Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Glia ; 70(2): 368-378, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34726298

RESUMEN

Alzheimer's disease (AD) is associated with senile plaques of beta-amyloid (Aß) that affect the function of neurons and astrocytes. Brain activity results from the coordinated function of neurons and astrocytes in astroglial-neuronal networks. However, the effects of Aß on astroglial and neuronal network function remains unknown. Simultaneously monitoring astrocyte calcium and electric neuronal activities, we quantified the impact of Aß on sensory-evoked cortical activity in a mouse model of AD. At rest, cortical astrocytes displayed spontaneous hyperactivity that was related to Aß density. Sensory-evoked astrocyte responsiveness was diminished in AD mice, depending on the density and distance of Aß, and the responses showed altered calcium dynamics. Hence, astrocytes were spontaneously hyperactive but hypo-responsive to sensory stimulation. Finally, AD mice showed sensory-evoked electrical cortical hyperresponsiveness associated with altered astrocyte-neuronal network interplay. Our findings suggest dysfunction of astrocyte networks in AD mice may dysregulate cortical electrical activity and contribute to cognitive decline.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/farmacología , Animales , Astrocitos , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Neuronas , Placa Amiloide
2.
Neuroendocrinology ; 112(8): 796-806, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34666336

RESUMEN

INTRODUCTION: Parvalbumin (PV)-positive cells are strategic elements of neuronal networks capable of influencing memory and learning processes. However, it is not known whether pituitary hormones may be related to PV expression in the hippocampus - a part of the limbic system with important functions in learning and memory. OBJECTIVE: Since previous studies indicate that prolactin (PRL) plays a significant role in hippocampal-dependent learning and synaptic plasticity, we hypothesized that a rise in PRL levels can modify PV expression in the hippocampus. METHODS: We employed biochemical, immunohistochemistry, and densitometry techniques - as well as a behavioural assay - in a hyperprolactinemia model using subcutaneous osmotic pumps in female mice. RESULTS: PRL treatment via osmotic pump induced an increase in PRL receptor (PRLR) expression in most regions of the hippocampus analysed by Western blotting and immunohistochemistry methods. Fluorescent densitometry analysis revealed that PV expression decreases in the same layers in the hippocampus following PRL treatment, while double labelling immunostaining indicated close localization of PV and PRLR in PV-positive interneurons. In addition, we found that PRL induced a reduction in the ß2/3 subunit of GABAA receptor (GABAAR) expression that was linearly correlated with the reduction in PV expression. This reduction in the ß2/3 subunit of GABAAR expression was maintained in trained animals in which PRL treatment improved the learning of a spatial memory task. CONCLUSIONS: These data show, for the first time, that an increase in PRL level is associated with changes in key constituent elements of inhibitory circuits in the hippocampus and may be of relevance for the alterations in cognitive function reported in hyperprolactinemia.


Asunto(s)
Hipocampo , Hiperprolactinemia , Parvalbúminas , Prolactina , Receptores de GABA-A , Animales , Femenino , Hipocampo/metabolismo , Ratones , Parvalbúminas/metabolismo , Prolactina/farmacología , Receptores de GABA-A/metabolismo , Receptores de Prolactina/metabolismo
3.
Hippocampus ; 31(3): 281-293, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33285014

RESUMEN

Dynamic signaling between the endocrine system (ES) and the nervous system (NS) is essential for brain and body homeostasis. In particular, reciprocal interaction occurs during pregnancy and motherhood that may involve changes in some brain plasticity processes. Prolactin (PRL), a hormone with pleiotropic effects on the NS, promotes maternal behavior and has been linked to modifications in brain circuits during motherhood; however, it is unclear whether PRL may regulate synaptic plasticity. Therefore, the main aim of the present work was to determine the cellular and molecular mechanisms triggered by PRL that regulate synaptic plasticity in the hippocampus. By analyzing extracellular recordings in CA3-CA1 synapses of hippocampal slices, we report that PRL modifies short and long-term synaptic plasticity in female mice of reproductive age, but not in sexually immature females or adult males. This effect is carried out through mechanisms that include participation of GABAA receptors and activation of the JAK2-mediated signaling pathway. These findings show for the first time how PRL enhances the synaptic strength in hippocampal circuits and that this effect is sexually dimorphic, which would influence complex brain processes in physiological conditions like pregnancy and lactation.


Asunto(s)
Plasticidad Neuronal , Prolactina , Animales , Femenino , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Plasticidad Neuronal/fisiología , Receptores de GABA-A , Sinapsis/fisiología , Transmisión Sináptica/fisiología
4.
Mov Disord ; 35(9): 1636-1648, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32666590

RESUMEN

BACKGROUND: Numerous studies indicate an association between neurodegenerative and metabolic diseases. Although still a matter of debate, growing evidence from epidemiological and animal studies indicate that preexisting diabetes increases the risk to develop Parkinson's disease. However, the mechanisms of such an association are unknown. OBJECTIVES: We investigated whether diabetes alters striatal dopamine neurotransmission and assessed the vulnerability of nigrostriatal neurons to neurodegeneration. METHODS: We used streptozotocin-treated and genetically diabetic db/db mice. Expression of oxidative stress and nigrostriatal neuronal markers and levels of dopamine and its metabolites were monitored. Dopamine release and uptake were assessed using fast-scan cyclic voltammetry. 6-Hydroxydopamine was unilaterally injected into the striatum using stereotaxic surgery. Motor performance was scored using specific tests. RESULTS: Diabetes resulted in oxidative stress and decreased levels of dopamine and its metabolites in the striatum. Levels of proteins regulating dopamine release and uptake, including the dopamine transporter, the Girk2 potassium channel, the vesicular monoamine transporter 2, and the presynaptic vesicle protein synaptobrevin-2, were decreased in diabetic mice. Electrically evoked levels of extracellular dopamine in the striatum were enhanced, and altered dopamine uptake was observed. Striatal microinjections of a subthreshold dose of the neurotoxin 6-hydroxydopamine in diabetic mice, insufficient to cause motor alterations in nondiabetic animals, resulted in motor impairment, higher loss of striatal dopaminergic axons, and decreased neuronal cell bodies in the substantia nigra. CONCLUSIONS: Our results indicate that diabetes promotes striatal oxidative stress, alters dopamine neurotransmission, and increases vulnerability to neurodegenerative damage leading to motor impairment. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Diabetes Mellitus Experimental , Dopamina , Animales , Cuerpo Estriado/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Ratones , Sustancia Negra/metabolismo , Transmisión Sináptica
5.
Hum Mol Genet ; 24(25): 7265-85, 2015 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-26464483

RESUMEN

Cognitive dysfunction is an early clinical hallmark of Huntington's disease (HD) preceding the appearance of motor symptoms by several years. Neuronal dysfunction and altered corticostriatal connectivity have been postulated to be fundamental to explain these early disturbances. However, no treatments to attenuate cognitive changes have been successful: the reason may rely on the idea that the temporal sequence of pathological changes is as critical as the changes per se when new therapies are in development. To this aim, it becomes critical to use HD mouse models in which cognitive impairments appear prior to motor symptoms. In this study, we demonstrate procedural memory and motor learning deficits in two different HD mice and at ages preceding motor disturbances. These impairments are associated with altered corticostriatal long-term potentiation (LTP) and specific reduction of dendritic spine density and postsynaptic density (PSD)-95 and spinophilin-positive clusters in the cortex of HD mice. As a potential mechanism, we described an early decrease of Kalirin-7 (Kal7), a guanine-nucleotide exchange factor for Rho-like small GTPases critical to maintain excitatory synapse, in the cortex of HD mice. Supporting a role for Kal7 in HD synaptic deficits, exogenous expression of Kal7 restores the reduction of excitatory synapses in HD cortical cultures. Altogether, our results suggest that cortical dysfunction precedes striatal disturbances in HD and underlie early corticostriatal LTP and cognitive defects. Moreover, we identified diminished Kal7 as a key contributor to HD cortical alterations, placing Kal7 as a molecular target for future therapies aimed to restore corticostriatal function in HD.


Asunto(s)
Cuerpo Estriado/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Enfermedad de Huntington/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Animales , Electrofisiología , Femenino , Factores de Intercambio de Guanina Nucleótido/genética , Inmunohistoquímica , Masculino , Ratones , Microscopía Confocal , Transmisión Sináptica/genética
6.
Addict Biol ; 22(6): 1706-1718, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27457910

RESUMEN

Caffeine has cognitive-enhancing properties with effects on learning and memory, concentration, arousal and mood. These effects imply changes at circuital and synaptic level, but the mechanism by which caffeine modifies synaptic plasticity remains elusive. Here we report that caffeine, at concentrations representing moderate to high levels of consumption in humans, induces an NMDA receptor-independent form of LTP (CAF LTP) in the CA1 region of the hippocampus by promoting calcium-dependent secretion of BDNF, which subsequently activates TrkB-mediated signaling required for the expression of CAF LTP. Our data include the novel observation that insulin receptor substrate 2 (IRS2) is phosphorylated during induction of CAF LTP, a process that requires cytosolic free Ca2+ . Consistent with the involvement of IRS2 signals in caffeine-mediated synaptic plasticity, phosphorylation of Akt (Ser473) in response to LTP induction is defective in Irs2-/- mice, demonstrating that these plasticity changes are associated with downstream targets of the phosphoinositide 3-kinase (PI3K) pathway. These findings indicate that TrkB-IRS2 signals are essential for activation of PI3K during the induction of LTP by caffeine.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cafeína/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Proteínas Sustrato del Receptor de Insulina/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Femenino , Proteínas Sustrato del Receptor de Insulina/efectos de los fármacos , Proteínas Sustrato del Receptor de Insulina/genética , Masculino , Ratones , Modelos Animales
7.
Glia ; 64(11): 1962-71, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27462832

RESUMEN

Previous findings indicate that reducing brain insulin-like growth factor I receptor (IGF-IR) activity promotes ample neuroprotection. We now examined a possible action of IGF-IR on brain glucose transport to explain its wide protective activity, as energy availability is crucial for healthy tissue function. Using (18) FGlucose PET we found that shRNA interference of IGF-IR in mouse somatosensory cortex significantly increased glucose uptake upon sensory stimulation. In vivo microscopy using astrocyte specific staining showed that after IGF-IR shRNA injection in somatosensory cortex, astrocytes displayed greater increases in glucose uptake as compared to astrocytes in the scramble-injected side. Further, mice with the IGF-IR knock down in astrocytes showed increased glucose uptake in somatosensory cortex upon sensory stimulation. Analysis of underlying mechanisms indicated that IGF-IR interacts with glucose transporter 1 (GLUT1), the main facilitative glucose transporter in astrocytes, through a mechanism involving interactions with the scaffolding protein GIPC and the multicargo transporter LRP1 to retain GLUT1 inside the cell. These findings identify IGF-IR as a key modulator of brain glucose metabolism through its inhibitory action on astrocytic GLUT1 activity. GLIA 2016;64:1962-1971.


Asunto(s)
Astrocitos/metabolismo , Glucosa/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , 4-Cloro-7-nitrobenzofurazano/análogos & derivados , 4-Cloro-7-nitrobenzofurazano/farmacología , Animales , Animales Recién Nacidos , Biotinilación , Encéfalo/citología , Encéfalo/diagnóstico por imagen , Células Cultivadas , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Glucosamina/análogos & derivados , Glucosamina/farmacología , Transportador de Glucosa de Tipo 1/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Factor I del Crecimiento Similar a la Insulina/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Estimulación Física , Transporte de Proteínas/genética , ARN Mensajero/metabolismo , Transfección , Vibrisas/fisiología
8.
J Neurosci ; 34(38): 12738-44, 2014 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-25232111

RESUMEN

Experience-dependent plasticity of synaptic transmission, which represents the cellular basis of learning, is accompanied by morphological changes in dendritic spines. Astrocytic processes are intimately associated with synapses, structurally enwrapping and functionally interacting with dendritic spines and synaptic terminals by responding to neurotransmitters and by releasing gliotransmitters that regulate synaptic function. While studies on structural synaptic plasticity have focused on neuronal elements, the structural-functional plasticity of astrocyte-neuron relationships remains poorly known. Here we show that stimuli inducing hippocampal synaptic LTP enhance the motility of synapse-associated astrocytic processes. This motility increase is relatively rapid, starting <5 min after the stimulus, and reaching a maximum in 20-30 min (t(1/2) = 10.7 min). It depends on presynaptic activity and requires G-protein-mediated Ca(2+) elevations in astrocytes. The structural remodeling is accompanied by changes in the ability of astrocytes to regulate synaptic transmission. Sensory stimuli that increase astrocyte Ca(2+) also induce similar plasticity in mouse somatosensory cortex in vivo. Therefore, structural relationships between astrocytic processes and dendritic spines undergo activity-dependent changes with metaplasticity consequences on synaptic regulation. These results reveal novel forms of synaptic plasticity based on structural-functional changes of astrocyte-neuron interactions.


Asunto(s)
Astrocitos/fisiología , Espinas Dendríticas/fisiología , Plasticidad Neuronal/fisiología , Potenciales de Acción/fisiología , Animales , Astrocitos/citología , Astrocitos/metabolismo , Calcio/metabolismo , Femenino , Hipocampo/fisiología , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Corteza Somatosensorial/fisiología , Transmisión Sináptica/fisiología
9.
PLoS Biol ; 10(2): e1001259, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22347811

RESUMEN

Long-term potentiation (LTP) of synaptic transmission represents the cellular basis of learning and memory. Astrocytes have been shown to regulate synaptic transmission and plasticity. However, their involvement in specific physiological processes that induce LTP in vivo remains unknown. Here we show that in vivo cholinergic activity evoked by sensory stimulation or electrical stimulation of the septal nucleus increases Ca²âº in hippocampal astrocytes and induces LTP of CA3-CA1 synapses, which requires cholinergic muscarinic (mAChR) and metabotropic glutamate receptor (mGluR) activation. Stimulation of cholinergic pathways in hippocampal slices evokes astrocyte Ca²âº elevations, postsynaptic depolarizations of CA1 pyramidal neurons, and LTP of transmitter release at single CA3-CA1 synapses. Like in vivo, these effects are mediated by mAChRs, and this cholinergic-induced LTP (c-LTP) also involves mGluR activation. Astrocyte Ca²âº elevations and LTP are absent in IP3R2 knock-out mice. Downregulating astrocyte Ca²âº signal by loading astrocytes with BAPTA or GDPßS also prevents LTP, which is restored by simultaneous astrocyte Ca²âº uncaging and postsynaptic depolarization. Therefore, cholinergic-induced LTP requires astrocyte Ca²âº elevations, which stimulate astrocyte glutamate release that activates mGluRs. The cholinergic-induced LTP results from the temporal coincidence of the postsynaptic activity and the astrocyte Ca²âº signal simultaneously evoked by cholinergic activity. Therefore, the astrocyte Ca²âº signal is necessary for cholinergic-induced synaptic plasticity, indicating that astrocytes are directly involved in brain storage information.


Asunto(s)
Astrocitos/fisiología , Neuronas Colinérgicas/fisiología , Potenciación a Largo Plazo , Sinapsis/fisiología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Atropina/farmacología , Señalización del Calcio , Neuronas Colinérgicas/efectos de los fármacos , Neuronas Colinérgicas/metabolismo , Ácido Glutámico/metabolismo , Glicina/análogos & derivados , Glicina/farmacología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antagonistas Muscarínicos/farmacología , Ratas , Ratas Wistar , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Transmisión Sináptica/efectos de los fármacos
10.
PLoS Biol ; 10(2): e1001262, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22363206

RESUMEN

Cell adhesion molecules and downstream growth factor-dependent signaling are critical for brain development and synaptic plasticity, and they have been linked to cognitive function in adult animals. We have previously developed a mimetic peptide (FGL) from the neural cell adhesion molecule (NCAM) that enhances spatial learning and memory in rats. We have now investigated the cellular and molecular basis of this cognitive enhancement, using biochemical, morphological, electrophysiological, and behavioral analyses. We have found that FGL triggers a long-lasting enhancement of synaptic transmission in hippocampal CA1 neurons. This effect is mediated by a facilitated synaptic delivery of AMPA receptors, which is accompanied by enhanced NMDA receptor-dependent long-term potentiation (LTP). Both LTP and cognitive enhancement are mediated by an initial PKC activation, which is followed by persistent CaMKII activation. These results provide a mechanistic link between facilitation of AMPA receptor synaptic delivery and improved hippocampal-dependent learning, induced by a pharmacological cognitive enhancer.


Asunto(s)
Cognición/fisiología , Hipocampo/citología , Potenciación a Largo Plazo/efectos de los fármacos , Moléculas de Adhesión de Célula Nerviosa/farmacología , Neuronas/efectos de los fármacos , Receptores AMPA/metabolismo , Transmisión Sináptica/efectos de los fármacos , Análisis de Varianza , Animales , Western Blotting , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Ensayo de Inmunoadsorción Enzimática , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Microscopía Electrónica , Microscopía Fluorescente , Neuronas/fisiología , Técnicas de Placa-Clamp , Fosforilación , Proteína Quinasa C/metabolismo , Ratas , Ratas Wistar , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología
11.
Brain Pathol ; : e13279, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38887180

RESUMEN

Voltage-gated CaV2.1 (P/Q-type) Ca2+ channels play a crucial role in regulating neurotransmitter release, thus contributing to synaptic plasticity and to processes such as learning and memory. Despite their recognized importance in neural function, there is limited information on their potential involvement in neurodegenerative conditions such as Alzheimer's disease (AD). Here, we aimed to explore the impact of AD pathology on the density and nanoscale compartmentalization of CaV2.1 channels in the hippocampus in association with GABAB receptors. Histoblotting experiments showed that the density of CaV2.1 channel was significantly reduced in the hippocampus of APP/PS1 mice in a laminar-dependent manner. CaV2.1 channel was enriched in the active zone of the axon terminals and was present at a very low density over the surface of dendritic tree of the CA1 pyramidal cells, as shown by quantitative SDS-digested freeze-fracture replica labelling (SDS-FRL). In APP/PS1 mice, the density of CaV2.1 channel in the active zone was significantly reduced in the strata radiatum and lacunosum-moleculare, while it remained unaltered in the stratum oriens. The decline in Cav2.1 channel density was found to be associated with a corresponding impairment in the GABAergic synaptic function, as evidenced by electrophysiological experiments carried out in the hippocampus of APP/PS1 mice. Remarkably, double SDS-FRL showed a co-clustering of CaV2.1 channel and GABAB1 receptor in nanodomains (~40-50 nm) in wild type mice, while in APP/PS1 mice this nanoarchitecture was absent. Together, these findings suggest that the AD pathology-induced reduction in CaV2.1 channel density and CaV2.1-GABAB1 de-clustering may play a role in the synaptic transmission alterations shown in the AD hippocampus. Therefore, uncovering these layer-dependent changes in P/Q calcium currents associated with AD pathology can benefit the development of future strategies for AD management.

12.
Neurobiol Dis ; 58: 231-41, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23800715

RESUMEN

Brain ischemia triggers excessive release of neurotransmitters that mediate neuronal damage following ischemic injury. The striatum is one of the areas most sensitive to ischemia. Release of dopamine (DA) from ischemic neurons is neurotoxic and directly contributes to the cell death in affected areas. Astrocytes are known to be critically involved in the physiopathology of cerebrovascular disease. However, their response to ischemia and their role in neuroprotection in striatum are not completely understood. In this study, we used an in vitro model to evaluate the mechanisms of ischemia-induced DA release, and to study whether astrocytes modulate the release of DA in response to short-term ischemic conditions. Using slices of adult mouse brain exposed to oxygen and glucose deprivation (OGD), we measured the OGD-evoked DA efflux using fast cyclic voltammetry and also assessed metabolic impairment by 2,3,5-triphenyltetrazolium chloride (TTC) and tissue viability by propidium iodide (PI) staining. Our data indicate that ischemia induces massive release of DA by dual mechanisms: one which operates via vesicular exocytosis and is action potential dependent and another involving reverse transport by the dopamine transporter (DAT). Simultaneous blockade of astrocyte glutamate transporters and DAT prevented the massive release of dopamine and reduced the brain tissue damage. The present results provide the first experimental evidence that astrocytes function as a key cellular element of ischemia-induced DA release in striatum, constituting a novel and promising therapeutic target in ischemia.


Asunto(s)
Astrocitos/metabolismo , Cuerpo Estriado/patología , Dopamina/metabolismo , Hipoxia-Isquemia Encefálica/patología , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Animales , Ácido Aspártico/análogos & derivados , Ácido Aspártico/farmacología , Astrocitos/efectos de los fármacos , Calcio/metabolismo , Cuerpo Estriado/efectos de los fármacos , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Lateralidad Funcional , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/patología , Oxidopamina/farmacología , Piperazinas/farmacología , Tetrodotoxina/farmacología , Factores de Tiempo
13.
Neurobiol Dis ; 52: 117-27, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23220201

RESUMEN

The cognitive dysfunctions of Down Syndrome (DS) individuals are the most disabling alterations caused by the trisomy of human chromosome 21 (HSA21). In trisomic Ts65Dn mice, a genetic model for DS, the overexpression of HSA21 homologous genes has been associated with strong visuo-spatial cognitive alterations, ascribed to hippocampal dysfunction. In the present study, we evaluated whether the normalization of the expression levels of Dyrk1A (Dual specificity tyrosine-phosphorylation-regulated kinase 1A), a candidate gene for DS, might correct hippocampal defects in Ts65Dn mice. In the hippocampus of 2 month-old Ts65Dn mice, such normalization was achieved through the stereotaxical injection of adeno-associated viruses containing a short hairpin RNA against Dyrk1A (AAV2/1-shDyrk1A) and a luciferase reporter gene. The injected hippocampi were efficiently transduced, as shown by bioluminescence in vivo imaging, luciferase activity quantification and immunohistochemical analysis. At the molecular level, viral infusion allowed the normalization of the targeted Dyrk1A expression, as well as of the key players of the MAPK/CREB pathway. The electrophysiological recordings of hippocampal slices from Ts65Dn mice injected with AAV2/1-shDyrk1A displayed attenuation of the synaptic plasticity defects of trisomic mice. In contrast, contralateral hippocampal injection with an AAV2/1 control virus containing a scrambled sequence, showed neither the normalization of Dyrk1A levels nor changes of synaptic plasticity. In the Morris water maze task, although long-term consolidation of the task was not achieved, treated Ts65Dn mice displayed initially a normalized thigmotactic behavior, similar to euploid littermates, indicating the partial improvement in their hippocampal-dependent search strategy. Taken together, these results show Dyrk1A as a critical player in the pathophysiology of DS and define Dyrk1A as a therapeutic target in adult trisomic mice.


Asunto(s)
Síndrome de Down/fisiopatología , Hipocampo/fisiopatología , Aprendizaje por Laberinto/fisiología , Plasticidad Neuronal/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Sinapsis/genética , Animales , Conducta Animal/fisiología , Dependovirus , Modelos Animales de Enfermedad , Síndrome de Down/genética , Síndrome de Down/metabolismo , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Quinasas DyrK
14.
Cereb Cortex ; 22(8): 1717-27, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21955917

RESUMEN

The beneficial effects of insulin and insulin-like growth factor I on cognition have been documented in humans and animal models. Conversely, obesity, hyperinsulinemia, and diabetes increase the risk for neurodegenerative disorders including Alzheimer's disease (AD). However, the mechanisms by which insulin regulates synaptic plasticity are not well understood. Here, we report that complete disruption of insulin receptor substrate 2 (Irs2) in mice impairs long-term potentiation (LTP) of synaptic transmission in the hippocampus. Basal synaptic transmission and paired-pulse facilitation were similar between the 2 groups of mice. Induction of LTP by high-frequency conditioning tetanus did not activate postsynaptic N-methyl-D-aspartate (NMDA) receptors in hippocampus slices from Irs2(-/-) mice, although the expression of NR2A, NR2B, and PSD95 was equivalent to wild-type controls. Activation of Fyn, AKT, and MAPK in response to tetanus stimulation was defective in Irs2(-/-) mice. Interestingly, IRS2 was phosphorylated during induction of LTP in control mice, revealing a potential new component of the signaling machinery which modulates synaptic plasticity. Given that IRS2 expression is diminished in Type 2 diabetics as well as in AD patients, these data may reveal an explanation for the prevalence of cognitive decline in humans with metabolic disorders by providing a mechanistic link between insulin resistance and impaired synaptic transmission.


Asunto(s)
Proteínas Sustrato del Receptor de Insulina/metabolismo , Potenciación a Largo Plazo/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/fisiología , Animales , Western Blotting , Femenino , Hipocampo/metabolismo , Inmunoprecipitación , Proteínas Sustrato del Receptor de Insulina/deficiencia , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp
15.
J Neuroimmune Pharmacol ; 18(3): 529-550, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37698780

RESUMEN

Sirtuin 2 (SIRT2) has been proposed to have a central role on aging, inflammation, cancer and neurodegenerative diseases; however, its specific function remains controversial. Recent studies propose SIRT2 pharmacological inhibition as a therapeutic strategy for several neurodegenerative diseases including Alzheimer's disease (AD). Surprisingly, none of these published studies regarding the potential interest of SIRT2 inhibition has assessed the peripheral adverse side consequences of this treatment. In this study, we demonstrate that the specific SIRT2 inhibitor, the compound 33i, does not exhibit genotoxic or mutagenic properties. Moreover, pharmacological treatment with 33i, improved cognitive dysfunction and long-term potentiation, reducing amyloid pathology and neuroinflammation in the APP/PS1 AD mouse model. However, this treatment increased peripheral levels of the inflammatory cytokines IL-1ß, TNF, IL-6 and MCP-1. Accordingly, peripheral SIRT2 inhibition with the blood brain barrier impermeable compound AGK-2, worsened the cognitive capacities and increased systemic inflammation. The analysis of human samples revealed that SIRT2 is increased in the brain but not in the serum of AD patients. These results suggest that, although SIRT2 pharmacological inhibition may have beneficial consequences in neurodegenerative diseases, its pharmacological inhibition at the periphery would not be recommended and the systemic adverse side effects should be considered. This information is essential to maximize the therapeutic potential of SIRT2 inhibition not only for AD but also for other neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer , Sirtuina 2 , Animales , Humanos , Ratones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Inflamación/inducido químicamente , Inflamación/patología , Ratones Transgénicos , Sirtuina 2/antagonistas & inhibidores , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/patología
16.
Neurobiol Dis ; 48(3): 271-81, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22820144

RESUMEN

Tyrosine hydroxylase (TH)-immunoreactive (ir) neurons have been found in the striatum after dopamine depletion; however, little is known about the mechanism underlying their appearance or their functional significance. We previously showed an increase in striatal TH-ir neurons after L-DOPA treatment in mice with unilateral 6-OHDA lesions in the striatum. In the present study, we further examined the time-course and persistence of the effects of chronic L-DOPA treatment on the appearance and regulation of TH-ir neurons as well as their possible function. We found that the L-DOPA-induced increase in striatal TH-ir neurons is dose-dependent and persists for days after L-DOPA withdrawal, decreasing significantly 10 days after L-DOPA treatment ends. Using hemiparkinsonian D1 receptor knock-out (D1R-/-) and D2 receptor knock-out (D2R-/-) mice, we found that the D1R, but not the D2R, is required for the L-DOPA-induced appearance of TH-ir neurons in the dopamine-depleted striatum. Interestingly, our experiments in aphakia mice, which lack Pitx3 expression in the brain, indicate that the L-DOPA-dependent increase in the number of TH-ir neurons is independent of Pitx3, a transcription factor necessary for the development of mesencephalic dopaminergic neurons. To explore the possible function of L-DOPA-induced TH-ir neurons in the striatum, we examined dopamine overflow and forelimb use in L-DOPA-treated parkinsonian mice. These studies revealed a tight spatio-temporal correlation between the presence of striatal TH-ir neurons, the recovery of electrically stimulated dopamine overflow in the lesioned striatum, and the recovery of contralateral forelimb use with chronic L-DOPA treatment. Our results suggest that the presence of TH-ir neurons in the striatum may underlie the long-duration response to L-DOPA following withdrawal. Promotion of these neurons in the early stages of Parkinson's disease, when dopamine denervation is incomplete, may be beneficial for maintaining motor function.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Dopaminérgicos/farmacología , Levodopa/farmacología , Neuronas/efectos de los fármacos , Trastornos Parkinsonianos/metabolismo , Animales , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/fisiología , Neuronas/citología , Neuronas/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
17.
Glia ; 59(1): 94-107, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20967884

RESUMEN

Astrocytes react to brain injury triggering neuroinflammatory processes that determine the degree of neuronal damage. However, the signaling events associated to astrocyte activation remain largely undefined. The nuclear factor of activated T-cells (NFAT) is a transcription factor family implicated in activation of immune cells. We previously characterized the expression of NFAT isoforms in cultured astrocytes, and NFAT activation in response to mechanical lesion. Here we analyze NFATc3 in two mouse models of inflammatory brain damage: hippocampal excitotoxicity induced by intracerebral kainic acid (KA) injection and cortical mechanical lesion. Immunofluorescence results demonstrated that NFATc3 is specifically induced in a subset of reactive astrocytes, and not in microglia or neurons. In KA-treated brains, NFATc3 expression is transient and NFATc3-positive astrocytes concentrate around damaged neurons in areas CA3 and CA1. Complementary Western blot and RT-PCR analysis revealed an NFAT-dependent induction of RCAN1-4 and COX-2 in hippocampus as soon as 6 h after KA exposure, indicating that NFAT activation precedes NFATc3 over-expression. Moreover, activation of NFAT by ATP increased NFATc3 mRNA levels in astrocyte cultures, suggesting that NFATc3 expression is controlled through an auto-regulatory loop. Meanwhile, stab wound enhanced NFATc3 expression specifically in a subclass of reactive astrocytes confined within the proximal layer of the glial scar, and GFAP immunoreactivity was attenuated in NFATc3-expressing astrocytes. In conclusion, our work establishes NFATc3 as a marker of activation for a specific population of astrocytes in response to brain damage, which may have consequences for neuronal survival.


Asunto(s)
Astrocitos/metabolismo , Lesiones Encefálicas/metabolismo , Factores de Transcripción NFATC/metabolismo , Análisis de Varianza , Animales , Astrocitos/patología , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Encefálicas/etiología , Lesiones Encefálicas/patología , Muerte Celular , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Ácido Kaínico/farmacología , Ratones , Microscopía Confocal , Factores de Transcripción NFATC/genética , Neuronas/metabolismo , Neuronas/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Neurobiol Dis ; 42(3): 391-403, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21303698

RESUMEN

Methamphetamine (METH) and 3,4-methylenedioxymethamphetamine (MDMA), amphetamine derivatives widely used as recreational drugs, induce similar neurotoxic effects in mice, including a marked loss of tyrosine hydroxylase (TH) and dopamine transporter (DAT) in the striatum. Although the role of dopamine in these neurotoxic effects is well established and pharmacological studies suggest involvement of a dopamine D2-like receptor, the specific dopamine receptor subtype involved has not been determined. In this study, we used dopamine D2 receptor knock-out mice (D2R(-/-)) to determine whether D2R is involved in METH- and MDMA-induced hyperthermia and neurotoxicity. In wild type animals, both drugs induced marked hyperthermia, decreased striatal dopamine content and TH- and DAT-immunoreactivity and increased striatal GFAP and Mac-1 expression as well as iNOS and interleukin 15 at 1 and 7days after drug exposure. They also caused dopaminergic cell loss in the SNpc. Inactivation of D2R blocked all these effects. Remarkably, D2R inactivation prevented METH-induced loss of dopaminergic neurons in the SNpc. In addition, striatal dopamine overflow, measured by fast scan cyclic voltammetry in the presence of METH, was significantly reduced in D2R(-/-) mice. Pre-treatment with reserpine indicated that the neuroprotective effect of D2R inactivation cannot be explained solely by its ability to prevent METH-induced hyperthermia: reserpine lowered body temperature in both genotypes, and potentiated METH toxicity in WT, but not D2R(-/-) mice. Our results demonstrate that the D2R is necessary for METH and MDMA neurotoxicity and that the neuroprotective effect of D2R inactivation is independent of its effect on body temperature.


Asunto(s)
Estimulantes del Sistema Nervioso Central/toxicidad , Dopamina/metabolismo , Metanfetamina/toxicidad , N-Metil-3,4-metilenodioxianfetamina/toxicidad , Neuronas/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Receptores de Dopamina D2/metabolismo , Análisis de Varianza , Animales , Temperatura Corporal/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Inmunohistoquímica , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/genética , Receptores de Dopamina D2/genética , Tirosina 3-Monooxigenasa/metabolismo
19.
Psychoneuroendocrinology ; 124: 105048, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33249333

RESUMEN

Beyond the direct physiological functions associated with motherhood in mammals, previous studies have suggested the potential role of prolactin (Prl) in distinct brain processes such as neuroprotection, neurogenesis, and stress responses. However, the cognitive influence of Prl remains unclear, particularly regarding the mechanisms of acquisition, consolidation and retrieval of information in the brain. Using chronic implanted electrodes in freely moving female mice combined with behavioral tests, we investigated the rhythmic activity changes induced by Prl in a model of hippocampus-dependent learning and memory. Our results show that Prl improves the learning of a spatial memory task in the acquisition stage. The main variations at the circuitry level were in the theta frequency band (4-8 Hz and 8-12 Hz), marked by a faster change in oscillatory activity with no modifications to higher frequencies. These results show that Prl plays a significant role in the acquisition of information during learning of a spatial memory task, suggesting that an increase in Prl levels may induce changes in circuital network plasticity.


Asunto(s)
Aprendizaje Espacial , Animales , Femenino , Hipocampo , Ratones , Neurogénesis , Prolactina
20.
Epilepsia ; 51(3): 384-90, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19732136

RESUMEN

PURPOSE: In this study, we explore the antiepileptic effects of flufenamic acid (FFA) in order to identify the cellular mechanisms that underlie the potential anticonvulsant properties of this nonsteroidal antiinflammatory compound. METHODS: The mechanisms of FFA action were analyzed using an in vitro model in which epileptiform activity was induced in hippocampal slices by perfusion with 100 microm 4-aminopyridine (4-AP) added to a modified Mg(2+)-free solution. The activity of CA1 pyramidal neurons as well as the synaptic connection between CA3 and CA1 was monitored using extracellular and patch-clamp recordings. RESULTS: Epileptiform activity was suppressed in hippocampal neurons by FFA at concentrations between 50 and 200 microm. Glutamatergic excitatory synaptic transmission was diminished by FFA without modifying recurrent gamma-aminobutyric acid (GABA)ergic synaptic inhibition. Several lines of evidence indicated that FFA did not decrease neurotransmitter release probability, implicating a postsynaptic mechanism of action. FFA also potently reduced neuronal excitability, but did not alter the amplitude, duration, or undershoot of action potentials. CONCLUSIONS: Our results suggest that FFA exerts an anticonvulsive effect on hippocampal pyramidal neurons by simultaneously decreasing glutamatergic excitatory synaptic activity and reducing neuronal excitability. Therefore, our study provides experimental evidence that FFA may represent an effective pharmacologic agent in the treatment of epilepsy in the mammalian central nervous system.


Asunto(s)
Antiinflamatorios/farmacología , Anticonvulsivantes/farmacología , Epilepsia/prevención & control , Epilepsia/fisiopatología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ácido Flufenámico/farmacología , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , 4-Aminopiridina/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/farmacología , Epilepsia/tratamiento farmacológico , Hipocampo/fisiología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Inhibición Neural/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp , Células Piramidales/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Ácido gamma-Aminobutírico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA