events in cultured neurons. Studies in our laboratory have connected this to oxidative stress and effects on fetal cortical neuron glutathione homeostasis. AIMS: The intent of the following studies is to address mechanisms underlying ethanol-associated DNA damage that may be connected to apoptotic death of neurons. METHODS: Cultures of fetal rat cerebral cortical neurons were utilized. Estimates of DNA damage was determined by Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining and nuclear condensation; Poly(ADP-ribose) polymerase-1 (PARP-1) expression was determined by immunostaining and Western blotting; and occurrence of parylation and AIF translocations were assessed by Western blotting. RESULTS: Ethanol treatment of the neurons generated increases in DNA damage by 4 hours while nuclear condensation was low at the short exposure period but increased markedly by 24 hours. This was temporally related to a marked up-regulation of PARP-1 expression. Activity of PARP-1, as assessed by PolyADP-ribose (PAR) formation, occurred within 15 minutes and peaked by 6 to 8 hours of ethanol treatment. An almost complete translocation of apoptosis inducing factor (AIF) from mitochondria to the nucleus occurred by 24 hours of ethanol treatment (4.0 mg/ml). Ethanol treatment for 4, 12, and 24 hours elicited an increasing caspase-mediated cleavage of PARP-1 to its 24 kDa fragment. CONCLUSIONS: These data illustrate the rapid occurrence of DNA damage following ethanol exposure and that PARP-1 pathways may play a role in the subsequent apoptotic death of these neurons.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Depresores del Sistema Nervioso Central/farmacología , Corteza Cerebral/metabolismo , Daño del ADN/efectos de los fármacos , Etanol/farmacología , Neuronas/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Núcleo Celular/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasa-1 , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Tiempo
9.
PLoS One ; 9(5): e98080, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24837604

RESUMEN

Ingestion of ethanol (ETOH) during pregnancy induces grave abnormalities in developing fetal brain. We have previously reported that ETOH induces programmed cell death 4 (PDCD4), a critical regulator of cell growth, in cultured fetal cerebral cortical neurons (PCNs) and in the cerebral cortex in vivo and affect protein synthesis as observed in Fetal Alcohol Spectrum Disorder (FASD). However, the mechanism which activates PDCD4 in neuronal systems is unclear and understanding this regulation may provide a counteractive strategy to correct the protein synthesis associated developmental changes seen in FASD. The present study investigates the molecular mechanism by which ethanol regulates PDCD4 in cortical neuroblasts, the immediate precursor of neurons. ETOH treatment significantly increased PDCD4 protein and transcript expression in spontaneously immortalized rat brain neuroblasts. Since PDCD4 is regulated at both the post-translational and post-transcriptional level, we assessed ETOH's effect on PDCD4 protein and mRNA stability. Chase experiments demonstrated that ETOH does not significantly impact either PDCD4 protein or mRNA stabilization. PDCD4 promoter-reporter assays confirmed that PDCD4 is transcriptionally regulated by ETOH in neuroblasts. Given a critical role of glycogen synthase kinase 3ß (GSK-3ß) signaling in regulating protein synthesis and neurotoxic mechanisms, we investigated the involvement of GSK-3ß and showed that multifunctional GSK-3ß was significantly activated in response to ETOH in neuroblasts. In addition, we found that ETOH-induced activation of PDCD4 was inhibited by pharmacologic blockade of GSK-3ß using inhibitors, lithium chloride (LiCl) and SB-216763 or siRNA mediated silencing of GSK-3ß. These results suggest that ethanol transcriptionally upregulates PDCD4 by enhancing GSK-3ß signaling in cortical neuroblasts. Further, we demonstrate that canonical Wnt-3a/GSK-3ß signaling is involved in regulating PDCD4 protein expression. Altogether, we provide evidence that GSK-3ß/PDCD4 network may represent a critical modulatory point to manage the protein synthetic anomalies and growth aberrations of neural cells seen in FASD.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Etanol/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Células-Madre Neurales/efectos de los fármacos , Transducción de Señal , Activación Transcripcional , Animales , Proteínas Reguladoras de la Apoptosis/genética , Línea Celular Tumoral , Células Cultivadas , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Células-Madre Neurales/metabolismo , Ratas
10.
Exp Biol Med (Maywood) ; 237(7): 740-7, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22829701

RESUMEN

Chronic alcohol abuse results in a variety of pathological effects including damage to the brain. The causes of alcohol-induced brain pathology are presently unclear. Several mechanisms of pathogenicity of chronic alcoholism have been proposed, including accumulation of DNA damage in the absence of repair, resulting in genomic instability and death of neurons. Genomic instability is a unified genetic mechanism leading to a variety of neurodegenerative disorders. Ethanol also likely interacts with various metabolic pathways, including one-carbon metabolism (OCM). OCM is critical for the synthesis of DNA precursors, essential for DNA repair, and as a methyl donor for various methylation events, including DNA methylation. Both DNA repair and DNA methylation are critical for maintaining genomic stability. In this review, we outline the role of DNA damage and DNA repair dysfunction in chronic alcohol-induced neurodegeneration.


Asunto(s)
Alcoholismo/patología , Encéfalo/patología , Daño del ADN , Alcoholismo/genética , Enfermedad Crónica , Metilación de ADN , Reparación del ADN , Inestabilidad Genómica , Humanos , Neuronas/patología
11.
Environ Toxicol Pharmacol ; 33(2): 353-60, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22301167

RESUMEN

Primary cultures of fetal rat cortical neurons and astrocytes were used to test the hypothesis that astrocyte-mediated control of neuronal glutathione (GSH) is a potent factor in neuroprotection against rotenone and paraquat. In neurons, rotenone (0.025-1 µM) for 4 and 24 h decreased viability as did paraquat (2-100 µM). Rotenone (30 nM) decreased neuronal viability and GSH by 24% and 30%, while ROS were increased by 56%. Paraquat (30 µM) decreased neuronal viability and GSH by 36% and 70%, while ROS were increased by 23%. When neurons were co-cultured with astrocytes, their GSH increased 1.5 fold and 5 fold at 12 and 24 h. Co-culturing with astrocytes blocked neuronal death and damage by rotenone and paraquat. Astrocyte-mediated neuroprotection was dependent on the activity of components of the γ-glutamyl cycle. These studies illustrate the importance of astrocyte-mediated glutathione homeostasis for protection of neurons from rotenone and paraquat and the role of the γ-glutamyl cycle in this neuroprotection.


Asunto(s)
Astrocitos/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Herbicidas/toxicidad , Insecticidas/toxicidad , Neuronas/efectos de los fármacos , Paraquat/toxicidad , Rotenona/toxicidad , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Antígenos CD13/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Técnicas de Cocultivo , Citoprotección , Relación Dosis-Respuesta a Droga , Glutatión/metabolismo , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , gamma-Glutamiltransferasa/metabolismo
12.
J Neurochem ; 96(5): 1289-300, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16464233

RESUMEN

Ethanol increases apoptotic neuron death in the developing brain and at least part of this may be mediated by oxidative stress. In cultured fetal rat cortical neurons, Ethanol increases levels of reactive oxygen species (ROS) within minutes of exposure and reduces total cellular glutathione (GSH) shortly thereafter. This is followed by onset of apoptotic cell death. These responses to Ethanol can be blocked by elevating neuron GSH with N-acetylcysteine or by co-culturing neurons with neonatal cortical astrocytes. We describe here mechanisms by which the astrocyte-neuron gamma-glutamyl cycle is up-regulated by Ethanol, enhancing control of neuron GSH in response to the pro-oxidant, Ethanol. Up to 6 days of Ethanol exposure had no consistent effects on activities of gamma-glutamyl cysteine ligase or glutathione synthetase, and GSH content remained unchanged (p < 0.05). However, glutathione reductase was increased with 1 and 2 day Ethanol exposures, 25% and 39% for 2.5 and 4.0 mg/mL Ethanol by 1 day, and 11% and 16% for 2.5 and 4.0 mg/mL at 2 days, respectively (p < 0.05). A 24 h exposure to 4.0 mg/mL Ethanol increased GSH efflux from astrocyte up to 517% (p < 0.05). Ethanol increased both gamma-glutamyl transpeptidase expression and activity on astrocyte within 24 h of exposure (40%, p = 0.05 with 4.0 mg/mL) and this continued for at least 4 days of Ethanol treatment. Aminopeptidase N activity on neurons increased by 62% and 55% within 1 h of Ethanol for 2.5 and 4.0 mg/mL concentration, respectively (p < 0.05), remaining elevated for 24 h of treatment. Thus, there are at least three key points of the gamma-glutamyl cycle that are up-regulated by Ethanol, the net effect being to enhance neuron GSH homeostasis, thereby protecting neurons from Ethanol-mediated oxidative stress and apoptotic death.


Asunto(s)
Astrocitos/fisiología , Depresores del Sistema Nervioso Central/farmacología , Corteza Cerebral/citología , Etanol/farmacología , Glutatión/metabolismo , Homeostasis/efectos de los fármacos , Neuronas/enzimología , Animales , Animales Recién Nacidos , Anexina A5/metabolismo , Western Blotting/métodos , Antígenos CD13/metabolismo , Muerte Celular/efectos de los fármacos , Técnicas de Cocultivo/métodos , Relación Dosis-Respuesta a Droga , Fluoresceínas/metabolismo , Glutatión Reductasa/metabolismo , Glutatión Sintasa/metabolismo , Unión Proteica/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , gamma-Glutamiltransferasa/metabolismo
13.
J Neurosci Res ; 80(5): 655-66, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15880562

RESUMEN

Ethanol induces oxidative stress in cultured fetal rat cortical neurons and this is followed by apoptotic death, which can be prevented by normalization of cell content of reduced glutathione (GSH). Because astrocytes can play a central role in maintenance of neuron GSH homeostasis, the following experiments utilized cocultures of neonatal rat cortical astrocytes and fetal cortical neurons to determine if astrocytes could protect neurons from ethanol-mediated apoptotic death via this mechanism. In cortical neurons cultured in the absence of astrocytes, ethanol (2.5 and 4 mg/ml; 6-, 12-, and 24-hr exposures) decreased trypan blue exclusion and the MTT viability measures by up to 45% (P < 0.05), increased levels of reactive oxygen species (ROS) by up to 81% (P < 0.05), and decreased GSH within 1 hr of treatment by 49 and 51% for 2.5 and 4 mg/ml, respectively (P < 0.05). This was followed by onset of apoptotic cell death as determined by increased Annexin V binding and DNA fragmentation by 12 hr of ethanol exposure. Coculturing neurons with astrocytes prevented GSH depletion by 2.5 mg/ml ethanol, whereas GSH content was increased over controls in neurons exposed to 4 mg/ml ethanol (by up to 341%; P < 0.05). Ethanol generated increases in neuron ROS and apoptosis; decreases in viability were also prevented by coculture. Astrocytes were largely insensitive to ethanol, using the same measures. Only exposure to 4.0 mg/ml ethanol decreased GSH content in astrocytes, concomitant with a 204% increase in GSH efflux (P < 0.05). These studies illustrate that astrocytes can protect neurons from ethanol-mediated apoptotic death and that this may be related to maintenance of neuron GSH.


Asunto(s)
Astrocitos/citología , Astrocitos/metabolismo , Comunicación Celular/fisiología , Neuronas/citología , Neuronas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Depresores del Sistema Nervioso Central/farmacología , Colorantes/farmacocinética , Etanol/farmacología , Femenino , Trastornos del Espectro Alcohólico Fetal/metabolismo , Trastornos del Espectro Alcohólico Fetal/patología , Glutatión/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Estrés Oxidativo/fisiología , Embarazo , Ratas , Ratas Sprague-Dawley , Azul de Tripano/farmacocinética
14.
Alcohol Clin Exp Res ; 26(8): 1252-8, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12198402

RESUMEN

BACKGROUND: 4-Hydroxynonenal (HNE), a toxic lipid peroxidation product, has been implicated in mitochondrial damage in rat liver by ethanol consumption. The present study assessed the effects of short-term in vivo ethanol exposure on HNE detoxification by mitochondrial glutathione S-transferase (GST). METHODS: Male Sprague Dawley rats were administered 5 doses of ethanol (4 g/kg) at 12 hr intervals by gavage. Pair-fed rats that received isocaloric dextrose instead of ethanol served as controls. Mitochondrial and submitochondrial fractions were prepared from the livers. Mitochondrial contents of HNE and HNE-glutathione conjugate were measured by high-performance liquid chromatography. GST isoforms were identified by Western blots in submitochondrial fractions. RESULTS: Whereas there was an 80% increase in mitochondrial HNE content after ethanol consumption, there was a 42% decrease in the content of HNE-glutathione conjugate, compared with controls (p< 0.05). After ethanol exposure, the GST activities toward HNE in intact mitochondria and in the membranous fraction were decreased by 37% and 45% (p< 0.05), respectively, whereas that in the aqueous fraction was unchanged. Kinetic analysis of HNE conjugation by the membrane-associated GST showed that ethanol decreased the V(max) nearly by half (p< 0.05), whereas it did not affect the K(m). HNE conjugation by the aqueous GST demonstrated a higher K(m) than that of the membrane-associated GST, although its kinetics were not significantly altered by ethanol. Immunochemical analysis with Western blots demonstrated that both the membranous and the aqueous fractions of mitochondria contain GST-alpha and GST-mu isoforms, whereas GST-pi was absent. CONCLUSIONS: HNE detoxification by mitochondrial GST is compromised by short-term ethanol consumption, which may contribute to elevated mitochondrial HNE content and hence its toxicity in the ethanol-exposed liver.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Aldehídos/metabolismo , Glutatión Transferasa/metabolismo , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/enzimología , Aldehídos/toxicidad , Animales , Etanol/farmacología , Inactivación Metabólica/fisiología , Masculino , Mitocondrias Hepáticas/metabolismo , Ratas , Ratas Sprague-Dawley
15.
Biochemistry ; 41(25): 8212-20, 2002 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-12069614

RESUMEN

Bovine heart cytochrome c oxidase (CcO) was inactivated by the lipid peroxidation product 4-hydroxy-2-nonenal (HNE) in a time- and concentration-dependent manner with pseudo-first-order kinetics. Cytochrome c oxidase electron transport activity decreased by as much as 50% when the enzyme was incubated for 2 h at room temperature with excess HNE (300-500 microM). HNE-modified CcO subunits were identified by two mass spectrometric methods: electrospray ionization mass spectrometry (ESI/MS) and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF/MS). All of the experimentally determined molecular masses were in excellent agreement with published sequence values with an accuracy of approximately 1 part per 10000 mass units for subunits smaller than 20 kDa and approximately 1 part per 1000 mass units for the three subunits larger than 20 kDa. Both MS methods detected six CcO subunits with an increased mass of 156 Da after reaction with HNE (subunits II, IV, Vb, VIIa, VIIc, and VIII); this result indicates a single Michael-type reaction site on either a lysine or histidine residue within each subunit. Reaction of HNE with either subunit VIIc or subunit VIII (modified approximately 30% and 50-75%, respectively) must be responsible for CcO inhibition. None of the other subunits were modified more than 5% and could not account for the observed loss of activity. Reaction of HNE with His-36 of subunit VIII is most consistent with the approximately 50% inhibition of CcO: (1) subunit VIII is modified more than any other subunit by HNE; (2) the time dependence of subunit VIII modification is consistent with the percent inhibition of CcO; (3) His-36 was identified as the HNE-modified amino acid residue within subunit VIII by tandem MS analysis.


Asunto(s)
Aldehídos/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Inhibidores Enzimáticos/metabolismo , Peroxidación de Lípido , Miocardio/enzimología , Aldehídos/farmacología , Secuencia de Aminoácidos , Animales , Bovinos , Cromatografía Líquida de Alta Presión , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Peroxidación de Lípido/efectos de los fármacos , Datos de Secuencia Molecular , Miocardio/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA