Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neurosci Lett ; 808: 137292, 2023 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-37156440

RESUMEN

Caffeic acid is a polyphenolic compound present in a vast array of dietary components. We previously showed that caffeic acid reduces the burden of brain ischemia joining evidence by others that it can attenuate different brain diseases. However, it is unknown if caffeic acid affects information processing in neuronal networks. Thus, we now used electrophysiological recordings in mouse hippocampal slices to test if caffeic acid directly affected synaptic transmission, plasticity and dysfunction caused by oxygen-glucose deprivation (OGD), an in vitro ischemia model. Caffeic acid (1-10 µM) was devoid of effect on synaptic transmission and paired-pulse facilitation in Schaffer collaterals-CA1 pyramidal synapses. Also, the magnitude of either hippocampal long-term potentiation (LTP) or the subsequent depotentiation were not significantly modified by 10 µM caffeic acid. However, caffeic acid (10 µM) increased the recovery of synaptic transmission upon re-oxygenation following 7 min of OGD. Furthermore, caffeic acid (10 µM) also recovered plasticity after OGD, as heralded by the increased magnitude of LTP after exposure. These findings show that caffeic acid does not directly affect synaptic transmission and plasticity but can indirectly affect other cellular targets to correct synaptic dysfunction. Unraveling the molecular mechanisms of action of caffeic acid may allow the design of hitherto unrecognized novel neuroprotective strategies.


Asunto(s)
Hipocampo , Transmisión Sináptica , Ratones , Animales , Transmisión Sináptica/fisiología , Potenciación a Largo Plazo/fisiología , Isquemia , Plasticidad Neuronal/fisiología
2.
Int J Mol Sci ; 23(22)2022 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-36430321

RESUMEN

Methylmercury (MeHg) is highly toxic to the human brain. Although much is known about MeHg neurotoxic effects, less is known about how chronic MeHg affects hippocampal amino acids and other neurochemical markers in adult mice. In this study, we evaluated the MeHg effects on systemic lipids and inflammation, hippocampal oxidative stress, amino acid levels, neuroinflammation, and behavior in adult male mice. Challenged mice received MeHg in drinking water (2 mg/L) for 30 days. We assessed weight gain, total plasma cholesterol (TC), triglycerides (TG), endotoxin, and TNF levels. Hippocampal myeloperoxidase (MPO), malondialdehyde (MDA), acetylcholinesterase (AChE), amino acid levels, and cytokine transcripts were evaluated. Mice underwent open field, object recognition, Y, and Barnes maze tests. MeHg-intoxicated mice had higher weight gain and increased the TG and TC plasma levels. Elevated circulating TNF and LPS confirmed systemic inflammation. Higher levels of MPO and MDA and a reduction in IL-4 transcripts were found in the hippocampus. MeHg-intoxication led to increased GABA and glycine, reduced hippocampal taurine levels, delayed acquisition in the Barnes maze, and poor locomotor activity. No significant changes were found in AChE activity and object recognition. Altogether, our findings highlight chronic MeHg-induced effects that may have long-term mental health consequences in prolonged exposed human populations.


Asunto(s)
Compuestos de Metilmercurio , Animales , Humanos , Masculino , Ratones , Acetilcolinesterasa/metabolismo , Aminoácidos , Hipocampo/metabolismo , Inflamación/inducido químicamente , Compuestos de Metilmercurio/toxicidad , Compuestos de Metilmercurio/metabolismo , Aumento de Peso , Ratones Endogámicos C57BL
3.
Sci Rep ; 11(1): 10488, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34006978

RESUMEN

The increased healthspan afforded by coffee intake provides novel opportunities to identify new therapeutic strategies. Caffeine has been proposed to afford benefits through adenosine A2A receptors, which can control synaptic dysfunction underlying some brain disease. However, decaffeinated coffee and other main components of coffee such as chlorogenic acids, also attenuate brain dysfunction, although it is unknown if they control synaptic function. We now used electrophysiological recordings in mouse hippocampal slices to test if realistic concentrations of chlorogenic acids directly affect synaptic transmission and plasticity. 3-(3,4-dihydroxycinnamoyl)quinic acid (CA, 1-10 µM) and 5-O-(trans-3,4-dihydroxycinnamoyl)-D-quinic acid (NCA, 1-10 µM) were devoid of effect on synaptic transmission, paired-pulse facilitation or long-term potentiation (LTP) and long-term depression (LTD) in Schaffer collaterals-CA1 pyramidal synapses. However, CA and NCA increased the recovery of synaptic transmission upon re-oxygenation following 7 min of oxygen/glucose deprivation, an in vitro ischemia model. Also, CA and NCA attenuated the shift of LTD into LTP observed in hippocampal slices from animals with hippocampal-dependent memory deterioration after exposure to ß-amyloid 1-42 (2 nmol, icv), in the context of Alzheimer's disease. These findings show that chlorogenic acids do not directly affect synaptic transmission and plasticity but can indirectly affect other cellular targets to correct synaptic dysfunction. Unraveling the molecular mechanisms of action of chlorogenic acids will allow the design of hitherto unrecognized novel neuroprotective strategies.


Asunto(s)
Ácido Clorogénico/farmacología , Hipocampo/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Neurotransmisores/farmacología , Transmisión Sináptica/efectos de los fármacos , Enfermedad de Alzheimer/patología , Animales , Modelos Animales de Enfermedad , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL
4.
Front Mol Neurosci ; 13: 111, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32848592

RESUMEN

Dopamine replacement therapy with L-3,4-dihydroxyphenylalanine (L-DOPA) is the only temporary therapy for Parkinson's disease (PD), but it triggers dyskinesia over time. Since dyskinesia is associated with increased neuronal firing that bolsters purinergic signaling, we now tested whether the selective and blood-brain barrier-permeable P2X7 receptor antagonist Brilliant Blue-G (BBG, 22.5-45 mg/kg ip) attenuated behavioral, neurochemical and biochemical alterations in rats turned hemiparkinsonian upon unilateral striatal injection of 6-hydroxydopamine (6-OHDA) and treated daily with L-DOPA (30 mg/kg by gavage) for 22 days. The blockade of P2X7 receptors decreased L-DOPA-induced dyskinesia and motor incoordination in hemiparkinsonian rats. In parallel, BBG treatment rebalanced the altered dopamine D1 and D2 receptor density and signaling as well as some neuroinflammation-associated parameters in the striatum and substantia nigra. These findings herald a hitherto unrecognized role for purinergic signaling in the etiopathology of dyskinesia and prompt P2X7 receptor antagonists as novel candidate anti-dyskinesia drugs.

5.
Br J Pharmacol ; 176(18): 3666-3680, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31220343

RESUMEN

BACKGROUND AND PURPOSE: Parkinson's disease (PD) involves an initial loss of striatal dopamine terminals evolving into degeneration of dopamine neurons in substantia nigra (SN), which can be modelled by 6-hydroxydopamine (6-OHDA) administration. Adenosine A2A receptor blockade attenuates PD features in animal models, but the source of the adenosine causing A2A receptor over-activation is unknown. As ATP is a stress signal, we have tested if extracellular catabolism of adenine nucleotides into adenosine (through ecto-5'-nucleotidase or CD73) leads to A2A receptor over-activation in PD. EXPERIMENTAL APPROACH: Effects of blocking CD73 with α,ß-methylene ADP (AOPCP) were assayed in 6-OHDA-treated rats and dopamine-differentiated neuroblastoma SH-SY5Y cells. KEY RESULTS: 6-OHDA increased ATP release and extracellular conversion into adenosine through CD73 up-regulation in SH-SY5Y cells. Removing extracellular adenosine with adenosine deaminase, blocking CD73 with AOPCP, or blocking A2A receptors with SCH58261 were equi-effective in preventing 6-OHDA-induced damage in SH-SY5Y cells. In vivo striatal exposure to 6-OHDA increased ATP release and extracellular formation of adenosine from adenosine nucleotides and up-regulated CD73 and A2A receptors in striatal synaptosomes. Intracerebroventricular administration of AOPCP phenocopied effects of SCH58261, attenuating 6-OHDA-induced (a) increase of contralateral rotations after apomorphine, (b) reduction of dopamine content in striatum and SN, (c) loss of TH staining in striatum and SN, (d) motor dysfunction in the cylinder test, and (e) short-term memory impairment in the object recognition test. CONCLUSION AND IMPLICATIONS: Our data indicate that increased ATP-derived adenosine formation is responsible for A2A receptor over-activation in PD, suggesting CD73 as a new target to manage PD.


Asunto(s)
5'-Nucleotidasa/metabolismo , Adenosina Trifosfato/metabolismo , Adenosina/metabolismo , Trastornos Parkinsonianos/metabolismo , Receptor de Adenosina A2A/metabolismo , 5'-Nucleotidasa/antagonistas & inhibidores , Antagonistas del Receptor de Adenosina A2/farmacología , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Animales , Línea Celular Tumoral , Humanos , Masculino , Oxidopamina/farmacología , Pirimidinas/farmacología , Ratas Wistar , Triazoles/farmacología
6.
J Neuroinflammation ; 15(1): 249, 2018 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-30170624

RESUMEN

BACKGROUND: The deficiency in 1α, 25-dihydroxyvitamin D3 (VD3) seems to increase the risk for neurodegenerative pathologies, including Parkinson's disease (PD). The majority of its actions are mediated by the transcription factor, VD3 receptor (VD3R). METHODS: The neuroprotective effects of VD3 were investigated on a PD model. Male Wistar rats were divided into the following groups: sham-operated (SO), 6-OHDA-lesioned (non-treated), and 6-OHDA-lesioned and treated with VD3 (7 days before the lesion, pre-treatment or for 14 days after the 6-OHDA striatal lesion, post-treatment). Afterwards, the animals were subjected to behavioral tests and euthanized for striatal neurochemical and immunohistochemical assays. The data were analyzed by ANOVA and the Tukey test and considered significant for p < 0.05. RESULTS: We showed that pre- or post-treatments with VD3 reversed behavioral changes and improved the decreased DA contents of the 6-OHDA group. In addition, VD3 reduced the oxidative stress, increased (TH and DAT), and reduced (TNF-alpha) immunostainings in the lesioned striata. While significant decreases in VD3R immunoreactivity were observed after the 6-OHDA lesion, these changes were blocked after VD3 pre- or post-treatments. We showed that VD3 offers neuroprotection, decreasing behavioral changes, DA depletion, and oxidative stress. In addition, it reverses partially or completely TH, DAT, TNF-alpha, and VD3R decreases of immunoreactivities in the non-treated 6-OHDA group. CONCLUSIONS: Taken together, VD3 effects could result from its anti-inflammatory and antioxidant actions and from its actions on VD3R. These findings should stimulate translational research towards the VD3 potential for prevention or treatment of neurodegenerative diseases, as PD.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Encefalitis/etiología , Encefalitis/patología , Estrés Oxidativo/efectos de los fármacos , Trastornos Parkinsonianos/complicaciones , Vitamina D/farmacología , Animales , Apomorfina/farmacología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Conducta Exploratoria/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Masculino , Oxidopamina/toxicidad , Trastornos Parkinsonianos/inducido químicamente , Ratas , Ratas Wistar , Conducta Estereotipada/efectos de los fármacos , Natación/fisiología , Simpaticolíticos/toxicidad , Tirosina 3-Monooxigenasa/metabolismo
7.
eNeuro ; 5(6)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30627646

RESUMEN

Neurodegeneration is a process transversal to neuropsychiatric diseases and the understanding of its mechanisms should allow devising strategies to prevent this irreversible step in brain diseases. Neurodegeneration caused by seizures is a critical step in the aggravation of temporal lobe epilepsy, but its mechanisms remain undetermined. Convulsions trigger an elevation of extracellular adenosine and upregulate adenosine A2A receptors (A2AR), which have been associated with the control of neurodegenerative diseases. Using the rat and mouse kainate model of temporal lobe epilepsy, we now tested whether A2AR control convulsions-induced hippocampal neurodegeneration. The pharmacological or genetic blockade of A2AR did not affect kainate-induced convulsions but dampened the subsequent neurotoxicity. This neurotoxicity began with a rapid A2AR upregulation within glutamatergic synapses (within 2 h), through local translation of synaptic A2AR mRNA. This bolstered A2AR-mediated facilitation of glutamate release and of long-term potentiation (LTP) in CA1 synapses (4 h), triggered a subsequent synaptotoxicity, heralded by decreased synaptic plasticity and loss of synaptic markers coupled to calpain activation (12 h), that predated overt neuronal loss (24 h). All modifications were prevented by the deletion of A2AR selectively in forebrain neurons. This shows that synaptic A2AR critically control synaptic excitotoxicity, which underlies the development of convulsions-induced neurodegeneration.


Asunto(s)
Convulsivantes/toxicidad , Ácido Kaínico/toxicidad , Degeneración Nerviosa/etiología , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Receptor de Adenosina A2A/metabolismo , Antagonistas del Receptor de Adenosina A2/uso terapéutico , Amígdala del Cerebelo/fisiología , Animales , Células Cultivadas , Epilepsia/complicaciones , Epilepsia/tratamiento farmacológico , Epilepsia/etiología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Excitación Neurológica/efectos de los fármacos , Excitación Neurológica/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/prevención & control , Neuronas/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Pirimidinas/uso terapéutico , Ratas , Ratas Wistar , Receptor de Adenosina A2A/genética , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética , Triazoles/uso terapéutico
8.
Behav Pharmacol ; 28(2 and 3-Spec Issue): 223-237, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27763892

RESUMEN

Inflammation plays a pivotal role in the development of ischemic brain damage. Astrocyte activation promotes the production of several proinflammatory mediators, such as TNF-α and iNOS. Eventually, neuronal death occurs, leading to the development of motor and memory deficits in patients. Boldine is the main alkaloid in the leaves and bark of the Peumus boldus Molina, and has anti-inflammatory and antioxidant properties. The aim of this work was to investigate the neuroprotective effect of boldine on neuroinflammation and memory deficits induced by permanent middle cerebral artery occlusion (pMCAO) in mice. Thirty minutes before pMCAO and during the next 5 days, animals received vehicle (0.025 µmol/l HCl) or boldine (8, 16 and 25 mg/kg, intraperitoneally). The extension of the infarct area, neurological scores, and myeloperoxidase activity were evaluated 24 h after pMCAO. Locomotor activity, working, and aversive memory were evaluated 72 h after pMCAO, object recognition memory was tested 96 h after pMCAO, and spatial memory was tested 120 h after pMCAO. Cresyl violet, Fluoro-Jade C staining, and immunohistochemical for GFAP, TNF-α, and iNOS were also carried out. The treatment with boldine significantly decreased the infarct area, improved the neurological scores, and increased cell viability. The vertical exploratory activity and aversive, spatial, object recognition, and working memory deficits induced by pMCAO were prevented by boldine. Moreover, myeloperoxidase activity and GFAP, TNF-α, and iNOS immunoreactivity were decreased significantly by boldine. Although various mechanisms such as its antioxidant activity should be considered, these results suggest that the neuroprotective effect of boldine might be related in part to its anti-inflammatory properties.


Asunto(s)
Aporfinas/farmacología , Inflamación/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Aporfinas/administración & dosificación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inflamación/patología , Inyecciones Intraperitoneales , Masculino , Trastornos de la Memoria/etiología , Trastornos de la Memoria/prevención & control , Ratones , Fármacos Neuroprotectores/administración & dosificación , Peumus/química , Accidente Cerebrovascular/complicaciones
9.
Oncotarget ; 7(39): 62862-62872, 2016 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-27590517

RESUMEN

Opportunistic gut infections and chronic inflammation, in particular due to overgrowth of Candida albicans present in the gut microbiota, are increasingly reported in the elder population. In aged, adult and young mice, we now compared the relative intestinal over-colonization by ingested C. albicans and their translocation to other organs, focusing on the role of adenosine A2A receptors that are a main stop signal of inflammation. We report that elderly mice are more prone to over-colonization by C. albicans than adult and young mice. This fungal over-growth seems to be related with higher growth rate in intestinal lumen, independent of gut tissues invasion, but resulting in higher GI tract inflammation. We observed a particularly high colonization of the stomach, with increased rate of yeast-to-hypha transition in aged mice. We found a correlation between A2A receptor density and tissue damage due to yeast infection: comparing with young and adults, aged mice have a lower gut A2A receptor density and C. albicans infection failed to increase it. In conclusion, this study shows that aged mice have a lower ability to cope with inflammation due to C. albicans over-colonization, associated with an inability to adaptively adjust adenosine A2A receptors density.


Asunto(s)
Envejecimiento , Candida albicans , Candidiasis/genética , Receptor de Adenosina A2A/genética , Animales , Candidiasis/metabolismo , Heces , Tracto Gastrointestinal/microbiología , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Estómago/microbiología
10.
Nutrition ; 32(9): 1019-27, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27157468

RESUMEN

OBJECTIVE: Prolonged maternal separation (PMS) in the first 2 wk of life has been associated with poor growth with lasting effects in brain structure and function. This study aimed to investigate whether PMS-induced undernutrition could cause systemic inflammation and changes in nutrition-related hormonal levels, affecting hippocampal structure and neurotransmission in C57BL/6J suckling mice. METHODS: This study assessed mouse growth parameters coupled with insulin-like growth factor-1 (IGF-1) serum levels. In addition, leptin, adiponectin, and corticosterone serum levels were measured following PMS. Hippocampal stereology and the amino acid levels were also assessed. Furthermore, we measured myelin basic protein and synapthophysin (SYN) expression in the overall brain tissue and hippocampal SYN immunolabeling. For behavioral tests, we analyzed the ontogeny of selected neonatal reflexes. PMS was induced by separating half the pups in each litter from their lactating dams for defined periods each day (4 h on day 1, 8 h on day 2, and 12 h thereafter). A total of 67 suckling pups were used in this study. RESULTS: PMS induced significant slowdown in weight gain and growth impairment. Significant reductions in serum leptin and IGF-1 levels were found following PMS. Total CA3 area and volume were reduced, specifically affecting the pyramidal layer in PMS mice. CA1 pyramidal layer area was also reduced. Overall hippocampal SYN immunolabeling was lower, especially in CA3 field and dentate gyrus. Furthermore, PMS reduced hippocampal aspartate, glutamate, and gamma-aminobutyric acid levels, as compared with unseparated controls. CONCLUSION: These findings suggest that PMS causes significant growth deficits and alterations in hippocampal morphology and neurotransmission.


Asunto(s)
Hipocampo/crecimiento & desarrollo , Inflamación/etiología , Desnutrición/etiología , Privación Materna , Aminoácidos/sangre , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Hipocampo/fisiopatología , Inflamación/sangre , Factor I del Crecimiento Similar a la Insulina/metabolismo , Desnutrición/sangre , Ratones , Ratones Endogámicos C57BL
11.
Neuropharmacology ; 81: 142-52, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24508709

RESUMEN

Parkinson's disease (PD) involves an initial loss of striatal dopaminergic terminals evolving into a degeneration of dopaminergic neurons in the substantia nigra (SN), which can be modeled by 6-hydroxydopamine (6-OHDA) administration. Since ATP is a danger signal acting through its P2X7 receptors (P2X7R), we now tested if a blood-brain barrier-permeable P2X7R antagonist, Brilliant Blue G (BBG), controlled the 6-OHDA-induced PD-like features in rats. BBG (45 mg/kg) attenuated the 6-OHDA-induced: 1) increase of contralateral rotations in the apomorphine test, an effect mimicked by another P2X7R antagonist A438079 applied intra-cerebroventricularly; 2) short-term memory impairment in the passive avoidance and cued version of the Morris Water maze; 3) reduction of dopamine content in the striatum and SN; 4) microgliosis and astrogliosis in the striatum. To grasp the mechanism of action of BBG, we used in vitro models exploring synaptotoxicity (striatal synaptosomes) and neurotoxicity (dopamine-differentiated neuroblastoma SH-SY5Y cells). P2X7R were present in striatal dopaminergic terminals, and BBG (100 nM) prevented the 6-OHDA-induced synaptosomal dysfunction. P2X7R were also co-localized with tyrosine hydroxylase in SH-SY5Y cells, where BBG (100 nM) attenuated the 6-OHDA-induced neurotoxicity. This suggests that P2X7R contribute to PD pathogenesis through a triple impact on synaptotoxicity, gliosis and neurotoxicity, highlighting the therapeutic potential of P2X7R antagonists in PD.


Asunto(s)
Lateralidad Funcional/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Síndromes de Neurotoxicidad/tratamiento farmacológico , Trastornos Parkinsonianos/tratamiento farmacológico , Antagonistas del Receptor Purinérgico P2X/uso terapéutico , Colorantes de Rosanilina/uso terapéutico , Animales , Reacción de Prevención/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Gliosis/tratamiento farmacológico , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Síndromes de Neurotoxicidad/etiología , Oxidopamina/toxicidad , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/fisiopatología , Antagonistas del Receptor Purinérgico P2X/farmacología , Ratas , Ratas Wistar , Tiempo de Reacción/efectos de los fármacos , Colorantes de Rosanilina/farmacología
12.
Eur J Neurosci ; 39(4): 614-22, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24304101

RESUMEN

ATP is a pleiotropic cell-to-cell signaling molecule in the brain that functions through activation of the P2 receptors (P2R), encompassing ionotropic P2XR or metabotropic P2YR. Noxious brain insults increase the extracellular levels of ATP and previous studies have implicated different P2R, namely P2Y1R, in the control of ischemic brain damage, but it remains to be defined if P2Y1R antagonists also alleviate the behavioral impairments associated with brain ischemia. Furthermore, as P2Y1R can control neuronal and glial functions, we explored if P2Y1R antagonist-mediated protection would mainly involve neuronal and/or glial processes. Adult male mice subject to permanent middle cerebral artery occlusion (pMCAO) displayed an infarcted cortical area (2,3,5-triphenyltetrazolium chloride staining), decreased neurological score with decreased working and reference memory performance (Y-maze, object recognition and aversive memory), accompanied by neuronal damage (FluoroJade C), astrogliosis (glial fibrillary acidic protein) and microgliosis (CD11b). All of these changes were attenuated by intracerebroventricular pre-treatment (10 min before pMCAO) with the generic P2R antagonist 4-[(E)-{4-formyl-5-hydroxy-6-methyl-3-[(phosphono-oxy)methyl]pyridin-2-yl}diazenyl]benzene-1,3-disulfonic acid (PPADS, 0.5-1.0 nmol/µL). In contrast, the selective P2Y1R antagonist (1R*,2S*)-4-[2-Iodo-6-(methylamino)-9H-purin-9-yl]-2-(phosphono-oxy)bicycle[3.1.0] hexane-1-methanol dihydrogen phosphate ester (MRS2500, 1.0-2.0 nmol/µL) afforded equivalent behavioral benefits but only prevented neuronal damage but not astrogliosis or microgliosis upon pMCAO. These results indicated that P2Y1R-associated neuroprotection mainly occurred through neuronal mechanisms, whereas other P2R were also involved in the control of astrocytic reactivity upon brain injury.


Asunto(s)
Astrocitos/metabolismo , Cognición , Infarto de la Arteria Cerebral Media/metabolismo , Neuronas/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Animales , Astrocitos/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Nucleótidos de Desoxiadenina/farmacología , Nucleótidos de Desoxiadenina/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Aprendizaje por Laberinto , Memoria , Ratones , Neuronas/patología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Antagonistas del Receptor Purinérgico P2Y/uso terapéutico , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacología , Fosfato de Piridoxal/uso terapéutico
13.
Pharmacol Rep ; 64(4): 848-56, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23087136

RESUMEN

BACKGROUND: Accumulating evidence indicates that mangiferin (MGF), a natural xanthone, by virtue of its antioxidant and antiinflammatory properties is neuroprotective. Here we sought to verify the cytoprotective role of MGF on cultured rat primary mesencephalic cells exposed to 6-hydroxydopamine (6-OHDA) in vitro, and the MGFs anti-inflammatory potential in mouse model of ketamine-induced schizophrenia in vivo. METHODS: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)-assay was performed tomeasure cell viability inmesencephalic cell cultures exposed to neurotoxin (6-OHDA, 40 µM). Schizophrenia was induced in mice by ketamine (50 mg/kg, ip, twice a day, for 7 days). The treatment effects of MGF (50 mg/kg, po, for 7 days) were verified on locomotor behavioral changes in open-field test, and on the oxidant stress-related increase in lipid-peroxidation (malondialdehyde) and interleukin-6 (IL-6) levels in brain tissues. RESULTS: MGF (10-100 µM) produced no per se effect on cell viability as measured by MTT assay, but significantly prevented the 6-OHDA-induced cell death in a concentration-dependent manner. Acridine orange/ethidium bromide (AO/EtBr) staining confirmed the absence of 6-OHDA-induced morphological changes characteristic of apoptosis/necrosis. In open-field test, ketamine-induced impaired locomotor activity and behavioral changes such as grooming and stereotyped but not rearing were effectively ameliorated by MGF pretreatment. Also, ketamine-associated increase in brain tissue levels of IL-6 and MDA were significantly lowered in MGF-pretreated mice. CONCLUSION: Mangiferin has a neurocytoprotective role related, at least in part, to an antioxidant and anti-inflammatory mechanism, which could be explored for more effective therapies of schizophrenia and other neurodegenerative diseases.


Asunto(s)
Neurotoxinas/efectos adversos , Estrés Oxidativo/efectos de los fármacos , Esquizofrenia/tratamiento farmacológico , Xantonas/farmacología , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Femenino , Interleucina-6/metabolismo , Ketamina/farmacología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Malondialdehído/metabolismo , Mesencéfalo/efectos de los fármacos , Mesencéfalo/metabolismo , Ratones , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Nitritos/metabolismo , Oxidopamina , Ratas , Ratas Wistar , Esquizofrenia/inducido químicamente , Esquizofrenia/metabolismo
14.
An Acad Bras Cienc ; 84(1): 113-22, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22441600

RESUMEN

Byrsonima sericea leaves are extensively used in folk medicine in Brazil against gastric disorders. This study investigated the chemical constituents of B. sericea leaf ethanolic extract (BSLE) and its potential gastroprotective activity, with its possible mechanism of the action using ethanol to induce gastric mucosal damage in mice. The phytochemical analysis was carried out to identify the active constituents present in the extract, and the HPLC analysis was performed for the identification of flavonoids. BSLE at oral doses of 125, 250 and 500 mg/kg markedly attenuated the ethanol-evoked gastric lesions by 53.2, 84.9 and 87.6 %, respectively. The BSLE (250 mg/kg) prevented the depletion of gastric mucus and gastric mucosal nonproteic-sulfhydryl groups, SOD and CAT, as well as the increase in the MDA content promoted by absolute ethanol. Moreover, the effect of BSLE against ethanol damage was found to be significantly reduced in mice pretreated with Capsazepine (i.p.), L-NAME (i.p.) or glibenclamide (i.p.), the respective blockers/inhibitors of TRPV1, NO synthase and K+ATP channel. The phytochemical investigation on BSLE revealed the presence of flavonoids rutin, isoquercitrin, kaempferol 3-O-rutinoside and quercetin, which are compounds well known for their antioxidant and gastroprotective properties. These results suggest that BSLE affords gastroprotection through multiple mechanisms, which may be helpful in the treatment of pathologies associated with gastric dysfunctions.


Asunto(s)
Antiulcerosos/uso terapéutico , Mucosa Gástrica/efectos de los fármacos , Malpighiaceae/química , Extractos Vegetales/uso terapéutico , Úlcera Gástrica/prevención & control , Animales , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Etanol/efectos adversos , Femenino , Ratones , Hojas de la Planta/química , Úlcera Gástrica/inducido químicamente
15.
J Neurochem ; 117(1): 100-11, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21235574

RESUMEN

The blockade of adenosine A(2A) receptors (A2AR) affords a robust neuroprotection in different noxious brain conditions. However, the mechanisms underlying this general neuroprotection are unknown. One possible mechanism could be the control of neuroinflammation that is associated with brain damage, especially because A2AR efficiently control peripheral inflammation. Thus, we tested if the intracerebroventricular injection of a selective A2AR antagonist (SCH58261) would attenuate the changes in the hippocampus triggered by intraperitoneal administration of lipopolysaccharide (LPS) that induces neuroinflammation through microglia activation. LPS administration triggers an increase in inflammatory mediators like interleukin-1ß that causes biochemical changes (p38 and c-jun N-terminal kinase phosphorylation and caspase 3 activation) contributing to neuronal dysfunction typified by decreased long-term potentiation, a form of synaptic plasticity. Long-term potentiation, measured 30 min after the tetanus, was significantly lower in LPS-treated rats compared with control-treated rats, while SCH58261 attenuated the LPS-induced change. The LPS-induced increases in phosphorylation of c-jun N-terminal kinase and p38 and activation of caspase 3 were also prevented by SCH58261. Significantly, SCH58261 also prevented the LPS-induced recruitment of activated microglial cells and the increase in interleukin-1ß concentration in the hippocampus, indicating that A2AR activation is a pivotal step in mediating the neuroinflammation triggered by LPS. These results indicate that A2AR antagonists prevent neuroinflammation and support the hypothesis that this mechanism might contribute for the ability of A2AR antagonists to control different neurodegenerative diseases known to involve neuroinflammation.


Asunto(s)
Hipocampo/patología , Mediadores de Inflamación/fisiología , Inhibición Neural/fisiología , Neuronas/patología , Receptor de Adenosina A2A/fisiología , Animales , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/fisiología , Masculino , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Wistar , Receptor de Adenosina A2A/metabolismo , Resorcinoles/farmacología , Resorcinoles/uso terapéutico , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/patología
16.
J Negat Results Biomed ; 9: 3, 2010 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-20546603

RESUMEN

BACKGROUND: The present study investigated the effects of venlafaxine, an antidepressant drug with immunoregulatory properties on the inflammatory response and bone loss associated with experimental periodontal disease (EPD). MATERIALS AND METHODS: Wistar rats were subjected to a ligature placement around the second upper left molar. The treated groups received orally venlafaxine (10 or 50 mg/kg) one hour before the experimental periodontal disease induction and daily for 10 days. Vehicle-treated experimental periodontal disease and a sham-operated (SO) controls were included. Bone loss was analyzed morphometrically and histopathological analysis was based on cell influx, alveolar bone, and cementum integrity. Lipid peroxidation quantification and immunohistochemistry to TNF-alpha and iNOS were performed. RESULTS: Experimental periodontal disease rats showed an intense bone loss compared to SO ones (SO = 1.61 +/- 1.36; EPD = 4.47 +/- 1.98 mm, p < 0.001) and evidenced increased cellular infiltration and immunoreactivity for TNF-alpha and iNOS. Venlafaxine treatment while at low dose (10 mg/kg) afforded no significant protection against bone loss (3.25 +/- 1.26 mm), a high dose (50 mg/kg) caused significantly enhanced bone loss (6.81 +/- 3.31 mm, p < 0.05). Venlafaxine effectively decreased the lipid peroxidation but showed no significant change in TNF-alpha or iNOS immunoreactivity. CONCLUSION: The increased bone loss associated with high dose venlafaxine may possibly be a result of synaptic inhibition of serotonin uptake.


Asunto(s)
Pérdida de Hueso Alveolar/complicaciones , Pérdida de Hueso Alveolar/tratamiento farmacológico , Ciclohexanoles/uso terapéutico , Periodontitis/complicaciones , Periodontitis/tratamiento farmacológico , Pérdida de Hueso Alveolar/enzimología , Animales , Ciclohexanoles/farmacología , Encía/efectos de los fármacos , Encía/patología , Inmunohistoquímica , Ligadura , Malondialdehído/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Periodontitis/enzimología , Periodontitis/patología , Ratas , Ratas Wistar , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Clorhidrato de Venlafaxina
17.
Nutrition ; 26(6): 662-70, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20371167

RESUMEN

OBJECTIVE: The effect of zinc and glutamine on brain development was investigated during the lactation period in Swiss mice. METHODS: Malnutrition was induced by clustering the litter size from 6-7 pups/dam (nourished control) to 12-14 pups/dam (undernourished control) following birth. Undernourished groups received daily supplementation with glutamine by subcutaneous injections starting at day 2 and continuing until day 14. Glutamine (100 mM, 40-80 microL) was used for morphological and behavioral studies. Zinc acetate was added in the drinking water (500 mg/L) to the lactating dams. Synaptophysin and myelin basic protein brain expressions were evaluated by immunoblot. Zinc serum and brain levels and hippocampal neurotransmitters were also evaluated. RESULTS: Zinc with or without glutamine improved weight gain as compared to untreated, undernourished controls. In addition, zinc supplementation improved cliff avoidance and head position during swim behaviors especially on days 9 and 10. Using design-based stereological methods, we found a significant increase in the volume of CA1 neuronal cells in undernourished control mice, which was not seen in mice receiving zinc or glutamine alone or in combination. Undernourished mice given glutamine showed increased CA1 layer volume as compared with the other groups, consistent with the trend toward increased number of neurons. Brain zinc levels were increased in the nourished and undernourished-glutamine treated mice as compared to the undernourished controls on day 7. Undernourished glutamine-treated mice showed increased hippocampal gamma-aminobutyric acid and synaptophysin levels on day 14. CONCLUSION: We conclude that glutamine or zinc protects against malnutrition-induced brain developmental impairments.


Asunto(s)
Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Glutamina/farmacología , Desnutrición/tratamiento farmacológico , Micronutrientes/farmacología , Aumento de Peso/efectos de los fármacos , Zinc/farmacología , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Animales Recién Nacidos/metabolismo , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Suplementos Dietéticos , Quimioterapia Combinada , Femenino , Glutamina/uso terapéutico , Lactancia , Desnutrición/sangre , Ratones , Micronutrientes/sangre , Neuronas/efectos de los fármacos , Embarazo , Natación , Sinaptofisina/metabolismo , Zinc/sangre , Zinc/uso terapéutico , Acetato de Zinc/farmacología , Ácido gamma-Aminobutírico/metabolismo
18.
Neurochem Res ; 34(6): 1066-75, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19005754

RESUMEN

In the present work, we showed that a chalcone-enriched fraction (CEF) isolated from the stem bark of a Brazilian medicinal plant, Myracrodruon urundeuva, presents neuroprotective actions on 6-hydroxydopamine (6-OHDA)-induced neuronal cell death, in rat mesencephalic cells. In the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium] assay, which is an index of cell viability, CEF (1-100 microg/ml) reversed in a concentration-dependent manner the 6-OHDA-induced cell death. While cells exposed to 6-OHDA (40 microM) showed an increased concentration of thiobarbituric acid reactive substances (TBARS), the pretreatment with CEF (10-100 microg/ml) significantly decreased the 6-OHDA-induced TBARS formation, indicative of a neuroprotection against lipoperoxidation. Furthermore, the drastic increase of nitrite levels induced by 6-OHDA, indicative of nitric oxide formation and free radicals production, was prevented by CEF. Double staining with acridine orange/ethidium bromide showed that cultures exposed to 6-OHDA (40 and 200 microM) presented an increase of apoptotic and necrotic cell numbers in a concentration-dependent manner. CEF (100 microg/ml) protected cells from apoptosis and necrosis and increased number of cells presenting a normal morphology. The immunohistochemical analysis for tyrosine hydroxylase (TH) positive neurons indicated that 6-OHDA (40 and 200 microM) caused a concentration-dependent loss of TH+ and TH- neurons. CEF protected both cells types from 6-OHDA-induced cell death. All together, our results demonstrated neuroprotective effects of chalcones, which are able to reduce oxidative stress and apoptotic injury caused by 6-OHDA. Our findings suggest that chalcones could provide benefits, along with other therapies, in neurodegenerative injuries, such as Parkinson's disease.


Asunto(s)
Anacardiaceae , Chalconas/farmacología , Citotoxinas/toxicidad , Mesencéfalo/citología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Oxidopamina/toxicidad , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Dopamina/metabolismo , Inmunohistoquímica , Peroxidación de Lípido/efectos de los fármacos , Neuronas/citología , Neuronas/metabolismo , Nitritos/metabolismo , Estrés Oxidativo , Ratas , Ratas Wistar , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...