Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 152(5): 1119-33, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23452857

RESUMEN

The activation of N-methyl-D-aspartate-receptors (NMDARs) in synapses provides plasticity and cell survival signals, whereas NMDARs residing in the neuronal membrane outside synapses trigger neurodegeneration. At present, it is unclear how these opposing signals are transduced to and discriminated by the nucleus. In this study, we demonstrate that Jacob is a protein messenger that encodes the origin of synaptic versus extrasynaptic NMDAR signals and delivers them to the nucleus. Exclusively synaptic, but not extrasynaptic, NMDAR activation induces phosphorylation of Jacob at serine-180 by ERK1/2. Long-distance trafficking of Jacob from synaptic, but not extrasynaptic, sites depends on ERK activity, and association with fragments of the intermediate filament α-internexin hinders dephosphorylation of the Jacob/ERK complex during nuclear transit. In the nucleus, the phosphorylation state of Jacob determines whether it induces cell death or promotes cell survival and enhances synaptic plasticity.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Animales , Supervivencia Celular , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Hipocampo/citología , Hipocampo/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Potenciación a Largo Plazo , Depresión Sináptica a Largo Plazo , Sistema de Señalización de MAP Quinasas , Ratones , Neuronas/citología , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Ratas
2.
EMBO J ; 42(4): e112453, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36594364

RESUMEN

Synaptic dysfunction caused by soluble ß-amyloid peptide (Aß) is a hallmark of early-stage Alzheimer's disease (AD), and is tightly linked to cognitive decline. By yet unknown mechanisms, Aß suppresses the transcriptional activity of cAMP-responsive element-binding protein (CREB), a master regulator of cell survival and plasticity-related gene expression. Here, we report that Aß elicits nucleocytoplasmic trafficking of Jacob, a protein that connects a NMDA-receptor-derived signalosome to CREB, in AD patient brains and mouse hippocampal neurons. Aß-regulated trafficking of Jacob induces transcriptional inactivation of CREB leading to impairment and loss of synapses in mouse models of AD. The small chemical compound Nitarsone selectively hinders the assembly of a Jacob/LIM-only 4 (LMO4)/ Protein phosphatase 1 (PP1) signalosome and thereby restores CREB transcriptional activity. Nitarsone prevents impairment of synaptic plasticity as well as cognitive decline in mouse models of AD. Collectively, the data suggest targeting Jacob protein-induced CREB shutoff as a therapeutic avenue against early synaptic dysfunction in AD.


Asunto(s)
Enfermedad de Alzheimer , Animales , Ratones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Ratones Transgénicos , Neuronas/metabolismo , Sinapsis/metabolismo
3.
PLoS Genet ; 12(3): e1005907, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26977770

RESUMEN

Jacob, the protein encoded by the Nsmf gene, is involved in synapto-nuclear signaling and docks an N-Methyl-D-Aspartate receptor (NMDAR)-derived signalosome to nuclear target sites like the transcription factor cAMP-response-element-binding protein (CREB). Several reports indicate that mutations in NSMF are related to Kallmann syndrome (KS), a neurodevelopmental disorder characterized by idiopathic hypogonadotropic hypogonadism (IHH) associated with anosmia or hyposmia. It has also been reported that a protein knockdown results in migration deficits of Gonadotropin-releasing hormone (GnRH) positive neurons from the olfactory bulb to the hypothalamus during early neuronal development. Here we show that mice that are constitutively deficient for the Nsmf gene do not present phenotypic characteristics related to KS. Instead, these mice exhibit hippocampal dysplasia with a reduced number of synapses and simplification of dendrites, reduced hippocampal long-term potentiation (LTP) at CA1 synapses and deficits in hippocampus-dependent learning. Brain-derived neurotrophic factor (BDNF) activation of CREB-activated gene expression plays a documented role in hippocampal CA1 synapse and dendrite formation. We found that BDNF induces the nuclear translocation of Jacob in an NMDAR-dependent manner in early development, which results in increased phosphorylation of CREB and enhanced CREB-dependent Bdnf gene transcription. Nsmf knockout (ko) mice show reduced hippocampal Bdnf mRNA and protein levels as well as reduced pCREB levels during dendritogenesis. Moreover, BDNF application can rescue the morphological deficits in hippocampal pyramidal neurons devoid of Jacob. Taken together, the data suggest that the absence of Jacob in early development interrupts a positive feedback loop between BDNF signaling, subsequent nuclear import of Jacob, activation of CREB and enhanced Bdnf gene transcription, ultimately leading to hippocampal dysplasia.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Dendritas/metabolismo , Hipocampo/crecimiento & desarrollo , Proteínas del Tejido Nervioso/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hormona Liberadora de Gonadotropina/metabolismo , Hipocampo/metabolismo , Ratones , Ratones Noqueados , Neuronas/metabolismo , Fosforilación , ARN Mensajero/biosíntesis , Transducción de Señal , Sinapsis/genética , Sinapsis/metabolismo
4.
Nature ; 486(7402): 256-60, 2012 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-22699619

RESUMEN

Autism spectrum disorders comprise a range of neurodevelopmental disorders characterized by deficits in social interaction and communication, and by repetitive behaviour. Mutations in synaptic proteins such as neuroligins, neurexins, GKAPs/SAPAPs and ProSAPs/Shanks were identified in patients with autism spectrum disorder, but the causative mechanisms remain largely unknown. ProSAPs/Shanks build large homo- and heteromeric protein complexes at excitatory synapses and organize the complex protein machinery of the postsynaptic density in a laminar fashion. Here we demonstrate that genetic deletion of ProSAP1/Shank2 results in an early, brain-region-specific upregulation of ionotropic glutamate receptors at the synapse and increased levels of ProSAP2/Shank3. Moreover, ProSAP1/Shank2(-/-) mutants exhibit fewer dendritic spines and show reduced basal synaptic transmission, a reduced frequency of miniature excitatory postsynaptic currents and enhanced N-methyl-d-aspartate receptor-mediated excitatory currents at the physiological level. Mutants are extremely hyperactive and display profound autistic-like behavioural alterations including repetitive grooming as well as abnormalities in vocal and social behaviours. By comparing the data on ProSAP1/Shank2(-/-) mutants with ProSAP2/Shank3αß(-/-) mice, we show that different abnormalities in synaptic glutamate receptor expression can cause alterations in social interactions and communication. Accordingly, we propose that appropriate therapies for autism spectrum disorders are to be carefully matched to the underlying synaptopathic phenotype.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Trastorno Autístico/genética , Conducta Animal/fisiología , Proteínas del Tejido Nervioso/genética , Agitación Psicomotora/genética , Animales , Trastorno Autístico/patología , Espinas Dendríticas/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Agitación Psicomotora/patología , Receptores Ionotrópicos de Glutamato/metabolismo , Sinapsis/metabolismo , Regulación hacia Arriba , Vocalización Animal/fisiología
5.
PLoS Biol ; 6(2): e34, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18303947

RESUMEN

NMDA (N-methyl-D-aspartate) receptors and calcium can exert multiple and very divergent effects within neuronal cells, thereby impacting opposing occurrences such as synaptic plasticity and neuronal degeneration. The neuronal Ca2+ sensor Caldendrin is a postsynaptic density component with high similarity to calmodulin. Jacob, a recently identified Caldendrin binding partner, is a novel protein abundantly expressed in limbic brain and cerebral cortex. Strictly depending upon activation of NMDA-type glutamate receptors, Jacob is recruited to neuronal nuclei, resulting in a rapid stripping of synaptic contacts and in a drastically altered morphology of the dendritic tree. Jacob's nuclear trafficking from distal dendrites crucially requires the classical Importin pathway. Caldendrin binds to Jacob's nuclear localization signal in a Ca2+-dependent manner, thereby controlling Jacob's extranuclear localization by competing with the binding of Importin-alpha to Jacob's nuclear localization signal. This competition requires sustained synapto-dendritic Ca2+ levels, which presumably cannot be achieved by activation of extrasynaptic NMDA receptors, but are confined to Ca2+ microdomains such as postsynaptic spines. Extrasynaptic NMDA receptors, as opposed to their synaptic counterparts, trigger the cAMP response element-binding protein (CREB) shut-off pathway, and cell death. We found that nuclear knockdown of Jacob prevents CREB shut-off after extrasynaptic NMDA receptor activation, whereas its nuclear overexpression induces CREB shut-off without NMDA receptor stimulation. Importantly, nuclear knockdown of Jacob attenuates NMDA-induced loss of synaptic contacts, and neuronal degeneration. This defines a novel mechanism of synapse-to-nucleus communication via a synaptic Ca2+-sensor protein, which links the activity of NMDA receptors to nuclear signalling events involved in modelling synapto-dendritic input and NMDA receptor-induced cellular degeneration.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Núcleo Celular/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal , Animales , Secuencia de Bases , Western Blotting , Proteínas de Unión al Calcio/genética , Cromatografía de Afinidad , Cartilla de ADN , ADN Complementario , Inmunohistoquímica , Señales de Localización Nuclear , Unión Proteica , Ratas , Técnicas del Sistema de Dos Híbridos
6.
Eur J Neurosci ; 32(1): 1-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20576033

RESUMEN

Small Rap guanosine-tri-phosphate (GTP)ases are crucially involved in many cellular processes, including cell proliferation, differentiation, survival, adhesion and movement. In line, it has been shown that Rap signalling is involved in various aspects of neuronal differentiation, like the establishment of neuronal polarity or axonal growth cone movement. Rap GTPases can be activated by a wide variety of external stimuli, and this is mediated by specific guanine nucleotide exchange factors (RapGEFs). Inactivation of RapGTP can be achieved with the aid of specific GTPase-activating proteins (RapGAPs). In the brain, the most prominent RapGAPs are Rap1GAP and those of the spine-associated RapGAP (SPAR) family. This latter family consists of three members (SPAR1-3), from which two of them, namely SPAR1 and 2, have been investigated in more detail. As such, the localization of RapGAPs is crucially important in regulating Rap signalling at various sites in the cell and, for both SPAR1 and 2, enrichment at synaptic sites has been demonstrated. In recent years particularly the role of SPAR1 in shaping dendritic spine morphology has attracted considerable interest. In this review we will summarize the described actions of different RapGAPs expressed in the brain, and we will focus in particular on the SPAR family members.


Asunto(s)
Encéfalo/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Transducción de Señal/fisiología , Proteínas de Unión al GTP rap1/metabolismo , Secuencia de Aminoácidos , Encéfalo/citología , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Activación Enzimática , Proteínas Activadoras de GTPasa/genética , Humanos , Datos de Secuencia Molecular , Neuronas/citología , Neuronas/metabolismo , Isoformas de Proteínas/metabolismo , Alineación de Secuencia , Proteínas de Unión al GTP rap1/genética
7.
Nat Commun ; 10(1): 5448, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31784514

RESUMEN

Amphisomes are organelles of the autophagy pathway that result from the fusion of autophagosomes with late endosomes. While biogenesis of autophagosomes and late endosomes occurs continuously at axon terminals, non-degradative roles of autophagy at boutons are barely described. Here, we show that in neurons BDNF/TrkB traffick in amphisomes that signal locally at presynaptic boutons during retrograde transport to the soma. This is orchestrated by the Rap GTPase-activating (RapGAP) protein SIPA1L2, which connects TrkB amphisomes to a dynein motor. The autophagosomal protein LC3 regulates RapGAP activity of SIPA1L2 and controls retrograde trafficking and local signaling of TrkB. Following induction of presynaptic plasticity, amphisomes dissociate from dynein at boutons enabling local signaling and promoting transmitter release. Accordingly, sipa1l2 knockout mice show impaired BDNF-dependent presynaptic plasticity. Taken together, the data suggest that in hippocampal neurons, TrkB-signaling endosomes are in fact amphisomes that during retrograde transport have local signaling capacity in the context of presynaptic plasticity.


Asunto(s)
Autofagosomas/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Endosomas/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Plasticidad Neuronal/genética , Neuronas/metabolismo , Terminales Presinápticos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Transporte Axonal , Axones/metabolismo , Dineínas/metabolismo , Proteínas Activadoras de GTPasa/genética , Hipocampo , Ratones , Ratones Noqueados , Transporte de Proteínas
8.
J Neurochem ; 104(1): 187-201, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17961154

RESUMEN

Spine-associated RapGAP 2 (SPAR2) is a novel GTPase activating protein (GAP) for the small GTPase Rap that shows significant sequence homology to SPAR, a synaptic RapGAP that was reported to regulate spine morphology in hippocampal neurons. SPAR2, like SPAR, interacts with the recently described synaptic scaffolding protein ProSAP-interacting protein (ProSAPiP), which in turn binds to the PDZ domain of ProSAP/Shank post-synaptic density proteins. In subcellular fractionation experiments, SPAR2 is enriched in synaptosomes and post-synaptic density fractions indicating that it is a synaptic protein. Furthermore, we could show using in vitro GAP assays that SPAR2 has GAP activity for Rap1 and Rap2. Expression in COS-7 cells, however, revealed different actin-binding properties of SPAR2 and SPAR. Additionally, over-expression of SPAR2 in cultured hippocampal neurons did not affect spine morphology as it was reported for SPAR. In situ hybridization studies also revealed a differential tissue distribution of SPAR and SPAR2 with SPAR2 transcripts being mainly expressed in cerebellar and hippocampal granule cells. Moreover, in the cerebellum SPAR2 is developmentally regulated with a peak of expression around the period of synapse formation. Our results imply that SPAR2 is a new RapGAP with specific functions in cerebellar and hippocampal granule cells.


Asunto(s)
Proteínas Activadoras de GTPasa/fisiología , Proteínas de Unión al GTP rap/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Animales , Animales Recién Nacidos , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Clonación Molecular/métodos , Embrión de Mamíferos , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Inmunoprecipitación/métodos , Hibridación in Situ/métodos , Neuronas/metabolismo , Ratas , Ratas Wistar , Fracciones Subcelulares/metabolismo , Sinapsis/metabolismo , Transfección/métodos
9.
Neuron ; 97(5): 1110-1125.e14, 2018 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-29478916

RESUMEN

Compartmentalization of calcium-dependent plasticity allows for rapid actin remodeling in dendritic spines. However, molecular mechanisms for the spatio-temporal regulation of filamentous actin (F-actin) dynamics by spinous Ca2+-transients are still poorly defined. We show that the postsynaptic Ca2+ sensor caldendrin orchestrates nano-domain actin dynamics that are essential for actin remodeling in the early phase of long-term potentiation (LTP). Steep elevation in spinous [Ca2+]i disrupts an intramolecular interaction of caldendrin and allows cortactin binding. The fast on and slow off rate of this interaction keeps cortactin in an active conformation, and protects F-actin at the spine base against cofilin-induced severing. Caldendrin gene knockout results in higher synaptic actin turnover, altered nanoscale organization of spinous F-actin, defects in structural spine plasticity, LTP, and hippocampus-dependent learning. Collectively, the data indicate that caldendrin-cortactin directly couple [Ca2+]i to preserve a minimal F-actin pool that is required for actin remodeling in the early phase of LTP.


Asunto(s)
Señalización del Calcio/fisiología , Proteínas de Unión al Calcio/deficiencia , Espinas Dendríticas/metabolismo , Potenciación a Largo Plazo/fisiología , Potenciales Sinápticos/fisiología , Animales , Células COS , Proteínas de Unión al Calcio/genética , Células Cultivadas , Chlorocebus aethiops , Espinas Dendríticas/química , Espinas Dendríticas/genética , Células HEK293 , Hipocampo/química , Hipocampo/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar
10.
Biochim Biophys Acta ; 1762(1): 66-72, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16257512

RESUMEN

The interaction between the EF-hand Ca(2+)-binding protein calmyrin and presenilin 2 (PS2) has been suggested to play a role in Alzheimer's disease (AD). We now report that calmyrin binds specifically endogenous PS2 and not PS1. However, binding appears to be Ca(2+)-independent and calmyrin does not exhibit a Ca(2+)-dependent translocation to intracellular membranes as demonstrated in a Ca(2+)-myristoyl switch assay. Moreover, calmyrin is only present at very low levels in brain areas associated with the onset of AD. In rat, forebrain calmyrin is localized only in a subset of principal neurons, similarly as in human forebrain. Finally, subcellular fractionation demonstrates only a limited overlap of calmyrin and PS2 at neuronal membranes. We therefore conclude that calmyrin will not contribute significantly as a Ca(2+)-sensor that transduces Ca(2+)-signaling events to PS2 in forebrain.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Señalización del Calcio , Proteínas de Unión al Calcio/metabolismo , Calcio/metabolismo , Perfilación de la Expresión Génica , Proteínas de la Membrana/metabolismo , Prosencéfalo/metabolismo , Animales , Células COS , Chlorocebus aethiops , Células HeLa , Humanos , Membranas Intracelulares/metabolismo , Presenilina-2 , Unión Proteica , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA