Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
EMBO J ; 43(4): 533-567, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38316990

RESUMEN

The phospholipid and free fatty acid (FFA) composition of neuronal membranes plays a crucial role in learning and memory, but the mechanisms through which neuronal activity affects the brain's lipid landscape remain largely unexplored. The levels of saturated FFAs, particularly of myristic acid (C14:0), strongly increase during neuronal stimulation and memory acquisition, suggesting the involvement of phospholipase A1 (PLA1) activity in synaptic plasticity. Here, we show that genetic ablation of the PLA1 isoform DDHD2 in mice dramatically reduces saturated FFA responses to memory acquisition across the brain. Furthermore, DDHD2 loss also decreases memory performance in reward-based learning and spatial memory models prior to the development of neuromuscular deficits that mirror human spastic paraplegia. Via pulldown-mass spectrometry analyses, we find that DDHD2 binds to the key synaptic protein STXBP1. Using STXBP1/2 knockout neurosecretory cells and a haploinsufficient STXBP1+/- mouse model of human early infantile encephalopathy associated with intellectual disability and motor dysfunction, we show that STXBP1 controls targeting of DDHD2 to the plasma membrane and generation of saturated FFAs in the brain. These findings suggest key roles for DDHD2 and STXBP1 in lipid metabolism and in the processes of synaptic plasticity, learning, and memory.


Asunto(s)
Ácidos Grasos no Esterificados , Memoria a Largo Plazo , Proteínas Munc18 , Fosfolipasas , Animales , Ratones , Encéfalo/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Memoria/fisiología , Proteínas Munc18/genética , Fosfolipasas/genética
2.
Hum Mol Genet ; 27(12): 2138-2153, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29659809

RESUMEN

The aristaless-related homeobox (ARX) transcription factor is involved in the development of GABAergic and cholinergic neurons in the forebrain. ARX mutations have been associated with a wide spectrum of neurodevelopmental disorders in humans, among which the most frequent, a 24 bp duplication in the polyalanine tract 2 (c.428_451dup24), gives rise to intellectual disability, fine motor defects with or without epilepsy. To understand the functional consequences of this mutation, we generated a partially humanized mouse model carrying the c.428_451dup24 duplication (Arxdup24/0) that we characterized at the behavior, neurological and molecular level. Arxdup24/0 males presented with hyperactivity, enhanced stereotypies and altered contextual fear memory. In addition, Arxdup24/0 males had fine motor defects with alteration of reaching and grasping abilities. Transcriptome analysis of Arxdup24/0 forebrains at E15.5 showed a down-regulation of genes specific to interneurons and an up-regulation of genes normally not expressed in this cell type, suggesting abnormal interneuron development. Accordingly, interneuron migration was altered in the cortex and striatum between E15.5 and P0 with consequences in adults, illustrated by the defect in the inhibitory/excitatory balance in Arxdup24/0 basolateral amygdala. Altogether, we showed that the c.428_451dup24 mutation disrupts Arx function with a direct consequence on interneuron development, leading to hyperactivity and defects in precise motor movement control and associative memory. Interestingly, we highlighted striking similarities between the mouse phenotype and a cohort of 33 male patients with ARX c.428_451dup24, suggesting that this new mutant mouse line is a good model for understanding the pathophysiology and evaluation of treatment.


Asunto(s)
Epilepsia/genética , Proteínas de Homeodominio/genética , Trastornos del Neurodesarrollo/genética , Factores de Transcripción/genética , Adolescente , Adulto , Animales , Niño , Preescolar , Neuronas Colinérgicas/metabolismo , Neuronas Colinérgicas/patología , Contractura , Modelos Animales de Enfermedad , Epilepsia/fisiopatología , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/patología , Regulación del Desarrollo de la Expresión Génica , Humanos , Lactante , Discapacidad Intelectual , Masculino , Ratones , Mutación , Trastornos del Neurodesarrollo/fisiopatología , Péptidos/genética , Prosencéfalo/fisiopatología , Paraplejía Espástica Hereditaria , Transcriptoma/genética , Adulto Joven
3.
PLoS Genet ; 13(7): e1006886, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28704368

RESUMEN

Koolen-de Vries syndrome (KdVS) is a multi-system disorder characterized by intellectual disability, friendly behavior, and congenital malformations. The syndrome is caused either by microdeletions in the 17q21.31 chromosomal region or by variants in the KANSL1 gene. The reciprocal 17q21.31 microduplication syndrome is associated with psychomotor delay, and reduced social interaction. To investigate the pathophysiology of 17q21.31 microdeletion and microduplication syndromes, we generated three mouse models: 1) the deletion (Del/+); or 2) the reciprocal duplication (Dup/+) of the 17q21.31 syntenic region; and 3) a heterozygous Kansl1 (Kans1+/-) model. We found altered weight, general activity, social behaviors, object recognition, and fear conditioning memory associated with craniofacial and brain structural changes observed in both Del/+ and Dup/+ animals. By investigating hippocampus function, we showed synaptic transmission defects in Del/+ and Dup/+ mice. Mutant mice with a heterozygous loss-of-function mutation in Kansl1 displayed similar behavioral and anatomical phenotypes compared to Del/+ mice with the exception of sociability phenotypes. Genes controlling chromatin organization, synaptic transmission and neurogenesis were upregulated in the hippocampus of Del/+ and Kansl1+/- animals. Our results demonstrate the implication of KANSL1 in the manifestation of KdVS phenotypes and extend substantially our knowledge about biological processes affected by these mutations. Clear differences in social behavior and gene expression profiles between Del/+ and Kansl1+/- mice suggested potential roles of other genes affected by the 17q21.31 deletion. Together, these novel mouse models provide new genetic tools valuable for the development of therapeutic approaches.


Asunto(s)
Anomalías Múltiples/genética , Duplicación Cromosómica/genética , Cognición , Discapacidad Intelectual/genética , Proteínas Nucleares/genética , Animales , Peso Corporal , Encéfalo/metabolismo , Encéfalo/ultraestructura , Deleción Cromosómica , Estructuras Cromosómicas/genética , Estructuras Cromosómicas/metabolismo , Cromosomas Humanos Par 17/genética , Variaciones en el Número de Copia de ADN , Modelos Animales de Enfermedad , Epigénesis Genética , Femenino , Eliminación de Gen , Reordenamiento Génico , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasticidad Neuronal/genética , Proteínas Nucleares/metabolismo , Transmisión Sináptica/genética , Regulación hacia Arriba
4.
J Neurosci ; 37(46): 11114-11126, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29030432

RESUMEN

Classical and systems genetics have identified wide networks of genes associated with cognitive and neurodevelopmental diseases. In parallel to deciphering the role of each of these genes in neuronal or synaptic function, evaluating the response of neuronal and molecular networks to gene loss of function could reveal some pathophysiological mechanisms potentially accessible to nongenetic therapies. Loss of function of the Rho-GAP oligophrenin-1 is associated with cognitive impairments in both human and mouse. Upregulation of both PKA and ROCK has been reported in Ophn1-/y mice, but it remains unclear whether kinase hyperactivity contributes to the behavioral phenotypes. In this study, we thoroughly characterized a prominent perseveration phenotype displayed by Ophn1-deficient mice using a Y-maze spatial working memory (SWM) test. We report that Ophn1 deficiency in the mouse generated severe cognitive impairments, characterized by both a high occurrence of perseverative behaviors and a lack of deliberation during the SWM test. In vivo and in vitro pharmacological experiments suggest that PKA dysregulation in the mPFC underlies cognitive dysfunction in Ophn1-deficient mice, as assessed using a delayed spatial alternation task results. Functionally, mPFC neuronal networks appeared to be affected in a PKA-dependent manner, whereas hippocampal-PFC projections involved in SWM were not affected in Ophn1-/y mice. Thus, we propose that discrete gene mutations in intellectual disability might generate "secondary" pathophysiological mechanisms, which are prone to become pharmacological targets for curative strategies in adult patients.SIGNIFICANCE STATEMENT Here we report that Ophn1 deficiency generates severe impairments in performance at spatial working memory tests, characterized by a high occurrence of perseverative behaviors and a lack of decision making. This cognitive deficit is consecutive to PKA deregulation in the mPFC that prevents Ophn1 KO mice to exploit a correctly acquired rule. Functionally, mPFC neuronal networks appear to be affected in a PKA-dependent manner, whereas behaviorally important hippocampal projections were preserved by the mutation. Thus, we propose that discrete gene mutations in intellectual disability can generate "secondary" pathophysiological mechanisms prone to become pharmacological targets for curative strategies in adults.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas del Citoesqueleto/deficiencia , Proteínas Activadoras de GTPasa/deficiencia , Trastornos de la Memoria/metabolismo , Memoria a Corto Plazo/fisiología , Proteínas Nucleares/deficiencia , Corteza Prefrontal/metabolismo , Animales , Masculino , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Red Nerviosa/metabolismo , Red Nerviosa/fisiopatología , Técnicas de Cultivo de Órganos , Corteza Prefrontal/fisiopatología , Distribución Aleatoria
5.
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
6.
Hum Mol Genet ; 24(4): 1106-18, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25305082

RESUMEN

Mutations in interleukin-1 receptor accessory protein like 1 (IL1RAPL1) gene have been associated with non-syndromic intellectual disability (ID) and autism spectrum disorder. This protein interacts with synaptic partners like PSD-95 and PTPδ, regulating the formation and function of excitatory synapses. The aim of this work was to characterize the synaptic consequences of three IL1RAPL1 mutations, two novel causing the deletion of exon 6 (Δex6) and one point mutation (C31R), identified in patients with ID. Using immunofluorescence and electrophysiological recordings, we examined the effects of IL1RAPL1 mutant over-expression on synapse formation and function in cultured rodent hippocampal neurons. Δex6 but not C31R mutation leads to IL1RAPL1 protein instability and mislocalization within dendrites. Analysis of different markers of excitatory synapses and sEPSC recording revealed that both mutants fail to induce pre- and post-synaptic differentiation, contrary to WT IL1RAPL1 protein. Cell aggregation and immunoprecipitation assays in HEK293 cells showed a reduction of the interaction between IL1RAPL1 mutants and PTPδ that could explain the observed synaptogenic defect in neurons. However, these mutants do not affect all cellular signaling because their over-expression still activates JNK pathway. We conclude that both mutations described in this study lead to a partial loss of function of the IL1RAPL1 protein through different mechanisms. Our work highlights the important function of the trans-synaptic PTPδ/IL1RAPL1 interaction in synaptogenesis and as such in ID in the patients.


Asunto(s)
Discapacidad Intelectual/genética , Proteína Accesoria del Receptor de Interleucina-1/genética , Mutación , Neurogénesis/genética , Sinapsis/genética , Adulto , Niño , Preescolar , Análisis Mutacional de ADN , Exones , Femenino , Humanos , Discapacidad Intelectual/metabolismo , Proteína Accesoria del Receptor de Interleucina-1/química , Proteína Accesoria del Receptor de Interleucina-1/metabolismo , Intrones , Masculino , Linaje , Polimorfismo de Nucleótido Simple , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Eliminación de Secuencia , Transducción de Señal , Sinapsis/metabolismo
7.
Hum Mol Genet ; 24(23): 6736-55, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26376863

RESUMEN

ATP6AP2, an essential accessory component of the vacuolar H+ ATPase (V-ATPase), has been associated with intellectual disability (ID) and Parkinsonism. ATP6AP2 has been implicated in several signalling pathways; however, little is known regarding its role in the nervous system. To decipher its function in behaviour and cognition, we generated and characterized conditional knockdowns of ATP6AP2 in the nervous system of Drosophila and mouse models. In Drosophila, ATP6AP2 knockdown induced defective phototaxis and vacuolated photoreceptor neurons and pigment cells when depleted in eyes and altered short- and long-term memory when depleted in the mushroom body. In mouse, conditional Atp6ap2 deletion in glutamatergic neurons (Atp6ap2(Camk2aCre/0) mice) caused increased spontaneous locomotor activity and altered fear memory. Both Drosophila ATP6AP2 knockdown and Atp6ap2(Camk2aCre/0) mice presented with presynaptic transmission defects, and with an abnormal number and morphology of synapses. In addition, Atp6ap2(Camk2aCre/0) mice showed autophagy defects that led to axonal and neuronal degeneration in the cortex and hippocampus. Surprisingly, axon myelination was affected in our mutant mice, and axonal transport alterations were observed in Drosophila. In accordance with the identified phenotypes across species, genome-wide transcriptome profiling of Atp6ap2(Camk2aCre/0) mouse hippocampi revealed dysregulation of genes involved in myelination, action potential, membrane-bound vesicles and motor behaviour. In summary, ATP6AP2 disruption in mouse and fly leads to cognitive impairment and neurodegeneration, mimicking aspects of the neuropathology associated with ATP6AP2 mutations in humans. Our results identify ATP6AP2 as an essential gene for the nervous system.


Asunto(s)
Trastornos del Conocimiento/etiología , Proteínas de Drosophila/genética , Proteínas de la Membrana/genética , Degeneración Nerviosa/etiología , ATPasas de Translocación de Protón/genética , Receptores de Superficie Celular/genética , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Drosophila , Femenino , Técnicas de Silenciamiento del Gen , Discapacidad Intelectual/genética , Masculino , Ratones , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/fisiología , Neuronas/ultraestructura , Trastornos Parkinsonianos/genética , Sinapsis/metabolismo , Sinapsis/fisiología , Sinapsis/ultraestructura
8.
PLoS Biol ; 12(3): e1001820, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24667537

RESUMEN

Cognitive and behavioral disorders are thought to be a result of neuronal dysfunction, but the underlying molecular defects remain largely unknown. An important signaling pathway involved in the regulation of neuronal function is the cyclic AMP/Protein kinase A pathway. We here show an essential role for coronin 1, which is encoded in a genomic region associated with neurobehavioral dysfunction, in the modulation of cyclic AMP/PKA signaling. We found that coronin 1 is specifically expressed in excitatory but not inhibitory neurons and that coronin 1 deficiency results in loss of excitatory synapses and severe neurobehavioral disabilities, including reduced anxiety, social deficits, increased aggression, and learning defects. Electrophysiological analysis of excitatory synaptic transmission in amygdala revealed that coronin 1 was essential for cyclic-AMP-protein kinase A-dependent presynaptic plasticity. We further show that upon cell surface stimulation, coronin 1 interacted with the G protein subtype Gαs to stimulate the cAMP/PKA pathway. The absence of coronin 1 or expression of coronin 1 mutants unable to interact with Gαs resulted in a marked reduction in cAMP signaling. Strikingly, synaptic plasticity and behavioral defects of coronin 1-deficient mice were restored by in vivo infusion of a membrane-permeable cAMP analogue. Together these results identify coronin 1 as being important for cognition and behavior through its activity in promoting cAMP/PKA-dependent synaptic plasticity and may open novel avenues for the dissection of signal transduction pathways involved in neurobehavioral processes.


Asunto(s)
Conducta Animal , Cognición/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Proteínas de Microfilamentos/fisiología , 4-Butirolactona/análogos & derivados , 4-Butirolactona/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Humanos , Memoria , Ratones , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Transducción de Señal , Conducta Social
9.
J Neurosci ; 33(50): 19470-9, 2013 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-24336713

RESUMEN

More than 80 human X-linked genes have been associated with mental retardation and deficits in learning and memory. However, most of the identified mutations induce limited morphological alterations in brain organization and the molecular bases underlying neuronal clinical features remain elusive. We show here that neurons cultured from mice lacking ribosomal S6 kinase 2 (Rsk2), a model for the Coffin-Lowry syndrome (CLS), exhibit a significant delay in growth in a similar way to that shown by neurons cultured from phospholipase D1 (Pld1) knock-out mice. We found that gene silencing of Pld1 or Rsk2 as well as acute pharmacological inhibition of PLD1 or RSK2 in PC12 cells strongly impaired neuronal growth factor (NGF)-induced neurite outgrowth. Expression of a phosphomimetic PLD1 mutant rescued the inhibition of neurite outgrowth in PC12 cells silenced for RSK2, revealing that PLD1 is a major target for RSK2 in neurite formation. NGF-triggered RSK2-dependent phosphorylation of PLD1 led to its activation and the synthesis of phosphatidic acid at sites of neurite growth. Additionally, total internal reflection fluorescence microscopy experiments revealed that RSK2 and PLD1 positively control fusion of tetanus neurotoxin insensitive vesicle-associated membrane protein (TiVAMP)/VAMP-7 vesicles at sites of neurite outgrowth. We propose that the loss of function mutations in RSK2 that leads to CLS and neuronal deficits are related to defects in neuronal growth due to impaired RSK2-dependent PLD1 activity resulting in a reduced vesicle fusion rate and membrane supply.


Asunto(s)
Neuritas/metabolismo , Ácidos Fosfatidicos/biosíntesis , Fosfolipasa D/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Animales , Células Cultivadas , Síndrome de Coffin-Lowry/genética , Síndrome de Coffin-Lowry/metabolismo , Ratones , Ratones Noqueados , Factor de Crecimiento Nervioso/farmacología , Neuritas/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Células PC12 , Fosforilación , Ratas , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética
10.
J Neurosci ; 33(34): 13805-19, 2013 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-23966701

RESUMEN

Intellectual disorders (IDs) have been regularly associated with morphological and functional deficits at glutamatergic synapses in both humans and rodents. How these synaptic deficits may lead to the variety of learning and memory deficits defining ID is still unknown. Here we studied the functional and behavioral consequences of the ID gene il1rapl1 deficiency in mice and reported that il1rapl1 constitutive deletion alters cued fear memory formation. Combined in vivo and in vitro approaches allowed us to unveil a causal relationship between a marked inhibitory/excitatory (I/E) imbalance in dedicated amygdala neuronal subcircuits and behavioral deficits. Cell-targeted recordings further demonstrated a morpho-functional impact of the mutation at thalamic projections contacting principal cells, whereas the same afferents on interneurons are unaffected by the lack of Il1rapl1. We thus propose that excitatory synapses have a heterogeneous vulnerability to il1rapl1 gene constitutive mutation and that alteration of a subset of excitatory synapses in neuronal circuits is sufficient to generate permanent cognitive deficits.


Asunto(s)
Potenciales Postsinápticos Excitadores/genética , Discapacidad Intelectual/complicaciones , Trastornos de la Memoria/etiología , Amígdala del Cerebelo/citología , Anestésicos Locales/farmacología , Animales , Aprendizaje por Asociación/fisiología , Corteza Cerebral/citología , Channelrhodopsins , Condicionamiento Psicológico/fisiología , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Miedo/fisiología , Antagonistas del GABA/farmacología , Glutamato Descarboxilasa/genética , Proteínas Fluorescentes Verdes/genética , Técnicas In Vitro , Discapacidad Intelectual/genética , Proteína Accesoria del Receptor de Interleucina-1/genética , Proteína Accesoria del Receptor de Interleucina-1/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Inhibición Neural/efectos de los fármacos , Inhibición Neural/genética , Neuronas/fisiología , Neuronas/ultraestructura
11.
Methods Cell Biol ; 188: 237-254, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38880526

RESUMEN

The prevalence of central nervous system (CNS) dysfunction as a result of disease or trauma remains a clinically unsolved problem which is raising increased awareness in our aging society. Human Dental Pulp Stem Cells (hDPSCs) are excellent candidates to be used in tissue engineering and regenerative therapies of the CNS due to their neural differentiation ability and lack of tumorigenicity. Accordingly, they have been successfully used in animal models of spinal cord injury, stroke and peripheral neuropathies. The ideal therapy in brain injury should combine strategies aiming to protect the damaged lesion and, at the same time, accelerate brain tissue regeneration, thus promoting fast recovery while minimizing side or long-term effects. The use of bioresorbable nanopatterned poly(lactide-co-ɛ-caprolactone) (PLCL) polymeric scaffolds as hDPCSs carriers can represent an advantage for tissue regeneration. In this chapter, we describe the surgical procedures to implant functionalized bioresorbable scaffolds loaded with hDPSCs to improve the brain lesion microenvironment in an intracranial stab wound injury model severing the rostral migratory stream (RMS) that connects the brain subventricular zone (SVZ) and the olfactory bulb in nude mice. Additionally, we also describe the technical steps after animal sacrifice for histological tissue observation and characterization.


Asunto(s)
Pulpa Dental , Modelos Animales de Enfermedad , Ratones Desnudos , Células Madre , Andamios del Tejido , Pulpa Dental/citología , Animales , Humanos , Andamios del Tejido/química , Ratones , Células Madre/citología , Trasplante de Células Madre/métodos , Heridas Punzantes/terapia , Implantes Absorbibles , Lesiones Encefálicas/terapia , Lesiones Encefálicas/patología , Ingeniería de Tejidos/métodos
12.
Semin Cell Dev Biol ; 22(4): 425-33, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21843652

RESUMEN

Data collected from the invertebrate models have allowed to establish several of the basic mechanisms of neuronal function and pioneered the studies on the molecular and cellular mechanisms involved in behavioral responses. In the 1970s, the first synaptic proteins--including synapsin--being identified, the first attempts to evaluate their synaptic function were done using available invertebrate preparations. Forty years later, it appears that deductions made from invertebrate synapsin were largely validated in vertebrates, probably reflecting the phylogenic conservation of some specific synapsin sub-domains. In this review, in light of insights got from invertebrate preparations, we discuss the role of synapsin in synaptogenesis and synaptic function, especially on short term plasticity.


Asunto(s)
Invertebrados/metabolismo , Sinapsinas/metabolismo , Animales , Humanos , Plasticidad Neuronal , Sinapsinas/química
13.
Front Cell Neurosci ; 17: 1212202, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37435048

RESUMEN

Imbalance between excitation and inhibition in the cerebral cortex is one of the main theories in neuropsychiatric disorder pathophysiology. Cortical inhibition is finely regulated by a variety of highly specialized GABAergic interneuron types, which are thought to organize neural network activities. Among interneurons, axo-axonic cells are unique in making synapses with the axon initial segment of pyramidal neurons. Alterations of axo-axonic cells have been proposed to be implicated in disorders including epilepsy, schizophrenia and autism spectrum disorder. However, evidence for the alteration of axo-axonic cells in disease has only been examined in narrative reviews. By performing a systematic review of studies investigating axo-axonic cells and axo-axonic communication in epilepsy, schizophrenia and autism spectrum disorder, we outline convergent findings and discrepancies in the literature. Overall, the implication of axo-axonic cells in neuropsychiatric disorders might have been overstated. Additional work is needed to assess initial, mostly indirect findings, and to unravel how defects in axo-axonic cells translates to cortical dysregulation and, in turn, to pathological states.

14.
Nat Commun ; 14(1): 8312, 2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-38097535

RESUMEN

The consolidation of recent memories depends on memory replays, also called ripples, generated within the hippocampus during slow-wave sleep, and whose inactivation leads to memory impairment. For now, the mobilisation, localisation and importance of synaptic plasticity events associated to ripples are largely unknown. To tackle this question, we used cell surface AMPAR immobilisation to block post-synaptic LTP within the hippocampal region of male mice during a spatial memory task, and show that: 1- hippocampal synaptic plasticity is engaged during consolidation, but is dispensable during encoding or retrieval. 2- Plasticity blockade during sleep results in apparent forgetting of the encoded rule. 3- In vivo ripple recordings show a strong effect of AMPAR immobilisation when a rule has been recently encoded. 4- In situ investigation suggests that plasticity at CA3-CA3 recurrent synapses supports ripple generation. We thus propose that post-synaptic AMPAR mobility at CA3 recurrent synapses is necessary for ripple-dependent rule consolidation.


Asunto(s)
Consolidación de la Memoria , Ratones , Masculino , Animales , Consolidación de la Memoria/fisiología , Hipocampo/fisiología , Plasticidad Neuronal/fisiología , Sueño/fisiología , Memoria Espacial , Región CA1 Hipocampal/fisiología , Región CA3 Hipocampal/fisiología
15.
Sci Adv ; 8(30): eabm5298, 2022 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-35895810

RESUMEN

Regulation of synaptic neurotransmitter receptor content is a fundamental mechanism for tuning synaptic efficacy during experience-dependent plasticity and behavioral adaptation. However, experimental approaches to track and modify receptor movements in integrated experimental systems are limited. Exploiting AMPA-type glutamate receptors (AMPARs) as a model, we generated a knock-in mouse expressing the biotin acceptor peptide (AP) tag on the GluA2 extracellular N-terminal. Cell-specific introduction of biotin ligase allows the use of monovalent or tetravalent avidin variants to respectively monitor or manipulate the surface mobility of endogenous AMPAR containing biotinylated AP-GluA2 in neuronal subsets. AMPAR immobilization precluded the expression of long-term potentiation and formation of contextual fear memory, allowing target-specific control of the expression of synaptic plasticity and animal behavior. The AP tag knock-in model offers unprecedented access to resolve and control the spatiotemporal dynamics of endogenous receptors, and opens new avenues to study the molecular mechanisms of synaptic plasticity and learning.

16.
J Neurosci ; 30(49): 16679-91, 2010 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-21148007

RESUMEN

Repetitive firing of neurons at a low frequency often leads to a decrease in synaptic strength. The mechanism of this low-frequency depression (LFD) is poorly understood. Here, LFD was studied at Aplysia cholinergic synapses. The absence of a significant change in the paired-pulse ratio during LFD, together with the facts that neither the time course nor the extent of LFD were affected by the initial release probability, suggests that LFD is not related to a depletion of the ready-to-fuse synaptic vesicles (SVs) or to a decrease in the release probability, but results from the silencing of a subpopulation of release sites. A subset of SVs or release sites, which acquired a high release probability status during LFD, permits synapses to rapidly and temporarily recover the initial synaptic strength when the stimulation is stopped. However, the recovery of the full capacity of the synapse to sustain repetitive stimulations is slow and involves spontaneous reactivation of the silent release sites. Application of tetanic stimulations accelerates this recovery by immediately switching on the silent sites. This high-frequency-dependent phenomenon underlies a new form of synaptic plasticity that allows resetting of presynaptic efficiency independently of the recent history of the synapse. Microinjection of a mutated Aplysia synapsin that cannot be phosphorylated by cAMP-dependent protein kinase (PKA), or a PKA inhibitor both prevented high-frequency-dependent awakening of release sites. Changes in the firing pattern of neurons appear to be able to regulate the on-off status of release sites via a molecular cascade involving PKA-dependent phosphorylation of synapsin.


Asunto(s)
Fenómenos Biofísicos/fisiología , Inhibición Neural/fisiología , Plasticidad Neuronal/fisiología , Neuronas/citología , Sinapsis/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Análisis de Varianza , Animales , Aplysia , Fenómenos Biofísicos/efectos de los fármacos , Calcio/metabolismo , Estimulación Eléctrica/métodos , Inhibidores Enzimáticos/farmacología , Ganglios de Invertebrados/citología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Magnesio/metabolismo , Microinyecciones/métodos , Modelos Neurológicos , Inhibición Neural/efectos de los fármacos , Neurotransmisores/metabolismo , Probabilidad , Sinapsis/efectos de los fármacos , Factores de Tiempo
17.
Proc Natl Acad Sci U S A ; 105(39): 15130-5, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18815362

RESUMEN

NMDA receptor-dependent long-term potentiation (LTP) of glutamatergic synaptic transmission in sensory pathways from auditory thalamus or cortex to the lateral amygdala (LA) underlies the acquisition of auditory fear conditioning. Whereas the mechanisms of postsynaptic LTP at thalamo-LA synapses are well understood, much less is known about the sequence of events mediating presynaptic NMDA receptor-dependent LTP at cortico-LA synapses. Here, we show that presynaptic cortico-LA LTP can be entirely accounted for by a persistent increase in the vesicular release probability. At the molecular level, we found that signaling via the cAMP/PKA pathway is necessary and sufficient for LTP induction. Moreover, by using mice lacking the active-zone protein and PKA target RIM1alpha (RIM1alpha(-/-)), we demonstrate that RIM1alpha is required for both chemically and synaptically induced presynaptic LTP. Further analysis of cortico-LA synaptic transmission in RIM1alpha(-/-) mice revealed a deficit in Ca(2+)-release coupling leading to a lower baseline release probability. Our results reveal the molecular mechanisms underlying the induction of presynaptic LTP at cortico-LA synapses and indicate that RIM1alpha-dependent LTP may involve changes in Ca(2+)-release coupling.


Asunto(s)
Amígdala del Cerebelo/fisiología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Proteínas de Unión al GTP/metabolismo , Potenciación a Largo Plazo/fisiología , Terminales Presinápticos/fisiología , Amígdala del Cerebelo/metabolismo , Animales , Calcio/metabolismo , Proteínas de Unión al GTP/genética , Masculino , Ratones , Ratones Mutantes , Terminales Presinápticos/metabolismo , Transducción de Señal , Transmisión Sináptica
18.
Nat Commun ; 12(1): 1557, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33692361

RESUMEN

Fragile X syndrome (FXS) is the most frequent form of inherited intellectual disability and the best-described monogenic cause of autism. CGG-repeat expansion in the FMR1 gene leads to FMR1 silencing, loss-of-expression of the Fragile X Mental Retardation Protein (FMRP), and is a common cause of FXS. Missense mutations in the FMR1 gene were also identified in FXS patients, including the recurrent FMRP-R138Q mutation. To investigate the mechanisms underlying FXS caused by this mutation, we generated a knock-in mouse model (Fmr1R138Q) expressing the FMRP-R138Q protein. We demonstrate that, in the hippocampus of the Fmr1R138Q mice, neurons show an increased spine density associated with synaptic ultrastructural defects and increased AMPA receptor-surface expression. Combining biochemical assays, high-resolution imaging, electrophysiological recordings, and behavioural testing, we also show that the R138Q mutation results in impaired hippocampal long-term potentiation and socio-cognitive deficits in mice. These findings reveal the functional impact of the FMRP-R138Q mutation in a mouse model of FXS.


Asunto(s)
Disfunción Cognitiva/genética , Disfunción Cognitiva/fisiopatología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Mutación Missense/fisiología , Receptores de Glutamato/metabolismo , Animales , Biotinilación , Encéfalo/metabolismo , Encéfalo/fisiopatología , Células Cultivadas , Disfunción Cognitiva/metabolismo , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Hipocampo/metabolismo , Hipocampo/fisiopatología , Humanos , Immunoblotting , Potenciación a Largo Plazo/genética , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Mutación Missense/genética , Técnicas de Placa-Clamp , Receptores de Glutamato/genética
19.
Nature ; 426(6968): 841-5, 2003 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-14685239

RESUMEN

The induction of associative synaptic plasticity in the mammalian central nervous system classically depends on coincident presynaptic and postsynaptic activity. According to this principle, associative homosynaptic long-term potentiation (LTP) of excitatory synaptic transmission can be induced only if synaptic release occurs during postsynaptic depolarization. In contrast, heterosynaptic plasticity in mammals is considered to rely on activity-independent, non-associative processes. Here we describe a novel mechanism underlying the induction of associative LTP in the lateral amygdala (LA). Simultaneous activation of converging cortical and thalamic afferents specifically induced associative, N-methyl-D-aspartate (NMDA)-receptor-dependent LTP at cortical, but not at thalamic, inputs. Surprisingly, the induction of associative LTP at cortical inputs was completely independent of postsynaptic activity, including depolarization, postsynaptic NMDA receptor activation or an increase in postsynaptic Ca2+ concentration, and did not require network activity. LTP expression was mediated by a persistent increase in the presynaptic probability of release at cortical afferents. Our study shows the presynaptic induction and expression of heterosynaptic and associative synaptic plasticity on simultaneous activity of converging afferents. Our data indicate that input specificity of associative LTP can be determined exclusively by presynaptic properties.


Asunto(s)
Amígdala del Cerebelo/fisiología , Plasticidad Neuronal , Sinapsis/metabolismo , Animales , Calcio/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica
20.
Nat Neurosci ; 9(8): 1028-35, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16819521

RESUMEN

Pavlovian fear conditioning, a simple form of associative learning, is thought to involve the induction of associative, NMDA receptor-dependent long-term potentiation (LTP) in the lateral amygdala. Using a combined genetic and electrophysiological approach, we show here that lack of a specific GABA(B) receptor subtype, GABA(B(1a,2)), unmasks a nonassociative, NMDA receptor-independent form of presynaptic LTP at cortico-amygdala afferents. Moreover, the level of presynaptic GABA(B(1a,2)) receptor activation, and hence the balance between associative and nonassociative forms of LTP, can be dynamically modulated by local inhibitory activity. At the behavioral level, genetic loss of GABA(B(1a)) results in a generalization of conditioned fear to nonconditioned stimuli. Our findings indicate that presynaptic inhibition through GABA(B(1a,2)) receptors serves as an activity-dependent constraint on the induction of homosynaptic plasticity, which may be important to prevent the generalization of conditioned fear.


Asunto(s)
Amígdala del Cerebelo/fisiología , Aprendizaje por Asociación/fisiología , Condicionamiento Clásico/fisiología , Miedo , Potenciación a Largo Plazo/fisiología , Receptores de GABA-B/metabolismo , Amígdala del Cerebelo/citología , Animales , Conducta Animal/fisiología , Antagonistas de Receptores de GABA-B , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Neuronas/ultraestructura , Técnicas de Placa-Clamp , Isoformas de Proteínas/metabolismo , Receptores de GABA-B/genética , Transmisión Sináptica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA