Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Mol Psychiatry ; 28(2): 871-882, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36280751

RESUMEN

Molecular and functional abnormalities of astrocytes have been implicated in the etiology and pathogenesis of schizophrenia (SCZ). In this study, we examined the proteome, inflammatory responses, and secretome effects on vascularization of human induced pluripotent stem cell (hiPSC)-derived astrocytes from patients with SCZ. Proteomic analysis revealed alterations in proteins related to immune function and vascularization. Reduced expression of the nuclear factor kappa B (NF-κB) p65 subunit was observed in these astrocytes, with no incremental secretion of cytokines after tumor necrosis factor alpha (TNF-α) stimulation. Among inflammatory cytokines, secretion of interleukin (IL)-8 was particularly elevated in SCZ-patient-derived-astrocyte-conditioned medium (ASCZCM). In a chicken chorioallantoic membrane (CAM) assay, ASCZCM reduced the diameter of newly grown vessels. This effect could be mimicked with exogenous addition of IL-8. Taken together, our results suggest that SCZ astrocytes are immunologically dysfunctional and may consequently affect vascularization through secreted factors.


Asunto(s)
Células Madre Pluripotentes Inducidas , Esquizofrenia , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Astrocitos/metabolismo , Proteómica , Esquizofrenia/metabolismo , Citocinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Fenotipo
2.
Neurochem Res ; 48(8): 2424-2435, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36995561

RESUMEN

Attention deficit hyperactivity disorder (ADHD) is a neurodevelopmental disorder characterized by inattention, hyperactivity, and impulsivity symptoms. Neuroimaging studies have revealed a delayed cortical and subcortical development pattern in children diagnosed with ADHD. This study followed up on the development in vitro of frontal cortical neurons from Spontaneously hypertensive rats (SHR), an ADHD rat model, and Wistar-Kyoto rats (WKY), control strain, over their time in culture, and in response to BDNF treatment at two different days in vitro (DIV). These neurons were also evaluated for synaptic proteins, brain-derived neurotrophic factor (BDNF), and related protein levels. Frontal cortical neurons from the ADHD rat model exhibited shorter dendrites and less dendritic branching over their time in culture. While pro- and mature BDNF levels were not altered, the cAMP-response element-binding (CREB) decreased at 1 DIV and SNAP-25 decreased at 5 DIV. Different from control cultures, exogenous BDNF promoted less dendritic branching in neurons from the ADHD model. Our data revealed that neurons from the ADHD model showed decreased levels of an important transcription factor at the beginning of their development, and their delayed outgrowth and maturation had consequences in the levels of SNAP-25 and may be associated with less response to BDNF. These findings provide an alternative tool for studies on synaptic dysfunctions in ADHD. They may also offer a valuable tool for investigating drug effects and new treatment opportunities.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Factor Neurotrófico Derivado del Encéfalo , Ratas , Animales , Ratas Endogámicas SHR , Factor Neurotrófico Derivado del Encéfalo/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ratas Endogámicas WKY , Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Trastorno por Déficit de Atención con Hiperactividad/metabolismo , Neuronas/metabolismo , Modelos Animales de Enfermedad
3.
Nutr Neurosci ; 25(5): 1066-1077, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-33107813

RESUMEN

OBJECTIVE: We investigated the influence of dietary omega-3 polyunsaturated fatty acids (n-3 PUFAs) on glutamatergic system modulation after a single episode of neonatal seizures and their possible effects on seizure-induced long-lasting behavioral deficits. METHODS: Male Wistar rats receiving an omega-3 diet (n-3) or an n-3 deficient diet (D) from the prenatal period were subjected to a kainate-induced seizure model at P7. Glutamate transporter activity and immunocontents (GLT-1 and GLAST) were assessed in the hippocampus at 12, 24, and 48 h after the seizure episode. Fluorescence intensity for glial cells (GFAP) and neurons (NeuN) was assessed 24 h after seizure in the hippocampus. Behavioral analysis (elevated-plus maze and inhibitory avoidance memory task) was performed at 60 days of age. RESULTS: The D group showed a decrease in glutamate uptake 24 h after seizure. In this group only, the GLT1 content increased at 12 h, followed by a decrease at 24 h. GLAST increased up to 24 h after seizure. GFAP fluorescence was higher, and NeuN fluorescence decreased, in the D group independent of seizures. In adulthood, the D group presented memory deficits independent of seizures, but short-term memory (1.5 h after a training session) was abolished in the D group treated with kainate. SIGNIFICANCE: N-3 PUFA positively influenced the glutamatergic system during seizure and prevented seizure-related memory deficits in adulthood.


Asunto(s)
Epilepsia , Ácidos Grasos Omega-3 , Animales , Dieta , Ácidos Grasos Omega-3/efectos adversos , Femenino , Ácido Glutámico , Hipocampo , Ácido Kaínico , Masculino , Trastornos de la Memoria/prevención & control , Embarazo , Ratas , Ratas Wistar , Convulsiones/inducido químicamente , Convulsiones/prevención & control
4.
Eur J Neurosci ; 49(12): 1673-1683, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30667546

RESUMEN

Attention deficit and hyperactivity disorder (ADHD) is characterized by impaired levels of hyperactivity, impulsivity, and inattention. Adenosine and endocannabinoid systems tightly interact in the modulation of dopamine signaling, involved in the neurobiology of ADHD. In this study, we evaluated the modulating effects of the cannabinoid and adenosine systems in a tolerance to delay of reward task using the most widely used animal model of ADHD. Spontaneous Hypertensive Rats (SHR) and Wistar-Kyoto rats were treated chronically or acutely with caffeine, a non-selective adenosine receptor antagonist, or acutely with a cannabinoid agonist (WIN55212-2, WIN) or antagonist (AM251). Subsequently, animals were tested in the tolerance to delay of reward task, in which they had to choose between a small, but immediate, or a large, but delayed, reward. Treatment with WIN decreased, whereas treatment with AM251 increased the choices of the large reward, selectively in SHR rats, indicating a CB1 receptor-mediated increase in impulsive behavior. An acute pre-treatment with caffeine blocked WIN effects. Conversely, a chronic treatment with caffeine increased the impulsive phenotype and potentiated the WIN effects. The results indicate that both cannabinoid and adenosine receptors modulate impulsive behavior in SHR: the antagonism of cannabinoid receptors might be effective in reducing impulsive symptoms present in ADHD; in addition, caffeine showed the opposite effects on impulsive behavior depending on the length of treatment. These observations are of particular importance to consider when therapeutic manipulation of CB1 receptors is applied to ADHD patients who consume coffee.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Cafeína/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Antagonistas de Receptores de Cannabinoides/farmacología , Conducta Impulsiva/efectos de los fármacos , Psicotrópicos/farmacología , Animales , Benzoxazinas/farmacología , Modelos Animales de Enfermedad , Masculino , Morfolinas/farmacología , Naftalenos/farmacología , Piperidinas/farmacología , Antagonistas de Receptores Purinérgicos P1/farmacología , Pirazoles/farmacología , Distribución Aleatoria , Ratas Endogámicas SHR , Ratas Endogámicas WKY
5.
Proc Natl Acad Sci U S A ; 112(25): 7833-8, 2015 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-26056314

RESUMEN

The consumption of caffeine (an adenosine receptor antagonist) correlates inversely with depression and memory deterioration, and adenosine A2A receptor (A2AR) antagonists emerge as candidate therapeutic targets because they control aberrant synaptic plasticity and afford neuroprotection. Therefore we tested the ability of A2AR to control the behavioral, electrophysiological, and neurochemical modifications caused by chronic unpredictable stress (CUS), which alters hippocampal circuits, dampens mood and memory performance, and enhances susceptibility to depression. CUS for 3 wk in adult mice induced anxiogenic and helpless-like behavior and decreased memory performance. These behavioral changes were accompanied by synaptic alterations, typified by a decrease in synaptic plasticity and a reduced density of synaptic proteins (synaptosomal-associated protein 25, syntaxin, and vesicular glutamate transporter type 1), together with an increased density of A2AR in glutamatergic terminals in the hippocampus. Except for anxiety, for which results were mixed, CUS-induced behavioral and synaptic alterations were prevented by (i) caffeine (1 g/L in the drinking water, starting 3 wk before and continued throughout CUS); (ii) the selective A2AR antagonist KW6002 (3 mg/kg, p.o.); (iii) global A2AR deletion; and (iv) selective A2AR deletion in forebrain neurons. Notably, A2AR blockade was not only prophylactic but also therapeutically efficacious, because a 3-wk treatment with the A2AR antagonist SCH58261 (0.1 mg/kg, i.p.) reversed the mood and synaptic dysfunction caused by CUS. These results herald a key role for synaptic A2AR in the control of chronic stress-induced modifications and suggest A2AR as candidate targets to alleviate the consequences of chronic stress on brain function.


Asunto(s)
Cafeína/farmacología , Trastornos de la Memoria/prevención & control , Trastornos del Humor/prevención & control , Neuronas/efectos de los fármacos , Receptor de Adenosina A2A/efectos de los fármacos , Estrés Psicológico/complicaciones , Animales , Masculino , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Trastornos del Humor/etiología , Neuronas/metabolismo
6.
Behav Brain Res ; 436: 114102, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36089101

RESUMEN

Attention Deficit Hyperactivity Disorder (ADHD) is a neurodevelopmental disorder that presents sex differences in the severity and presentation of symptoms, whose neurobiological basis is still unknown. Both Growth-associated Protein 43 (GAP-43) and Sonic hedgehog (Shh) are considered essential proteins for the appropriate brain development, but their participation in ADHD neurobiology have not been investigated yet. In this study, we hypothesized that alterations in these proteins could be related to behavioral traits to ADHD phenotype. Thus, both sexes of infant Spontaneously hypertensive rats (SHR, used as ADHD animal model) were evaluated for developmental milestones, locomotor activity, olfactory and recognition memory. Both GAP-43 and Shh were assessed in the olfactory bulb, frontal cortex and hippocampus in early and late infancy. During early infancy, SHR reached three developmental milestones later, and females showed olfactory memory impairment accompanied by increased levels of Shh in the olfactory bulb. In later infancy, hyperlocomotion, impaired recognition memory, and decreased Shh in the hippocampus were observed in SHR from both sexes. While in early infancy GAP-43 was not altered, it was decreased in the frontal cortex and hippocampus of female SHR in late infancy. Therefore, both Shh and GAP-43 are involved in the sex-dependent behavioral alterations showed by infant SHR. Despite the disorder's complexity and heterogeneity, our findings reveal important developmental parameters during SHR development and also emphasizes the relevance of studying sex differences in the ADHD context.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Proteínas Hedgehog , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína GAP-43/metabolismo , Proteínas Hedgehog/metabolismo , Masculino , Trastornos de la Memoria/metabolismo , Odorantes , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Caracteres Sexuales
7.
Front Neurosci ; 15: 674563, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34483818

RESUMEN

Over the past years, brain development has been investigated in rodent models, which were particularly relevant to establish the role of specific genes in this process. However, the cytoarchitectonic features, which determine neuronal network formation complexity, are unique to humans. This implies that the developmental program of the human brain and neurological disorders can only partly be reproduced in rodents. Advancement in the study of the human brain surged with cultures of human brain tissue in the lab, generated from induced pluripotent cells reprogrammed from human somatic tissue. These cultures, termed brain organoids, offer an invaluable model for the study of the human brain. Brain organoids reproduce the cytoarchitecture of the cortex and can develop multiple brain regions and cell types. Integration of functional activity of neural cells within brain organoids with genetic, cellular, and morphological data in a comprehensive model for human development and disease is key to advance in the field. Because the functional activity of neural cells within brain organoids relies on cell repertoire and time in culture, here, we review data supporting the gradual formation of complex neural networks in light of cell maturity within brain organoids. In this context, we discuss how the technology behind brain organoids brought advances in understanding neurodevelopmental, pathogen-induced, and neurodegenerative diseases.

8.
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
9.
Behav Brain Res ; 404: 113163, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33549686

RESUMEN

Anxiety disorders are linked to mitochondrial dysfunction and decreased neurotrophic support. Since anxiolytic drugs target mitochondria, non-pharmacological approaches to improve mitochondrial metabolism such as intermittent fasting (IF) may cause parallel behavioral benefits against anxiety disorders. Here, we investigated whether a chronic IF regimen could induce anxiolytic-like effects concomitantly to modulation in mitochondrial bioenergetics and trophic signaling in mice brain. A total of 44 Male C57BL/6 J mice (180 days old) were assigned to two dietary regimens: a normal, ad libitum diet (AL group) and an alternate-day fasting (IF group), where animals underwent 10 cycles of 24 h food restriction followed by 24 h ad libitum access. Animals underwent the open field test, dark/light box and elevated plus maze tasks. Isolated nerve terminals were obtained from mice brain and used for mitochondrial respirometry, hydrogen peroxide production and assessment of membrane potential dynamics, calcium handling and western blotting. We showed that IF significantly alters total daily food intake and food consumption patterns but not body weight. There were no differences in the exploratory and locomotory parameters. Remarkably, animals from IF showed decreased anxiety-like behavior. Mitochondrial metabolic responses in different coupling states and parameters linked with H2O2 production, Ca2+ buffering and electric gradient were not different between groups. Finally, no alterations in molecular indicators of apoptotic death (Bax/Bcl-2 ratio) and neuroplasticity (proBDNF/BDNF and synaptophysin were observed). In conclusion, IF exerts anxiolytic-like effect not associated with modulation in synaptic neuronergetics or expression of neurotrophic proteins. These results highlight a potential benefit of intermittent fasting as a nutritional intervention in anxiety-related disorders.


Asunto(s)
Ansiedad/etiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ayuno/efectos adversos , Mitocondrias/metabolismo , Sinapsis/metabolismo , Animales , Ansiedad/metabolismo , Ansiedad/fisiopatología , Glucemia/análisis , Western Blotting , Encéfalo/metabolismo , Encéfalo/fisiología , Factor Neurotrófico Derivado del Encéfalo/fisiología , Prueba de Laberinto Elevado , Ayuno/metabolismo , Ayuno/psicología , Peróxido de Hidrógeno/metabolismo , Cetonas/sangre , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Mitocondrias/fisiología , Prueba de Campo Abierto , Consumo de Oxígeno , Sinapsis/fisiología , Sinaptosomas/metabolismo , Sinaptosomas/fisiología
10.
J Neurosci ; 29(47): 14741-51, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19940169

RESUMEN

Alzheimer's disease (AD) is characterized by memory impairment, neurochemically by accumulation of beta-amyloid peptide (namely Abeta(1-42)) and morphologically by an initial loss of nerve terminals. Caffeine consumption prevents memory dysfunction in different models, which is mimicked by antagonists of adenosine A(2A) receptors (A(2A)Rs), which are located in synapses. Thus, we now tested whether A(2A)R blockade prevents the early Abeta(1-42)-induced synaptotoxicity and memory dysfunction and what are the underlying signaling pathways. The intracerebral administration of soluble Abeta(1-42) (2 nmol) in rats or mice caused, 2 weeks later, memory impairment (decreased performance in the Y-maze and object recognition tests) and a loss of nerve terminal markers (synaptophysin, SNAP-25) without overt neuronal loss, astrogliosis, or microgliosis. These were prevented by pharmacological blockade [5-amino-7-(2-phenylethyl)-2-(2-furyl)-pyrazolo[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidine (SCH58261); 0.05 mg . kg(-1) . d(-1), i.p.; for 15 d] in rats, and genetic inactivation of A(2A)Rs in mice. Moreover, these were synaptic events since purified nerve terminals acutely exposed to Abeta(1-42) (500 nm) displayed mitochondrial dysfunction, which was prevented by A(2A)R blockade. SCH58261 (50 nm) also prevented the initial synaptotoxicity (loss of MAP-2, synaptophysin, and SNAP-25 immunoreactivity) and subsequent loss of viability of cultured hippocampal neurons exposed to Abeta(1-42) (500 nm). This A(2A)R-mediated control of neurotoxicity involved the control of Abeta(1-42)-induced p38 phosphorylation and was independent from cAMP/PKA (protein kinase A) pathway. Together, these results show that A(2A)Rs play a crucial role in the development of Abeta-induced synaptotoxicity leading to memory dysfunction through a p38 MAPK (mitogen-activated protein kinase)-dependent pathway and provide a molecular basis for the benefits of caffeine consumption in AD.


Asunto(s)
Antagonistas del Receptor de Adenosina A2 , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Trastornos de la Memoria/tratamiento farmacológico , Degeneración Nerviosa/tratamiento farmacológico , Fragmentos de Péptidos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/toxicidad , Animales , Cafeína/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Regulación hacia Abajo/genética , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/fisiopatología , Degeneración Nerviosa/prevención & control , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/toxicidad , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación/efectos de los fármacos , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/metabolismo , Terminales Presinápticos/patología , Pirimidinas/farmacología , Ratas , Ratas Wistar , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2A/metabolismo , Membranas Sinápticas/efectos de los fármacos , Membranas Sinápticas/metabolismo , Triazoles/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Artículo en Inglés | MEDLINE | ID: mdl-31707092

RESUMEN

Longitudinal and some experimental studies have showed the potential of caffeine to counteract some depressive behaviors and synaptic dysfunctions. In this study, we investigated the potential of caffeine in preventing behavioral outcomes, neurodegeneration and synaptic proteins alterations in a mice model of agitated depression by bilateral olfactory bulbectomy (OB). For this purpose, bulbectomized mice received caffeine (0.3 g/L and 1.0 g/L, drinking water), during the active cycle, for seven weeks (two before the surgery and throughout five weeks after OB). Caffeine prevented OB-induced hyperactivity and recognition memory impairment and rescue self care and motivational behavior. In the frontal cortex, bulbectomized mice presented increase in the adenosine A1 receptors (A1R) and GFAP, while adenosine A2A receptors (A2AR) increased in the hippocampus and striatum and SNAP-25 was decreased in frontal cortex and striatum. Caffeine increased A1R in the striatum of bulbectomized mice and in SHAM-water group caffeine increased A2AR in the striatum and decreased SNAP-25 in the frontal cortex. Astrogliosis observed in the polymorphic layer of the dentate gyrus of OB mice was prevented by caffeine as well as the neurodegeneration in the striatum and piriform cortex. Based on these behavioral and neurochemical evidences, caffeine confirms its efficacy in preventing neurodegeneration associated with memory impairment and may be considered as a promising therapeutic tool in the prophylaxis and/or treatment of depression.


Asunto(s)
Cafeína/uso terapéutico , Estimulantes del Sistema Nervioso Central/uso terapéutico , Depresión/prevención & control , Depresión/psicología , Enfermedades Neurodegenerativas/prevención & control , Agitación Psicomotora/prevención & control , Agitación Psicomotora/psicología , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/patología , Gliosis/patología , Masculino , Trastornos de la Memoria/prevención & control , Trastornos de la Memoria/psicología , Ratones , Enfermedades Neurodegenerativas/patología , Bulbo Olfatorio , Receptor de Adenosina A1/efectos de los fármacos , Receptor de Adenosina A2A/efectos de los fármacos , Reconocimiento en Psicología/efectos de los fármacos , Proteína 25 Asociada a Sinaptosomas/metabolismo
12.
Neuropharmacology ; 166: 107782, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31756336

RESUMEN

Although some studies have supported the effects of caffeine for treatment of Attention deficit and hyperactivity disorder (ADHD), there were no evidences about its effects at the neuronal level. In this study, we sought to find morphological alterations during in vitro development of frontal cortical neurons from Spontaneoulsy hypertensive rats (SHR, an ADHD rat model) and Wistar-Kyoto rats (WKY, control strain). Further, we investigated the effects of caffeine and adenosine A1 and A2A receptors (A1R and A2AR) signaling. Cultured cortical neurons from WKY and SHR were analyzed by immunostaining of microtubule-associated protein 2 (MAP-2) and tau protein after treatment with either caffeine, or A1R and A2AR agonists or antagonists. Besides, the involvement of PI3K and not PKA signaling was also assessed. Neurons from ADHD model displayed less neurite branching, shorter maximal neurite length and decreased axonal outgrowth. While caffeine recovered neurite branching and elongation from ADHD neurons via both PKA and PI3K signaling, A2AR agonist (CGS 21680) promoted more neurite branching via PKA signaling. The selective A2AR antagonist (SCH 58261) was efficient in recovering axonal outgrowth from ADHD neurons through PI3K and not PKA signaling. For the first time, frontal cortical neurons were isolated from ADHD model and they presented disturbances in the differentiation and outgrowth. By showing that caffeine and A2AR may act at neuronal level rescuing ADHD neurons outgrowth, our findings strengthen the potential of caffeine and A2AR receptors as an adjuvant for ADHD treatment.


Asunto(s)
Agonistas del Receptor de Adenosina A2/uso terapéutico , Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Cafeína/farmacología , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/embriología , Neuronas/efectos de los fármacos , Antagonistas del Receptor de Adenosina A1/farmacología , Agonistas del Receptor de Adenosina A2/farmacología , Animales , Trastorno por Déficit de Atención con Hiperactividad/patología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Lóbulo Frontal/patología , Neuronas/patología , Embarazo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptor de Adenosina A2A , Xantinas/farmacología
13.
Mol Neurobiol ; 56(5): 3145-3158, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30105669

RESUMEN

Traumatic brain injury (TBI) is a leading cause of disability worldwide, triggering chronic neurodegeneration underlying cognitive and mood disorder still without therapeutic prospects. Based on our previous observations that guanosine (GUO) attenuates short-term neurochemical alterations caused by TBI, this study investigated the effects of chronical GUO treatment in behavioral, molecular, and morphological disturbances 21 days after trauma. Rats subject to TBI displayed mood (anxiety-like) and memory dysfunction. This was accompanied by a decreased expression of both synaptic (synaptophysin) and plasticity proteins (BDNF and CREB), a loss of cresyl violet-stained neurons, and increased astrogliosis and microgliosis in the hippocampus. Notably, chronic GUO treatment (7.5 mg/kg i.p. daily starting 1 h after TBI) prevented all these TBI-induced long-term behavioral, neurochemical, and morphological modifications. This neuroprotective effect of GUO was abrogated in the presence of the adenosine A1 receptor antagonist DPCPX (1 mg/kg) but unaltered by the adenosine A2A receptor antagonist SCH58261 (0.05 mg/kg). These findings show that a chronic GUO treatment prevents the long-term mood and memory dysfunction triggered by TBI, which involves adenosinergic receptors.


Asunto(s)
Conducta Animal/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Guanosina/uso terapéutico , Receptores Purinérgicos P1/metabolismo , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/etiología , Biomarcadores/metabolismo , Lesiones Traumáticas del Encéfalo/complicaciones , Gliosis/complicaciones , Gliosis/patología , Guanosina/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Trastornos de la Memoria/complicaciones , Microglía/efectos de los fármacos , Microglía/patología , Modelos Biológicos , Actividad Motora/efectos de los fármacos , Plasticidad Neuronal/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Ratas Wistar
14.
Neurochem Int ; 53(3-4): 89-94, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18620014

RESUMEN

Caffeine is one of the most psychostimulants consumed all over the world that usually presents positive effects on cognition. In this study, effects of caffeine on mice performance in the object recognition task were tested in different intertrial intervals. In addition, it was analyzed the effects of caffeine on brain derived neurotrophic factor (BDNF) and its receptor, TrkB, immunocontent to try to establish a connection between the behavioral finding and BDNF, one of the neurotrophins strictly involved in memory and learning process. CF1 mice were treated during 4 consecutive days with saline (0.9g%, i.p.) or caffeine (10mg/kg, i.p., equivalent dose corresponding to 2-3 cups of coffee). Caffeine treatment was interrupted 24h before the object recognition task analysis. In the test session performed 15min after training session, caffeine-treated mice recognized more efficiently both the familiar and the novel object. In the test session performed 90min and 24h after training session, caffeine did not change the time spent in the familiar object but increased the object recognition index, when compared to control group. Western blotting analysis of hippocampus from caffeine-treated mice revealed an increase in BDNF and TrkB immunocontent, compared to their saline-matched controls. Phospho-CREB immunocontent did not change with caffeine treatment. Our results suggest that acute treatment with caffeine improves recognition memory, and this effect may be related to an increase of the BDNF and TrkB immunocontent in the hippocampus.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cafeína/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Hipocampo/metabolismo , Reconocimiento Visual de Modelos/efectos de los fármacos , Receptor trkB/metabolismo , Animales , Estimulantes del Sistema Nervioso Central/farmacología , Cognición/efectos de los fármacos , Cognición/fisiología , Relación Dosis-Respuesta a Droga , Memoria/efectos de los fármacos , Memoria/fisiología , Ratones , Pruebas Neuropsicológicas , Reconocimiento Visual de Modelos/fisiología , Fosforilación/efectos de los fármacos , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
15.
Brain Res ; 1221: 134-40, 2008 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-18554575

RESUMEN

Glutamate is the main excitatory neurotransmitter in brain involved in pathophysiology of several brain injuries. In this context, glutamate showed to stimulate ecto-nucleotidase activities in cerebellar granule cells increasing extracellular adenosine levels, an important neuromodulator in the CNS able to prevent cell damage. The organoselenium compounds, such as ebselen and diphenyl diselenide [(PhSe)(2)], display neuroprotective activities mediated at least in part by their antioxidant and anti-inflammatory properties. Ebselen was described to prevent glutamate-induced lipid peroxidation and cell death in cerebellar granule cells and (PhSe)(2) modify glutamatergic synapse parameters in vitro and in vivo. In the present study, we investigated the effects of ebselen or (PhSe)(2) on glutamate-induced stimulation of ecto-nucleotidase activities in rat cultured cerebellar granule cells. Glutamate increased nucleotide hydrolysis at lower concentrations (10 and 100 microM) than described in the literature and this effect was counteracted by both organoselenium compounds tested. Based on these results, we investigated the association of organoselenium effects with their antioxidant properties searching for redox site modulation by using the alkylant agent N-ethylmaleimide (NEM). Our results suggest that selenium compounds, as well as the well-known antioxidant trolox, can avoid the increase on glutamate-induced stimulation of ecto-nucleotidase activities probably due to their antioxidant properties.


Asunto(s)
Adenosina Trifosfatasas/efectos de los fármacos , Daño Encefálico Crónico/tratamiento farmacológico , Degeneración Nerviosa/tratamiento farmacológico , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Selenio/farmacología , Adenosina Trifosfatasas/metabolismo , Animales , Antioxidantes/farmacología , Azoles/farmacología , Daño Encefálico Crónico/enzimología , Daño Encefálico Crónico/fisiopatología , Células Cultivadas , Cerebelo/citología , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Cromanos/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Etilmaleimida/farmacología , Ácido Glutámico/metabolismo , Isoindoles , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/fisiopatología , Neuronas/enzimología , Fármacos Neuroprotectores/metabolismo , Nucleótidos/metabolismo , Compuestos de Organoselenio/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Selenio/metabolismo
16.
Prog Neuropsychopharmacol Biol Psychiatry ; 32(6): 1508-15, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18579279

RESUMEN

Diphenyl diselenide [(PhSe)2] is an organoselenium compound which presents pharmacological antioxidant, anti-inflammatory, antinociceptive and antidepressant properties. The present study was designed to investigate the anxiolytic effect of (PhSe)2 in rats, employing the elevated plus maze task. The involvement of 5HT and GABA receptors in the anxiolytic-like effect was also evaluated. (PhSe)2 (5, 25 and 50 micromol/kg, i.p.) did not affect locomotor activity as evaluated in the open open-field test, and learning and memory when assessed in the inhibitory foot-shock avoidance task. However, (PhSe)2 at the 50 micromol/kg dose produced signs of an anxiolytic action, namely a decreased number of fecal boli in the open-field arena and an increased time spent in as well as an increased number of entries to the open arms of the elevated plus maze test. To evaluate the role of GABA and 5HT receptors in the anxiolytic-like effect of (PhSe)2, a selective GABAA receptor antagonist bicuculline, (0.75 mg/kg, i.p.), a non-selective 5HT2A/2C receptor antagonist, ritanserin (2 mg/kg, i.p.), a selective 5HT2A receptor antagonist, ketanserin (1 mg/kg, i.p.), and a selective 5HT1A receptor antagonist, WAY100635 (0.1 mg/kg, i.p.) were used. All the antagonists used were able to abolish the anxiolytic effect of (PhSe)2 suggesting that GABAA and 5HT receptors may play a role in the pharmacological property of this selenocompound in the central nervous system.


Asunto(s)
Ansiolíticos , Derivados del Benceno/farmacología , Compuestos de Organoselenio/farmacología , Receptores de GABA-A/efectos de los fármacos , Receptores de Serotonina/efectos de los fármacos , Animales , Reacción de Prevención/efectos de los fármacos , Bicuculina/farmacología , Defecación/efectos de los fármacos , Electrochoque , Conducta Exploratoria/efectos de los fármacos , Antagonistas del GABA/farmacología , Ketanserina/farmacología , Masculino , Piperazinas/farmacología , Piridinas/farmacología , Ratas , Ratas Wistar , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Receptor de Serotonina 5-HT2A/efectos de los fármacos , Receptor de Serotonina 5-HT2C/efectos de los fármacos , Ritanserina/farmacología , Antagonistas de la Serotonina/farmacología
17.
Artículo en Inglés | MEDLINE | ID: mdl-28826637

RESUMEN

Few studies have addressed the effects of caffeine in the puberty and/or adolescence in a sex dependent manner. Considering that caffeine intake has increased in this population, we investigated the behavioral and synaptic proteins changes in pubescent male and female rats after maternal consumption of caffeine. Adult female Wistar rats started to receive water or caffeine (0.1 and 0.3g/L in drinking water; low and moderate dose, respectively) during the active cycle at weekdays, two weeks before mating. The treatment lasted up to weaning and the offspring received caffeine until the onset of puberty (30-34days old). Behavioral tasks were performed to evaluate locomotor activity (open field task), anxious-like behavior (elevated plus maze task) and recognition memory (object recognition task) and synaptic proteins levels (proBDNF, BDNF, GFAP and SNAP-25) were verified in the hippocampus and cerebral cortex. While hyperlocomotion was observed in both sexes after caffeine treatment, anxiety-related behavior was attenuated by caffeine (0.3g/L) only in females. While moderate caffeine worsened recognition memory in females, an improvement in the long-term memory was observed in male rats for both doses. Coincident with memory improvement in males, caffeine increased pro- and BDNF in the hippocampus and cortex. Females presented increased proBDNF levels in both brain regions, with no effects of caffeine. While GFAP was not altered, moderate caffeine intake increased SNAP-25 in the cortex of female rats. Our findings revealed that caffeine promoted cognitive benefits in males associated with increased BDNF levels, while females showed less anxiety. Our findings revealed that caffeine promotes distinct behavioral outcomes and alterations in synaptic proteins during brain development in a sex dependent manner.


Asunto(s)
Ansiedad , Encéfalo/crecimiento & desarrollo , Cafeína/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Memoria/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Animales , Ansiedad/etiología , Ansiedad/metabolismo , Ansiedad/patología , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Agua Potable , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Memoria/fisiología , Actividad Motora/fisiología , Ratas Wistar , Caracteres Sexuales , Maduración Sexual , Proteína 25 Asociada a Sinaptosomas/metabolismo
18.
Cell Death Dis ; 9(3): 297, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29463792

RESUMEN

Despite the characteristic etiologies and phenotypes, different brain disorders rely on common pathogenic events. Glutamate-induced neurotoxicity is a pathogenic event shared by different brain disorders. Another event occurring in different brain pathological conditions is the increase of the extracellular ATP levels, which is now recognized as a danger and harmful signal in the brain, as heralded by the ability of P2 receptors (P2Rs) to affect a wide range of brain disorders. Yet, how ATP and P2R contribute to neurodegeneration remains poorly defined. For that purpose, we now examined the contribution of extracellular ATP and P2Rs to glutamate-induced neurodegeneration. We found both in vitro and in vivo that ATP/ADP through the activation of P2Y1R contributes to glutamate-induced neuronal death in the rat hippocampus. We found in cultured rat hippocampal neurons that the exposure to glutamate (100 µM) for 30 min triggers a sustained increase of extracellular ATP levels, which contributes to NMDA receptor (NMDAR)-mediated hippocampal neuronal death through the activation of P2Y1R. We also determined that P2Y1R is involved in excitotoxicity in vivo as the blockade of P2Y1R significantly attenuated rat hippocampal neuronal death upon the systemic administration of kainic acid or upon the intrahippocampal injection of quinolinic acid. This contribution of P2Y1R fades with increasing intensity of excitotoxic conditions, which indicates that P2Y1R is not contributing directly to neurodegeneration, rather behaving as a catalyst decreasing the threshold from which glutamate becomes neurotoxic. Moreover, we unraveled that such excitotoxicity process began with an early synaptotoxicity that was also prevented/attenuated by the antagonism of P2Y1R, both in vitro and in vivo. This should rely on the observed glutamate-induced calpain-mediated axonal cytoskeleton damage, most likely favored by a P2Y1R-driven increase of NMDAR-mediated Ca2+ entry selectively in axons. This may constitute a degenerative mechanism shared by different brain diseases, particularly relevant at initial pathogenic stages.


Asunto(s)
Ácido Glutámico/toxicidad , Enfermedades Neurodegenerativas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Muerte Celular , Femenino , Ácido Glutámico/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Masculino , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/citología , Neuronas/metabolismo , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/genética , Receptores Purinérgicos P2Y1/genética
19.
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
20.
Life Sci ; 81(25-26): 1668-76, 2007 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-17963786

RESUMEN

The role of excitotoxicity in the cerebral damage of glutaryl-CoA dehydrogenase deficiency (GDD) is under intense debate. We therefore investigated the in vitro effect of glutaric (GA) and 3-hydroxyglutaric (3-OHGA) acids, which accumulate in GDD, on [(3)H]glutamate uptake by slices and synaptosomal preparations from cerebral cortex and striatum of rats aged 7, 15 and 30 days. Glutamate uptake was significantly decreased by high concentrations of GA in cortical slices of 7-day-old rats, but not in cerebral cortex from 15- and 30-day-old rats and in striatum from all studied ages. Furthermore, this effect was not due to cellular death and was prevented by N-acetylcysteine preadministration, suggesting the involvement of oxidative damage. In contrast, glutamate uptake by brain slices was not affected by 3-OHGA exposure. Immunoblot analysis revealed that GLAST transporters were more abundant in the cerebral cortex compared to the striatum of 7-day-old rats. Moreover, the simultaneous addition of GA and dihydrokainate (DHK), a specific inhibitor of GLT1, resulted in a significantly higher inhibition of [(3)H]glutamate uptake by cortical slices of 7-day-old rats than that induced by the sole presence of DHK. We also observed that both GA and 3-OHGA exposure did not alter the incorporation of glutamate into synaptosomal preparations from cerebral cortex and striatum of rats aged 7, 15 and 30 days. Finally, GA in vivo administration did not alter glutamate uptake into cortical slices from 7-day-old rats. Our findings may explain at least in part why cortical neurons are more vulnerable to damage at birth as evidenced by the frontotemporal cortical atrophy observed in newborns affected by GDD.


Asunto(s)
Animales Recién Nacidos/metabolismo , Corteza Cerebral/metabolismo , Glutamatos/farmacocinética , Glutaratos/administración & dosificación , Glutaratos/metabolismo , Acetilcisteína/administración & dosificación , Acetilcisteína/metabolismo , Animales , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Glutamatos/metabolismo , Glutaril-CoA Deshidrogenasa/deficiencia , Técnicas In Vitro , Ácido Kaínico/análogos & derivados , Ácido Kaínico/metabolismo , Neostriado/metabolismo , Ratas , Ratas Wistar , Sinaptosomas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA