Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(34): e2312511121, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39141354

RESUMEN

Schizophrenia phenotypes are suggestive of impaired cortical plasticity in the disease, but the mechanisms of these deficits are unknown. Genomic association studies have implicated a large number of genes that regulate neuromodulation and plasticity, indicating that the plasticity deficits have a genetic origin. Here, we used biochemically detailed computational modeling of postsynaptic plasticity to investigate how schizophrenia-associated genes regulate long-term potentiation (LTP) and depression (LTD). We combined our model with data from postmortem RNA expression studies (CommonMind gene-expression datasets) to assess the consequences of altered expression of plasticity-regulating genes for the amplitude of LTP and LTD. Our results show that the expression alterations observed post mortem, especially those in the anterior cingulate cortex, lead to impaired protein kinase A (PKA)-pathway-mediated LTP in synapses containing GluR1 receptors. We validated these findings using a genotyped electroencephalogram (EEG) dataset where polygenic risk scores for synaptic and ion channel-encoding genes as well as modulation of visual evoked potentials were determined for 286 healthy controls. Our results provide a possible genetic mechanism for plasticity impairments in schizophrenia, which can lead to improved understanding and, ultimately, treatment of the disorder.


Asunto(s)
Plasticidad Neuronal , Esquizofrenia , Esquizofrenia/genética , Esquizofrenia/fisiopatología , Esquizofrenia/metabolismo , Humanos , Plasticidad Neuronal/genética , Simulación por Computador , Potenciación a Largo Plazo/genética , Receptores AMPA/genética , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Sinapsis/genética , Electroencefalografía , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Modelos Neurológicos , Depresión Sináptica a Largo Plazo/genética , Masculino , Potenciales Evocados Visuales/fisiología
2.
J Endocrinol ; 258(3)2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37399525

RESUMEN

Estrogens regulate synaptic properties and influence hippocampus-related learning and memory via estrogen receptors, which include the G-protein-coupled estrogen receptor 1 (GPER1). Studying mice, in which the GPER1 gene is dysfunctional (GPER1-KO), we here provide evidence for sex-specific roles of GPER1 in these processes. GPER1-KO males showed reduced anxiety in the elevated plus maze, whereas the fear response ('freezing') was specifically increased in GPER1-KO females in a contextual fear conditioning paradigm. In the Morris water maze, spatial learning and memory consolidation was impaired by GPER1 deficiency in both sexes. Notably, in the females, spatial learning deficits and the fear response were more pronounced if mice were in a stage of the estrous cycle, in which E2 serum levels are high (proestrus) or rising (diestrus). On the physiological level, excitability at Schaffer collateral synapses in CA1 increased in GPER1-deficient males and in proestrus/diestrus ('E2 high') females, concordant with an increased hippocampal expression of the AMPA-receptor subunit GluA1 in GPER1-KO males and females as compared to wildtype males. Further changes included an augmented early long-term potentiation (E-LTP) maintenance specifically in GPER1-KO females and an increased hippocampal expression of spinophilin in metestrus/estrus ('E2 low') GPER1-KO females. Our findings suggest modulatory and sex-specific functions of GPER1 in the hippocampal network, which reduce rather than increase neuronal excitability. Dysregulation of these functions may underlie sex-specific cognitive deficits or mood disorders.


Asunto(s)
Hipocampo , Receptores de Estrógenos , Masculino , Femenino , Ratones , Animales , Receptores de Estrógenos/genética , Potenciación a Largo Plazo/genética , Sinapsis/fisiología , Cognición , Plasticidad Neuronal/genética
3.
Glia ; 71(9): 2137-2153, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37183905

RESUMEN

Astrocytes are wired to bidirectionally communicate with neurons namely with synapses, thus shaping synaptic plasticity, which in the hippocampus is considered to underlie learning and memory. Adenosine A2A receptors (A2A R) are a potential candidate to modulate this bidirectional communication, since A2A R regulate synaptic plasticity and memory and also control key astrocytic functions. Nonetheless, little is known about the role of astrocytic A2A R in synaptic plasticity and hippocampal-dependent memory. Here, we investigated the impact of genetic silencing astrocytic A2A R on hippocampal synaptic plasticity and memory of adult mice. The genetic A2A R silencing in astrocytes was accomplished by a bilateral injection into the CA1 hippocampal area of a viral construct (AAV5-GFAP-GFP-Cre) that inactivate A2A R expression in astrocytes of male adult mice carrying "floxed" A2A R gene, as confirmed by A2A R binding assays. Astrocytic A2A R silencing alters astrocytic morphology, typified by an increment of astrocytic arbor complexity, and led to deficits in spatial reference memory and compromised hippocampal synaptic plasticity, typified by a reduction of LTP magnitude and a shift of synaptic long-term depression (LTD) toward LTP. These data indicate that astrocytic A2A R control astrocytic morphology and influence hippocampal synaptic plasticity and memory of adult mice in a manner different from neuronal A2A R.


Asunto(s)
Astrocitos , Hipocampo , Ratones , Masculino , Animales , Astrocitos/metabolismo , Hipocampo/metabolismo , Plasticidad Neuronal/genética , Sinapsis/metabolismo , Memoria Espacial , Ratones Endogámicos C57BL , Potenciación a Largo Plazo/genética
4.
Mol Biol (Mosk) ; 55(6): 1021-1029, 2021.
Artículo en Ruso | MEDLINE | ID: mdl-34837706

RESUMEN

The PBAF chromatin remodeling complex interacts with many transcriptional activators and is recruited to target chromatin regions. PBAF plays an important role in maintaining and modifying the chromatin structure in mammalian cells. A subunit of the PBAF complex, the PHF10 transcription factor, is required for proliferation of neuronal precursors in the early stages of mouse brain development and gene expression in differentiated neurons. We showed that PHF10 interacts with the protein product of the early response gene c-FOS, the c-FOS transcriptional activator, which is expressed in response to the induction of long-term potentiation (LTP). LTP induction triggers the transcription of genes and the synthesis of proteins that provide changes that lead to the establishment of long-term contacts between neurons. We showed that in cells in differentiated neuronal culture, after the induction of LTP, expression of c-FOS, which is initially localized in the cytoplasm and then moves to the nucleus, begins. PHF10 is expressed in neuronal cells prior to LTP induction and has nuclear localization. However, 1 h after LTP induction, PHF10 is detected in the cytoplasm together with c-FOS, and then moves into the nucleus with it. Importantly, this behavior of PHF10 in response to KC1 stimulation is specific for neuronal cultures. It is assumed that during LTP, PHF10 together with c-FOS participates in the activation of secondary response genes that regulate the maintenance of plastic modifications and homeostasis of neuronal synapses. The PHF10 export from the nucleus and its rapid return together with c-FOS to the nucleus is possibly necessary for the rapid modulation of expression of target secondary response genes during LTP.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona , Animales , Proteínas Cromosómicas no Histona/genética , Hipocampo/metabolismo , Potenciación a Largo Plazo/genética , Ratones , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética
5.
Int J Mol Sci ; 22(9)2021 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-34064454

RESUMEN

The γ-aminobutyric acid type A receptor (GABAAR) plays a major role in fast inhibitory synaptic transmission and is highly regulated by the neuromodulator dopamine. In this aspect, most of the attention has been focused on the classical intracellular signaling cascades following dopamine G-protein-coupled receptor activation. Interestingly, the GABAAR and dopamine D5 receptor (D5R) have been shown to physically interact in the hippocampus, but whether a functional cross-talk occurs is still debated. In the present study, we use a combination of imaging and single nanoparticle tracking in live hippocampal neurons to provide evidence that GABAARs and D5Rs form dynamic surface clusters. Disrupting the GABAAR-D5R interaction with a competing peptide leads to an increase in the diffusion coefficient and the explored area of both receptors, and a drop in immobile synaptic GABAARs. By means of patch-clamp recordings, we show that this fast lateral redistribution of surface GABAARs correlates with a robust depression in the evoked GABAergic currents. Strikingly, it also shifts in time the expression of long-term potentiation at glutamatergic synapses. Together, our data both set the plasma membrane as the primary stage of a functional interplay between GABAAR and D5R, and uncover a non-canonical role in regulating synaptic transmission.


Asunto(s)
Potenciación a Largo Plazo/genética , Neuronas/metabolismo , Receptor Cross-Talk , Receptores de Dopamina D5/genética , Receptores de GABA-A/genética , Transmisión Sináptica/genética , Animales , Unión Competitiva , Membrana Celular/metabolismo , Embrión de Mamíferos , Regulación de la Expresión Génica , Hipocampo/citología , Hipocampo/metabolismo , Neuronas/citología , Técnicas de Placa-Clamp , Péptidos/síntesis química , Péptidos/metabolismo , Cultivo Primario de Células , Unión Proteica , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D5/metabolismo , Receptores de GABA-A/metabolismo , Sinapsis/genética , Sinapsis/metabolismo
6.
Neurobiol Learn Mem ; 179: 107397, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33524570

RESUMEN

Human genetic studies established MET gene as a risk factor for autism spectrum disorders. We have previously shown that signaling mediated by MET receptor tyrosine kinase, expressed in early postnatal developing forebrain circuits, controls glutamatergic neuron morphological development, synapse maturation, and cortical critical period plasticity. Here we investigated how MET signaling affects synaptic plasticity, learning and memory behavior, and whether these effects are age-dependent. We found that in young adult (postnatal 2-3 months) Met conditional knockout (Metfx/fx:emx1cre, cKO) mice, the hippocampus exhibits elevated plasticity, measured by increased magnitude of long-term potentiation (LTP) and depression (LTD) in hippocampal slices. Surprisingly, in older adult cKO mice (10-12 months), LTP and LTD magnitudes were diminished. We further conducted a battery of behavioral tests to assess learning and memory function in cKO mice and littermate controls. Consistent with age-dependent LTP/LTD findings, we observed enhanced spatial memory learning in 2-3 months old young adult mice, assessed by hippocampus-dependent Morris water maze test, but impaired spatial learning in 10-12 months mice. Contextual and cued learning were further assessed using a Pavlovian fear conditioning test, which also revealed enhanced associative fear acquisition and extinction in young adult mice, but impaired fear learning in older adult mice. Lastly, young cKO mice also exhibited enhanced motor learning. Our results suggest that a shift in the window of synaptic plasticity and an age-dependent early cognitive decline may be novel circuit pathophysiology for a well-established autism genetic risk factor.


Asunto(s)
Envejecimiento/genética , Disfunción Cognitiva/genética , Memoria/fisiología , Plasticidad Neuronal/genética , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Factores de Edad , Animales , Conducta Animal , Corteza Cerebral , Condicionamiento Clásico/fisiología , Extinción Psicológica , Miedo , Hipocampo/metabolismo , Aprendizaje/fisiología , Potenciación a Largo Plazo/genética , Depresión Sináptica a Largo Plazo/genética , Ratones , Ratones Noqueados , Prueba del Laberinto Acuático de Morris , Aprendizaje Espacial/fisiología
7.
Aging Cell ; 19(3): e13113, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32061032

RESUMEN

Alzheimer's disease (AD) is one of the most common causes of neurodegenerative diseases in the elderly. The accumulation of amyloid-ß (Aß) peptides is one of the pathological hallmarks of AD and leads to the impairments of synaptic plasticity and cognitive function. The transient receptor potential vanilloid 1 (TRPV1), a nonselective cation channel, is involved in synaptic plasticity and memory. However, the role of TRPV1 in AD pathogenesis remains largely elusive. Here, we reported that the expression of TRPV1 was decreased in the brain of APP23/PS45 double transgenic AD model mice. Genetic upregulation of TRPV1 by adeno-associated virus (AAV) inhibited the APP processing and Aß deposition in AD model mice. Meanwhile, upregulation of TRPV1 ameliorated the deficits of hippocampal CA1 long-term potentiation (LTP) and spatial learning and memory through inhibiting GluA2-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) endocytosis. Furthermore, pharmacological activation of TRPV1 by capsaicin (1 mg/kg, i.p.), an agonist of TRPV1, dramatically reversed the impairments of hippocampal CA1 LTP and spatial learning and memory in AD model mice. Taken together, these results indicate that TRPV1 activation effectively ameliorates cognitive and synaptic functions through inhibiting AMPAR endocytosis in AD model mice and could be a novel molecule for AD treatment.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Disfunción Cognitiva/metabolismo , Endocitosis/genética , Potenciación a Largo Plazo/genética , Receptores AMPA/metabolismo , Canales Catiónicos TRPV/metabolismo , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Animales , Región CA1 Hipocampal/metabolismo , Disfunción Cognitiva/genética , Modelos Animales de Enfermedad , Aprendizaje por Laberinto , Memoria , Ratones , Ratones Transgénicos , Transducción de Señal/efectos de los fármacos , Aprendizaje Espacial , Sinapsis/metabolismo , Canales Catiónicos TRPV/genética
8.
Neuropharmacology ; 162: 107834, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31682853

RESUMEN

Resilience to stress is critical for the development of depression. Enhanced adenosine A1 receptor (A1R) signaling mediates the antidepressant effects of acute sleep deprivation (SD). However, chronic SD causes long-lasting upregulation of brain A1R and increases the risk of depression. To investigate the effects of A1R on mood, we utilized two transgenic mouse lines with inducible A1R overexpression in forebrain neurons. These two lines have identical levels of A1R increase in the cortex, but differ in the transgenic A1R expression in the hippocampus. Switching on the transgene promotes robust antidepressant and anxiolytic effects in both lines. The mice of the line without transgenic A1R overexpression in the hippocampus (A1Hipp-) show very strong resistance towards development of stress-induced chronic depression-like behavior. In contrast, the mice of the line in which A1R upregulation extends to the hippocampus (A1Hipp+), exhibit decreased resilience to depression as compared to A1Hipp-. Similarly, automatic analysis of reward behavior of the two lines reveals that depression resistant A1Hipp-transgenic mice exhibit high sucrose preference, while mice of the vulnerable A1Hipp + line developed stress-induced anhedonic phenotype. The A1Hipp + mice have increased Homer1a expression in hippocampus, correlating with impaired long-term potentiation in the CA1 region, mimicking the stressed mice. Furthermore, virus-mediated overexpression of Homer1a in the hippocampus decreases stress resilience. Taken together our data indicate for first time that increased expression of A1R and Homer1a in the hippocampus modulates the resilience to stress-induced depression and thus might potentially mediate the detrimental effects of chronic sleep restriction on mood.


Asunto(s)
Corteza Cerebral/metabolismo , Depresión/genética , Hipocampo/metabolismo , Proteínas de Andamiaje Homer/genética , Receptor de Adenosina A1/genética , Resiliencia Psicológica , Privación de Sueño/metabolismo , Estrés Psicológico/genética , Animales , Conducta Animal , Región CA1 Hipocampal/metabolismo , Depresión/metabolismo , Depresión/psicología , Prueba de Laberinto Elevado , Potenciales Postsinápticos Excitadores , Suspensión Trasera , Proteínas de Andamiaje Homer/metabolismo , Potenciación a Largo Plazo/genética , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Prueba de Campo Abierto , Prosencéfalo , Receptor de Adenosina A1/metabolismo , Recompensa , Privación de Sueño/psicología
9.
J Neurosci ; 39(15): 2792-2809, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30728170

RESUMEN

17ß-estradiol (E2) is produced from androgens via the action of the enzyme aromatase. E2 is known to be made in neurons in the brain, but its precise functions in the brain are unclear. Here, we used a forebrain-neuron-specific aromatase knock-out (FBN-ARO-KO) mouse model to deplete neuron-derived E2 in the forebrain of mice and thereby elucidate its functions. FBN-ARO-KO mice showed a 70-80% decrease in aromatase and forebrain E2 levels compared with FLOX controls. Male and female FBN-ARO-KO mice exhibited significant deficits in forebrain spine and synaptic density, as well as hippocampal-dependent spatial reference memory, recognition memory, and contextual fear memory, but had normal locomotor function and anxiety levels. Reinstating forebrain E2 levels via exogenous in vivo E2 administration was able to rescue both the molecular and behavioral defects in FBN-ARO-KO mice. Furthermore, in vitro studies using FBN-ARO-KO hippocampal slices revealed that, whereas induction of long-term potentiation (LTP) was normal, the amplitude was significantly decreased. Intriguingly, the LTP defect could be fully rescued by acute E2 treatment in vitro Mechanistic studies revealed that FBN-ARO-KO mice had compromised rapid kinase (AKT, ERK) and CREB-BDNF signaling in the hippocampus and cerebral cortex. In addition, acute E2 rescue of LTP in hippocampal FBN-ARO-KO slices could be blocked by administration of a MEK/ERK inhibitor, further suggesting a key role for rapid ERK signaling in neuronal E2 effects. In conclusion, the findings provide evidence of a critical role for neuron-derived E2 in regulating synaptic plasticity and cognitive function in the male and female brain.SIGNIFICANCE STATEMENT The steroid hormone 17ß-estradiol (E2) is well known to be produced in the ovaries in females. Intriguingly, forebrain neurons also express aromatase, the E2 biosynthetic enzyme, but the precise functions of neuron-derived E2 is unclear. Using a novel forebrain-neuron-specific aromatase knock-out mouse model to deplete neuron-derived E2, the current study provides direct genetic evidence of a critical role for neuron-derived E2 in the regulation of rapid AKT-ERK and CREB-BDNF signaling in the mouse forebrain and demonstrates that neuron-derived E2 is essential for normal expression of LTP, synaptic plasticity, and cognitive function in both the male and female brain. These findings suggest that neuron-derived E2 functions as a novel neuromodulator in the forebrain to control synaptic plasticity and cognitive function.


Asunto(s)
Estradiol/fisiología , Memoria/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Animales , Ansiedad/genética , Ansiedad/psicología , Aromatasa/genética , Cognición , Espinas Dendríticas , Estradiol/metabolismo , Estradiol/farmacología , Femenino , Hipocampo , Potenciación a Largo Plazo/genética , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Prosencéfalo/enzimología , Prosencéfalo/metabolismo , Desempeño Psicomotor/fisiología , Aprendizaje Espacial
10.
J Alzheimers Dis ; 64(3): 957-971, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29991137

RESUMEN

Alzheimer's disease (AD) is characterized by neuritic plaques and neurofibrillary tangles. It is reported that enzymatic degradation of amyloid-ß (Aß) plays a pivotal role in Aß accumulation and type-2 cannabinoid receptor (CB2R) participates in Aß processing in the brain; however, the underlying mechanisms remain unclear. We determined that Aß degradation-related proteins are significantly different between CB2R-/- mice and wild-type (WT) mice via proteomic analysis. Moreover, the data demonstrated that the angiotensin converting enzyme (ACE) and insulin-degrading enzyme (IDE) levels are substantially attenuated, and the Aß level is significantly enhanced in CB2R-/--Aß1 - 42 mice compared with that of WT-Aß1 - 42 mice. Furthermore, Aß-mediated synaptic dysfunction, the loss of memory associated proteins, and the suppression of glutamatergic transmission are more severe in CB2R-/--Aß1 - 42 mice than that in WT-Aß1 - 42 mice. CB2R activation could decrease Aß1 - 40 and Aß1 - 42 levels and enhance ACE and IDE levels with its selective agonist JWH133; however, AM630 (CB2R antagonist) abrogates all changes induced by JWH133 in N2a cells with AßPP overexpression. Taken together, our study demonstrated that the deletion of CB2R reduces exogenous Aß degradation and aggravates the toxicity of Aß via the reduction of ACE and IDE, which suggests that CB2R is involved in the onset of AD and a potential therapeutic target for AD.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Regulación hacia Abajo/efectos de los fármacos , Insulisina/metabolismo , Síndromes de Neurotoxicidad/etiología , Fragmentos de Péptidos/toxicidad , Peptidil-Dipeptidasa A/metabolismo , Receptor Cannabinoide CB2/deficiencia , Péptidos beta-Amiloides/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Estimulación Eléctrica , Inyecciones Intraventriculares , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Fragmentos de Péptidos/metabolismo , Proteómica , Receptor Cannabinoide CB2/genética , Estadísticas no Paramétricas
11.
Nat Commun ; 9(1): 1726, 2018 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-29712923

RESUMEN

The ribosome is indispensable for precisely controlling the capacity of protein synthesis. However, how translational machinery is coordinated to meet the translational demands remains elusive. Here, we identify a nucleolar-specific lncRNA (LoNA), its 5' portion binds and sequesters nucleolin to suppress rRNA transcription, and its snoRNA like 3' end recruits and diminishes fibrillarin activity to reduce rRNA methylation. Activity-dependent decrease of LoNA leads to elevated rRNA and ribosome levels, an increased proportion of polysomes, mRNA polysome loading, and protein translation. In addition, transport of ribosomes to synapses is particularly promoted, resulting in increased levels of AMPA/NMDA receptor, enhanced synaptic plasticity, long-term potentiation and consolidated memory. Strikingly, hippocampal LoNA deficiency not only enhances long-term memory in WT mice, but also restores impaired memory function in APP/PS1 transgenic mice. Together, these findings reveal the multifaceted role of LoNA in modulating ribosome biogenesis to meet the translational demands of long-term memory.


Asunto(s)
Hipocampo/metabolismo , Potenciación a Largo Plazo/genética , Memoria a Largo Plazo/fisiología , ARN Largo no Codificante/genética , ARN Ribosómico/genética , ARN Nucleolar Pequeño/genética , Regiones no Traducidas 5' , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Hipocampo/citología , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Biosíntesis de Proteínas , ARN Largo no Codificante/metabolismo , ARN Ribosómico/metabolismo , ARN Nucleolar Pequeño/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores AMPA/genética , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Sinapsis/genética , Transgenes , Nucleolina
12.
Brain ; 140(12): 3215-3232, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29096020

RESUMEN

Fragile X syndrome is an inheritable form of intellectual disability caused by loss of fragile X mental retardation protein (FMRP, encoded by the FMR1 gene). Absence of FMRP caused overexpression of progranulin (PGRN, encoded by GRN), a putative tumour necrosis factor receptor ligand. In the present study, we found that progranulin mRNA and protein were upregulated in the medial prefrontal cortex of Fmr1 knock-out mice. In Fmr1 knock-out mice, elevated progranulin caused insufficient dendritic spine pruning and late-phase long-term potentiation in the medial prefrontal cortex of Fmr1 knock-out mice. Partial progranulin knock-down restored spine morphology and reversed behavioural deficits, including impaired fear memory, hyperactivity, and motor inflexibility in Fmr1 knock-out mice. Progranulin increased levels of phosphorylated glutamate ionotropic receptor GluA1 and nuclear factor kappa B in cultured wild-type neurons. Tumour necrosis factor receptor 2 antibody perfusion blocked the effects of progranulin on GluA1 phosphorylation; this result indicates that tumour necrosis factor receptor 2 is required for progranulin-mediated GluA1 phosphorylation and late-phase long-term potentiation expression. However, high basal level of progranulin in Fmr1 knock-out mice prevented further facilitation of synaptic plasticity by exogenous progranulin. Partial downregulation of progranulin or tumour necrosis factor receptor 2/nuclear factor kappa B signalling restored synaptic plasticity and memory deficits in Fmr1 knock-out mice. These findings suggest that elevated PGRN is linked to cognitive deficits of fragile X syndrome, and the progranulin/tumour necrosis factor receptor 2 signalling pathway may be a putative therapeutic target for improving cognitive deficits in fragile X syndrome.


Asunto(s)
Conducta Animal , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Aprendizaje , Sinapsis/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Western Blotting , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Granulinas , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Ratones , Ratones Noqueados , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Corteza Prefrontal/metabolismo , Progranulinas , ARN Mensajero/metabolismo , Receptores AMPA/efectos de los fármacos , Receptores AMPA/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Transducción de Señal
13.
Neuroscience ; 367: 200-210, 2017 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-29104031

RESUMEN

The neuroimmune factor IL-6 has been shown to regulate hippocampal long-term potentiation (LTP), an activity-dependent enhancement of synaptic transmission that plays a central role in memory and learning. This IL-6 action was demonstrated with relatively short IL-6 exposure, and may reflect physiological actions of IL-6. IL-6 is also expressed chronically at elevated levels in the central nervous system (CNS) under pathological conditions such as neurological disorders. Little is known about the effects IL-6 on LTP under such conditions, an issue that we are addressing by electrophysiological recordings from CA1 pyramidal neurons of hippocampal slices from transgenic mice that persistently express elevated levels of IL-6 in the CNS (IL-6 tg). The current studies examined the long-lasting phase of LTP (late LTP; L-LTP) and the potential involvement mammalian target of rapamycin (mTOR), a known regulator of L-LTP and a downstream partner of IL-6 signal transduction pathways. Results show that basal synaptic transmission and L-LTP were increased in hippocampal slices from IL-6 tg mice compared to slices from non-transgenic (non-tg) control mice. An inhibitor of mTOR, rapamycin, reduced L-LTP in slices from both genotypes, and eliminated the difference in magnitude of L-LTP between IL-6 and non-tg hippocampus. There were no genotypic effect of rapamycin on basal synaptic transmission, but synaptic responses during the LTP induction protocol were reduced in IL-6 tg slices, an effect that could contribute to the reduction of L-LTP in the IL-6 tg slices. These results indicate that persistently increased levels of IL-6 can lead to alterations in mTOR regulation of L-LTP, possibly affecting learning and memory.


Asunto(s)
Hipocampo/citología , Inmunosupresores/farmacología , Interleucina-6/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Neuronas/efectos de los fármacos , Sirolimus/farmacología , Animales , Estimulación Eléctrica , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipocampo/efectos de los fármacos , Técnicas In Vitro , Interleucina-6/genética , Potenciación a Largo Plazo/genética , Masculino , Ratones , Ratones Transgénicos , Estadísticas no Paramétricas , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo
14.
J Neurosci ; 37(41): 9917-9924, 2017 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-28912154

RESUMEN

Increased p25, a proteolytic fragment of the regulatory subunit p35, is known to induce aberrant activity of cyclin-dependent kinase 5 (Cdk5), which is associated with neurodegenerative disorders, including Alzheimer's disease. Previously, we showed that replacing endogenous p35 with the noncleavable mutant p35 (Δp35) attenuated amyloidosis and improved cognitive function in a familial Alzheimer's disease mouse model. Here, to address the role of p25/Cdk5 in tauopathy, we generated double-transgenic mice by crossing mice overexpressing mutant human tau (P301S) with Δp35KI mice. We observed significant reduction of phosphorylated tau and its seeding activity in the brain of double transgenic mice compared with the P301S mice. Furthermore, synaptic loss and impaired LTP at hippocampal CA3 region of P301S mice were attenuated by blocking p25 generation. To further validate the role of p25/Cdk5 in tauopathy, we used frontotemporal dementia patient-derived induced pluripotent stem cells (iPSCs) carrying the Tau P301L mutation and generated P301L:Δp35KI isogenic iPSC lines using CRISPR/Cas9 genome editing. We created cerebral organoids from the isogenic iPSCs and found that blockade of p25 generation reduced levels of phosphorylated tau and increased expression of synaptophysin. Together, these data demonstrate a crucial role for p25/Cdk5 in mediating tau-associated pathology and suggest that inhibition of this kinase can remedy neurodegenerative processes in the presence of pathogenic tau mutation.SIGNIFICANCE STATEMENT Accumulation of p25 results in aberrant Cdk5 activation and induction of numerous pathological phenotypes, such as neuroinflammation, synaptic loss, Aß accumulation, and tau hyperphosphorylation. However, it was not clear whether p25/Cdk5 activity is necessary for the progression of these pathological changes. We recently developed the Δp35KI transgenic mouse that is deficient in p25 generation and Cdk5 hyperactivation. In this study, we used this mouse model to elucidate the role of p25/Cdk5 in FTD mutant tau-mediated pathology. We also used a frontotemporal dementia patient-derived induced pluripotent stem cell carrying the Tau P301L mutation and generated isogenic lines in which p35 is replaced with noncleavable mutant Δp35. Our data suggest that p25/Cdk5 plays an important role in tauopathy in both mouse and human model systems.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/genética , Demencia Frontotemporal/genética , Fosfotransferasas/genética , Células Madre Pluripotentes , Tauopatías/genética , Animales , Región CA3 Hipocampal/patología , Región CA3 Hipocampal/fisiopatología , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Demencia Frontotemporal/prevención & control , Humanos , Potenciación a Largo Plazo/genética , Ratones , Ratones Transgénicos , Fibras Musgosas del Hipocampo/patología , Fosforilación , Fosfotransferasas/antagonistas & inhibidores , Trasplante de Células Madre , Sinapsis/patología , Sinaptofisina/genética , Tauopatías/prevención & control
15.
Neuropharmacology ; 121: 231-246, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28390893

RESUMEN

Alzheimer's disease amyloid-ß (Aß) oligomers are synaptotoxic, inappropriately increasing extracellular glutamate concentration and glutamate receptor activation to thereby rapidly disrupt synaptic plasticity. Thus, acutely promoting brain glutamate homeostasis with a blood-based scavenging system, glutamate-oxaloacetate transaminase (GOT), and blocking metabotropic glutamate 5 (mGlu5) receptor or its co-receptor cellular prion protein (PrP), prevent the acute inhibition of long-term potentiation (LTP) by exogenous Aß. Here, we evaluated the time course of the effects of such interventions in the persistent disruptive effects of Aß oligomers, either exogenously injected in wild type rats or endogenously generated in transgenic rats that model Alzheimer's disease amyloidosis. We report that repeated, but not acute, systemic administration of recombinant GOT type 1, with or without the glutamate co-substrate oxaloacetate, reversed the persistent deleterious effect of exogenous Aß on synaptic plasticity. Moreover, similar repetitive treatment reversibly abrogated the inhibition of LTP monitored longitudinally in freely behaving transgenic rats. Remarkably, brief repeated treatment with an mGlu5 receptor antagonist, basimglurant, or an antibody that prevents Aß oligomer binding to PrP, ICSM35, also had similar reversible ameliorative effects in the transgenic rat model. Overall, the present findings support the ongoing development of therapeutics for early Alzheimer's disease based on these complementary approaches.


Asunto(s)
Amiloidosis/patología , Amiloidosis/fisiopatología , Región CA1 Hipocampal/fisiología , Potenciación a Largo Plazo/efectos de los fármacos , Proteínas Priónicas/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Péptidos beta-Amiloides/inmunología , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidosis/sangre , Amiloidosis/genética , Animales , Anticuerpos/farmacología , Aspartato Aminotransferasa Citoplasmática/farmacología , Región CA1 Hipocampal/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Imidazoles/farmacología , Potenciación a Largo Plazo/genética , Masculino , Mutación/genética , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/farmacología , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Piridinas/farmacología , Ratas , Ratas Transgénicas , Receptor del Glutamato Metabotropico 5/antagonistas & inhibidores , Vigilia
16.
J Alzheimers Dis ; 56(1): 335-349, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28085018

RESUMEN

It has been reported that cyclin-dependent kinase 5 (cdk5), a critical neuronal kinase, is hyperactivated in Alzheimer's disease (AD) and may be, in part, responsible for the hallmark pathology of amyloid plaques and neurofibrillary tangles (NFTs). It has been proposed by several laboratories that hyperactive cdk5 results from the overexpression of p25 (a truncated fragment of p35, the normal cdk5 regulator), which, when complexed to cdk5, induces hyperactivity, hyperphosphorylated tau/NFTs, amyloid-ß plaques, and neuronal death. It has previously been shown that intraperitoneal (i.p.) injections of a modified truncated 24-aa peptide (TFP5), derived from the cdk5 activator p35, penetrated the blood-brain barrier and significantly rescued AD-like pathology in 5XFAD model mice. The principal pathology in the 5XFAD mutant, however, is extensive amyloid plaques; hence, as a proof of concept, we believe it is essential to demonstrate the peptide's efficacy in a mouse model expressing high levels of p25, such as the inducible CK-p25Tg model mouse that overexpresses p25 in CamKII positive neurons. Using a modified TFP5 treatment, here we show that peptide i.p. injections in these mice decrease cdk5 hyperactivity, tau, neurofilament-M/H hyperphosphorylation, and restore synaptic function and behavior (i.e., spatial working memory, motor deficit using Rota-rod). It is noteworthy that TFP5 does not inhibit endogenous cdk5/p35 activity, nor other cdks in vivo suggesting it might have no toxic side effects, and may serve as an excellent therapeutic candidate for neurodegenerative disorders expressing abnormally high brain levels of p25 and hyperactive cdk5.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Potenciación a Largo Plazo/efectos de los fármacos , Péptidos/farmacología , Péptidos/uso terapéutico , Fosfotransferasas/metabolismo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Animales , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Modelos Animales de Enfermedad , Doxiciclina/administración & dosificación , Agonistas de Aminoácidos Excitadores/farmacología , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Hipercinesia/tratamiento farmacológico , Hipercinesia/etiología , Potenciación a Largo Plazo/genética , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , N-Metilaspartato/farmacología , Fosfotransferasas/genética , Proteínas tau/metabolismo
17.
J Proteome Res ; 16(4): 1445-1459, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-27998058

RESUMEN

The neural circuit of the dorsal hippocampus (dHip) and nucleus accumbens (NAc) contributes to cue-induced learning and addictive behaviors, as demonstrated by the escalation of ethanol-seeking behaviors observed following deletion of the adenosine equilibrative nucleoside transporter 1 (ENT1-/-) in mice. Here we perform quantitative LC-MS/MS neuroproteomics in the dHip and NAc of ENT1-/- mice. Using Ingenuity Pathway Analysis, we identified proteins associated with increased long-term potentiation, ARP2/3-mediated actin cytoskeleton signaling and protein expression patterns suggesting deficits in glutamate degradation, GABAergic signaling, as well as significant changes in bioenergetics and energy homeostasis (oxidative phosphorylation, TCA cycle, and glycolysis). These pathways are consistent with previously reported behavioral and biochemical phenotypes that typify mice lacking ENT1. Moreover, we validated decreased expression of the SNARE complex protein VAMP1 (synaptobrevin-1) in the dHip as well as decreased expression of pro-dynorphin (PDYN), neuroendocrine convertase (PCSK1), and Leu-Enkephalin (dynorphin-A) in the NAc. Taken together, our proteomic approach provides novel pathways indicating that ENT1-regulated signaling is essential for neurotransmitter release and neuropeptide processing, both of which underlie learning and reward-seeking behaviors.


Asunto(s)
Encefalinas/genética , Tranportador Equilibrativo 1 de Nucleósido/genética , Proproteína Convertasa 1/genética , Precursores de Proteínas/genética , Proteómica , Proteína 1 de Membrana Asociada a Vesículas/genética , Consumo de Bebidas Alcohólicas/genética , Consumo de Bebidas Alcohólicas/patología , Animales , Etanol/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Potenciación a Largo Plazo/genética , Ratones , Neuropéptidos/biosíntesis , Neuropéptidos/genética , Neurotransmisores/biosíntesis , Neurotransmisores/genética , Núcleo Accumbens/metabolismo , Núcleo Accumbens/patología , Transducción de Señal/genética , Espectrometría de Masas en Tándem
18.
Sci Rep ; 6: 18668, 2016 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-26733416

RESUMEN

Alzheimer's disease (AD) is the most common form of dementia in the elderly. It is generally believed that ß-amyloidogenesis, tau-hyperphosphorylation, and synaptic loss underlie cognitive decline in AD. Rps23rg1, a functional retroposed mouse gene, has been shown to reduce Alzheimer's ß-amyloid (Aß) production and tau phosphorylation. In this study, we have identified its human homolog, and demonstrated that RPS23RG1 regulates synaptic plasticity, thus counteracting Aß oligomer (oAß)-induced cognitive deficits in mice. The level of RPS23RG1 mRNA is significantly lower in the brains of AD compared to non-AD patients, suggesting its potential role in the pathogenesis of the disease. Similar to its mouse counterpart, human RPS23RG1 interacts with adenylate cyclase, activating PKA/CREB, and inhibiting GSK-3. Furthermore, we show that human RPS23RG1 promotes synaptic plasticity and offsets oAß-induced synaptic loss in a PKA-dependent manner in cultured primary neurons. Overexpression of Rps23rg1 in transgenic mice consistently prevented oAß-induced PKA inactivation, synaptic deficits, suppression of long-term potentiation, and cognitive impairment as compared to wild type littermates. Our study demonstrates that RPS23RG1 may reduce the occurrence of key elements of AD pathology and enhance synaptic functions to counteract oAß-induced synaptic and cognitive deficits in AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Disfunción Cognitiva/metabolismo , Agregado de Proteínas , Agregación Patológica de Proteínas/metabolismo , Proteínas Ribosómicas/metabolismo , Sinapsis/metabolismo , Adenilil Ciclasas/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Secuencia de Bases , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Clonación Molecular , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Disfunción Cognitiva/fisiopatología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Potenciación a Largo Plazo/genética , Ratones , Ratones Transgénicos , Plasticidad Neuronal , Neuronas/metabolismo , Fosforilación , Unión Proteica , ARN Mensajero/genética , Proteínas Ribosómicas/química , Proteínas Ribosómicas/genética , Proteínas tau/genética , Proteínas tau/metabolismo
19.
J Alzheimers Dis ; 49(3): 707-21, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26484927

RESUMEN

Alzheimer's disease, one of the most important brain pathologies associated with neurodegenerative processes, is related to overactivation of calpain-mediated proteolysis. Previous data showed a compelling efficacy of calpain inhibition against abnormal synaptic plasticity and memory produced by the excess of amyloid-ß, a distinctive marker of the disease. Moreover, a beneficial effect of calpain inhibitors in Alzheimer's disease is predictable by the occurrence of calpain hyperactivation leading to impairment of memory-related pathways following abnormal calcium influxes that might ensue independently of amyloid-ß elevation. However, molecules currently available as effective calpain inhibitors lack adequate selectivity. This work is aimed at characterizing the efficacy of a novel class of epoxide-based inhibitors, synthesized to display improved selectivity and potency towards calpain 1 compared to the prototype epoxide-based generic calpain inhibitor E64. Both functional and preliminary toxicological investigations proved the efficacy, potency, and safety of the novel and selective calpain inhibitors NYC438 and NYC488 as possible therapeutics against the disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Glicoproteínas/uso terapéutico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Inhibidores de Cisteína Proteinasa/farmacología , Inhibidores de Cisteína Proteinasa/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Miedo/efectos de los fármacos , Glicoproteínas/química , Glicoproteínas/farmacología , Hipocampo/citología , Humanos , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , Mutación/genética , Técnicas de Placa-Clamp , Fragmentos de Péptidos/metabolismo , Presenilina-1/genética , Espectrina/metabolismo
20.
Sci Rep ; 5: 15301, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26477583

RESUMEN

Bv8/Prokineticin 2 (PROK2) is a bioactive peptide initially discovered as a regulator of gastrointestinal motility. Among multiple biological roles demonstrated for PROK2, it was recently established that PROK2 is an insult-inducible endangering mediator for cerebral damage. Aim of the present study was to evaluate the PROK2 and its receptors' potential involvement in amyloid beta (Aß) neurotoxicity, a hallmark of Alzheimer's disease (AD) and various forms of traumatic brain injury (TBI). Analyzing primary cortical cultures (CNs) and cortex and hippocampus from Aß treated rats, we found that PROK2 and its receptors PKR1 and PKR2 mRNA are up-regulated by Aß, suggesting their potential involvement in AD. Hence we evaluated if impairing the prokineticin system activation might have protective effect against neuronal death induced by Aß. We found that a PKR antagonist concentration-dependently protects CNs against Aß(1-42)-induced neurotoxicity, by reducing the Aß-induced PROK2 neuronal up-regulation. Moreover, the antagonist completely rescued LTP impairment in hippocampal slices from 6 month-old Tg2576 AD mice without affecting basal synaptic transmission and paired pulse-facilitation paradigms. These results indicate that PROK2 plays a role in cerebral amyloidosis and that PROK2 antagonists may represent a new approach for ameliorating the defining pathology of AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Hormonas Gastrointestinales/metabolismo , Neuronas/metabolismo , Neuropéptidos/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Apoptosis/efectos de los fármacos , Hormonas Gastrointestinales/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Masculino , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuropéptidos/genética , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Transporte de Proteínas , Ratas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA