Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Vitam Horm ; 111: 313-337, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31421706

RESUMO

Endogenous opioids (enkephalins, endorphins and dynorphins) are small peptides that play a main role in pain perception and analgesia, as well as in alcohol (ethanol) reinforcement and reward. Alcohol reinforcement involves the ethanol-induced activation of the endogenous opioid system, a process that may augment the hedonic value and the reinforcing properties of the drug, which in turn increases substance consumption. Changes in opioidergic transmission may contribute to alcohol intoxication and to the neuroadaptive responses produced by the long-lasting exposure to ethanol. Opioidergic transmission may be altered by ethanol at distinct levels, including the expression of precursor mRNAs, biosynthesis and release of opioid peptides, as well as ligand binding to opioid receptors. In adult rats, ß-endorphinergic and enkephalinergic transmission, through activation of mu and delta opioid receptors, mediate ethanol reinforcement and high alcohol drinking behavior. Prenatal ethanol exposure (PEE) selectively modifies Methionine-enkephalin (Met-enk) content in several brain regions of infant and adolescent rats, particularly those of the reward circuits. In preweanling rats, Met-enk content is decreased in the ventral tegmental area but is increased in the prefrontal cortex and the nucleus accumbens and other brain areas, as a consequence of a short and moderate ethanol exposure during late gestation. PEE also increases Met-enk levels in the prefrontal cortex and other brain regions of 30-day-old adolescent rats. These findings suggest that mesocorticolimbic enkephalins are essential in ethanol reinforcement in offspring, as previously reported in adult rats.


Assuntos
Encefalinas/análise , Encefalinas/fisiologia , Etanol/farmacologia , Efeitos Tardios da Exposição Pré-Natal , Transmissão Sináptica/efeitos dos fármacos , Consumo de Bebidas Alcoólicas , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Química Encefálica/efeitos dos fármacos , Dopamina/fisiologia , Etanol/administração & dosagem , Etanol/efeitos adversos , Feminino , Núcleo Accumbens/metabolismo , Peptídeos Opioides/metabolismo , Córtex Pré-Frontal/metabolismo , Gravidez , Ratos , Receptores Opioides/metabolismo , Reforço Psicológico , Área Tegmentar Ventral/metabolismo
2.
Behav Brain Res ; 368: 111908, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-30986490

RESUMO

Fetal ethanol experience generates learning and memories capable to increase ethanol consummatory behaviors during infancy. Opioid system seems to be involved in mediating those alcohol-related behaviors. In this work, we proposed to study the impact of prenatal exposure to a moderate ethanol dose, upon ingestion of the drug and possible ethanol-induced molecular changes on opioid precursor peptides (POMC, Pro-enk and Pro-DYN) and receptors (MOR, DOR and KOR) mRNA expression, in hypothalamus. Pregnant rats received during gestational days (GDs) 17-20, a daily intragastric (i.g.) administration with 2g/kg ethanol or water. A third group of dams was left undisturbed during pregnancy (Unmanipulated group). Intake test was conducted at postnatal days (PDs) 14-15. Three groups of pups were performed: control (no intake test), water (vehicle) and 5% ethanol. At the end of intake test blood samples were taken to quantify blood ethanol concentrations (BECs) and hypothalamus sections were obtained to perform qRT-PRC assessment of opioid precursor peptides and receptors. The analysis of the consummatory responses (% of consumption) and pharmacokinetic profiles (BECs) suggested that maternal manipulation induced by i.g. intubations, during the last four days of gestation (whenever ethanol or water), are sufficient to induce infantile ethanol intake during infancy. Gene expression from the hypothalamus of unmanipulated group revealed that infantile ingestive experiences with ethanol can down-regulate expression of mRNA Pro-Dyn and up-regulate mRNA expression of MOR and KOR. Finally, MOR mRNA expression was attenuated by prenatal i.g. manipulation in pups exposed to 5% ethanol.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Receptores Opioides/efeitos dos fármacos , Analgésicos Opioides/metabolismo , Animais , Animais Recém-Nascidos , Etanol/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Peptídeos Opioides , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores Opioides/genética , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética
3.
Salud ment ; 38(3): 167-176, may.-jun. 2015. ilus
Artigo em Espanhol | LILACS-Express | LILACS | ID: lil-759191

RESUMO

Antecedentes: La exposición crónica al alcohol se asocia con procesos neurotóxicos y neurodegenerativos relacionados con disfunciones cognitivas y de memoria. El daño inducido por alcohol depende de los patrones de consumo de etanol. La exposición prolongada al alcohol induce daño en distintas regiones cerebrales (cortezas prefrontal, perirrinal, entorrinal y parahipocampal, tálamo, hipotálamo, hipocampo y cerebelo) en pacientes alcohólicos y modelos animales de alcoholismo. Sin embargo, no se han estudiado las regiones cerebrales asociadas con el circuito de reforzamiento y recompensa de drogas de abuso.Objetivo: Investigar si la exposición crónica al alcohol induce daño neurodegenerativo en el cerebro de la rata, en particular en el sistema mesocorticolímbico y la amígdala.Método: Ratas Wistar macho fueron expuestas a etanol (10% v/v) o agua por consumo oral durante 30 días y se les privó de la droga por 0, 24 y 48h. Los animales fueron sacrificados y se les extrajo la sangre troncal y el cerebro. Para evaluar el daño neurodegenerativo, se utilizó el marcador fluorescente Fluoro-Jade B. La concentración de alcohol en sangre se determinó por espectrofotometría.Resultados: Se observó un escaso número de células positivas a Fluoro-Jade en las cortezas piriforme y frontal de asociación, el caudado-putamen y el tálamo dorsal. No se encontraron diferencias entre el tratamiento crónico o la privación de alcohol versus el grupo control.Discusión y conclusión: La exposición crónica al alcohol no indujo neurodegeneración en las condiciones utilizadas en este estudio. Probablemente, las concentraciones de alcohol en sangre alcanzadas durante el tratamiento no fueron suficientes para inducir muerte celular.


Background: Chronic alcohol exposure is associated to neurotoxic and neurodegenerative mechanisms that lead to several cognitive and memory dysfunctions. Alcohol-induced damage depends on ethanol consumption patterns. Prolonged alcohol exposure induces damage in distinct brain regions (prefrontal, perirhinal, entorhinal and parahippocampal cortices, thalamus, hypothalamus, hippocampus and cerebellum) in both alcoholic patients and animal models of alcoholism. However, brain areas of the drug reinforcement and reward circuit have not been investigated.Objective: To investigate if chronic alcohol exposure induces neurodegenerative damage in the rat brain, particularly in the mesocorticolimbic system and the amygdala.Method: Male Wistar rats were exposed to ethanol (10% v/v) or water by oral consumption during 30 days. In another set of experiments, animals similarly treated with ethanol were withdrawn from the drug for 24 and 48 h. At the end of the treatments, animals were sacrificed, whole blood samples were obtained and the brains were removed. A fluorescence marker (Fluoro-Jade B) was used to assess neurodegenerative damage in the brain. Blood alcohol concentration was evaluated by spectrophotometry.Results: We observed a low number of Fluoro-Jade B positive cells in different brain regions, including the piriform cortex, frontal cortex of association, caudate-putamen and dorsal thalamus. No differences were found between chronic alcohol or ethanol withdrawn groups versus control animals.Discussion and conclusion: Our results suggest that chronic alcohol exposure does not induce neurodegeneration under the present experimental conditions. Alcohol blood concentrations attained during treatment may not be sufficient to induce cell death.

4.
Apoptosis ; 19(3): 414-27, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24185830

RESUMO

Glucose is the main energy source in brain and it is critical for correct brain functioning. Type 1 diabetic patients might suffer from severe hypoglycemia if exceeding insulin administration, which can lead to acute brain injury if not opportunely corrected. The mechanisms leading to hypoglycemic brain damage are not completely understood and the role of endoplasmic reticulum (ER) stress has not been studied. ER stress resulting from the accumulation of unfolded or misfolded proteins in the ER is counteracted by the unfolded protein response (UPR). When the UPR is sustained, apoptotic death might take place. We have examined UPR activation during glucose deprivation (GD) in hippocampal cultured neurons and its role in the induction of apoptosis. Activation of the PERK pathway of the UPR was observed, as increased phosphorylation of eIF2α and elevated levels of the transcription factor ATF4, occurred 30 min after GD and the levels of the chaperone protein, GRP78 and the transcription factor CHOP, increased after 2 h of GD. In addition, we observed an early activation of caspase-7 and 12 during GD, while caspase-3 activity increased only transiently during glucose reintroduction. Inhibition of caspase-3/7 and the calcium-dependent protease, calpain, significantly decreased caspase-12 activity. The ER stress inhibitor, salubrinal prevented neuronal death and caspase-12 activity. Results suggest that the PERK pathway of the UPR is involved in GD-induced apoptotic neuronal death through the activation of caspase-12, rather than the mitochondrial-dependent caspase pathway. In addition, we show that calpain and caspase-7 are soon activated after GD and mediate caspase-12 activation and neuronal death.


Assuntos
Calpaína/metabolismo , Caspase 12/metabolismo , Caspase 7/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Glucose/deficiência , eIF-2 Quinase/metabolismo , Animais , Apoptose , Calpaína/genética , Células Cultivadas , Chaperona BiP do Retículo Endoplasmático , Ativação Enzimática , Glucose/metabolismo , Humanos , Técnicas In Vitro , Neurônios/citologia , Neurônios/metabolismo , Ratos Wistar , Transdução de Sinais , Resposta a Proteínas não Dobradas
5.
Neuroendocrinology ; 96(1): 41-50, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22213775

RESUMO

Studies have reported the protective effect of estradiol (E(2)) against neuronal death induced by several insults including oxygen deprivation, mitochondrial toxins and activation of glutamate receptors. Glucose deprivation (GD) is associated with ischemia and hypoglycemia, and to date there is no effective therapeutic agent able to prevent neuronal damage induced by these conditions. In this study, we have investigated the effects of 17ß-E(2) and the selective agonists of the alpha (ERα) and beta (ERß) estrogen receptors, propyl pyrazole triol (PPT) and diarylpropionitrile (DPN), respectively, on neuronal death induced by GD in cultured rat hippocampal neurons. We have also analyzed the expression of both ER isoforms after GD. Results show that GD for 2 and 4 h reduces cell survival by 42 and 55%, respectively. Treatment with 17ß-E(2) (10 nM to 10 µM) induces a dose-dependent protective effect that is blocked by ICI 182,780, an ER antagonist, and by 1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(-piperidinylethoxy)phenol]-1H'pyrazole dihydrochloride (MPP) and 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl]phenol (PHTPP), selective ERα and ERß antagonists, respectively. The ERα and ERß agonists PPT and DPN show a similar neuroprotective effect to that of 17ß-E(2), but DPN is more efficient. In addition, hippocampal neurons under normal conditions show a higher expression of the ERß isoform. When exposed to GD during 4 h, the expression of both ER isoforms is increased, while only that of the ERß isoform significantly increases after 2 h of GD. Results demonstrate that E(2) prevents neuronal death induced by GD through its interaction with ER, although the ERß isoform might have a predominant role. Results also suggest that GD differentially alters the expression of ERα and ERß in hippocampal neurons.


Assuntos
Estradiol/farmacologia , Glucose/metabolismo , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Hipocampo/citologia , Hipocampo/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Nitrilas/farmacologia , Fenóis , Propionatos/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Wistar
6.
Neurochem Res ; 33(8): 1599-609, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18461448

RESUMO

Glycogen synthase kinase GSK-3beta has been identified as one of the major candidates mediating tau hyperphosphorylation at the same sites as those present in tau protein in brain from Alzheimer's disease (AD) patients. However, the signal transduction pathways involved in the abnormal activation of GSK-3beta, have not been completely elucidated. GSK-3beta activity is repressed by the canonical Wnt signaling pathway, but it is also modulated through the PI3K/Akt route. Recent studies have suggested that Wnt signaling might be involved in the pathophysiology of AD. On the other hand, modulators of the PI3K pathway might be reduced during aging leading to a sustained activation of GSK-3beta, which in turn would increase the risk of tau hyperphosphorylation. The role of Wnt and PI3K signaling inhibition on the extent of tau phosphorylation and neuronal morphology has not been completely elucidated. Thus, in the present investigation we analyzed the effects of different negative modulators of the Wnt and the PI3K pathways on GSK-3beta activation and phosphorylation of tau at the PHF-1 epitope in cortical cultured neurons and hippocampal slices from adult rat brain. Changes in the microtubule network were also studied. We found that a variety of Wnt and PI3K inhibitors, significantly increased tau phosphorylation at the PHF-1 site, induced the disarrangement of the microtubule network and the accumulation of tau within cell bodies. These changes correlated with alterations in neuronal morphology.


Assuntos
Córtex Cerebral/citologia , Quinase 3 da Glicogênio Sintase/metabolismo , Neurônios , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/fisiologia , Proteínas Wnt/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Androstadienos/metabolismo , Animais , Forma Celular , Ativação Enzimática , Glicogênio Sintase Quinase 3 beta , Glicoproteínas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Proteínas de Membrana/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/metabolismo , Ratos , Ratos Wistar , Receptores de Superfície Celular/metabolismo , Wortmanina , Proteínas tau/genética
7.
Exp Neurol ; 211(1): 85-96, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18339375

RESUMO

Ketone bodies play a key role in mammalian energy metabolism during the suckling period. Normally ketone bodies' blood concentration during adulthood is very low, although it can rise during starvation, an exogenous infusion or a ketogenic diet. Whenever ketone bodies' levels increase, their oxidation in the brain rises. For this reason they have been used as protective molecules against refractory epilepsy and in experimental models of ischemia and excitotoxicity. The mechanisms underlying the protective effect of these compounds are not completely understood. Here, we studied a possible antioxidant capacity of ketone bodies and whether it contributes to the protection against oxidative damage induced during hypoglycemia. We report for the first time the scavenging capacity of the ketone bodies, acetoacetate (AcAc) and both the physiological and non-physiological isomers of beta-hydroxybutyrate (D- and L-BHB, respectively), for diverse reactive oxygen species (ROS). Hydroxyl radicals (.OH) were effectively scavenged by D- and L-BHB. In addition, the three ketone bodies were able to reduce cell death and ROS production induced by the glycolysis inhibitor, iodoacetate (IOA), while only D-BHB and AcAc prevented neuronal ATP decline. Finally, in an in vivo model of insulin-induced hypoglycemia, the administration of D- or L-BHB, but not of AcAc, was able to prevent the hypoglycemia-induced increase in lipid peroxidation in the rat hippocampus. Our data suggest that the antioxidant capacity contributes to protection of ketone bodies against oxidative damage in in vitro and in vivo models associated with free radical production and energy impairment.


Assuntos
Antioxidantes/farmacologia , Hipoglicemia/patologia , Hipoglicemia/prevenção & controle , Corpos Cetônicos/farmacologia , Neurônios/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Alquilantes/toxicidade , Análise de Variância , Animais , Contagem de Células , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Hipocampo/citologia , Hipoglicemia/induzido quimicamente , Insulina , Iodoacetatos/toxicidade , Corpos Cetônicos/metabolismo , Masculino , Neurônios/metabolismo , Oxirredução/efeitos dos fármacos , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
8.
J Neurosci Res ; 86(8): 1768-80, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18293416

RESUMO

Neuronal damage associated with in vivo hypoglycemia has been suggested to be excitotoxic due to the release of excitatory amino acids and the protective effect of glutamate receptor antagonists. The production of reactive oxygen species (ROS) has been also implicated in hypoglycemic damage. Excitotoxicity involves oxidative stress, insofar as the influx of calcium through N-methyl-D-aspartate (NMDA) receptors stimulates ROS production. We have studied the participation of NMDA receptors and intracellular calcium in ROS production and cell death triggered during moderate and severe glycolysis inhibition in cultured hippocampal neurons. Iodoacetate (IOA), an inhibitor of the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH), dose dependently reduces ATP levels and cell survival and increases the intracellular concentration of calcium. During mild glycolysis inhibition, the increases in intracellular calcium, ROS production, and cell death are dependent on NMDA receptor activation. In contrast, during severe glycolysis impairment, these processes are not inhibited by NMDA receptor blockade. BAPTA-AM and vitamin E efficiently reduce ROS generation and cell death under both conditions. Results suggest that calcium influx through NMDA receptors is involved in ROS production and neuronal damage resulting from moderate energy depletion, whereas intracellular calcium increase and ROS generation during severe glycolysis inhibition are more related to energy depletion.


Assuntos
Cálcio/metabolismo , Glicólise/fisiologia , Hipocampo/metabolismo , Neurônios/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Glicólise/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Iodoacetatos/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ratos , Ratos Wistar
9.
J Neurosci Res ; 82(2): 196-205, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16175570

RESUMO

Disturbances in neuronal calcium homeostasis have been implicated in a variety of neuropathological conditions, including cerebral ischemia, hypoglycemia, and epilepsy, and possibly constitute part of the cell death process associated with chronic neurodegenerative disorders. We investigated if endoplasmic reticulum (ER) calcium stores participate in neuronal death triggered by moderate glycolysis inhibition induced by iodoacetate, an inhibitor of glyceraldehyde-3-phosphate dehydrogenase, in cultured hippocampal neurons. Results show that exposure to iodoacetate leads to a slow partial decrease in cell survival, which is significantly prevented in the absence of Ca(2+) or in the presence of the calcium chelator BAPTA-AM. Treatment with caffeine and a low (1 microM) concentration of ryanodine, which activates the ryanodine receptor (RyR), exacerbates neuronal death, whereas dantrolene and 25 microM ryanodine, which antagonizes RyR, prevents damage. Xestospongin C (XeC), an antagonist of the inositol-3-phosphate (IP(3)) receptor (IP(3)R) also prevents neuronal damage. Inhibitors of the ER calcium ATPase (sarcoendoplasmic reticulum Ca(2+) ATPase; SERCA) have no effect. The decrease in ATP levels induced by iodoacetate is potentiated by caffeine and prevented by dantrolene. Although only a slight increase in glutamate extracellular levels is observed 3.5 hr after iodoacetate exposure, the N-methyl-D-aspartate (NMDA) glutamate receptor antagonist, MK-801, efficiently prevents neuronal damage. Taken together, the data suggest that neuronal death induced during moderate glycolysis inhibition involves calcium influx through NMDA receptors and calcium release from intracellular ER stores. These results might be relevant to the understanding the mechanisms involved in neuronal damage related to aging and chronic neurodegenerative diseases, which have been associated with decreased glucose metabolism.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Glicólise/fisiologia , Degeneração Neural/metabolismo , Neurônios/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Canais de Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Quelantes/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Retículo Endoplasmático/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Gliceraldeído 3-Fosfato Desidrogenase (NADP+)/antagonistas & inibidores , Gliceraldeído 3-Fosfato Desidrogenase (NADP+)/metabolismo , Glicólise/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Receptores de Inositol 1,4,5-Trifosfato , Iodoacetatos/farmacologia , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/fisiopatologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ratos , Ratos Wistar , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...