Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
J Neurosci ; 43(50): 8744-8755, 2023 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-37857485

RESUMEN

Mammalian target of rapamycin (mTOR) pathway has emerged as a key molecular mechanism underlying memory processes. Although mTOR inhibition is known to block memory processes, it remains elusive whether and how an enhancement of mTOR signaling may improve memory processes. Here we found in male mice that the administration of VO-OHpic, an inhibitor of the phosphatase and tensin homolog (PTEN) that negatively modulates AKT-mTOR pathway, enhanced auditory fear memory for days and weeks, while it left short-term memory unchanged. Memory enhancement was associated with a long-lasting increase in immature-type dendritic spines of pyramidal neurons into the auditory cortex. The persistence of spine remodeling over time arose by the interplay between PTEN inhibition and memory processes, as VO-OHpic induced only a transient immature spine growth in the somatosensory cortex, a region not involved in long-term auditory memory. Both the potentiation of fear memories and increase in immature spines were hampered by rapamycin, a selective inhibitor of mTORC1. These data revealed that memory can be potentiated over time by the administration of a selective PTEN inhibitor. In addition to disclosing new information on the cellular mechanisms underlying long-term memory maintenance, our study provides new insights on the molecular processes that aid enhancing memories over time.SIGNIFICANCE STATEMENT The neuronal mechanisms that may help improve the maintenance of long-term memories are still elusive. The inhibition of mammalian-target of rapamycin (mTOR) signaling shows that this pathway plays a crucial role in synaptic plasticity and memory formation. However, whether its activation may strengthen long-term memory storage is unclear. We assessed the consequences of positive modulation of AKT-mTOR pathway obtained by VO-OHpic administration, a phosphatase and tensin homolog inhibitor, on memory retention and underlying synaptic modifications. We found that mTOR activation greatly enhanced memory maintenance for weeks by producing a long-lasting increase of immature-type dendritic spines in pyramidal neurons of the auditory cortex. These results offer new insights on the cellular and molecular mechanisms that can aid enhancing memories over time.


Asunto(s)
Corteza Auditiva , Proteínas Proto-Oncogénicas c-akt , Masculino , Ratones , Animales , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Corteza Auditiva/metabolismo , Espinas Dendríticas/metabolismo , Tensinas/metabolismo , Memoria a Largo Plazo/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Memoria a Corto Plazo/fisiología , Sirolimus/farmacología , Miedo/fisiología , Monoéster Fosfórico Hidrolasas/metabolismo , Mamíferos
2.
J Neurosci ; 2022 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-35953295

RESUMEN

The N-Methyl-D-aspartate receptors (NMDAR) are key players in both physiological and pathological synaptic plasticity because of their involvement in many aspects of neuronal transmission as well as learning and memory. The contribution in these events of different types of GluN2A-interacting proteins is still unclear. The p140Cap scaffold protein acts as a hub for postsynaptic complexes relevant to psychiatric and neurological disorders and regulates synaptic functions like the stabilization of mature dendritic spine, memory consolidation, long-term potentiation, and depression. Here we demonstrate that p140Cap directly binds the GluN2A subunit of NMDAR and modulates GluN2A-associated molecular network. Indeed, in p140Cap knockout male mice, GluN2A is less associated with PSD95 both in ex vivo synaptosomes and in cultured hippocampal neurons and p140Cap expression in knockout neurons can rescue GluN2A and PSD95 colocalization. p140Cap is crucial in the recruitment of GluN2A-containing NMDARs and, consequently, in regulating NMDARs intrinsic properties. p140Cap is associated to synaptic lipid-raft (LR) and to soluble postsynaptic membranes and GluN2A and PSD95 are less recruited into synaptic LR of p140Cap knockout male mice. g-STED microscopy on hippocampal neurons confirmed that p140Cap is required for embedding GluN2A clusters in LR in an activity-dependent fashion. In the synaptic compartment p140Cap influences the association between GluN2A and PSD95 and modulates GluN2A enrichment into LR. Overall, such increase in these membrane domains rich in signalling molecules results in improved signal transduction efficiency.SIGNIFICANT STATEMENTHere we originally show that the adaptor protein p140Cap directly binds the GluN2A subunit of NMDAR and modulates the GluN2A-associated molecular network. Moreover, we show for the first time that p140Cap also associates to synaptic lipid rafts and controls the selective recruitment of GluN2A and PSD95 to this specific compartment. Finally, g-STED microscopy on hippocampal neurons confirmed that p140Cap is required for embedding GluN2A clusters in lipid rafts in an activity-dependent fashion. Overall, our findings provide the molecular and functional dissection of p140Cap as a new active member of a highly dynamic synaptic network involved in memory consolidation, LTP and LTD that are known to be altered in neurological and psychiatric disorders.

3.
Genet Med ; 25(11): 100922, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37403762

RESUMEN

PURPOSE: RPH3A encodes a protein involved in the stabilization of GluN2A subunit of N-methyl-D-aspartate (NMDA)-type glutamate receptors at the cell surface, forming a complex essential for synaptic plasticity and cognition. We investigated the effect of variants in RPH3A in patients with neurodevelopmental disorders. METHODS: By using trio-based exome sequencing, GeneMatcher, and screening of 100,000 Genomes Project data, we identified 6 heterozygous variants in RPH3A. In silico and in vitro models, including rat hippocampal neuronal cultures, have been used to characterize the effect of the variants. RESULTS: Four cases had a neurodevelopmental disorder with untreatable epileptic seizures [p.(Gln73His)dn; p.(Arg209Lys); p.(Thr450Ser)dn; p.(Gln508His)], and 2 cases [p.(Arg235Ser); p.(Asn618Ser)dn] showed high-functioning autism spectrum disorder. Using neuronal cultures, we demonstrated that p.(Thr450Ser) and p.(Asn618Ser) reduce the synaptic localization of GluN2A; p.(Thr450Ser) also increased the surface levels of GluN2A. Electrophysiological recordings showed increased GluN2A-dependent NMDA ionotropic glutamate receptor currents for both variants and alteration of postsynaptic calcium levels. Finally, expression of the Rph3AThr450Ser variant in neurons affected dendritic spine morphology. CONCLUSION: Overall, we provide evidence that missense gain-of-function variants in RPH3A increase GluN2A-containing NMDA ionotropic glutamate receptors at extrasynaptic sites, altering synaptic function and leading to a clinically variable neurodevelopmental presentation ranging from untreatable epilepsy to autism spectrum disorder.


Asunto(s)
Trastorno del Espectro Autista , Epilepsia , Animales , Humanos , Ratas , Trastorno del Espectro Autista/genética , Epilepsia/genética , Mutación Missense/genética , N-Metilaspartato/metabolismo , Neuronas/metabolismo , Rabfilina-3A
4.
J Physiol ; 600(24): 5295-5309, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36284365

RESUMEN

The oligomeric form of the peptide amyloid beta 42 (Abeta42) contributes to the development of synaptic abnormalities and cognitive impairments associated with Alzheimer's disease (AD). To date, there is a gap in knowledge regarding how Abeta42 alters the elementary parameters of GABAergic synaptic function. Here we found that Abeta42 increased the frequency and amplitude of miniature GABAergic currents as well as the amplitude of evoked inhibitory postsynaptic currents. When we focused on paired pulse depression (PPD) to establish whether GABA release probability was affected by Abeta42, we did not observe any significant change. On the other hand, a more detailed investigation of the presynaptic effects induced by Abeta42 by means of multiple probability fluctuation analysis and cumulative amplitude analysis showed an increase in both the size of the readily releasable pool responsible for synchronous release and the number of release sites. We further explored whether ryanodine receptors (RyRs) contributed to exacerbating these changes by stabilizing the interaction between RyRs and the accessory protein calstabin. We observed that the RyR-calstabin interaction stabilizer S107 restored the synaptic parameters to values comparable to those measured in control conditions. In conclusion, our results clarify the mechanisms of potentiation of GABAergic synapses induced by Abeta42. We further suggest that RyRs are involved in the control of synaptic activity during the early stage of AD onset and that their stabilization could represent a new therapeutical approach for AD treatment. KEY POINTS: Accumulation of the peptide amyloid beta 42 (Abeta42) is a key characteristic of Alzheimer's disease (AD) and causes synaptic dysfunctions. To date, the effects of Abeta42 accumulation on GABAergic synapses are poorly understood. The findings reported here suggest that, similarly to what is observed on glutamatergic synapses, Abeta42 modifies GABAergic synapses by targeting ryanodine receptors and causing calcium dysregulation. The GABAergic impairments can be restored by the ryanodine receptor-calstabin interaction stabilizer S107. Based on this research, RyRs stabilization may represent a novel pharmaceutical strategy for preventing or delaying AD.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Humanos , Péptidos beta-Amiloides/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Rianodina/farmacología , Enfermedad de Alzheimer/metabolismo , Hipocampo/fisiología , Neuronas/metabolismo , Sinapsis/fisiología , Transmisión Sináptica/fisiología
5.
BMC Biol ; 19(1): 256, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34911542

RESUMEN

BACKGROUND: Rett syndrome (RTT) is a monogenic X-linked neurodevelopmental disorder characterized by loss-of-function mutations in the MECP2 gene, which lead to structural and functional changes in synapse communication, and impairments of neural activity at the basis of cognitive deficits that progress from an early age. While the restoration of MECP2 in animal models has been shown to rescue some RTT symptoms, gene therapy intervention presents potential side effects, and with gene- and RNA-editing approaches still far from clinical application, strategies focusing on signaling pathways downstream of MeCP2 may provide alternatives for the development of more effective therapies in vivo. Here, we investigate the role of the c-Jun N-terminal kinase (JNK) stress pathway in the pathogenesis of RTT using different animal and cell models and evaluate JNK inhibition as a potential therapeutic approach. RESULTS: We discovered that the c-Jun N-terminal kinase (JNK) stress pathway is activated in Mecp2-knockout, Mecp2-heterozygous mice, and in human MECP2-mutated iPSC neurons. The specific JNK inhibitor, D-JNKI1, promotes recovery of body weight and locomotor impairments in two mouse models of RTT and rescues their dendritic spine alterations. Mecp2-knockout presents intermittent crises of apnea/hypopnea, one of the most invalidating RTT pathological symptoms, and D-JNKI1 powerfully reduces this breathing dysfunction. Importantly, we discovered that also neurons derived from hiPSC-MECP2 mut show JNK activation, high-phosphorylated c-Jun levels, and cell death, which is not observed in the isogenic control wt allele hiPSCs. Treatment with D-JNKI1 inhibits neuronal death induced by MECP2 mutation in hiPSCs mut neurons. CONCLUSIONS: As a summary, we found altered JNK signaling in models of RTT and suggest that D-JNKI1 treatment prevents clinical symptoms, with coherent results at the cellular, molecular, and functional levels. This is the first proof of concept that JNK plays a key role in RTT and its specific inhibition offers a new and potential therapeutic tool to tackle RTT.


Asunto(s)
Síndrome de Rett , Animales , Modelos Animales de Enfermedad , Sistema de Señalización de MAP Quinasas , Ratones , Neuronas/metabolismo , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/terapia , Sinapsis/metabolismo
6.
J Neurochem ; 157(4): 1253-1269, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33448385

RESUMEN

Mutations in the X-linked CDKL5 gene cause CDKL5 deficiency disorder (CDD), a severe neurodevelopmental condition mainly characterized by infantile epileptic encephalopathy, intellectual disability, and autistic features. The molecular mechanisms underlying the clinical symptoms remain largely unknown and the identification of reliable biomarkers in animal models will certainly contribute to increase our comprehension of CDD as well as to assess the efficacy of therapeutic strategies. Here, we used different Magnetic Resonance (MR) methods to disclose structural, functional, or metabolic signatures of Cdkl5 deficiency in the brain of adult mice. We found that loss of Cdkl5 does not cause cerebral atrophy but affects distinct brain areas, particularly the hippocampus. By in vivo proton-MR spectroscopy (MRS), we revealed in the Cdkl5 null brain a metabolic dysregulation indicative of mitochondrial dysfunctions. Accordingly, we unveiled a significant reduction in ATP levels and a decrease in the expression of complex IV of mitochondrial electron transport chain. Conversely, the number of mitochondria appeared preserved. Importantly, we reported a significant defect in the activation of one of the major regulators of cellular energy balance, the adenosine monophosphate-activated protein kinase (AMPK), that might contribute to the observed metabolic impairment and become an interesting therapeutic target for future preclinical trials. In conclusion, MRS revealed in the Cdkl5 null brain the presence of a metabolic dysregulation suggestive of a mitochondrial dysfunction that permitted to foster our comprehension of Cdkl5 deficiency and brought our interest towards targeting mitochondria as therapeutic strategy for CDD.


Asunto(s)
Encéfalo/metabolismo , Síndromes Epilépticos , Mitocondrias/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Espasmos Infantiles , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Síndromes Epilépticos/metabolismo , Síndromes Epilépticos/patología , Espectroscopía de Resonancia Magnética , Metaboloma , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/patología , Espasmos Infantiles/metabolismo , Espasmos Infantiles/patología
7.
Neurobiol Dis ; 153: 105304, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33621640

RESUMEN

CDKL5 (cyclin-dependent kinase-like 5) deficiency disorder (CDD) is a severe neurodevelopmental encephalopathy characterized by early-onset epilepsy and intellectual disability. Studies in mouse models have linked CDKL5 deficiency to defects in neuronal maturation and synaptic plasticity, and disruption of the excitatory/inhibitory balance. Interestingly, increased density of both GABAergic synaptic terminals and parvalbumin inhibitory interneurons was recently observed in the primary visual cortex of Cdkl5 knockout (KO) mice, suggesting that excessive GABAergic transmission might contribute to the visual deficits characteristic of CDD. However, the functional relevance of cortical GABAergic circuits abnormalities in these mutant mice has not been investigated so far. Here we examined GABAergic circuits in the perirhinal cortex (PRC) of Cdkl5 KO mice, where we previously observed impaired long-term potentiation (LTP) associated with deficits in novel object recognition (NOR) memory. We found a higher number of GABAergic (VGAT)-immunopositive terminals in the PRC of Cdkl5 KO compared to wild-type mice, suggesting that increased inhibitory transmission might contribute to LTP impairment. Interestingly, while exposure of PRC slices to the GABAA receptor antagonist picrotoxin had no positive effects on LTP in Cdkl5 KO mice, the selective GABAB receptor antagonist CGP55845 restored LTP magnitude, suggesting that exaggerated GABAB receptor-mediated inhibition contributes to LTP impairment in mutants. Moreover, acute in vivo treatment with CGP55845 increased the number of PSD95 positive puncta as well as density and maturation of dendritic spines in PRC, and restored NOR memory in Cdkl5 KO mice. The present data show the efficacy of limiting excessive GABAB receptor-mediated signaling in improving synaptic plasticity and cognition in CDD mice.


Asunto(s)
Síndromes Epilépticos/metabolismo , Antagonistas de Receptores de GABA-B/farmacología , Neuronas GABAérgicas/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Corteza Perirrinal/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Receptores de GABA-B/metabolismo , Espasmos Infantiles/metabolismo , Animales , Modelos Animales de Enfermedad , Síndromes Epilépticos/genética , Antagonistas de Receptores de GABA-A/farmacología , Potenciación a Largo Plazo/genética , Ratones , Ratones Noqueados , Plasticidad Neuronal , Prueba de Campo Abierto , Corteza Perirrinal/metabolismo , Ácidos Fosfínicos/farmacología , Picrotoxina/farmacología , Propanolaminas/farmacología , Espasmos Infantiles/genética
8.
J Physiol ; 598(11): 2183-2197, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32246769

RESUMEN

KEY POINTS: NMDA receptors (NMDARs) are key molecules for controlling neuronal plasticity, learning and memory processes. Their function is impaired during Alzheimer's disease (AD) but the exact consequence on synaptic function is not yet fully identified. An important hallmark of AD onset is represented by the neuronal accumulation of Amyloid Beta42 oligomers (Abeta42) that we have recently shown to be responsible for the increased intracellular Ca2+ concentration through ryanodine receptors (RyRs). Here we characterized the effects of Abeta42 on NMDA synapses showing specific pre- and post-synaptic functional changes that lead to a potentiation of basal and synchronous NMDA synaptic transmission. These overall effects can be abolished by decreasing Ca2+ release from RyRs with specific inhibitors that we propose as new pharmacological tools for AD treatment. ABSTRACT: We have recently shown that Amyloid Beta42 oligomers (Abeta42) cause calcium dysregulation in hippocampal neurons by stimulating Ca2+ release from ryanodine receptors (RyRs) and inhibiting Ca2+ entry through NMDA receptors (NMDARs). Here, we found that Abeta42 decrease the average NMDA-activated inward current and that Ca2+ entry through NMDARs is accompanied by Ca2+ release from the stores. The overall amount of intraellular Ca2+ concentration([Ca2+ ]i ) increase during NMDA application is 50% associated with RyR opening and 50% with NMDARs activation. Addition of Abeta42 does not change this proportion. We estimated the number of NMDARs expressed in hippocampal neurons and their unitary current. We found that Abeta42 decrease the number of NMDARs without altering their unitary current. Paradoxically, the oligomer increases the size of electrically evoked eEPSCs induced by NMDARs activation. We found that this is the consequence of the increased release probability (p) of glutamate and the number of release sites (N) of NMDA synapses, while the quantal size (q) is significantly decreased as expected from the decreased number of NMDARs. An increased number of release sites induced by Abeta42 is also supported by the increased size of the ready releasable pool (RRPsyn) and by the enhanced percentage of paired pulse depression (PPD). Interestingly, the RyRs inhibitor dantrolene prevents the increase of PPD induced by Abeta42 oligomers. In conclusion, Abeta42 up-regulates NMDA synaptic responses with a mechanism involving RyRs that occurs during the early stages of Alzheimer's disease (AD) onset. This suggests that new selective modulators of RyRs may be useful for designing effective therapies to treat AD patients.


Asunto(s)
Péptidos beta-Amiloides , Receptores de N-Metil-D-Aspartato , Péptidos beta-Amiloides/metabolismo , Humanos , Fragmentos de Péptidos , Sinapsis/metabolismo
9.
Cereb Cortex ; 29(1): 91-105, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29161354

RESUMEN

The neuronal scaffold protein p140Cap was investigated during hippocampal network formation. p140Cap is present in presynaptic GABAergic terminals and its genetic depletion results in a marked alteration of inhibitory synaptic activity. p140Cap-/- cultured neurons display higher frequency of miniature inhibitory postsynaptic currents (mIPSCs) with no changes of their mean amplitude. Consistent with a potential presynaptic alteration of basal GABA release, p140Cap-/- neurons exhibit a larger synaptic vesicle readily releasable pool, without any variation of single GABAA receptor unitary currents and number of postsynaptic channels. Furthermore, p140Cap-/- neurons show a premature and enhanced network synchronization and appear more susceptible to 4-aminopyridine-induced seizures in vitro and to kainate-induced seizures in vivo. The hippocampus of p140Cap-/- mice showed a significant increase in the number of both inhibitory synapses and of parvalbumin- and somatostatin-expressing interneurons. Specific deletion of p140Cap in forebrain interneurons resulted in increased susceptibility to in vitro epileptic events and increased inhibitory synaptogenesis, comparable to those observed in p140Cap-/- mice. Altogether, our data demonstrate that p140Cap finely tunes inhibitory synaptogenesis and GABAergic neurotransmission, thus regulating the establishment and maintenance of the proper hippocampal excitatory/inhibitory balance.


Asunto(s)
Proteínas Portadoras/fisiología , Neuronas GABAérgicas/fisiología , Hipocampo/fisiología , Red Nerviosa/fisiología , Inhibición Neural/fisiología , Sinapsis/fisiología , Animales , Células Cultivadas , Potenciales Postsinápticos Inhibidores/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
10.
Arch Toxicol ; 94(5): 1703-1723, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32067069

RESUMEN

Glyphosate-based herbicides (GBH) are the most widely used pesticides worldwide. Despite considerable progress in describing the neurotoxic potential of GBH, the harmful effects on brain cytoarchitecture and behavior are still unclear. Here, we addressed the developmental impact of GBH by exposing female mice to 250 or 500 mg/kg doses of GBH during both pregnancy and lactation and then examined the downstream effects at the behavioral, neurochemical and molecular levels. We show that pre- and neonatal exposure to GBH impairs fertility and reproduction parameters as well as maternal behavior of exposed mothers. In offspring, GBH was responsible for a global delay in innate reflexes and a deficit in motor development. At the adult age, exposed animals showed a decrease of locomotor activity, sociability, learning and short- and long-term memory associated with alterations of cholinergic and dopaminergic systems. Furthermore, GBH-activated microglia and astrocytes, sign of neuroinflammation event in the medial prefrontal cortex and hippocampus. At the molecular level, a down-regulation of brain-derived neurotrophic factor (BDNF) expression and an up-regulation of tyrosine-related kinase receptor (TrkB), NR1 subunit of NMDA receptor as well as tumor necrosis factor α (TNFα) were found in the brain of GBH-exposed mice. The present work demonstrates that GBH induces numerous behavioral and cognitive abnormalities closely associated with significant histological, neurochemical and molecular impairments. It also raises fundamental concerns about the ability of current safety testing to assess risks of pesticide exposure during developmental periods of central nervous system.


Asunto(s)
Glicina/análogos & derivados , Herbicidas/toxicidad , Efectos Tardíos de la Exposición Prenatal , Animales , Astrocitos , Disfunción Cognitiva , Femenino , Glicina/toxicidad , Hipocampo , Lactancia , Aprendizaje , Ratones , Síndromes de Neurotoxicidad , Embarazo , Receptores de N-Metil-D-Aspartato , Reproducción , Transducción de Señal , Glifosato
11.
Hum Mol Genet ; 25(11): 2314-2323, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27146843

RESUMEN

Loss of function mutations in human Oligophrenin1 (OPHN1) gene are responsible for syndromic intellectual disability (ID) associated with cerebellar hypoplasia and cerebral ventricles enlargement. Functional studies in rodent models suggest that OPHN1 linked ID is a consequence of abnormal synaptic transmission and shares common pathophysiological mechanisms with other cognitive disorders. Variants of this gene have been also identified in autism spectrum disorder and schizophrenia. The advanced understanding of the mechanisms underlying OPHN1-related ID, allowed us to develop a therapeutic approach targeting the Ras homolog gene family, member A (RHOA) signalling pathway and repurpose Fasudil- a well-tolerated Rho Kinase (ROCK) and Protein Kinase A (PKA) inhibitor- as a treatment of ID. We have previously shown ex-vivo its beneficial effect on synaptic transmission and plasticity in a mouse model of the OPHN1 loss of function. Here, we report that chronic treatment in adult mouse with Fasudil, is able to counteract vertical and horizontal hyperactivities, restores recognition memory and limits the brain ventricular dilatation observed in Ophn1-/y However, deficits in working and spatial memories are partially or not rescued by the treatment. These results highlight the potential of Fasudil treatment in synaptopathies and also the need for multiple therapeutic approaches especially in adult where brain plasticity is reduced.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Encéfalo/fisiopatología , Proteínas del Citoesqueleto/genética , Proteínas Activadoras de GTPasa/genética , Discapacidad Intelectual/tratamiento farmacológico , Proteínas Nucleares/genética , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/administración & dosificación , Adulto , Animales , Trastorno del Espectro Autista , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Ratones , Transmisión Sináptica
12.
J Neurosci ; 34(4): 1542-53, 2014 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-24453341

RESUMEN

A major challenge in the neuroscience field is the identification of molecules and pathways that control synaptic plasticity and memory. Dendritic spines play a pivotal role in these processes, as the major sites of excitatory synapses in neuronal communication. Previous studies have shown that the scaffold protein p140Cap localizes into dendritic spines and that its knockdown negatively modulates spine shape in culture. However, so far, there is no information on its in vivo relevance. By using a knock-out mouse model, we here demonstrate that p140Cap is a key element for both learning and synaptic plasticity. Indeed, p140Cap(-/-) mice are impaired in object recognition test, as well as in LTP and in LTD measurements. The in vivo effects of p140Cap loss are presumably attenuated by noncell-autonomous events, since primary neurons obtained from p140Cap(-/-) mice show a strong reduction in number of mushroom spines and abnormal organization of synapse-associated F-actin. These phenotypes are most likely caused by a local reduction of the inhibitory control of RhoA and of cortactin toward the actin-depolymerizing factor cofilin. These events can be controlled by p140Cap through its capability to directly inhibit the activation of Src kinase and by its binding to the scaffold protein Citron-N. Altogether, our results provide new insight into how protein associated with dynamic microtubules may regulate spine actin organization through interaction with postsynaptic density components.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Memoria/fisiología , Plasticidad Neuronal/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Familia-src Quinasas/metabolismo , Actinas/metabolismo , Animales , Western Blotting , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Potenciales Postsinápticos Excitadores/fisiología , Técnica del Anticuerpo Fluorescente , Hipocampo/metabolismo , Aprendizaje/fisiología , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Ratas , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología
13.
EMBO J ; 30(20): 4287-98, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21847097

RESUMEN

The epigenetic changes of the chromatin represent an attractive molecular substrate for adaptation to the environment. We examined here the role of CREB-binding protein (CBP), a histone acetyltransferase involved in mental retardation, in the genesis and maintenance of long-lasting systemic and behavioural adaptations to environmental enrichment (EE). Morphological and behavioural analyses demonstrated that EE ameliorates deficits associated to CBP deficiency. However, CBP-deficient mice also showed a strong defect in environment-induced neurogenesis and impaired EE-mediated enhancement of spatial navigation and pattern separation ability. These defects correlated with an attenuation of the transcriptional programme induced in response to EE and with deficits in histone acetylation at the promoters of EE-regulated, neurogenesis-related genes. Additional experiments in CBP restricted and inducible knockout mice indicated that environment-induced adult neurogenesis is extrinsically regulated by CBP function in mature granule cells. Overall, our experiments demonstrate that the environment alters gene expression by impinging on activities involved in modifying the epigenome and identify CBP-dependent transcriptional neuroadaptation as an important mediator of EE-induced benefits, a finding with important implications for mental retardation therapeutics.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Cognición , Neurogénesis/fisiología , Acetilación , Animales , Conducta Animal , Proteína de Unión a CREB/genética , Femenino , Expresión Génica , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Neurogénesis/genética , Neuronas/metabolismo , Regiones Promotoras Genéticas , Transcripción Genética
14.
J Neurochem ; 131(1): 115-27, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24978323

RESUMEN

Rett syndrome (RTT, MIM312750), a neurodevelopmental disorder predominantly occurring in females, is caused in the majority of cases by sporadic mutations in the gene encoding the transcriptional modulator methyl-CpG-binding protein 2 (MECP2). In mice, impaired MeCP2 function results in severe motor, cognitive, and emotional defects. The lack of Mecp2 in γ-aminobutyric acid-(GABA) releasing forebrain interneurons (INs) recapitulate many RTT features, however, the role of this gene in the development of the cortical inhibitory system is still unknown. Here, we found that MeCP2 expression varies among the three major classes of cortical INs and its nuclear localization differs between neuronal types. The density of calretinin(+) and parvalbumin(+) INs increases in Mecp2 knockout mice (Mecp2(-/y) ) already at early post-natal developmental stages. In contrast, the density of somatostatin(+) INs is not affected. We also found that the development of multipolar-calretinin(+) INs is selectively affected by the absence of Mecp2. Additionally, we show that in Mecp2 heterozygous female mice, a model closely mimicking human RTT condition, IN abnormalities are similar to those observed in Mecp2(-/y) mice. Together, our study indicates that loss of function of Mecp2 strongly interferes with the correct establishment of the neocortical inhibitory system producing effects that are specific to different IN subtypes.


Asunto(s)
Modelos Animales de Enfermedad , Interneuronas/patología , Proteína 2 de Unión a Metil-CpG/deficiencia , Síndrome de Rett/patología , Corteza Somatosensorial/patología , Animales , Animales Recién Nacidos , Femenino , Interneuronas/metabolismo , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Corteza Somatosensorial/crecimiento & desarrollo , Corteza Somatosensorial/metabolismo
15.
EMBO J ; 29(11): 1889-902, 2010 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-20407421

RESUMEN

Neuronal plasticity is an important process for learning, memory and complex behaviour. Rapid remodelling of the actin cytoskeleton in the postsynaptic compartment is thought to have an important function for synaptic plasticity. However, the actin-binding proteins involved and the molecular mechanisms that in vivo link actin dynamics to postsynaptic physiology are not well understood. Here, we show that the actin filament depolymerizing protein n-cofilin is controlling dendritic spine morphology and postsynaptic parameters such as late long-term potentiation and long-term depression. Loss of n-cofilin-mediated synaptic actin dynamics in the forebrain specifically leads to impairment of all types of associative learning, whereas exploratory learning is not affected. We provide evidence for a novel function of n-cofilin function in synaptic plasticity and in the control of extrasynaptic excitatory AMPA receptors diffusion. These results suggest a critical function of actin dynamics in associative learning and postsynaptic receptor availability.


Asunto(s)
Actinas/fisiología , Cofilina 1/metabolismo , Aprendizaje , Plasticidad Neuronal/fisiología , Receptores AMPA/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Animales , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Espinas Dendríticas/metabolismo , Espinas Dendríticas/fisiología , Potenciación a Largo Plazo/fisiología , Memoria , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo
16.
Neurosci Lett ; 837: 137912, 2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-39032801

RESUMEN

Glyphosate (Gly) is the active ingredient of several widely used herbicide formulations. Studies on Gly and glyphosate-based herbicide (GBH) exposure in different experimental models have suggested that the nervous system represented a key target for its toxicity, especially the prefrontal cortex (PFC). However, it is still unknown whether exposure to GBH affects higher brain functions dependent on PFC circuitry. The present work aimed to examine the effects of subtoxic doses of GBH on social cognition and cognitive flexibility as two functions belonging to higher brain function in mice. To do so, adult male mice were exposed daily to GBH by gavage at doses of 250 or 500 mg/kg for a sub-chronic period lasting 6 weeks. Then, mice were subjected to behavioral testing using the three-chamber and the Barnes maze paradigms. Our results indicate that GBH did not affect sociability. However, we found that GBH affects social cognition expressed by a lower discrimination index in the three-chamber test. Moreover, spatial memories evaluated during the probe trial, and cognitive flexibility evaluated during the reversal probe, were affected in mice exposed to GBH. Based on these results, exposure to subtoxic doses of GBH led to neurobehavioral alterations affecting the integrity of social cognition and cognitive flexibility functions. Finally, these data urge a thorough investigation of the cellular and molecular mechanisms underlying these alterations.


Asunto(s)
Cognición , Glicina , Glifosato , Herbicidas , Animales , Glicina/análogos & derivados , Glicina/toxicidad , Herbicidas/toxicidad , Masculino , Ratones , Cognición/efectos de los fármacos , Cognición Social , Aprendizaje por Laberinto/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Conducta Social
17.
Hum Mol Genet ; 20(6): 1182-96, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21212100

RESUMEN

Rett syndrome (RTT) is a neurodevelopmental disorder with no efficient treatment that is caused in the majority of cases by mutations in the gene methyl-CpG binding-protein 2 (MECP2). RTT becomes manifest after a period of apparently normal development and causes growth deceleration, severe psychomotor impairment and mental retardation. Effective animal models for RTT are available and show morphofunctional abnormalities of synaptic connectivity. However, the molecular consequences of MeCP2 disruption leading to neuronal and synaptic alterations are not known. Protein synthesis regulation via the mammalian target of the rapamycin (mTOR) pathway is crucial for synaptic organization, and its disruption is involved in a number of neurodevelopmental diseases. We investigated the phosphorylation of the ribosomal protein (rp) S6, whose activation is highly dependent from mTOR activity. Immunohistochemistry showed that rpS6 phosphorylation is severely affected in neurons across the cortical areas of Mecp2 mutants and that this alteration precedes the severe symptomatic phase of the disease. Moreover, we found a severe defect of the initiation of protein synthesis in the brain of presymptomatic Mecp2 mutant that was not restricted to a specific subset of transcripts. Finally, we provide evidence for a general dysfunction of the Akt/mTOR, but not extracellular-regulated kinase, signaling associated with the disease progression in mutant brains. Our results indicate that defects in the AKT/mTOR pathway are responsible for the altered translational control in Mecp2 mutant neurons and disclosed a novel putative biomarker of the pathological process. Importantly, this study provides a novel context of therapeutic interventions that can be designed to successfully restrain or ameliorate the development of RTT.


Asunto(s)
Regulación hacia Abajo , Proteína Oncogénica v-akt/metabolismo , Biosíntesis de Proteínas , Síndrome de Rett/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Proteína Oncogénica v-akt/genética , Síndrome de Rett/genética , Serina-Treonina Quinasas TOR/genética
18.
Neuropsychopharmacology ; 48(6): 877-886, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-35945276

RESUMEN

Cyclin-dependent kinase-like 5 (CDKL5) deficiency disorder (CDD) is a devastating rare neurodevelopmental disease without a cure, caused by mutations of the serine/threonine kinase CDKL5 highly expressed in the forebrain. CDD is characterized by early-onset seizures, severe intellectual disabilities, autistic-like traits, sensorimotor and cortical visual impairments (CVI). The lack of an effective therapeutic strategy for CDD urgently demands the identification of novel druggable targets potentially relevant for CDD pathophysiology. To this aim, we studied Class I metabotropic glutamate receptors 5 (mGluR5) because of their important role in the neuropathological signs produced by the lack of CDKL5 in-vivo, such as defective synaptogenesis, dendritic spines formation/maturation, synaptic transmission and plasticity. Importantly, mGluR5 function strictly depends on the correct expression of the postsynaptic protein Homer1bc that we previously found atypical in the cerebral cortex of Cdkl5-/y mice. In this study, we reveal that CDKL5 loss tampers with (i) the binding strength of Homer1bc-mGluR5 complexes, (ii) the synaptic localization of mGluR5 and (iii) the mGluR5-mediated enhancement of NMDA-induced neuronal responses. Importantly, we showed that the stimulation of mGluR5 activity by administering in mice specific positive-allosteric-modulators (PAMs), i.e., 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) or RO6807794, corrected the synaptic, functional and behavioral defects shown by Cdkl5-/y mice. Notably, in the visual cortex of 2 CDD patients we found changes in synaptic organization that recapitulate those of mutant CDKL5 mice, including the reduced expression of mGluR5, suggesting that these receptors represent a promising therapeutic target for CDD.


Asunto(s)
Síndromes Epilépticos , Espasmos Infantiles , Ratones , Animales , Espasmos Infantiles/tratamiento farmacológico , Espasmos Infantiles/genética , Espasmos Infantiles/metabolismo , Síndromes Epilépticos/tratamiento farmacológico , Síndromes Epilépticos/genética , Síndromes Epilépticos/metabolismo , Neuronas/metabolismo , Modelos Animales de Enfermedad , Corteza Cerebral/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/uso terapéutico
19.
EMBO Mol Med ; 15(11): e15984, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37792911

RESUMEN

Cell signaling is central to neuronal activity and its dysregulation may lead to neurodegeneration and cognitive decline. Here, we show that selective genetic potentiation of neuronal ERK signaling prevents cell death in vitro and in vivo in the mouse brain, while attenuation of ERK signaling does the opposite. This neuroprotective effect mediated by an enhanced nuclear ERK activity can also be induced by the novel cell penetrating peptide RB5. In vitro administration of RB5 disrupts the preferential interaction of ERK1 MAP kinase with importinα1/KPNA2 over ERK2, facilitates ERK1/2 nuclear translocation, and enhances global ERK activity. Importantly, RB5 treatment in vivo promotes neuroprotection in mouse models of Huntington's (HD), Alzheimer's (AD), and Parkinson's (PD) disease, and enhances ERK signaling in a human cellular model of HD. Additionally, RB5-mediated potentiation of ERK nuclear signaling facilitates synaptic plasticity, enhances cognition in healthy rodents, and rescues cognitive impairments in AD and HD models. The reported molecular mechanism shared across multiple neurodegenerative disorders reveals a potential new therapeutic target approach based on the modulation of KPNA2-ERK1/2 interactions.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Neuroprotección , Animales , Humanos , Ratones , alfa Carioferinas/farmacología , Cognición , Fosforilación , Transducción de Señal
20.
Proc Natl Acad Sci U S A ; 106(24): 9872-7, 2009 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-19487674

RESUMEN

Activity-dependent changes in the strength of synaptic connections in the hippocampus are central for cognitive processes such as learning and memory storage. In this study, we reveal an activity-dependent presynaptic mechanism that is related to the modulation of synaptic plasticity. In acute mouse hippocampal slices, high-frequency stimulation (HFS) of the mossy fiber (MF)-CA3 pathway induced a strong and transient activation of extracellular-regulated kinase (ERK) in MF giant presynaptic terminals. Remarkably, pharmacological blockade of ERK disclosed a negative role of this kinase in the regulation of a presynaptic form of plasticity at MF-CA3 contacts. This ERK-mediated inhibition of post-tetanic enhancement (PTE) of MF-CA3 synapses was both frequency- and pathway-specific and was observed only with HFS at 50 Hz. Importantly, blockade of ERK was virtually ineffective on PTE of MF-CA3 synapses in mice lacking synapsin I, 1 of the major presynaptic ERK substrates, and triple knockout mice lacking all synapsin isoforms displayed PTE kinetics resembling that of wild-type mice under ERK inhibition. These findings reveal a form of short-term synaptic plasticity that depends on ERK and is finely tuned by the firing frequency of presynaptic neurons. Our results also demonstrate that presynaptic activation of the ERK signaling pathway plays part in the activity-dependent modulation of synaptic vesicle mobilization and transmitter release.


Asunto(s)
Axones , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipocampo/fisiología , Plasticidad Neuronal , Sinapsis/fisiología , Sinapsinas/fisiología , Animales , Activación Enzimática , Hipocampo/citología , Inmunohistoquímica , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA