Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Purinergic Signal ; 17(2): 247-254, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33548045

RESUMEN

6-Hydroxydopamine (6-OHDA) is the most used toxin in experimental Parkinson's disease (PD) models. 6-OHDA shows high affinity for the dopamine transporter and once inside the neuron, it accumulates and undergoes non-enzymatic auto-oxidation, promoting reactive oxygen species (ROS) formation and selective damage of catecholaminergic neurons. In this way, our group has established a 6-OHDA in vitro protocol with rat striatal slices as a rapid and effective model for screening of new drugs with protective effects against PD. We have shown that co-incubation with guanosine (GUO, 100 µM) prevented the 6-OHDA-induced damage in striatal slices. As the exact GUO mechanism of action remains unknown, the aim of this study was to investigate if adenosine A1 (A1R) and/or A2A receptors (A2AR) are involved on GUO protective effects on striatal slices. Pre-incubation with DPCPX, an A1R antagonist prevented guanosine effects on 6-OHDA-induced ROS formation and mitochondrial membrane potential depolarization, while CCPA, an A1R agonist, did not alter GUO effects. Regarding A2AR, the antagonist SCH58261 had similar protective effect as GUO in ROS formation and mitochondrial membrane potential. Additionally, SCH58261 did not affect GUO protective effects. The A2AR agonist CGS21680, although, completely blocked GUO effects. Finally, the A1R antagonist DPCPX, and the A2AR agonist CGS21680 also abolished the preventive guanosine effect on 6-OHDA-induced ATP levels decrease. These results reinforce previous evidence for a putative interaction of GUO with A1R-A2AR heteromer as its molecular target and clearly indicate a dependence on adenosine receptors modulation to GUO protective effect.


Asunto(s)
Guanosina/farmacología , Enfermedades Mitocondriales/prevención & control , Neostriado/metabolismo , Fármacos Neuroprotectores/farmacología , Oxidopamina/toxicidad , Receptor de Adenosina A1/efectos de los fármacos , Receptor de Adenosina A2A/efectos de los fármacos , Estallido Respiratorio/efectos de los fármacos , Antagonistas del Receptor de Adenosina A1/farmacología , Animales , Evaluación Preclínica de Medicamentos , Técnicas In Vitro , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Neostriado/efectos de los fármacos , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Xantinas/uso terapéutico
3.
Purinergic Signal ; 16(3): 379-387, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32725400

RESUMEN

Parkinson's disease (PD) signs and symptoms regularly include tremor. Interestingly, the nucleoside guanosine (GUO) has already proven to be effective in reducing reserpine-induced tremulous jaw movements (TJMs) in rodent models, thus becoming a promising antiparkinsonian drug. Here, we aimed at revealing the mechanism behind GUO antiparkinsonian efficacy by assessing the role of adenosine A1 and A2A receptors (A1R and A2AR) on GUO-mediated anti-tremor effects in the reserpinized mouse model of PD. Reserpinized mice showed elevated reactive oxygen species (ROS) production and cellular membrane damage in striatal slices assessed ex vivo and GUO treatment reversed ROS production. Interestingly, while the simultaneous administration of sub-effective doses of GUO (5 mg/kg) and SCH58261 (0.01 mg/kg), an A2AR antagonist, precluded reserpine-induced TJMs, these were ineffective on reverting ROS production in ex vivo experiments. Importantly, GUO was able to reduce TJM and ROS production in reserpinized mouse lacking the A2AR, thus suggesting an A2AR-independent mechanism of GUO-mediated effects. Conversely, the administration of DPCPX (0.75 mg/kg), an A1R antagonist, completely abolished both GUO-mediated anti-tremor effects and blockade of ROS production. Overall, these results indicated that GUO anti-tremor and antioxidant effects in reserpinized mice were A1R dependent but A2AR independent, thus suggesting a differential participation of adenosine receptors in GUO-mediated effects.


Asunto(s)
Guanosina/uso terapéutico , Enfermedad de Parkinson Secundaria/metabolismo , Receptor de Adenosina A1/metabolismo , Receptor de Adenosina A2A/metabolismo , Temblor/metabolismo , Antagonistas del Receptor de Adenosina A1/farmacología , Antagonistas del Receptor de Adenosina A2 , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Guanosina/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratones , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Temblor/inducido químicamente , Temblor/tratamiento farmacológico , Xantinas/farmacología
4.
Neurochem Res ; 41(8): 2017-28, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27084771

RESUMEN

Statins have been shown to promote neuroprotection in a wide range of neurological disorders. However, the mechanisms involved in such effects of statins are not fully understood. Quinolinic acid (QA) is a neurotoxin that induces seizures when infused in vivo and promotes glutamatergic excitotoxicity in the central nervous system. The aim of this study was to evaluate the putative glutamatergic mechanisms and the intracellular signaling pathways involved in the atorvastatin neuroprotective effects against QA toxicity. Atorvastatin (10 mg/kg) treatment for 7 days prevented the QA-induced decrease in glutamate uptake, but had no effect on increased glutamate release induced by QA. Moreover, atorvastatin treatment increased the phosphorylation of ERK1 and prevented the decrease in Akt phosphorylation induced by QA. Neither atorvastatin treatment nor QA infusion altered glutamine synthetase activity or the levels of phosphorylation of p38(MAPK) or JNK1/2 during the evaluation. Inhibition of MEK/ERK signaling pathway, but not PI3K/Akt signaling, abolished the neuroprotective effect of atorvastatin against QA-induced decrease in glutamate uptake. Our data suggest that atorvastatin protective effects against QA toxicity are related to modulation of glutamate transporters via MAPK/ERK signaling pathway.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Atorvastatina/farmacología , Ácido Glutámico/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ácido Quinolínico/toxicidad , Animales , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones
5.
Neurotox Res ; 27(2): 118-28, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25367806

RESUMEN

The search for new therapeutic strategies through modulation of glutamatergic transmission using effective neuroprotective agents is essential. Glutamatergic excitotoxicity is a major factor common to neurodegenerative diseases and in acute events such as cerebral ischemia, traumatic brain injury and epilepsy. We have previously demonstrated that N-methyl-D-aspartate (NMDA) preconditioning in mice showed 50 % of protection against seizures and full protection against damage to neuronal tissue induced by quinolinic acid (QA). In this study, cellular and molecular mechanisms involved on NMDA preconditioning and neuroprotection were investigated in mice treated with NMDA 24 h before QA insult. Calcium uptake and D-aspartate release from hippocampal slices obtained from mice treated with NMDA plus QA and not displaying seizures (protected mice) were similar to control (saline) or NMDA preconditioned mice. Increased calcium uptake and glutamate release is evidenced in unprotected (convulsed) mice as well as QA control, demonstrating that calcium and glutamate are involved in NMDA-induced preconditioning. Increased glutamate release evoked by QA was blocked by MK-801, whereas increased calcium uptake was abolished by voltage-dependent calcium channels inhibitors, but not MK-801. NMDA preconditioning is effective in normalizing the deregulation of glutamate transport and calcium homeostasis evoked by QA due to aberrant NMDA receptors activation that culminates in seizures and hippocampal cells damage.


Asunto(s)
Calcio/metabolismo , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/metabolismo , Hipocampo/efectos de los fármacos , Homeostasis/efectos de los fármacos , N-Metilaspartato/farmacología , Animales , Ácido D-Aspártico/metabolismo , Agonistas de Aminoácidos Excitadores/administración & dosificación , Hipocampo/metabolismo , Masculino , Ratones , N-Metilaspartato/administración & dosificación , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Ácido Quinolínico/administración & dosificación , Convulsiones/inducido químicamente , Convulsiones/metabolismo
6.
Neuroscience ; 215: 79-89, 2012 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-22537845

RESUMEN

The cellular prion protein (PrP(C)) is a neuronal-anchored glycoprotein that has been associated with several functions in the CNS such as synaptic plasticity, learning and memory and neuroprotection. There is great interest in understanding the role of PrP(C) in the deleterious effects induced by the central accumulation of amyloid-ß (Aß) peptides, a pathological hallmark of Alzheimer's disease, but the existent results are still controversial. Here we compared the effects of a single intracerebroventricular (i.c.v.) injection of aggregated Aß(1-40) peptide (400pmol/mouse) on the spatial learning and memory performance as well as hippocampal cell death biomarkers in adult wild type (Prnp(+/+)), PrP(C) knockout (Prnp(0/0)) and the PrP(C) overexpressing Tg-20 mice. Tg-20 mice, which present a fivefold increase in PrP(C) expression in comparison to wild type mice, were resistant to the Aß(1-40)-induced spatial learning and memory impairments as indicated by reduced escape latencies to find the platform and higher percentage of time spent in the correct quadrant during training and probe test sessions of the water maze task. The protection against Aß(1-40)-induced cognitive impairments observed in Tg-20 mice was accompanied by a significant decrease in the hippocampal expression of the activated caspase-3 protein and Bax/Bcl-2 ratio as well as reduced hippocampal cell damage assessed by MTT and propidium iodide incorporation assays. These findings indicate that the overexpression of PrP(C) prevents Aß(1-40)-induced spatial learning and memory deficits in mice and that this response is mediated, at least in part, by the modulation of programed cell death pathways.


Asunto(s)
Péptidos beta-Amiloides/administración & dosificación , Apoptosis/efectos de los fármacos , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/patología , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Priones/metabolismo , Análisis de Varianza , Animales , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/patología , Técnicas In Vitro , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Priónicas , Priones/genética , Propidio , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Tiempo de Reacción/efectos de los fármacos , Sales de Tetrazolio , Tiazoles , Proteína X Asociada a bcl-2/metabolismo
7.
Neuroscience ; 192: 631-41, 2011 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-21756976

RESUMEN

The toxicity of amyloid ß (Aß) is highly associated with Alzheimer's disease (AD), which has a high incidence in elderly people worldwide. While the current treatment for moderate and severe AD includes blockage of the N-methyl-d-aspartate receptor (NMDAR), the molecular mechanisms of its effect are still poorly understood. Herein, we report that a single i.p. administration of the selective and competitive (NMDAR) antagonist LY235959 reduced Aß neurotoxicity by preventing the down-regulation of glial glutamate transporters (glutamate-aspartate transporter (GLAST) and glutamate transporter-1 (GLT-1)), the decrease in glutamate uptake, and the production of reactive oxygen species (ROS) induced by Aß(1-40). Importantly, the blockage of NMDAR restored the Aß(1-40)-induced synaptic dysfunction and cognitive impairment. However, LY235959 failed to prevent the inflammatory response associated with Aß(1-40) treatment. Altogether, our data indicate that the acute administration of Aß promotes oxidative stress, a decrease in glutamate transporter expression, and neurotoxicity. Our results reinforce the idea that NMDAR plays a critical regulatory action in Aß toxicity and they provide further pre-clinical evidence for the potential role of the selective and competitive NMDAR antagonists in the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/toxicidad , Antagonistas de Aminoácidos Excitadores/farmacología , Isoquinolinas/farmacología , Sistema de Transporte de Aminoácidos X-AG/efectos de los fármacos , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Masculino , Ratones , Especies Reactivas de Oxígeno , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Sinapsis/patología , Sinaptofisina/biosíntesis
8.
Neuroscience ; 190: 346-53, 2011 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-21640799

RESUMEN

Folic acid (folate) is a vitamin of the B-complex group that is essential for cell replication. Folate is a major determinant of one-carbon metabolism, in which S-adenosylmethionine donates methyl groups that are crucial for neurological function. Many roles for folic acid have been reported, including neuroprotective and antidepressant properties. On the other hand, increased concentrations of corticoids have proven neurotoxic effects and hypersecretion of glucocorticoids has been linked to different mood disorders. The purpose of this study was to investigate the potential protective effect of folic acid on dexamethasone-induced cellular death in SH-SY5Y neuroblastoma cell line and the possible intracellular signaling pathway involved in such effect. Exposure to 1 mM dexamethasone for 48 h caused a significant reduction of cell viability measured as 3-[4,5 dimethylthiazol-2-yl]-2,5-diphenyl-tetrazolium bromide (MTT) reduction. Exposure of SH-SY5Y cells for 72 h to increasing concentrations of folate (1-300 µM) was not cytotoxic. However, pretreatment with folate (10-300 µM) reduced dexamethasone-induced toxicity in a significant manner. To explore the putative intracellular signaling pathways implicated in the protective effect of folate we used different protein kinase inhibitors. The protective effect of folic acid on dexamethasone-induced neurotoxicity was reversed by the phosphatidylinositol-3 kinase/Akt (PI3K/Akt, LY294002), Ca²âº/Calmodulin-dependent protein kinase II (CaMKII, KN-93), and protein kinase A (PKA, H-89) inhibitors, but not the mitogen-activated protein/extracellular signal-regulated kinase (MEK1/2, PD98059) and protein kinase C (PKC, chelerythrine) inhibitors. In conclusion, the results of this study show that folic acid can protect against dexamethasone-induced neurotoxicity and its protective mechanism is related to a signaling pathway that involves PI3K/Akt, CaMKII, and PKA.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Dexametasona/toxicidad , Ácido Fólico/farmacología , Glucocorticoides/toxicidad , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuronas/metabolismo , Neuronas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos
9.
Neuroscience ; 183: 212-20, 2011 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-21435378

RESUMEN

Guanine derivatives (GD) have been implicated in many relevant brain extracellular roles, such as modulation of glutamate transmission and neuronal protection against excitotoxic damage. GD are spontaneously released to the extracellular space from cultured astrocytes and during oxygen/glucose deprivation (OGD). The aim of this study has been to evaluate the potassium channels and phosphatidilinositol-3 kinase (PI3K) pathway involvement in the mechanisms related to the neuroprotective role of guanosine in rat hippocampal slices subjected to OGD. The addition of guanosine (100 µM) to hippocampal slices subjected to 15 min of OGD and followed by 2 h of re-oxygenation is neuroprotective. The presence of K+ channel blockers, glibenclamide (20 µM) or apamin (300 nM), revealed that neuroprotective effect of guanosine was not dependent on ATP-sensitive K+ channels or small conductance Ca²+-activated K+ channels. The presence of charybdotoxin (100 nM), a large conductance Ca²+-activated K+ channel (BK) blocker, inhibited the neuroprotective effect of guanosine. Hippocampal slices subjected to OGD and re-oxygenation showed a significant reduction of glutamate uptake. Addition of guanosine in the re-oxygenation period has blocked the reduction of glutamate uptake. This guanosine effect was inhibited when hippocampal slices were pre-incubated with charybdotoxin or wortmanin (a PI3K inhibitor, 1 µM) in the re-oxygenation period. Guanosine promoted an increase in Akt protein phosphorylation. However, the presence of charybdotoxin blocked such effect. In conclusion, the neuroprotective effect of guanosine involves augmentation of glutamate uptake, which is modulated by BK channels and the activation of PI3K pathway. Moreover, neuroprotection caused by guanosine depends on the increased expression of phospho-Akt protein.


Asunto(s)
Guanosina/farmacología , Hipocampo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Interacciones Farmacológicas , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glucosa/deficiencia , Ácido Glutámico/metabolismo , Hipocampo/enzimología , Hipoxia/tratamiento farmacológico , Técnicas In Vitro , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Estadísticas no Paramétricas , Tritio/metabolismo
10.
Exp Neurol ; 172(1): 250-4, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11681858

RESUMEN

The exact mechanisms by which 3-nitropropionic acid (3-NP), a naturally occurring plant and fungal neurotoxin, exerts its neurotoxic effects are not fully understood. However, blockage of ATP synthesis by the irreversible inhibition of succinate dehydrogenase activity, increased production of free radicals, and secondary excitotoxicity have been implicated in its actions. In the present study, synaptic vesicle preparations from brain of adult rats were incubated with 3-NP at final concentrations ranging from 0.01 to 10 mM for the determination of glutamate uptake. The effect of 3-NP on gamma-aminobutyric acid (GABA) and glycine uptake was also studied. Glutamate incorporation into vesicles was inhibited by 3-NP in a dose-dependent manner, whereas doses of up to 10 mM neurotoxin did not affect GABA or glycine uptake. Moreover, 3-NP did not inhibit the ATPase activity of synaptic vesicles. These findings indicate that low concentrations of 3-NP are able to selectively prevent vesicular glutamate storage, and this may represent at least one of the mechanisms responsible for the neurotoxic effects of 3-NP.


Asunto(s)
Química Encefálica , Ácido Glutámico/farmacocinética , Propionatos/farmacología , Vesículas Sinápticas/efectos de los fármacos , Vesículas Sinápticas/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Glicina/farmacocinética , Masculino , Neurotoxinas/farmacología , Nitrocompuestos , Ratas , Ratas Wistar , Vesículas Sinápticas/química , Ácido gamma-Aminobutírico/farmacocinética
11.
J Neurol Sci ; 181(1-2): 44-9, 2000 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11099711

RESUMEN

Maple syrup urine disease is an inherited metabolic disorder characterized by tissue accumulation of branched-chain amino acids and their corresponding keto acids in the affected children. Although this disorder is predominantly characterized by neurological symptoms, only few studies were carried out to investigate its neuropathology. In this study we investigated the effect of the metabolites accumulating in maple syrup urine disease on the in vitro uptake of [3H]glutamate by synaptic vesicles of rat brain. Synaptic vesicle preparations from whole brain of male adult Wistar rats (200-250 g) were incubated with the branched-chain amino acids and their corresponding keto acids at final concentrations ranging from 0.25 to 10 mM for the determination of glutamate uptake. Glutamate uptake was significantly inhibited by L-leucine, L-isoleucine, L-2-ketoisocaproic acid and L-2-keto-3-methylvaleric acid by approximately 60%, whereas L-valine and L-2-ketoisovaleric acid showed no effect. We also verified that the metabolites probably act by competitive inhibition. Therefore, it is possible that extracellular glutamate levels may be increased in maple syrup urine disease and that excitotoxicity may be involved in the neuropathology of this disorder.


Asunto(s)
Encéfalo/efectos de los fármacos , Ácido Glutámico/metabolismo , Enfermedad de la Orina de Jarabe de Arce/metabolismo , Neuronas/efectos de los fármacos , Vesículas Sinápticas/efectos de los fármacos , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Hemiterpenos , Isoleucina/metabolismo , Isoleucina/farmacología , Cetoácidos/metabolismo , Cetoácidos/farmacología , Leucina/metabolismo , Leucina/farmacología , Masculino , Enfermedad de la Orina de Jarabe de Arce/complicaciones , Enfermedad de la Orina de Jarabe de Arce/fisiopatología , Neuronas/metabolismo , Ratas , Ratas Wistar , Vesículas Sinápticas/metabolismo , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo , Valina/metabolismo , Valina/farmacología
12.
Neurochem Res ; 25(2): 181-8, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10786700

RESUMEN

Guanine nucleotides (GN) have been implicated in many intracellular mechanisms. Extracellular actions, probably as glutamate receptor antagonists, have also been recently attributed to these compounds. GN may have a neuroprotective role by inhibiting excitotoxic events evoked by glutamate. Effects of extracellular GN on adenosine-evoked cellular responses have also been reported. However, the exact mechanism of such interaction is not known. In the present study, we showed that GN potentiated adenosine-induced cAMP accumulation in slices of hippocampus from young rats. However, neither GMP nor the metabotropic glutamate receptor agonist, 1S,3R-ACPD, inhibited the binding of the adenosine receptor agonist [3H]NECA (when binding to adenosine A2 receptors), or the binding of the adenosine A2a receptor agonist [3H]CGS 21680 in hippocampal membrane preparations. GppNHp, probably by interacting with G-proteins, decreased [3H]CGS 21680 binding. [3H]GMP binding was assayed in order to evaluate the GN sites which are not G-proteins. [3H]GMP binding was inhibited by GMP and GppNHp, but not by IS,3R-ACPD. The interaction of endogenous adenosine with the GMP-binding sites was determined by incubating membranes in the presence or absence of adenosine deaminase (ADA). NECA, CADO, CGS 21680 and CPA (only at the highest concentration used) increased GMP binding in the presence of ADA. However, in the absence of ADA, the control levels of GMP binding were as high as in the presence of added ADA plus adenosine agonists, indicating that endogenous adenosine modulates the binding of GMP. If this site has a neuroprotective role, adenosine may be increasing its neuromodulator and proposed protective action.


Asunto(s)
Adenosina/farmacología , AMP Cíclico/metabolismo , Guanina/farmacología , Guanosina Monofosfato/metabolismo , Hipocampo/efectos de los fármacos , Adenosina/análogos & derivados , Adenosina/metabolismo , Animales , Sitios de Unión , Guanina/análogos & derivados , Guanina/metabolismo , Hipocampo/metabolismo , Agonistas del Receptor Purinérgico P1 , Ensayo de Unión Radioligante , Ratas , Ratas Wistar
13.
Neuroreport ; 11(2): 249-53, 2000 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-10674464

RESUMEN

Quinolinic acid (QA) is an endogenous and potent neurotoxin associated with the neurotoxicity of various common diseases. The uptake of neurotransmitters into synaptic vesicles is an important event involved in the storage and release of neurotransmitters by vesicles. The influence of QA on the uptake of glutamate, GABA and glycine into rat brain synaptic vesicles was investigated. QA (0.3-10 mM) significantly inhibited (>50%) the uptake of glutamate into synaptic vesicles, whereas QA at concentrations up to 10 mM had no significant effect on GABA or glycine uptake. Such results indicate that QA is able to selectively inhibit the vesicular uptake of glutamate, without interfering with the uptake of the inhibitory neurotransmitters GABA and glycine. These findings might be related to the neurotoxic effects of QA in the brain.


Asunto(s)
Encéfalo/metabolismo , Ácido Glutámico/farmacocinética , Ácido Quinolínico/toxicidad , Vesículas Sinápticas/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Glicina/farmacocinética , Masculino , Ratas , Ratas Wistar , Vesículas Sinápticas/efectos de los fármacos , Ácido gamma-Aminobutírico/farmacocinética
14.
Neurochem Res ; 24(8): 1067-74, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10478947

RESUMEN

Binding properties of the subtypes of adenosine A2 receptors in membrane preparations and the effects of adenosine receptor ligands on cAMP accumulation in slices from the optic tectum of neonatal chicks have been investigated. [3H]2-[4-(2-p-carboxyethyl)phenylamino]-5'-N-ethylcarboxaminoadenosin e (CGS 21680), a selective ligand for adenosine A2a receptors, did not bind to optic tectal membranes, as observed with rat striatal membranes. CGS 21680 also did not induce cyclic AMP accumulation in optic tectum slices. However, 5'-N-ethylcarboxamidoadenosine (NECA), 2-chloro-adenosine or adenosine induced a 2.5- to 3-fold increase on cyclic AMP accumulation in this preparation. [3H]NECA binds to fresh non-washed-membranes obtained from optic tectum of chicks, displaying one population of binding sites, which can be displaced by NECA, 8-phenyltheophylline, 2-chloro-adenosine, but is not affected by CGS 21680. The estimated K(D) value was 400.90 +/- 80.50 nM and the Bmax was estimated to be 2.51 +/- 0.54 pmol/mg protein. Guanine nucleotides, which modulate G-proteins activity intracellularly, are also involved in the inhibition of glutamate responses by acting extracellularly. Moreover, we have previously reported that guanine nucleotides potentiate, while glutamate inhibits, adenosine-induced cyclic AMP accumulation in slices from optic tectum of chicks. However, the guanine nucleotides, GMP or GppNHp and the metabotropic glutamate receptors agonist, 1S,3R-ACPD did not alter the [3H]NECA binding observed in fresh non-washed-membranes. Therefore, the adenosine A2 receptor found in the optic tectum must be the adenosine A2b receptor which is available only in fresh membrane preparations, and its not modulated by guanine nucleotides or glutamate analogs.


Asunto(s)
Receptores Purinérgicos P1/metabolismo , Colículos Superiores/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina-5'-(N-etilcarboxamida)/metabolismo , Animales , Membrana Celular/metabolismo , Pollos , AMP Cíclico/metabolismo , Cicloleucina/análogos & derivados , Cicloleucina/farmacología , Técnicas In Vitro , Fenetilaminas/metabolismo , Ensayo de Unión Radioligante , Ratas , Colículos Superiores/efectos de los fármacos
15.
Neuroreport ; 10(9): 1981-3, 1999 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-10501544

RESUMEN

Chick kainate binding protein was solubilized from cerebellar membranes and purified (x19) by use of two chromatographic steps. Measurements of [3H]kainate binding and GTPase activity in the different fractions reveal a consistent decrease of GTPase activity as the purification proceeds so that no GTPase is detectable after the final purification step. This fact, in the context of the differential involvement in nucleotide recognition of some critical amino acid residues in the p-loop motif of GTPases and in the guanine nucleotide-binding sequence of ionotropic glutamate receptors, together with significant discrepancies concerning the activity of individual nucleotides, suggests that both guanine nucleotide-recognizing sequences are unlikely to be alternative expressions of the same functional domain.


Asunto(s)
Cerebelo/enzimología , GTP Fosfohidrolasas/metabolismo , Receptores de Ácido Kaínico/metabolismo , Animales , Unión Competitiva , Cerebelo/química , Pollos , Concanavalina A , Citidina Monofosfato/metabolismo , Etanolaminas , Receptores de Ácido Kaínico/aislamiento & purificación , Sefarosa , Tritio
16.
Neurochem Int ; 34(3): 213-20, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10355488

RESUMEN

Glutamate and adenosine both modulate adenylyl cyclase activity through interaction of their specific receptors with stimulatory or inhibitory G-proteins. Guanine nucleotides (GN), which modulate G-protein activity intracellularly, are also involved in the inhibition of glutamate responses, acting from the outside of the cells. We had previously reported that glutamate inhibits adenosine-induced cyclic AMP (cAMP) accumulation in slices obtained from the optic tectum of chicks. In the present study we investigated the interaction of GN with these two neurotransmitters and found that GN inhibit the inhibitory effect of glutamate on adenosine-induced cAMP accumulation and potentiate adenosine-induced cAMP accumulation. These effects were observed with 5'-guanylylimidodiphosphate (GppNHp) or GMP, but not with guanosine (the nucleoside). Besides, these interactions of GN occur via a metabotropic glutamate receptor (mGluR) sensitive to (1 S,3R)-1-aminocyclopentane-1,3-dicarboxylic acid (1 S,3R-ACPD) but not to L-2-amino-4-phosphonobutyrate (L-AP4). These effects were partially modulated by a mGluR antagonist, (RS)-alpha-methyl-4-carboxyphenylglycine ((RS)M-CPG), and by an adenosine receptor antagonist, 8-phenyltheophylline. GN only potentiated the adenosine response when adenosine was acting through its receptor positively linked to adenylyl cyclase. Therefore, the data show that guanine nucleotides not only inhibit glutamate-induced responses, but also stimulate adenosine-induced responses, a fact that may contribute to the understanding of the physiological functions of guanine nucleotides.


Asunto(s)
Adenosina/fisiología , AMP Cíclico/metabolismo , Glutamatos/fisiología , Nucleótidos de Guanina/fisiología , Colículos Superiores/efectos de los fármacos , Animales , Benzoatos/farmacología , Pollos , Antagonistas de Aminoácidos Excitadores/farmacología , Glicina/análogos & derivados , Glicina/farmacología , Técnicas In Vitro , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/fisiología , Colículos Superiores/metabolismo
17.
Neurochem Res ; 23(2): 183-8, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9475513

RESUMEN

Metabotropic glutamate receptors (mGluRs) have been shown to modulate adenylate cyclase activity via G-proteins. In the present study we report similar results to the previously observed in the literature, showing that glutamate and the metabotropic agonists, 1S,3R-ACPD or quisqualate induced cAMP accumulation in hippocampal slices of young rats. Moreover, guanine nucleotides GTP, GDP or GMP, inhibited the glutamate-induced cAMP accumulation. By measuring LDH activity in the buffer surrounding the slices, we showed that the integrity of the slices was maintained, indicating that the effect of guanine nucleotides was extracellular. GMP, GDPbeta-S or Gpp(NH)p abolished quisqualate-induced cAMP accumulation. GDPbeta-S or Gpp(NH)p but not GMP inhibited 1S,3R-ACPD-induced cAMP accumulation. The response evoked by glutamate was also abolished by the mGluR antagonists: L-AP3 abolished glutamate-induced cAMP accumulation in a dose-dependent manner and MCPG was effective only at the 2 mM dose. DNQX was ineffective. We are reporting here, an inhibition induced by guanine nucleotides, via an extracellular site (s), similar to the observed with classical glutamate antagonists on a cellular response evoked by mGluR agonists.


Asunto(s)
AMP Cíclico/antagonistas & inhibidores , Nucleótidos de Guanina/farmacología , Receptores de Glutamato Metabotrópico/metabolismo , Animales , AMP Cíclico/metabolismo , Cicloleucina/análogos & derivados , Cicloleucina/farmacología , Ácido Glutámico/farmacología , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacología , Guanosina Monofosfato/farmacología , Guanosina Trifosfato/farmacología , Guanilil Imidodifosfato/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Neurotoxinas/farmacología , Ácido Quiscuálico/farmacología , Ratas , Ratas Wistar , Tionucleótidos/farmacología
18.
Int J Dev Neurosci ; 13(6): 545-53, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8553889

RESUMEN

Accumulation of cyclic adenosine monophosphate (cAMP) elicited by adenosine was studied in slices and membrane preparations of optic tectum from chicks aged 1-13 days post-hatch. Accumulation of cAMP promoted by adenosine declined with age, the highest value being observed in three-day-old chicks and the lowest in 11-day-old chicks. However, when the slices were incubated with adenosine and the phosphodiesterase inhibitor-Ro 20-1724 the differences between the two ages were abolished, suggesting a higher phosphodiesterase activity in 11-day-old chicks. In membrane preparations, although basal adenylate cyclase activity was lower in three-day-old chicks, the guanylyl-imidodiphosphate (Gpp(NH)p) concentration curves for stimulation of adenylate cyclase activity indicated a higher sensitivity of G protein to Gpp(NH)p at this age. This hypothesis was reinforced by the observation that the binding of [3H]Gpp(NH)p to the membrane preparation was greater in three-day-old animals. In spite of these differences, the percentage of adenylate cyclase activity stimulation by 2-chloroadenosine (2CADO)+Gpp(NH)p was the same at both ages. These findings suggest that the decreased response evoked by adenosine during development is probably due to increased phosphodiesterase activity and a lower sensitivity of adenylate cyclase activity to Gpp(NH)p.


Asunto(s)
Adenosina/farmacología , AMP Cíclico/biosíntesis , Transducción de Señal/efectos de los fármacos , Colículos Superiores/efectos de los fármacos , 3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Adenilil Ciclasas/metabolismo , Animales , Animales Recién Nacidos , Pollos , Guanilil Imidodifosfato/farmacología , Técnicas In Vitro , Masculino , Membranas/efectos de los fármacos , Membranas/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Ensayo de Unión Radioligante , Colículos Superiores/crecimiento & desarrollo , Colículos Superiores/metabolismo
19.
Neurochem Res ; 20(9): 1033-9, 1995 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8570007

RESUMEN

Changes on cyclic adenosine monophosphate (cAMP) levels in response to adenosine and glutamate and the subtype of glutamate receptors involved in this interaction were studied in slices of optic tectum from 3-day-old chicks. cAMP accumulation mediated by adenosine (100 microM) was abolished by 8-phenyltheophylline (15 microM). Glutamate and the glutamatergic agonists kainate or trans-D, L-1-aminocyclopentane-1,3-dicarboxylic acid (trans-ACPD) did not evoke cAMP accumulation. Glutamate blocked the adenosine response in a dose-dependent manner. At 100 microM, glutamate did not inhibit the effect of adenosine. The 1 mM and 10 mM doses of glutamate inhibited adenosine-induced cAMP accumulation by 55% and 100%, respectively. When glutamatergic antagonists were used, this inhibitory effect was not affected by 200 microM 6,7-dihydroxy-2,3,dinitroquinoxaline (DNQX), an ionotropic antagonist, and was partially antagonized by 1 mM (RS)-alpha-methyl-4-carboxyphenylglycine [(RS)M-CPG], a metabotropic antagonist, while 1 mM L-2-amino-3-phosphonopropionate (L-AP3) alone, another metabotropic antagonist, presented the same inhibitory effect of glutamate. Kainate (10 mM) and trans-ACPD (100 microM and 1 mM) partially blocked the adenosine response. This study indicates the involvement of metabotropic glutamate receptors in adenylate cyclase inhibition induced by glutamate and its agonists trans-ACPD and kainate.


Asunto(s)
Adenosina/farmacología , AMP Cíclico/metabolismo , Ácido Glutámico/farmacología , Receptores de Glutamato Metabotrópico/fisiología , Colículos Superiores/efectos de los fármacos , Alanina/análogos & derivados , Alanina/farmacología , Animales , Benzoatos , Pollos , Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/análogos & derivados , Glicina/análogos & derivados , Técnicas In Vitro , Masculino , Quinoxalinas/farmacología
20.
Neuroreport ; 6(2): 249-52, 1995 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-7756603

RESUMEN

Phosphorylation of the astrocytic marker protein glial fibrillary acidic protein (GFAP) in hippocampal slices from immature rats is stimulated by glutamate agonists via a metabotropic receptor. In this study we investigated the modulation of this stimulation by guanine nucleotides. Recent work has shown that guanine nucleotides inhibit the binding of kainate to its receptors in a manner independent of G proteins. Gpp(NH)p, GDP-beta-S and GMP inhibited by approximately 50% the stimulation of GFAP phosphorylation by glutamate or 1S,3R-ACPD. In the case of glutamate and Gpp(NH)p it was shown that the inhibition was dose dependent. These results indicate that guanine nucleotides can inhibit glutamate-stimulated phosphorylation responses by interaction with a cell surface metabotropic receptor.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Nucleótidos de Guanina/farmacología , Hipocampo/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Hipocampo/metabolismo , Técnicas In Vitro , Datos de Secuencia Molecular , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...