Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Brain Sci ; 11(9)2021 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-34573232

RESUMEN

Alzheimer's disease (AD), the most common neurodegenerative disease, is characterized by progressive cognitive impairment. The deposition of amyloid beta (Aß) and hyperphosphorylated tau is considered the hallmark of AD pathology. Many therapeutic approaches such as Food and Drug Administration-approved cholinesterase inhibitors and N-methyl-D-aspartate receptor antagonists have been used to intervene in AD pathology. However, current therapies only provide limited symptomatic relief and are ineffective in preventing AD progression. Cannabidiol (CBD), a phytocannabinoid devoid of psychoactive responses, provides neuroprotective effects through both cannabinoid and noncannabinoid receptors. Recent studies using an AD mouse model have suggested that CBD can reverse cognitive deficits along with Aß-induced neuroinflammatory, oxidative responses, and neuronal death. Furthermore, CBD can reduce the accumulation of Aß and hyperphosphorylation of tau, suggesting the possibility of delaying AD progression. Particularly, the noncannabinoid receptor, peroxisome proliferator-activated receptor gamma, has been suggested to be involved in multiple functions of CBD. Therefore, understanding the underlying mechanisms of CBD is necessary for intervening in AD pathology in depth and for the translation of preclinical studies into clinical settings. In this review, we summarize recent studies on the effect of CBD in AD and suggest problems to be overcome for the therapeutic use of CBD.

2.
Mol Psychiatry ; 26(12): 7538-7549, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34253863

RESUMEN

Heterogeneity in the etiopathology of autism spectrum disorders (ASD) limits the development of generic remedies, requires individualistic and patient-specific research. Recent progress in human-induced pluripotent stem cell (iPSC) technology provides a novel platform for modeling ASDs for studying complex neuronal phenotypes. In this study, we generated telencephalic induced neuronal (iN) cells from iPSCs derived from an ASD patient with a heterozygous point mutation in the DSCAM gene. The mRNA of DSCAM and the density of DSCAM in dendrites were significantly decreased in ASD compared to control iN cells. RNA sequencing analysis revealed that several synaptic function-related genes including NMDA receptor subunits were downregulated in ASD iN cells. Moreover, NMDA receptor (R)-mediated currents were significantly reduced in ASD compared to control iN cells. Normal NMDA-R-mediated current levels were rescued by expressing wild-type DSCAM in ASD iN cells, and reduced currents were observed by truncated DSCAM expression in control iN cells. shRNA-mediated DSCAM knockdown in control iN cells resulted in the downregulation of an NMDA-R subunit, which was rescued by the overexpression of shRNA-resistant DSCAM. Furthermore, DSCAM was co-localized with NMDA-R components in the dendritic spines of iN cells whereas their co-localizations were significantly reduced in ASD iN cells. Levels of phospho-ERK1/2 were significantly lower in ASD iN cells, suggesting a potential mechanism. A neural stem cell-specific Dscam heterozygous knockout mouse model, showing deficits in social interaction and social memory with reduced NMDA-R currents. These data suggest that DSCAM mutation causes pathological symptoms of ASD by dysregulating NMDA-R function.


Asunto(s)
Trastorno del Espectro Autista , Moléculas de Adhesión Celular/genética , Receptores de N-Metil-D-Aspartato , Animales , Trastorno del Espectro Autista/metabolismo , Humanos , Ratones , Ratones Noqueados , Mutación/genética , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
3.
Neurosci Res ; 161: 8-17, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33007326

RESUMEN

Successfully navigating dynamic environments requires balancing the decision to stay at an optimal choice with that to switch to an alternative to acquire new knowledge. However, the genetic factors and cellular activity shaping this "stay or switch" action decision remains largely unidentified. Here we find that mice carrying a deletion of the exchange protein directly activated by cAMP 2 (Epac2) gene, a putative autism locus, exhibit perseverative "stay" behavior in a dynamic foraging task. Anatomical analysis found that the loss of Epac2 resulted in a significant decrease in the density of PV-expressing interneurons in the ventrolateral orbitofrontal cortex (OFC) and dorsal striatum (dSTR). Further, in vitro whole cell patch clamp recordings of PV+ GABAergic interneurons in the dSTR revealed altered neural activity in Epac2 KO mice in response to dopamine. Our findings highlight a potential role of Epac2 in structural changes and neural responses of PV-expressing GABAergic interneurons in the ventrolateral OFC and dSTR during value-based reinforcement learning and link Epac2 function to abnormal decision-making processes and perseverative behaviors seen in autism.


Asunto(s)
Interneuronas , Recompensa , Animales , Toma de Decisiones , Dopamina , Ratones , Técnicas de Placa-Clamp , Corteza Prefrontal
4.
J Clin Med ; 9(6)2020 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-32560273

RESUMEN

Cohen syndrome (CS), a rare autosomal recessive disorder, has been associated with genetic mutations in the VPS13B gene, which regulates vesicle-mediated protein sorting and transport. However, the cellular mechanism underlying CS pathogenesis in patient-derived human neurons remains unknown. We identified a novel compound heterozygous mutation, due to homozygous variation of biparental origin and heterozygous variation inherited from the father, in the VPS13B gene in a 20-month-old female patient. To understand the cellular pathogenic mechanisms, we generated induced pluripotent stem cells (iPSCs) from the fibroblasts of the CS patient. The iPSCs were differentiated into forebrain-like functional glutamatergic neurons or neurospheres. Functional annotation from transcriptomic analysis using CS iPSC-derived neurons revealed that synapse-related functions were enriched among the upregulated and downregulated genes in the CS neurons, whereas processes associated with neurodevelopment were enriched in the downregulated genes. The developing CS neurospheres were small in size compared to control neurospheres, likely due to the reduced proliferation of SOX2-positive neural stem cells. Moreover, the number of SV2B-positive puncta and spine-like structures was significantly reduced in the CS neurons, suggesting synaptic dysfunction. Taking these findings together, for the first time, we report a potential cellular pathogenic mechanism which reveals the alteration of neurodevelopment-related genes and the dysregulation of synaptic function in the human induced neurons differentiated from iPSCs and neurospheres of a CS patient.

5.
Mol Brain ; 13(1): 69, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32375900

RESUMEN

Significant clinical symptoms of Cohen syndrome (CS), a rare autosomal recessive disorder, include intellectual disability, facial dysmorphism, postnatal microcephaly, retinal dystrophy, and intermittent neutropenia. CS has been associated with mutations in the VPS13B (vacuolar protein sorting 13 homolog B) gene, which regulates vesicle-mediated protein sorting and transport; however, the cellular mechanism underlying CS pathogenesis in patient-derived neurons remains uncertain. This report states that autophagic vacuoles accumulate in CS fibroblasts and the axonal terminals of CS patient-specific induced pluripotent stem cells (CS iPSC)-derived neurons; additionally, autophagic flux was significantly increased in CS-derived neurons compared to control neurons. VPS13B knockout HeLa cell lines generated using the CRISPR/Cas9 genome editing system showed significant upregulation of autophagic flux, indicating that VSP13B may be associated with autophagy in CS. Transcriptomic analysis focusing on the autophagy pathway revealed that genes associated with autophagosome organization were dysregulated in CS-derived neurons. ATG4C is a mammalian ATG4 paralog and a crucial regulatory component of the autophagosome biogenesis/recycling pathway. ATG4C was significantly upregulated in CS-derived neurons, indicating that autophagy is upregulated in CS neurons. The autophagy pathway in CS neurons may be associated with the pathophysiology exhibited in the neural network of CS patients.


Asunto(s)
Autofagosomas/metabolismo , Autofagia/genética , Fibroblastos/metabolismo , Dedos/anomalías , Células Madre Pluripotentes Inducidas/metabolismo , Discapacidad Intelectual/metabolismo , Microcefalia/metabolismo , Hipotonía Muscular/metabolismo , Miopía/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Degeneración Retiniana/metabolismo , Proteínas de Transporte Vesicular/genética , Autofagosomas/genética , Autofagosomas/ultraestructura , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Axones/metabolismo , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Discapacidades del Desarrollo/metabolismo , Discapacidades del Desarrollo/fisiopatología , Fibroblastos/patología , Fibroblastos/ultraestructura , Dedos/fisiopatología , Técnicas de Inactivación de Genes , Células HeLa , Humanos , Células Madre Pluripotentes Inducidas/patología , Discapacidad Intelectual/fisiopatología , Microcefalia/fisiopatología , Microscopía Electrónica , Hipotonía Muscular/fisiopatología , Mutación Missense , Miopía/fisiopatología , Red Nerviosa/fisiología , Neuronas/patología , Obesidad/fisiopatología , Degeneración Retiniana/fisiopatología , Regulación hacia Arriba , Vacuolas/metabolismo
6.
Mol Brain ; 13(1): 54, 2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-32252796

RESUMEN

Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders that are highly heterogeneous in clinical symptoms as well as etiologies. Mutations in SHANK2 are associated with ASD and accordingly, Shank2 knockout mouse shows ASD-like behavioral phenotypes, including social deficits. Intriguingly, two lines of Shank2 knockout (KO) mouse generated by deleting different exons (exon 6-7 or exon 7) showed distinct cellular phenotypes. Previously, we compared gene expressions between Shank2 KOs lacking exon 6-7 (e6-7 KO) and KOs lacking exon 7 (e7 KO) by performing RNA-seq. In this study, we expanded transcriptomic analyses to identify novel transcriptional variants in the KO mice. We found prominent expression of a novel exon (exon 4' or e4') between the existing exons 4 and 5 in the Shank2 e6-7 KO model. Expression of the transcriptional variant harboring this novel exon was confirmed by RT-PCR and western blotting. These findings suggest that the novel variant may function as a modifier gene, which contributes to the differences between the two Shank2 mutant lines. Furthermore, our result further represents an example of genetic compensation that may lead to phenotypic heterogeneity among ASD patients with mutations in the same gene.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Transcripción Genética , Animales , Encéfalo/metabolismo , Exones/genética , Regulación de la Expresión Génica , Genoma , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
7.
Exp Neurobiol ; 28(4): 485-494, 2019 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-31495077

RESUMEN

Vacuolar protein sorting-associated protein 13B (VPS13B), also known as COH1, is one of the VPS13 family members which is involved in transmembrane transport, Golgi integrity, and neuritogenesis. Mutations in the VPS13B gene are associated with Cohen syndrome and other cognitive disorders such as intellectual disabilities and autism spectrum disorder (ASD). However, the pathophysiology of VPS13B-associated cognitive deficits is unclear, in part, due to the lack of animal models. Here, we generated a Vps13b exon 2 deletion mutant mouse and analyzed the behavioral phenotypes. We found that Vps13b mutant mice showed reduced activity in open field test and significantly shorter latency to fall in the rotarod test, suggesting that the mutants have motor deficits. In addition, we found that Vps13b mutant mice showed deficits in spatial learning in the hidden platform version of the Morris water maze. The Vps13b mutant mice were normal in other behaviors such as anxiety-like behaviors, working memory and social behaviors. Our results suggest that Vps13b mutant mice may recapitulate key clinical symptoms in Cohen syndrome such as intellectual disability and hypotonia. Vps13b mutant mice may serve as a useful model to investigate the pathophysiology of Vps13b-associated disorders.

8.
Int J Mol Sci ; 20(18)2019 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-31491992

RESUMEN

Hepatocyte death is critical for the pathogenesis of liver disease progression, which is closely associated with endoplasmic reticulum (ER) stress responses. However, the molecular basis for ER stress-mediated hepatocyte injury remains largely unknown. This study investigated the effect of ER stress on dual-specificity phosphatase 5 (DUSP5) expression and its role in hepatocyte death. Analysis of Gene Expression Omnibus (GEO) database showed that hepatic DUSP5 levels increased in the patients with liver fibrosis, which was verified in mouse models of liver diseases with ER stress. DUSP5 expression was elevated in both fibrotic and acutely injured liver of mice treated with liver toxicants. Treatment of ER stress inducers enhanced DUSP5 expression in hepatocytes, which was validated in vivo condition. The induction of DUSP5 by ER stress was blocked by either treatment with a chemical inhibitor of the protein kinase RNA-like endoplasmic reticulum kinase (PERK) pathway, or knockdown of C/EBP homologous protein (CHOP), whereas it was not affected by the silencing of IRE1 or ATF6. In addition, DUSP5 overexpression decreased extracellular-signal-regulated kinase (ERK) phosphorylation, but increased cleaved caspase-3 levels. Moreover, the reduction of cell viability under ER stress condition was attenuated by DUSP5 knockdown. In conclusion, DUSP5 expression is elevated in hepatocytes by ER stress through the PERK-CHOP pathway, contributing to hepatocyte death possibly through ERK inhibition.


Asunto(s)
Fosfatasas de Especificidad Dual/genética , Estrés del Retículo Endoplásmico , Hepatocitos/metabolismo , Transducción de Señal , Factor de Transcripción CHOP/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Apoptosis/genética , Muerte Celular/genética , Expresión Génica , Hepatocitos/patología , Humanos , Hepatopatías/etiología , Hepatopatías/metabolismo , Ratones
9.
Sci Signal ; 12(571)2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30837304

RESUMEN

Mutations in RAS signaling pathway components cause diverse neurodevelopmental disorders, collectively called RASopathies. Previous studies have suggested that dysregulation in RAS-extracellular signal-regulated kinase (ERK) activation is restricted to distinct cell types in different RASopathies. Some cases of Noonan syndrome (NS) are associated with gain-of-function mutations in the phosphatase SHP2 (encoded by PTPN11); however, SHP2 is abundant in multiple cell types, so it is unclear which cell type(s) contribute to NS phenotypes. Here, we found that expressing the NS-associated mutant SHP2D61G in excitatory, but not inhibitory, hippocampal neurons increased ERK signaling and impaired both long-term potentiation (LTP) and spatial memory in mice, although endogenous SHP2 was expressed in both neuronal types. Transcriptomic analyses revealed that the genes encoding SHP2-interacting proteins that are critical for ERK activation, such as GAB1 and GRB2, were enriched in excitatory neurons. Accordingly, expressing a dominant-negative mutant of GAB1, which reduced its interaction with SHP2D61G, selectively in excitatory neurons, reversed SHP2D61G-mediated deficits. Moreover, ectopic expression of GAB1 and GRB2 together with SHP2D61G in inhibitory neurons resulted in ERK activation. These results demonstrate that RAS-ERK signaling networks are notably different between excitatory and inhibitory neurons, accounting for the cell type-specific pathophysiology of NS and perhaps other RASopathies.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipocampo/metabolismo , Potenciación a Largo Plazo , Sistema de Señalización de MAP Quinasas , Memoria , Mutación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Animales , Quinasas MAP Reguladas por Señal Extracelular/genética , Ratones , Ratones Transgénicos , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética
10.
Biol Psychiatry ; 85(7): 534-543, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30466882

RESUMEN

BACKGROUND: Autism spectrum disorder involves neurodevelopmental dysregulations that lead to visible symptoms at early stages of life. Many autism spectrum disorder-related mechanisms suggested by animal studies are supported by demonstrated improvement in autistic-like phenotypes in adult animals following experimental reversal of dysregulated mechanisms. However, whether such mechanisms also act at earlier stages to cause autistic-like phenotypes is unclear. METHODS: We used Shank2-/- mice carrying a mutation identified in human autism spectrum disorder (exons 6 and 7 deletion) and combined electrophysiological and behavioral analyses to see whether early pathophysiology at pup stages is different from late pathophysiology at juvenile and adult stages and whether correcting early pathophysiology can normalize late pathophysiology and abnormal behaviors in juvenile and adult mice. RESULTS: Early correction of a dysregulated mechanism in young mice prevents manifestation of autistic-like social behaviors in adult mice. Shank2-/- mice, known to display N-methyl-D-aspartate receptor (NMDAR) hypofunction and autistic-like behaviors at postweaning stages after postnatal day 21 (P21), show the opposite synaptic phenotype-NMDAR hyperfunction-at an earlier preweaning stage (∼P14). Moreover, this NMDAR hyperfunction at P14 rapidly shifts to NMDAR hypofunction after weaning (∼P24). Chronic suppression of the early NMDAR hyperfunction by the NMDAR antagonist memantine (P7-P21) prevents NMDAR hypofunction and autistic-like social behaviors from manifesting at later stages (∼P28 and P56). CONCLUSIONS: Early NMDAR hyperfunction leads to late NMDAR hypofunction and autistic-like social behaviors in Shank2-/- mice, and early correction of NMDAR dysfunction has the long-lasting effect of preventing autistic-like social behaviors from developing at later stages.


Asunto(s)
Trastorno del Espectro Autista/tratamiento farmacológico , Trastorno del Espectro Autista/fisiopatología , Conducta Animal/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Memantina/farmacología , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Conducta Social , Factores de Edad , Animales , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/fisiología
11.
J Neurochem ; 147(5): 595-608, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30125942

RESUMEN

Guanine nucleotide exchange factors (GEFs) play important roles in many cellular processes, including regulation of the structural plasticity of dendritic spines. A GEF protein, adenomatous polyposis coli-stimulated GEF 1 (Asef1, ARHGEF4) is highly expressed in the nervous system. However, the function of Asef1 has not been investigated in neurons. Here, we present evidence showing that Asef1 negatively regulates the synaptic localization of postsynaptic density protein 95 (PSD-95) in the excitatory synapse by inhibiting Staufen-mediated synaptic localization of PSD-95. Accordingly, Asef1 expression impairs synaptic transmission in hippocampal cultured neurons. In addition, neuronal activity facilitates the dissociation of Asef1 from Staufen in a phosphoinositide 3 kinase (PI3K)-dependent manner. Taken together, our data reveal Asef1 functions as a negative regulator of synaptic localization of PSD-95 and synaptic transmission.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Complejos de Clasificación Endosomal Requeridos para el Transporte/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Fosfoproteínas/fisiología , Sinapsis/fisiología , Adenosina Trifosfatasas/genética , Animales , Dendritas/fisiología , Dendritas/ultraestructura , Homólogo 4 de la Proteína Discs Large/biosíntesis , Homólogo 4 de la Proteína Discs Large/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Hipocampo/citología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Técnicas de Placa-Clamp , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología , Ratas , Transmisión Sináptica/fisiología
12.
eNeuro ; 5(3)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30023428

RESUMEN

Autism spectrum disorders (ASDs) are neurodevelopmental disorders with a strong genetic etiology. Since mutations in human SHANK genes have been found in patients with autism, genetic mouse models are used for a mechanistic understanding of ASDs and the development of therapeutic strategies. SHANKs are scaffold proteins in the postsynaptic density of mammalian excitatory synapses with proposed functions in synaptogenesis, regulation of dendritic spine morphology, and instruction of structural synaptic plasticity. In contrast to all studies so far on the function of SHANK proteins, we have previously observed enhanced synaptic plasticity in Shank2 Δex7-/- mice. In a series of experiments, we now reproduce these results, further explore the synaptic phenotype, and directly compare our model to the independently generated Shank2 Δex6-7-/- mice. Minimal stimulation experiments reveal that Shank2 Δex7-/- mice possess an excessive fraction of silent (i.e., α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, short, AMPA receptor lacking) synapses. The synaptic maturation deficit emerges during the third postnatal week and constitutes a plausible mechanistic explanation for the mutants' increased capacity for long-term potentiation, both in vivo and in vitro. A direct comparison with Shank2 Δex6-7-/- mice adds weight to the hypothesis that both mouse models show a different set of synaptic phenotypes, possibly due to differences in their genetic background. These findings add to the diversity of synaptic phenotypes in neurodevelopmental disorders and further support the supposed existence of "modifier genes" in the expression and inheritance of ASDs.


Asunto(s)
Trastorno del Espectro Autista/fisiopatología , Potenciación a Largo Plazo , Proteínas del Tejido Nervioso/fisiología , Sinapsis/fisiología , Animales , Trastorno del Espectro Autista/genética , Modelos Animales de Enfermedad , Hipocampo/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Receptores AMPA/fisiología
13.
Mol Pain ; 14: 1744806918783943, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29923456

RESUMEN

Protein kinase M ζ is well known for its role in maintaining memory and pain. Previously, we revealed that the activation of protein kinase M ζ in the anterior cingulate cortex plays a role in sustaining neuropathic pain. However, the mechanism by which protein kinase M ζ is expressed in the anterior cingulate cortex by peripheral nerve injury, and whether blocking of protein kinase M ζ using its inhibitor, zeta inhibitory peptide, produces analgesic effects in neuropathic pain maintained chronically after injury, have not previously been resolved. In this study, we show that protein kinase M ζ expression in the anterior cingulate cortex is enhanced by peripheral nerve injury in a transcription-independent manner. We also reveal that the inhibition of protein kinase M ζ through zeta inhibitory peptide treatment is enough to reduce mechanical allodynia responses in mice with one-month-old nerve injuries. However, the zeta inhibitory peptide treatment was only effective for a limited time.


Asunto(s)
Dolor Crónico/enzimología , Dolor Crónico/genética , Giro del Cíngulo/enzimología , Neuralgia/enzimología , Neuralgia/genética , Proteína Quinasa C/metabolismo , Transcripción Genética , Animales , Péptidos de Penetración Celular , Dolor Crónico/patología , Giro del Cíngulo/patología , Lipopéptidos/farmacología , Potenciación a Largo Plazo , Masculino , Ratones Endogámicos C57BL , Neuralgia/patología , Nervios Periféricos/patología , Receptores AMPA , Sinapsis/metabolismo , Transcripción Genética/efectos de los fármacos
14.
Brain Res Bull ; 141: 50-57, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29680772

RESUMEN

Memory reconsolidation is the process by which previously consolidated memories reenter a labile state through reactivation of the memory trace and are actively consolidated through de novo protein synthesis. Although extensive studies have shown that ß-adrenergic signaling plays a critical role in the restabilization of reactivated memory, its role in the destabilization of long-term memory is not well-studied. In this study, we found that membrane excitability increased in hippocampal CA1 neurons immediately after the retrieval of contextual fear memory. Interestingly, this increase in membrane excitability diminished after treatment with propranolol (a ß-adrenergic receptor antagonist), an NMDA receptor antagonist, and a PKA inhibitor. In addition, we found that administration of propranolol prior to, but not after, the retrieval of fear memory ameliorated the memory impairment caused by anisomycin, indicating that inhibition of ß-adrenergic signaling blocks the destabilization of contextual fear memory. Taken together, these results indicate that ß-adrenergic signaling via NMDA receptors and PKA signaling pathway induces a labile state of long-term memory through increased neuronal membrane excitability.


Asunto(s)
Región CA1 Hipocampal/fisiología , Potenciales de la Membrana/fisiología , Consolidación de la Memoria/fisiología , Neuronas/fisiología , Receptores Adrenérgicos beta/metabolismo , Animales , Región CA1 Hipocampal/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Miedo/efectos de los fármacos , Miedo/fisiología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Consolidación de la Memoria/efectos de los fármacos , Recuerdo Mental/efectos de los fármacos , Recuerdo Mental/fisiología , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neurotransmisores/farmacología , Propranolol/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/efectos de los fármacos , Técnicas de Cultivo de Tejidos
15.
Science ; 360(6387): 430-435, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29700265

RESUMEN

Memory resides in engram cells distributed across the brain. However, the site-specific substrate within these engram cells remains theoretical, even though it is generally accepted that synaptic plasticity encodes memories. We developed the dual-eGRASP (green fluorescent protein reconstitution across synaptic partners) technique to examine synapses between engram cells to identify the specific neuronal site for memory storage. We found an increased number and size of spines on CA1 engram cells receiving input from CA3 engram cells. In contextual fear conditioning, this enhanced connectivity between engram cells encoded memory strength. CA3 engram to CA1 engram projections strongly occluded long-term potentiation. These results indicate that enhanced structural and functional connectivity between engram cells across two directly connected brain regions forms the synaptic correlate for memory formation.


Asunto(s)
Región CA1 Hipocampal/fisiología , Región CA3 Hipocampal/fisiología , Memoria/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Animales , Región CA1 Hipocampal/citología , Región CA3 Hipocampal/citología , Condicionamiento Clásico , Miedo , Proteínas Fluorescentes Verdes/análisis , Células HEK293 , Humanos , Potenciación a Largo Plazo , Masculino , Ratones Endogámicos C57BL , Neuroimagen/métodos , Plasticidad Neuronal
16.
Cell Rep ; 22(3): 748-759, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29346771

RESUMEN

Peripheral nerve injury can induce pathological conditions that lead to persistent sensitized nociception. Although there is evidence that plastic changes in the cortex contribute to this process, the underlying molecular mechanisms are unclear. Here, we find that activation of the anterior cingulate cortex (ACC) induced by peripheral nerve injury increases the turnover of specific synaptic proteins in a persistent manner. We demonstrate that neural cell adhesion molecule 1 (NCAM1) is one of the molecules involved and show that it mediates spine reorganization and contributes to the behavioral sensitization. We show striking parallels in the underlying mechanism with the maintenance of NMDA-receptor- and protein-synthesis-dependent long-term potentiation (LTP) in the ACC. Our results, therefore, demonstrate a synaptic mechanism for cortical reorganization and suggest potential avenues for neuropathic pain treatment.


Asunto(s)
Antígeno CD56/metabolismo , Giro del Cíngulo/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Animales , Giro del Cíngulo/patología , Masculino , Ratones , Traumatismos de los Nervios Periféricos/patología , Sinapsis/patología
17.
Small ; 13(40)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28809097

RESUMEN

Intermolecular interactions dominate the behavior of signal transduction in various physiological and pathological cell processes, yet assessing these interactions remains a challenging task. Here, this study reports a single-molecule force spectroscopic method that enables functional delineation of two interaction sites (≈35 pN and ≈90 pN) between signaling effectors Ras and BRaf in the canonical mitogen-activated protein kinase (MAPK) pathway. This analysis reveals mutations on BRaf at Q257 and A246, two sites frequently linked to cardio-faciocutaneous syndrome, result in ≈10-30 pN alterations in RasBRaf intermolecular binding force. The magnitude of changes in RasBRaf binding force correlates with the size of alterations in protein affinity and in α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-sensitive glutamate receptor (-R)-mediated synaptic transmission in neurons expressing replacement BRaf mutants, and predicts the extent of learning impairments in animals expressing replacement BRaf mutants. These results establish single-molecule force spectroscopy as an effective platform for evaluating the piconewton-level interaction of signaling molecules and predicting the behavior outcome of signal transduction.


Asunto(s)
Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo , Animales , Células Cultivadas , Humanos , Trastornos Mentales/genética , Trastornos Mentales/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutación , Pinzas Ópticas , Transducción de Señal/genética , Transducción de Señal/fisiología
18.
Sci Rep ; 7(1): 4912, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28687800

RESUMEN

Lysine-specific demethylase 1 (LSD1) is a histone demethylase that participates in transcriptional repression or activation. Recent studies reported that LSD1 is involved in learning and memory. Although LSD1 phosphorylation by PKCα was implicated in circadian rhythmicity, the importance of LSD1 phosphorylation in learning and memory is unknown. In this study, we examined the roles of LSD1 in synaptic plasticity and memory using Lsd1 SA/SA knock-in (KI) mice, in which a PKCα phosphorylation site is mutated. Interestingly, short-term and long-term contextual fear memory as well as spatial memory were impaired in Lsd1 KI mice. In addition, short-term synaptic plasticity, such as paired pulse ratio and post-tetanic potentiation was impaired, whereas long-term synaptic plasticity, including long-term potentiation and long-term depression, was normal. Moreover, the frequency of miniature excitatory postsynaptic current was significantly increased, suggesting presynaptic dysfunction in Lsd1 KI mice. Consistent with this, RNA-seq analysis using the hippocampus of Lsd1 KI mice showed significant alterations in the expressions of presynaptic function-related genes. Intriguingly, LSD1n-SA mutant showed diminished binding to histone deacetylase 1 (HDAC1) compared to LSD1n-WT in SH-SY5Y cells. These results suggest that LSD1 is involved in the regulation of presynaptic gene expression and subsequently regulates the hippocampus-dependent memory in phosphorylation-dependent manner.


Asunto(s)
Hipocampo/metabolismo , Histona Demetilasas/genética , Memoria a Largo Plazo/fisiología , Memoria a Corto Plazo/fisiología , Proteína Quinasa C-alfa/genética , Animales , Animales Modificados Genéticamente , Línea Celular Tumoral , Miedo/fisiología , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Hipocampo/fisiopatología , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histona Demetilasas/metabolismo , Humanos , Aprendizaje/fisiología , Potenciación a Largo Plazo/fisiología , Depresión Sináptica a Largo Plazo/fisiología , Masculino , Ratones , Mutagénesis Sitio-Dirigida , Mutación , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Unión Proteica , Proteína Quinasa C-alfa/metabolismo , Transducción de Señal
20.
Genes Dev ; 31(6): 537-552, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28404629

RESUMEN

Rapid advances in genetics are linking mutations on genes to diseases at an exponential rate, yet characterizing the gene-mutation-cell-behavior relationships essential for precision medicine remains a daunting task. More than 350 mutations on small GTPase BRaf are associated with various tumors, and ∼40 mutations are associated with the neurodevelopmental disorder cardio-facio-cutaneous syndrome (CFC). We developed a fast cost-effective lentivirus-based rapid gene replacement method to interrogate the physiopathology of BRaf and ∼50 disease-linked BRaf mutants, including all CFC-linked mutants. Analysis of simultaneous multiple patch-clamp recordings from 6068 pairs of rat neurons with validation in additional mouse and human neurons and multiple learning tests from 1486 rats identified BRaf as the key missing signaling effector in the common synaptic NMDA-R-CaMKII-SynGap-Ras-BRaf-MEK-ERK transduction cascade. Moreover, the analysis creates the original big data unveiling three general features of BRaf signaling. This study establishes the first efficient procedure that permits large-scale functional analysis of human disease-linked mutations essential for precision medicine.


Asunto(s)
Sistema de Señalización de MAP Quinasas/genética , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Transmisión Sináptica/genética , Animales , Células Cultivadas , Enfermedad/genética , Femenino , Técnicas de Transferencia de Gen , Humanos , Lentivirus/genética , Masculino , Ratones Endogámicos C57BL , Neuronas/fisiología , Ratas Sprague-Dawley , Técnicas de Cultivo de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...