Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.373
Filtrar
1.
Molecules ; 27(9)2022 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-35565989

RESUMEN

The neuroprotective properties of piperine, the major alkaloid extracted from black pepper, have been under investigation, but its mechanism of action in excitotoxicity is still poorly understood. This study aimed to evaluate the protective effects of piperine with a focus on nerve growth factor (NGF) signalling in a kainic acid (KA) rat model of excitotoxicity. Rats were administered intraperitoneally (i.p.) piperine (10 or 50 mg/kg) before KA injection (15 mg/kg, i.p.). Our results show that KA exposure in rats caused seizure behaviour, intrinsic neuronal hyperactivity, glutamate elevation, hippocampal neuronal damage, and cognitive impairment. These KA-induced alterations could be restored to the normal state by piperine treatment. In addition, piperine decreased the expression of the NGF precursor proNGF and NGF-degrading protease matrix metalloproteinase 9, whereas it increased the expression of proNGF processing enzyme matrix metalloproteinase 7, NGF, and NGF-activated receptor TrkA in the hippocampus of KA-treated rats. Furthermore, KA decreased phosphorylation of the protein kinase B (Akt) and glycogen synthase kinase 3ß (GSK3ß) in the hippocampus, and piperine reversed these changes. Our data suggest that piperine protects hippocampal neurons against KA-induced excitotoxicity by upregulating the NGF/TrkA/Akt/GSK3ß signalling pathways.


Asunto(s)
Alcaloides , Fármacos Neuroprotectores , Síndromes de Neurotoxicidad , Alcaloides/metabolismo , Alcaloides/farmacología , Animales , Benzodioxoles , Agonistas de Aminoácidos Excitadores/toxicidad , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hipocampo/metabolismo , Ácido Kaínico/toxicidad , Factor de Crecimiento Nervioso/metabolismo , Neuroprotección , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Síndromes de Neurotoxicidad/metabolismo , Piperidinas , Alcamidas Poliinsaturadas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas
2.
Neural Plast ; 2021: 7174287, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721570

RESUMEN

Epilepsy is the most common childhood neurologic disorder. Status epilepticus (SE), which refers to continuous epileptic seizures, occurs more frequently in children than in adults, and approximately 40-50% of all cases occur in children under 2 years of age. Conventional antiepileptic drugs currently used in clinical practice have a number of adverse side effects. Drug-resistant epilepsy (DRE) can progressively develop in children with persistent SE, necessitating the development of novel therapeutic drugs. During SE, the persistent activation of neurons leads to decreased glutamate clearance with corresponding glutamate accumulation in the synaptic extracellular space, increasing the chance of neuronal excitotoxicity. Our previous study demonstrated that after developmental seizures in rats, E-64d exerts a neuroprotective effect on the seizure-induced brain damage by modulating lipid metabolism enzymes, especially ApoE and ApoJ/clusterin. In this study, we investigated the impact and mechanisms of E-64d administration on neuronal excitotoxicity. To test our hypothesis that E-64d confers neuroprotective effects by regulating autophagy and mitochondrial pathway activity, we simulated neuronal excitotoxicity in vitro using an immortalized hippocampal neuron cell line (HT22). We found that E-64d improved cell viability while reducing oxidative stress and neuronal apoptosis. In addition, E-64d treatment regulated mitochondrial pathway activity and inhibited chaperone-mediated autophagy in HT22 cells. Our findings indicate that E-64d may alleviate glutamate-induced damage via regulation of mitochondrial fission and apoptosis, as well as inhibition of chaperone-mediated autophagy. Thus, E-64d may be a promising therapeutic treatment for hippocampal injury associated with SE.


Asunto(s)
Agonistas de Aminoácidos Excitadores/toxicidad , Ácido Glutámico/toxicidad , Hipocampo/efectos de los fármacos , Leucina/análogos & derivados , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Hipocampo/fisiología , Leucina/farmacología , Ratones , Ratones Endogámicos C57BL , Neuronas/fisiología
3.
Cell Rep ; 37(6): 109960, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34758303

RESUMEN

Tonic inhibition mediated by extrasynaptic GABAARs regulates various brain functions. However, the mechanisms that regulate tonic inhibition remain largely unclear. Here, we report distinct actions of GluN2A- and GluN2B-NMDA receptors (NMDARs) on tonic inhibition in hippocampal neurons under basal and high activity conditions. Specifically, overexpression of GluN2B, but not GluN2A, reduces α5-GABAAR surface expression and tonic currents. Additionally, knockout of GluN2A and GluN2B decreases and increases tonic currents, respectively. Mechanistically, GluN2A-NMDARs inhibit and GluN2B-NMDARs promote α5-GABAAR internalization, resulting in increased and decreased surface α5-GABAAR expression, respectively. Furthermore, GluN2A-NMDARs, but not GluN2B-NMDARs, are required for homeostatic potentiation of tonic inhibition induced by prolonged increase of neuronal activity. Last, tonic inhibition decreases during acute seizures, whereas it increases 24 h later, involving GluN2-NMDAR-dependent signaling. Collectively, these data reveal an NMDAR subunit-specific regulation of tonic inhibition in physiological and pathological conditions and provide mechanistic insight into activity-dependent modulation of tonic inhibition.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Hipocampo/metabolismo , Inhibición Neural , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsiones/patología , Animales , Agonistas de Aminoácidos Excitadores/toxicidad , Ácido Kaínico/toxicidad , Masculino , Ratones , Transporte de Proteínas , Receptores de N-Metil-D-Aspartato/genética , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Transducción de Señal , Sinapsis
4.
Toxins (Basel) ; 13(10)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34678990

RESUMEN

Dolphins are well-regarded sentinels for toxin exposure and can bioaccumulate a cyanotoxin called ß-N-methylamino-l-alanine (BMAA) that has been linked to human neurodegenerative disease. The same dolphins also possessed hallmarks of Alzheimer's disease (AD), suggesting a possible association between toxin exposure and neuropathology. However, the mechanisms of neurodegeneration in dolphins and the impact cyanotoxins have on these processes are unknown. Here, we evaluate BMAA exposure by investigating transcription signatures using PCR for dolphin genes homologous to those implicated in AD and related dementias: APP, PSEN1, PSEN2, MAPT, GRN, TARDBP, and C9orf72. Immunohistochemistry and Sevier Münger silver staining were used to validate neuropathology. Methylmercury (MeHg), a synergistic neurotoxicant with BMAA, was also measured using PT-GC-AFS. We report that dolphins have up to a three-fold increase in gene transcription related to Aß+ plaques, neurofibrillary tangles, neuritic plaques, and TDP-43+ intracytoplasmic inclusions. The upregulation of gene transcription in our dolphin cohort paralleled increasing BMAA concentration. In addition, dolphins with BMAA exposures equivalent to those reported in AD patients displayed up to a 14-fold increase in AD-type neuropathology. MeHg was detected (0.16-0.41 µg/g) and toxicity associated with exposure was also observed in the brain. These results demonstrate that dolphins develop neuropathology associated with AD and exposure to BMAA and MeHg may augment these processes.


Asunto(s)
Aminoácidos Diaminos/toxicidad , Delfín Común , Toxinas de Cianobacterias/toxicidad , Agonistas de Aminoácidos Excitadores/toxicidad , Compuestos de Metilmercurio/toxicidad , Enfermedades Neurodegenerativas/veterinaria , Contaminantes Químicos del Agua/toxicidad , Animales , Femenino , Masculino , Massachusetts , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/patología
5.
J Neuroinflammation ; 18(1): 222, 2021 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-34565380

RESUMEN

BACKGROUND: During inflammatory demyelination, TNF receptor 1 (TNFR1) mediates detrimental proinflammatory effects of soluble TNF (solTNF), whereas TNFR2 mediates beneficial effects of transmembrane TNF (tmTNF) through oligodendroglia, microglia, and possibly other cell types. This model supports the use of selective inhibitors of solTNF/TNFR1 as anti-inflammatory drugs for central nervous system (CNS) diseases. A potential obstacle is the neuroprotective effect of solTNF pretreatment described in cultured neurons, but the relevance in vivo is unknown. METHODS: To address this question, we generated mice with neuron-specific depletion of TNFR1, TNFR2, or inhibitor of NF-κB kinase subunit ß (IKKß), a main downstream mediator of TNFR signaling, and applied experimental models of inflammatory demyelination and acute and preconditioning glutamate excitotoxicity. We also investigated the molecular and cellular requirements of solTNF neuroprotection by generating astrocyte-neuron co-cultures with different combinations of wild-type (WT) and TNF and TNFR knockout cells and measuring N-methyl-D-aspartate (NMDA) excitotoxicity in vitro. RESULTS: Neither neuronal TNFR1 nor TNFR2 protected mice during inflammatory demyelination. In fact, both neuronal TNFR1 and neuronal IKKß promoted microglial responses and tissue injury, and TNFR1 was further required for oligodendrocyte loss and axonal damage in cuprizone-induced demyelination. In contrast, neuronal TNFR2 increased preconditioning protection in a kainic acid (KA) excitotoxicity model in mice and limited hippocampal neuron death. The protective effects of neuronal TNFR2 observed in vivo were further investigated in vitro. As previously described, pretreatment of astrocyte-neuron co-cultures with solTNF (and therefore TNFR1) protected them against NMDA excitotoxicity. However, protection was dependent on astrocyte, not neuronal TNFR1, on astrocyte tmTNF-neuronal TNFR2 interactions, and was reproduced by a TNFR2 agonist. CONCLUSIONS: These results demonstrate that neuronal TNF receptors perform fundamentally different roles in CNS pathology in vivo, with neuronal TNFR1 and IKKß promoting microglial inflammation and neurotoxicity in demyelination, and neuronal TNFR2 mediating neuroprotection in excitotoxicity. They also reveal that previously described neuroprotective effects of solTNF against glutamate excitotoxicity in vitro are indirect and mediated via astrocyte tmTNF-neuron TNFR2 interactions. These results consolidate the concept that selective inhibition of solTNF/TNFR1 with maintenance of TNFR2 function would have combined anti-inflammatory and neuroprotective properties required for safe treatment of CNS diseases.


Asunto(s)
Quinasa I-kappa B/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Animales , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Agonistas de Aminoácidos Excitadores/toxicidad , Femenino , Ácido Kaínico/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/patología , Neuronas/patología , Neuroprotección/fisiología , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Convulsiones/patología
6.
Neurobiol Dis ; 159: 105505, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34520843

RESUMEN

OBJECTIVE: This study aimed to prospectively examine cardiac structure and function in the kainic acid-induced post-status epilepticus (post-KA SE) model of chronic acquired temporal lobe epilepsy (TLE), specifically to examine for changes between the pre-epileptic, early epileptogenesis and the chronic epilepsy stages. We also aimed to examine whether any changes related to the seizure frequency in individual animals. METHODS: Four hours of SE was induced in 9 male Wistar rats at 10 weeks of age, with 8 saline treated matched control rats. Echocardiography was performed prior to the induction of SE, two- and 10-weeks post-SE. Two weeks of continuous video-EEG and simultaneous ECG recordings were acquired for two weeks from 11 weeks post-KA SE. The video-EEG recordings were analyzed blindly to quantify the number and severity of spontaneous seizures, and the ECG recordings analyzed for measures of heart rate variability (HRV). PicroSirius red histology was performed to assess cardiac fibrosis, and intracellular Ca2+ levels and cell contractility were measured by microfluorimetry. RESULTS: All 9 post-KA SE rats were demonstrated to have spontaneous recurrent seizures on the two-week video-EEG recording acquired from 11 weeks SE (seizure frequency ranging from 0.3 to 10.6 seizures/day with the seizure durations from 11 to 62 s), and none of the 8 control rats. Left ventricular wall thickness was thinner, left ventricular internal dimension was shorter, and ejection fraction was significantly decreased in chronically epileptic rats, and was negatively correlated to seizure frequency in individual rats. Diastolic dysfunction was evident in chronically epileptic rats by a decrease in mitral valve deceleration time and an increase in E/E` ratio. Measures of HRV were reduced in the chronically epileptic rats, indicating abnormalities of cardiac autonomic function. Cardiac fibrosis was significantly increased in epileptic rats, positively correlated to seizure frequency, and negatively correlated to ejection fraction. The cardiac fibrosis was not a consequence of direct effect of KA toxicity, as it was not seen in the 6/10 rats from separate cohort that received similar doses of KA but did not go into SE. Cardiomyocyte length, width, volume, and rate of cell lengthening and shortening were significantly reduced in epileptic rats. SIGNIFICANCE: The results from this study demonstrate that chronic epilepsy in the post-KA SE rat model of TLE is associated with a progressive deterioration in cardiac structure and function, with a restrictive cardiomyopathy associated with myocardial fibrosis. Positive correlations between seizure frequency and the severity of the cardiac changes were identified. These results provide new insights into the pathophysiology of cardiac disease in chronic epilepsy, and may have relevance for the heterogeneous mechanisms that place these people at risk of sudden unexplained death.


Asunto(s)
Epilepsia del Lóbulo Temporal/fisiopatología , Válvula Mitral/fisiopatología , Miocardio/patología , Estado Epiléptico/fisiopatología , Disfunción Ventricular/fisiopatología , Remodelación Ventricular/fisiología , Animales , Enfermedad Crónica , Diástole , Modelos Animales de Enfermedad , Ecocardiografía , Electrocardiografía , Electroencefalografía , Epilepsia del Lóbulo Temporal/inducido químicamente , Agonistas de Aminoácidos Excitadores/toxicidad , Fibrosis , Frecuencia Cardíaca/fisiología , Ácido Kaínico/toxicidad , Válvula Mitral/diagnóstico por imagen , Ratas , Estado Epiléptico/inducido químicamente , Muerte Súbita e Inesperada en la Epilepsia , Disfunción Ventricular/diagnóstico por imagen , Disfunción Ventricular/patología , Grabación en Video
7.
Neurobiol Dis ; 157: 105441, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34224862

RESUMEN

Extracellular ATP is a danger signal to the brain and contributes to neurodegeneration in animal models of Alzheimer's disease through its extracellular catabolism by CD73 to generate adenosine, bolstering the activation of adenosine A2A receptors (A2AR). Convulsive activity leads to increased ATP release, with the resulting morphological alterations being eliminated by A2AR blockade. However, it is not known if upon convulsions there is a CD73-mediated coupling between ATP release and A2AR overactivation, causing neurodegeneration. We now show that kainate-induced convulsions trigger a parallel increase of ATP release and of CD73 and A2AR densities in synapses and astrocytes of the mouse hippocampus. Notably, the genetic deletion of CD73 attenuates neuronal degeneration but has no impact on astrocytic modifications in the hippocampus upon kainate-induced convulsions. Furthermore, kainate-induced convulsions cause a parallel deterioration of hippocampal long-term potentiation (LTP) and hippocampal-dependent memory performance, which is eliminated by knocking out CD73. This demonstrates the key role of the ATP release/CD73/A2AR pathway to selectively control synaptic dysfunction and neurodegeneration following an acute brain insult, paving the way to consider CD73 as a new therapeutic target to prevent neuronal damage upon acute brain damage.


Asunto(s)
5'-Nucleotidasa/metabolismo , Adenosina Trifosfato/metabolismo , Astrocitos/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Receptor de Adenosina A2A/metabolismo , Convulsiones/metabolismo , Sinapsis/metabolismo , 5'-Nucleotidasa/genética , Animales , Astrocitos/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/toxicidad , Hipocampo/efectos de los fármacos , Ácido Kaínico/toxicidad , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Memoria/efectos de los fármacos , Memoria/fisiología , Ratones , Ratones Noqueados , Enfermedades Neurodegenerativas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Convulsiones/inducido químicamente , Sinapsis/efectos de los fármacos
8.
Int J Mol Sci ; 22(12)2021 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-34200611

RESUMEN

One of the causes of nervous system degeneration is an excess of glutamate released upon several diseases. Glutamate analogs, like N-methyl-DL-aspartate (NMDA) and kainic acid (KA), have been shown to induce experimental retinal neurotoxicity. Previous results have shown that NMDA/KA neurotoxicity induces significant changes in the full field electroretinogram response, a thinning on the inner retinal layers, and retinal ganglion cell death. However, not all types of retinal neurons experience the same degree of injury in response to the excitotoxic stimulus. The goal of the present work is to address the effect of intraocular injection of different doses of NMDA/KA on the structure and function of several types of retinal cells and their functionality. To globally analyze the effect of glutamate receptor activation in the retina after the intraocular injection of excitotoxic agents, a combination of histological, electrophysiological, and functional tools has been employed to assess the changes in the retinal structure and function. Retinal excitotoxicity caused by the intraocular injection of a mixture of NMDA/KA causes a harmful effect characterized by a great loss of bipolar, amacrine, and retinal ganglion cells, as well as the degeneration of the inner retina. This process leads to a loss of retinal cell functionality characterized by an impairment of light sensitivity and visual acuity, with a strong effect on the retinal OFF pathway. The structural and functional injury suffered by the retina suggests the importance of the glutamate receptors expressed by different types of retinal cells. The effect of glutamate agonists on the OFF pathway represents one of the main findings of the study, as the evaluation of the retinal lesions caused by excitotoxicity could be specifically explored using tests that evaluate the OFF pathway.


Asunto(s)
Células Amacrinas/patología , Agonistas de Aminoácidos Excitadores/toxicidad , Ácido Glutámico/metabolismo , N-Metilaspartato/análogos & derivados , Células Ganglionares de la Retina/patología , Trastornos de la Visión/patología , Células Amacrinas/efectos de los fármacos , Células Amacrinas/metabolismo , Animales , Apoptosis , Ratones , Ratones Endogámicos C57BL , N-Metilaspartato/metabolismo , Receptores de Glutamato/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Trastornos de la Visión/inducido químicamente , Trastornos de la Visión/metabolismo
9.
J Neurosci Res ; 99(10): 2573-2591, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34197000

RESUMEN

Glutamate excitotoxicity and endoplasmic reticulum (ER) recently have been found to be instrumental in the pathogenesis of various neurodegenerative diseases. However, the paucity of literature deciphering the inter-linkage among glutamate receptors, behavioral alterations, and ER demands thorough exploration. Reckoning the aforesaid concerns, a prospective study was outlined to delineate the influence of ER stress inhibition via 4-phenylbutyric acid (PBA) on α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) excitotoxicity-induced behavioral aspects and possible ER stress-glutamate linkage. Male SD rats were randomly divided into four groups namely sham (surgical control+vehicle, group 1), AMPA-induced excitotoxic group 2 receive a single intra-hippocampal injection of 10 mM AMPA, group 3 received AMPA along with PBA (i.p, 100 mg/kg body weight) for 15 days, and group 4 received PBA alone. Behavioral analyses were performed prior to the sacrifice of animals and hippocampus was extracted thereafter for further analysis. AMPA-induced excitotoxicity exhibited significant impairment of locomotion as well as cognitive functions. The levels of neurotransmitters such as dopamine, homo vanillic acid (HVA), norepinephrine, and serotonin were reduced accompanied by reduced expression of GLUR1 and GLUR4 (glutamate receptor) as well as loss of neurons in different layers of hippocampus. ER stress markers were upregulated upon AMPA excitotoxicity. However, chemical chaperone PBA supplementation remarkably mitigated the behavioral alterations along with expression of glutamate and ER stress intermediates/markers in AMPA excitotoxic animals. Therefore, the present exploration convincingly emphasizes the significance of ER stress and its inhibition via PBA in combating cognitive impairment as well as improving locomotion in excitotoxic animals.


Asunto(s)
Butilaminas/farmacología , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/prevención & control , Estrés del Retículo Endoplásmico/fisiología , Agonistas de Aminoácidos Excitadores/toxicidad , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/toxicidad , Animales , Butilaminas/uso terapéutico , Disfunción Cognitiva/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ácido Glutámico/metabolismo , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratas , Ratas Sprague-Dawley
10.
Neurobiol Dis ; 157: 105443, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34246771

RESUMEN

Astrocytic glutamate transporters are crucial for glutamate homeostasis in the brain, and dysregulation of these transporters can contribute to the development of epilepsy. Glutamate transporter-1 (GLT-1) is responsible for the majority of glutamate uptake in the dorsal forebrain and has been shown to be reduced at epileptic foci in patients and preclinical models of temporal lobe epilepsy (TLE). Current antiepileptic drugs (AEDs) work primarily by targeting neurons directly through suppression of excitatory neurotransmission or enhancement of inhibitory neurotransmission, which can lead to both behavioral and psychiatric side effects. This study investigates the therapeutic capacity of astrocyte-specific AAV-mediated GLT-1 expression in the intrahippocampal kainic acid (IHKA) model of TLE. In this study, we used Western blot analysis, immunohistochemistry, and long-term-video EEG monitoring to demonstrate that cell-type-specific upregulation of GLT-1 in astrocytes is neuroprotective at early time points during epileptogenesis, reduces seizure frequency and total time spent in seizures, and eliminates large behavioral seizures in the IHKA model of epilepsy. Our findings suggest that targeting glutamate uptake is a promising therapeutic strategy for the treatment of epilepsy.


Asunto(s)
Astrocitos/metabolismo , Epilepsia del Lóbulo Temporal/genética , Transportador 2 de Aminoácidos Excitadores/genética , Hipocampo/metabolismo , Convulsiones/genética , Animales , Modelos Animales de Enfermedad , Electroencefalografía , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/fisiopatología , Agonistas de Aminoácidos Excitadores/toxicidad , Técnicas de Sustitución del Gen , Ácido Kaínico/toxicidad , Ratones , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Convulsiones/fisiopatología , Regulación hacia Arriba
11.
Neuropharmacology ; 196: 108654, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34119518

RESUMEN

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that regulates redox homeostasis of the cell through regulation of the antioxidant response element genes transcription. Nrf2 also regulates the antiapoptotic Bcl-2 gene. Nrf2 degradation and nuclear translocation is regulated by upstream kinases Akt and GSK3ß. Glutamate excitotoxicity is a process of neuronal cells death due to excessive activation of glutamate receptors. Glutamate excitotoxicity participates in the pathophysiology of several acute and chronic neurological conditions. In addition, glutamate excitotoxicity interrupts the PI3K/Akt prosurvival pathway so GSK3ß remains active. Active GSK3ß increases Nrf2 degradation, decreases Nrf2 nuclear translocation and increases Nrf2 nuclear export which decreases the ARE genes transcription such as, SOD, GSH synthesis enzyme and HO-1. Also, Bcl-2 transcription decreases. Flurbiprofen is a COX inhibitor. Previous studies showed that it has a neuroprotective effect in neurodegeneration and in focal cerebral ischemia/reperfusion model. In our research we aimed to test the hypothesis that flurbiprofen may have a neuroprotective effect in a rat model of glutamate-induced excitotoxicity and this neuroprotection may occur through modulation of (Akt/GSK3ß/Nrf2/HO-1) pathway. Rats were divided into 4 groups; control, MSG (2.5 g/Kg, i.p), low dose FB (5 mg/kg, i.p) and high dose FB (10 mg/kg, i.p). We found that low and high doses FB decreased COX-2, PGE2, NO and MDA and increased SOD and GSH in brain compared to MSG group. High dose was more effective than low dose. Western blotting analysis in hippocampus tissue showed that high dose FB increased p-Akt, p-GSK3ß, nuclear Nrf2 and HO-1 and decreased cytosolic Nrf2 level in comparison with MSG group. Immunohistochemical analysis in hippocampus and cerebral cortex showed that high dose FB increased Bcl-2 and decreased Bax compared to MSG group. In addition, FB increased the number of intact neurons in hippocampus areas and cerebral cortex neurons and showed an anxiolytic-like action in OF and EPM tests. These findings suggest that FB has a neuroprotective effect in glutamate-induced excitotoxicity model through reduction of the glutamate excitotoxicity damage and activation of the survival pathway. These may occur due to modulation the survival pathway (Akt/GSK3ß/Nrf2/HO-1) and inhibition of COX-2.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Apoptosis/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/toxicidad , Flurbiprofeno/farmacología , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Glutamato de Sodio/toxicidad , Animales , Antioxidantes/farmacología , Ansiedad , Modelos Animales de Enfermedad , Prueba de Laberinto Elevado , Ácido Glutámico , Glucógeno Sintasa Quinasa 3 beta , Hemo Oxigenasa (Desciclizante) , Hipocampo/metabolismo , Factor 2 Relacionado con NF-E2 , Neuronas/metabolismo , Prueba de Campo Abierto , Proteínas Proto-Oncogénicas c-akt , Ratas , Transducción de Señal
12.
Biochem Biophys Res Commun ; 558: 175-182, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33932777

RESUMEN

The structure of the brain is dramatically altered during the critical period. Physiological substances (neurotransmitters, hormones, etc.) in the body fluctuate significantly before and after sexual maturation. Therefore, the effect of chemical exposure on the central nervous system often differs depending on the developmental stage and sex. We aimed to compare the behavioural effects that emerged from the administration of chemicals to mice of different life stages (immature or mature) and different sex (male or female). We administered mice with domoic acid (DA), a marine poison, and ibotenic acid (IA), found in poisonous mushrooms. These excitatory amino acids act as agonists for glutamate and are potent neurotoxins. Interestingly, the behavioural effects of these chemicals were completely different. Following DA administration, we observed memory deficits only in groups of male mice treated at maturity. Following IA administration, we observed deviations in emotional behaviour in groups of male mice treated at both immaturity and maturity. In contrast, few characteristic changes were detected in all groups of females. Our results support the theory that the behavioural effects of chemical administration vary considerably with developmental stages and sex. In conclusion, our findings promote better understanding of individual differences in excitatory chemical-induced neurotoxicity and provide evidence for future risk strategies and treatments.


Asunto(s)
Conducta Animal/efectos de los fármacos , Ácido Iboténico/toxicidad , Ácido Kaínico/análogos & derivados , Administración Oral , Animales , Conducta Animal/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiología , Agonistas de Aminoácidos Excitadores/administración & dosificación , Agonistas de Aminoácidos Excitadores/toxicidad , Femenino , Ácido Iboténico/administración & dosificación , Ácido Kaínico/administración & dosificación , Ácido Kaínico/toxicidad , Masculino , Toxinas Marinas/administración & dosificación , Toxinas Marinas/toxicidad , Ratones , Ratones Endogámicos C57BL , Neurotoxinas/administración & dosificación , Neurotoxinas/toxicidad , Factores Sexuales , Maduración Sexual/fisiología
13.
Int J Mol Sci ; 22(9)2021 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-33922757

RESUMEN

Metformin, an anti-hyperglycemic drug of the biguanide class, exerts positive effects in several non-diabetes-related diseases. In this study, we aimed to examine the protective effects of metformin against N-methyl-D-aspartic acid (NMDA)-induced excitotoxic retinal damage in rats and determine the mechanisms of its protective effects. Male Sprague-Dawley rats (7 to 9 weeks old) were used in this study. Following intravitreal injection of NMDA (200 nmol/eye), the number of neuronal cells in the ganglion cell layer and parvalbumin-positive amacrine cells decreased, whereas the number of CD45-positive leukocytes and Iba1-positive microglia increased. Metformin attenuated these NMDA-induced responses. The neuroprotective effect of metformin was abolished by compound C, an inhibitor of AMP-activated protein kinase (AMPK). The AMPK activator, AICAR, exerted a neuroprotective effect in NMDA-induced retinal injury. The MEK1/2 inhibitor, U0126, reduced the neuroprotective effect of metformin. These results suggest that metformin protects against NMDA-induced retinal neurotoxicity through activation of the AMPK and MEK/extracellular signal-regulated kinase (ERK) signaling pathways. This neuroprotective effect could be partially attributable to the inhibitory effects on inflammatory responses.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Metformina/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , N-Metilaspartato/toxicidad , Fármacos Neuroprotectores/farmacología , Enfermedades de la Retina/prevención & control , Animales , Agonistas de Aminoácidos Excitadores/toxicidad , Hipoglucemiantes/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Enfermedades de la Retina/inducido químicamente , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Transducción de Señal
14.
Molecules ; 26(6)2021 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-33805696

RESUMEN

Experimental evidence indicates that the activation of ionotropic glutamate receptors plays an important role in neurological disorders' models such as epilepsy, cerebral ischemia and trauma. The glutamate receptor agonist kainic acid (KA) induces seizures and excitotoxic cell death in the CA3 region of the hippocampus. Thymoquinone (TQ) is the most important component of the essential oil obtained from black cumin (Nigella sativa L.) seeds. It has many pharmacological actions including antioxidant, anti-inflammatory, and anti-apoptotic effects. TQ was used in an in vitro experimental model of primary cultures where excitotoxicity was induced. Briefly, rat organotypic hippocampal slices were exposed to 5 µM KA for 24 h. Cell death in the CA3 subregions of slices was quantified by measuring propidium iodide fluorescence. The cross-talk between TQ, ER stress and apoptotic pathways was investigated by Western blot. In untreated slices TQ (10 µM) induced a significant increase on the PSD95 levels and it decreased the excitotoxic injury induced by KA. Additionally, TQ was able to ameliorate the KA-induced increase in unfolded proteins GRP78 and GRP94 expression. Finally, TQ was able to partially rescue the reduction of the KA-induced apoptotic pathway activation. Our results suggest that TQ modulates the processes leading to post-kainate neuronal death in the CA3 hippocampal area.


Asunto(s)
Benzoquinonas/farmacología , Región CA3 Hipocampal/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Región CA3 Hipocampal/patología , Región CA3 Hipocampal/fisiopatología , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico , Epilepsia/fisiopatología , Agonistas de Aminoácidos Excitadores/toxicidad , Femenino , Técnicas In Vitro , Ácido Kaínico/toxicidad , Masculino , Plasticidad Neuronal/efectos de los fármacos , Ratas , Ratas Wistar
15.
FASEB J ; 35(5): e21568, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33817875

RESUMEN

The neuronal Na+ -activated K+ channel Slack (aka Slo2.2, KNa 1.1, or Kcnt1) has been implicated in setting and maintaining the resting membrane potential and defining excitability and firing patterns, as well as in the generation of the slow afterhyperpolarization following bursts of action potentials. Slack activity increases significantly under conditions of high intracellular Na+ levels, suggesting this channel may exert important pathophysiological functions. To address these putative roles, we studied whether Slack K+ channels contribute to pathological changes and excitotoxic cell death caused by glutamatergic overstimulation of Ca2+ - and Na+ -permeable N-methyl-D-aspartic acid receptors (NMDAR). Slack-deficient (Slack KO) and wild-type (WT) mice were subjected to intrastriatal microinjections of the NMDAR agonist NMDA. NMDA-induced brain lesions were significantly increased in Slack KO vs WT mice, suggesting that the lack of Slack renders neurons particularly susceptible to excitotoxicity. Accordingly, excessive neuronal cell death was seen in Slack-deficient primary cerebellar granule cell (CGC) cultures exposed to glutamate and NMDA. Differences in neuronal survival between WT and Slack KO CGCs were largely abolished by the NMDAR antagonist MK-801, but not by NBQX, a potent and highly selective competitive antagonist of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type ionotropic glutamate receptors. Interestingly, NMDAR-evoked Ca2+ signals did not differ with regard to Slack genotype in CGCs. However, real-time monitoring of K+ following NMDAR activation revealed a significant contribution of this channel to the intracellular drop in K+ . Finally, TrkB and TrkC neurotrophin receptor transcript levels were elevated in NMDA-exposed Slack-proficient CGCs, suggesting a mechanism by which this K+ channel contributes to the activation of the extracellular-signal-regulated kinase (Erk) pathway and thereby to neuroprotection. Combined, our findings suggest that Slack-dependent K+ signals oppose the NMDAR-mediated excitotoxic neuronal injury by promoting pro-survival signaling via the BDNF/TrkB and Erk axis.


Asunto(s)
Potenciales de Acción , Encefalopatías/prevención & control , Muerte Celular , N-Metilaspartato/toxicidad , Proteínas del Tejido Nervioso/fisiología , Neuronas/citología , Canales de potasio activados por Sodio/fisiología , Animales , Encefalopatías/inducido químicamente , Encefalopatías/metabolismo , Encefalopatías/patología , Células Cultivadas , Agonistas de Aminoácidos Excitadores/toxicidad , Ácido Glutámico/metabolismo , Masculino , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Transducción de Señal
16.
Neurobiol Dis ; 153: 105317, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33639207

RESUMEN

Epilepsy is a neurological condition associated to significant brain damage produced by status epilepticus (SE) including neurodegeneration, gliosis and ectopic neurogenesis. Reduction of these processes constitutes a useful strategy to improve recovery and ameliorate negative outcomes after an initial insult. SGK1.1, the neuronal isoform of the serum and glucocorticoids-regulated kinase 1 (SGK1), has been shown to increase M-current density in neurons, leading to reduced excitability and protection against seizures. For this study, we used 4-5 months old male transgenic C57BL/6 J and FVB/NJ mice expressing near physiological levels of a constitutively active form of the kinase controlled by its endogenous promoter. Here we show that SGK1.1 activation potently reduces levels of neuronal death (assessed using Fluoro-Jade C staining) and reactive glial activation (reported by GFAP and Iba-1 markers) in limbic regions and cortex, 72 h after SE induced by kainate, even in the context of high seizure activity. This neuroprotective effect is not exclusively through M-current activation but is also directly linked to decreased apoptosis levels assessed by TUNEL assays and quantification of Bim and Bcl-xL by western blot of hippocampal protein extracts. Our results demonstrate that this newly described antiapoptotic role of SGK1.1 activation acts synergistically with the regulation of cellular excitability, resulting in a significant reduction of SE-induced brain damage in areas relevant to epileptogenesis.


Asunto(s)
Apoptosis/genética , Gliosis/genética , Proteínas Inmediatas-Precoces/genética , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Estado Epiléptico/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Supervivencia Celular , Agonistas de Aminoácidos Excitadores/toxicidad , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/metabolismo , Gliosis/patología , Ácido Kaínico/toxicidad , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Neuroglía/metabolismo , Neuronas/patología , Estado Epiléptico/inducido químicamente , Estado Epiléptico/patología
17.
Int J Mol Sci ; 22(4)2021 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-33562628

RESUMEN

Increased oxidative damage in the brain, which increases with age, is the cause of abnormal brain function and various diseases. Ascorbic acid (AA) is known as an endogenous antioxidant that provides neuronal protection against oxidative damage. However, with aging, its extracellular concentrations and uptake decrease in the brain. Few studies have dealt with age-related functional changes in the brain to sustained ascorbate supplementation. This study aimed to investigate the susceptibility of hippocampal neurons to oxidative injury following acute and chronic AA administration. Oxidative stress was induced by kainic acid (KA, 5 µM) for 18 h in hippocampal slice cultures. After KA exposure, less neuronal cell death was observed in the 3 w cultured slice compared to the 9 w cultured slice. In the chronic AA treatment (6 w), the 9 w-daily group showed reduced neuronal cell death and increased superoxide dismutase (SOD) and Nrf2 expressions compared to the 9 w. In addition, the 9 w group showed delayed latencies and reduced signal activity compared to the 3 w, while the 9 w-daily group showed shorter latencies and increased signal activity than the 9 w. These results suggest that the maintenance of the antioxidant system by chronic AA treatment during aging could preserve redox capacity to protect hippocampal neurons from age-related oxidative stress.


Asunto(s)
Ácido Ascórbico/administración & dosificación , Hipocampo/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Animales , Antioxidantes/administración & dosificación , Agonistas de Aminoácidos Excitadores/toxicidad , Hipocampo/lesiones , Hipocampo/metabolismo , Ácido Kaínico/toxicidad , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuroprotección/efectos de los fármacos , Técnicas de Cultivo de Órganos , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
18.
Neurotoxicology ; 83: 69-76, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33400970

RESUMEN

On the basis of the evidence that extracellular Zn2+ influx induced with AMPA causes Parkinson's syndrome in rats that apomorphine-induced movement disorder emerges, here we used a low dose of AMPA, which does not increase intracellular Zn2+ level in the substantia nigra pars compacta (SNpc) of young adult rats, and tested whether intracellular Zn2+ dysregulation induced with AMPA is accelerated in the SNpc of aged rats, resulting in age-related vulnerability to Parkinson's syndrome. When AMPA (1 mM) was injected at the rate of 0.05 µl/min for 20 min into the SNpc, intracellular Zn2+ level was increased in the SNpc of aged rats followed by increase in turning behavior in response to apomorphine and nigral dopaminergic degeneration. In contrast, young adult rats do not show movement disorder and nigral dopaminergic degeneration, in addition to no increase in intracellular Zn2+. In aged rats, movement disorder and nigral dopaminergic degeneration were rescued by co-injection of either extracellular (CaEDTA) or intracellular (ZnAF-2DA) Zn2+ chelators. 1-Naphthyl acetyl spermine (NASPM), a selective blocker of Ca2+- and Zn2+-permeable GluR2-lacking AMPA receptors blocked increase in intracellular Zn2+ in the SNpc of aged rats followed by rescuing nigral dopaminergic degeneration. The present study indicates that intracellular Zn2+ dysregulation is accelerated by Ca2+- and Zn2+-permeable GluR2-lacking AMPA receptor activation in the SNpc of aged rats, resulting in age-related vulnerability to Parkinson's syndrome.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/toxicidad , Degeneración Nerviosa , Enfermedad de Parkinson Secundaria/inducido químicamente , Porción Compacta de la Sustancia Negra/efectos de los fármacos , Receptores AMPA/agonistas , Zinc/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/toxicidad , Factores de Edad , Animales , Conducta Animal/efectos de los fármacos , Calcio/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Masculino , Actividad Motora/efectos de los fármacos , Enfermedad de Parkinson Secundaria/metabolismo , Enfermedad de Parkinson Secundaria/patología , Enfermedad de Parkinson Secundaria/fisiopatología , Porción Compacta de la Sustancia Negra/metabolismo , Porción Compacta de la Sustancia Negra/patología , Porción Compacta de la Sustancia Negra/fisiopatología , Ratas Wistar , Receptores AMPA/metabolismo
19.
Epilepsia ; 62(1): 163-175, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33258489

RESUMEN

OBJECTIVE: Following surgery, focal seizures relapse in 20% to 50% of cases due to the difficulty of delimiting the epileptogenic zone (EZ) by current imaging or electrophysiological techniques. Here, we evaluate an unbiased metabolomics approach based on ex vivo and in vivo nuclear magnetic resonance spectroscopy (MRS) methods to discriminate the EZ in a mouse model of mesiotemporal lobe epilepsy (MTLE). METHODS: Four weeks after unilateral injection of kainic acid (KA) into the dorsal hippocampus of mice (KA-MTLE model), we analyzed hippocampal and cortical samples with high-resolution magic angle spinning (HRMAS) magnetic resonance spectroscopy (MRS). Using advanced multivariate statistics, we identified the metabolites that best discriminate the injected dorsal hippocampus (EZ) and developed an in vivo MEGAPRESS MRS method to focus on the detection of these metabolites in the same mouse model. RESULTS: Multivariate analysis of HRMAS data provided evidence that γ-aminobutyric acid (GABA) is largely increased in the EZ of KA-MTLE mice and is the metabolite that best discriminates the EZ when compared to sham and, more importantly, when compared to adjacent brain regions. These results were confirmed by capillary electrophoresis analysis and were not reversed by a chronic exposition to an antiepileptic drug (carbamazepine). Then, using in vivo noninvasive GABA-edited MRS, we confirmed that a high GABA increase is specific to the injected hippocampus of KA-MTLE mice. SIGNIFICANCE: Our strategy using ex vivo MRS-based untargeted metabolomics to select the most discriminant metabolite(s), followed by in vivo MRS-based targeted metabolomics, is an unbiased approach to accurately define the EZ in a mouse model of focal epilepsy. Results suggest that GABA is a specific biomarker of the EZ in MTLE.


Asunto(s)
Epilepsia del Lóbulo Temporal/metabolismo , Hipocampo/metabolismo , Metabolómica , Ácido gamma-Aminobutírico/metabolismo , Animales , Anticonvulsivantes/farmacología , Carbamazepina/farmacología , Modelos Animales de Enfermedad , Electroforesis Capilar , Epilepsia del Lóbulo Temporal/inducido químicamente , Agonistas de Aminoácidos Excitadores/toxicidad , Hipocampo/diagnóstico por imagen , Hipocampo/efectos de los fármacos , Hipocampo/patología , Ácido Kaínico/toxicidad , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética/métodos , Masculino , Ratones , Análisis Multivariante , Espectroscopía de Protones por Resonancia Magnética/métodos , Esclerosis , Ácido gamma-Aminobutírico/efectos de los fármacos
20.
Exp Eye Res ; 202: 108342, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33144094

RESUMEN

B-N-methylamino-L-alanine (BMAA), a cyanotoxin produced by most cyanobacteria, has been proposed to cause long term damages leading to neurodegenerative diseases, including Amyotrophic Lateral Sclerosis/Parkinsonism Dementia complex (ALS/PDC) and retinal pathologies. Previous work has shown diverse mechanisms leading to BMAA-induced degeneration; however, the underlying mechanisms of toxicity affecting retina cells are not fully elucidated. We here show that BMAA treatment of rat retina neurons in vitro induced nuclear fragmentation and cell death in both photoreceptors (PHRs) and amacrine neurons, provoking mitochondrial membrane depolarization. Pretreatment with the N-Methyl-D-aspartate (NMDA) receptor antagonist MK-801 prevented BMAA-induced death of amacrine neurons, but not that of PHRs, implying activation of NMDA receptors participated only in amacrine cell death. Noteworthy, BMAA stimulated a selective axonal outgrowth in amacrine neurons, simultaneously promoting growth cone destabilization. BMAA partially decreased the viability of Müller glial cells (MGC), the main glial cell type in the retina, induced marked alterations in their actin cytoskeleton and impaired their capacity to protect retinal neurons. BMAA also induced cell death and promoted axonal outgrowth in differentiated rat pheochromocytoma (PC12) cells, implying these effects were not limited to amacrine neurons. These results suggest that BMAA is toxic for retina neurons and MGC and point to the involvement of NMDA receptors in amacrine cell death, providing new insight into the mechanisms involved in BMAA neurotoxic effects in the retina.


Asunto(s)
Aminoácidos Diaminos/toxicidad , Células Ependimogliales/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/toxicidad , Enfermedades de la Retina/inducido químicamente , Neuronas Retinianas/efectos de los fármacos , Animales , Animales Recién Nacidos , Supervivencia Celular/efectos de los fármacos , Toxinas de Cianobacterias , Fragmentación del ADN/efectos de los fármacos , Maleato de Dizocilpina/farmacología , Células Ependimogliales/patología , Antagonistas de Aminoácidos Excitadores/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/prevención & control , Neuronas Retinianas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...