Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Acta Neuropathol Commun ; 11(1): 199, 2023 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-38105257

RESUMO

The hypomethylation of fused in sarcoma (FUS) in frontotemporal lobar degeneration promotes the formation of irreversible condensates of FUS. However, the mechanisms by which these hypomethylated FUS condensates cause neuronal dysfunction are unknown. Here we report that expression of FUS constructs mimicking hypomethylated FUS causes aberrant dendritic FUS condensates in CA1 neurons. These hypomethylated FUS condensates exhibit spontaneous, and activity induced movement within the dendrite. They impair excitatory synaptic transmission, postsynaptic density-95 expression, and dendritic spine plasticity. These neurophysiological defects are dependent upon both the dendritic localisation of the condensates, and their ability to undergo liquid-liquid phase separation. These results indicate that the irreversible liquid-liquid phase separation is a key component of hypomethylated FUS pathophysiology in sporadic FTLD, and this can cause synapse dysfunction in sporadic FTLD.


Assuntos
Demência Frontotemporal , Degeneração Lobar Frontotemporal , Humanos , Separação de Fases , Proteína FUS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/metabolismo , Degeneração Lobar Frontotemporal/genética , Metilação de DNA
2.
Brain Commun ; 5(2): fcad114, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37124945

RESUMO

This scientific commentary refers to 'Human stem cell-derived astrocytes exhibit region-specific heterogeneity in their secretory profiles', by Clarke et al. (https://doi.org/10.1093/brain/awaa258) in Brain.

3.
Brain Stimul ; 16(2): 540-552, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36731773

RESUMO

BACKGROUND: Focused ultrasound stimulation (FUS) has the potential to provide non-invasive neuromodulation of deep brain regions with unparalleled spatial precision. However, the cellular and molecular consequences of ultrasound stimulation on neurons remains poorly understood. We previously reported that ultrasound stimulation induces increases in neuronal excitability that persist for hours following stimulation in vitro. In the present study we sought to further elucidate the molecular mechanisms by which ultrasound regulates neuronal excitability and synaptic function. OBJECTIVES: To determine the effect of ultrasound stimulation on voltage-gated ion channel function and synaptic plasticity. METHODS: Primary rat cortical neurons were exposed to a 40 s, 200 kHz pulsed ultrasound stimulus or sham-stimulus. Whole-cell patch clamp electrophysiology, quantitative proteomics and high-resolution confocal microscopy were employed to determine the effects of ultrasound stimulation on molecular regulators of neuronal excitability and synaptic function. RESULTS: We find that ultrasound exposure elicits sustained but reversible increases in whole-cell potassium currents. In addition, we find that ultrasound exposure activates synaptic signalling cascades that result in marked increases in excitatory synaptic transmission. Finally, we demonstrate the requirement of ionotropic glutamate receptor (AMPAR/NMDAR) activation for ultrasound-induced modulation of neuronal potassium currents. CONCLUSION: These results suggest specific patterns of pulsed ultrasound can induce contemporaneous enhancement of both neuronal excitability and synaptic function, with implications for the application of FUS in experimental and therapeutic settings. Further study is now required to deduce the precise molecular mechanisms through which these changes occur.


Assuntos
Potássio , Receptores Ionotrópicos de Glutamato , Ratos , Animais , Potássio/metabolismo , Potássio/farmacologia , Ratos Sprague-Dawley , Neurônios/fisiologia , Transmissão Sináptica/fisiologia , Plasticidade Neuronal
4.
Brain Sci ; 12(2)2022 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-35204052

RESUMO

Non-invasive focused ultrasound stimulation (FUS) is a non-ionising neuromodulatory technique that employs acoustic energy to acutely and reversibly modulate brain activity of deep-brain structures. It is currently being investigated as a potential novel treatment for Parkinson's disease (PD). This scoping review was carried out to map available evidence pertaining to the provision of FUS as a PD neuromodulatory tool. In accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Extension for Scoping Reviews, a search was applied to Ovid MEDLINE, Embase, Web of Science and Cochrane Central Register of Controlled Trials on 13 January 2022, with no limits applied. In total, 11 studies were included: 8 were from China and 1 each from Belgium, South Korea and Taiwan. All 11 studies were preclinical (6 in vivo, 2 in vitro, 2 mix of in vivo and in vitro and 1 in silico). The preclinical evidence indicates that FUS is safe and has beneficial neuromodulatory effects on motor behaviour in PD. FUS appears to have a therapeutic role in influencing the disease processes of PD, and therefore holds great promise as an attractive and powerful neuromodulatory tool for PD. Though these initial studies are encouraging, further study to understand the underlying cellular and molecular mechanisms is required before FUS can be routinely used in PD.

5.
Nat Commun ; 13(1): 27, 2022 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-35031607

RESUMO

Coordinated programs of gene expression drive brain development. It is unclear which transcriptional programs, in which cell-types, are affected in neuropsychiatric disorders such as schizophrenia. Here we integrate human genetics with transcriptomic data from differentiation of human embryonic stem cells into cortical excitatory neurons. We identify transcriptional programs expressed during early neurogenesis in vitro and in human foetal cortex that are down-regulated in DLG2-/- lines. Down-regulation impacted neuronal differentiation and maturation, impairing migration, morphology and action potential generation. Genetic variation in these programs is associated with neuropsychiatric disorders and cognitive function, with associated variants predominantly concentrated in loss-of-function intolerant genes. Neurogenic programs also overlap schizophrenia GWAS enrichment previously identified in mature excitatory neurons, suggesting that pathways active during prenatal cortical development may also be associated with mature neuronal dysfunction. Our data from human embryonic stem cells, when combined with analysis of available foetal cortical gene expression data, de novo rare variants and GWAS statistics for neuropsychiatric disorders and cognition, reveal a convergence on transcriptional programs regulating excitatory cortical neurogenesis.


Assuntos
Córtex Cerebral/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Guanilato Quinases/genética , Neurogênese , Proteínas Supressoras de Tumor/genética , Animais , Diferenciação Celular , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Predisposição Genética para Doença , Guanilato Quinases/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Transtornos Mentais/genética , Neurogênese/genética , Neurogênese/fisiologia , Neurônios , Gravidez , Esquizofrenia/genética , Transcriptoma , Proteínas Supressoras de Tumor/metabolismo
6.
Brain Stimul ; 14(2): 217-225, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33444809

RESUMO

BACKGROUND: Transcranial ultrasound stimulation can acutely modulate brain activity, but the lasting effects on neurons are unknown. OBJECTIVE: To assess the excitability profile of neurons in the hours following transient ultrasound stimulation. METHODS: Primary rat cortical neurons were stimulated with a 40 s, 200 kHz pulsed ultrasound stimulation or sham-stimulation. Intrinsic firing properties were investigated through whole-cell patch-clamp recording by evoking action potentials in response to somatic current injection. Recordings were taken at set timepoints following ultrasound stimulation: 0-2 h, 6-8 h, 12-14 h and 24-26 h. Transmission electron microscopy was used to assess synaptic ultrastructure at the same timepoints. RESULTS: In the 0-2 h window, neurons stimulated with ultrasound displayed an increase in the mean frequency of evoked action potentials of 32% above control cell levels (p = 0.023). After 4-6 h this increase was measured as 44% (p = 0.0043). By 12-14 h this effect was eliminated and remained absent 24-26 h post-stimulation. These changes to action potential firing occurred in conjunction with statistically significant differences between control and ultrasound-stimulated neurons in action potential half-width, depolarisation rate, and repolarisation rate, that were similarly eliminated by 24 h following stimulation. These effects occurred in the absence of alterations to intrinsic membrane properties or synaptic ultrastructure. CONCLUSION: We report that stimulating neurons with 40 s of ultrasound enhances their excitability for up to 8 h in conjunction with modifications to action potential kinetics. This occurs in the absence of major ultrastructural change or modification of intrinsic membrane properties. These results can inform the application of transcranial ultrasound in experimental and therapeutic settings.


Assuntos
Axônios , Neurônios , Potenciais de Ação , Animais , Estimulação Elétrica , Masculino , Técnicas de Patch-Clamp , Ratos , Ratos Wistar
7.
Brain Commun ; 2(2): fcaa058, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32766549

RESUMO

Aggregation of amyloid beta and loss of cholinergic innervation in the brain are predominant components of Alzheimer's disease pathology and likely underlie cognitive impairment. Acetylcholinesterase inhibitors are one of the few treatment options for Alzheimer's disease, where levels of available acetylcholine are enhanced to counteract the cholinergic loss. However, these inhibitors show limited clinical efficacy. One potential explanation for this is a concomitant dysregulation of cholinergic receptors themselves as a consequence of the amyloid beta pathology. We tested this hypothesis by examining levels of M1 muscarinic acetylcholine receptors in the temporal cortex from seven Alzheimer's disease and seven non-disease age-matched control brain tissue samples (control: 85 ± 2.63 years old, moderate Alzheimer's disease: 84 ± 2.32 years old, P-value = 0.721; eight female and six male patients). The samples were categorized into two groups: 'control' (Consortium to Establish a Registry for Alzheimer's Disease diagnosis of 'No Alzheimer's disease', and Braak staging pathology of I-II) and 'moderate Alzheimer's disease' (Consortium to Establish a Registry for Alzheimer's Disease diagnosis of 'possible/probable Alzheimer's disease', and Braak staging pathology of IV). We find that in comparison to age-matched controls, there is a loss of M1 muscarinic acetylcholine receptors in moderate Alzheimer's disease tissue (control: 2.17 ± 0.27 arbitrary units, n = 7, Mod-AD: 0.83 ± 0.16 arbitrary units, n = 7, two-tailed t-test, t = 4.248, P = 0.00113). Using a functional rat cortical brain slice model, we find that postsynaptic muscarinic acetylcholine receptor function is dysregulated by aberrant amyloid beta-mediated activation of metabotropic glutamate receptor 5. Crucially, blocking metabotropic glutamate receptor 5 restores muscarinic acetylcholine receptor function and object recognition memory in 5XFAD transgenic mice. This indicates that the amyloid beta-mediated activation of metabotropic glutamate receptor 5 negatively regulates muscarinic acetylcholine receptor and illustrates the importance of muscarinic acetylcholine receptors as a potential disease-modifying target in the moderate pathological stages of Alzheimer's disease.

8.
J Am Soc Nephrol ; 30(10): 1910-1924, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31416818

RESUMO

BACKGROUND: Mutations in the transient receptor potential channel 6 (TRPC6) gene are associated with an inherited form of FSGS. Despite widespread expression, patients with TRPC6 mutations do not present with any other pathologic phenotype, suggesting that this protein has a unique yet unidentified role within the target cell for FSGS, the kidney podocyte. METHODS: We generated a stable TRPC6 knockout podocyte cell line from TRPC6 knockout mice. These cells were engineered to express wild-type TRPC6, a dominant negative TRPC6 mutation, or either of two disease-causing mutations of TRPC6, G109S or K874*. We extensively characterized these cells using motility, detachment, and calpain activity assays; immunofluorescence; confocal or total internal reflection fluorescence microscopy; and western blotting. RESULTS: Compared with wild-type cells, TRPC6-/- podocytes are less motile and more adhesive, with an altered actin cytoskeleton. We found that TRPC6 binds to ERK1/2 and the actin regulatory proteins, caldesmon (a calmodulin- and actin-binding protein) and calpain 1 and 2 (calcium-dependent cysteine proteases that control the podocyte cytoskeleton, cell adhesion, and motility via cleavage of paxillin, focal adhesion kinase, and talin). Knockdown or expression of the truncated K874* mutation (but not expression of the gain-of-function G019S mutation or dominant negative mutant of TRPC6) results in the mislocalization of calpain 1 and 2 and significant downregulation of calpain activity; this leads to altered podocyte cytoskeleton, motility, and adhesion-characteristics of TRPC6-/- podocytes. CONCLUSIONS: Our data demonstrate that independent of TRPC6 channel activity, the physical interaction between TRPC6 and calpain in the podocyte is important for cell motility and detachment and demonstrates a scaffolding role of the TRPC6 protein in disease.


Assuntos
Calpaína/fisiologia , Adesão Celular , Movimento Celular , Citoesqueleto/fisiologia , Podócitos/fisiologia , Podócitos/ultraestrutura , Canal de Cátion TRPC6/fisiologia , Animais , Camundongos , Camundongos Knockout
9.
Cell Discov ; 4: 44, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30181899

RESUMO

It is well documented that reactive oxygen species (ROS) affects neurodegeneration in the brain. Several studies also implicate ROS in the regulation of synapse function and learning and memory processes, although the precise source of ROS generation within these contexts remains to be further explored. Here we show that postsynaptic superoxide generation through PKCζ-activated NADPH oxidase 2 (NOX2) is critical for long-term depression (LTD) of synaptic transmission in the CA1-Shaffer collateral synapse of the rat hippocampus. Specifically, PKCζ-dependent phosphorylation of p47phox at serine 316, a NOX2 regulatory subunit, is required for LTD but is not necessary for long-term potentiation (LTP). Our data suggest that postsynaptic p47phox phosphorylation at serine 316 is a key upstream determinant for LTD and synapse weakening.

10.
Mol Brain ; 10(1): 59, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29221469

RESUMO

CORRECTION TO: MOLECULAR BRAIN (2017) 10:35 DOI: 10.1186/S13041-017-0315-X: In the original version of this article [1], published on 1 August 2017, Fig. 3 contains a typo. In this Correction the incorrect and correct version of Fig. 3 are shown. - Fig. 3 was originally published like this: - The correct version of Fig. 3 looks like this.

11.
Cell Death Dis ; 8(10): e3102, 2017 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-29022894

RESUMO

Alzheimer's disease (AD) is the most common neurodegenerative disease, characterized by excessive beta amyloid (Aß) deposition in brain, leading to blood-brain barrier (BBB) disruption. The mechanisms of BBB disruption in AD are still unclear, despite considerable research. The adipokine adiponectin is known to regulate various metabolic functions and reduce inflammation. Though adiponectin receptors have been reported in the brain, its role in the central nervous system has not been fully characterized. In the present study, we investigate whether adiponectin contributes to the tight junction integrity and cell death of brain endothelial cells under Aß-induced toxicity conditions. We measured the expression of adiponectin receptors (AdipoR1 and AdipoR2) and the alteration of tight junction proteins in in vivo 5xFAD mouse brain. Moreover, we examined the production of reactive oxygen species (ROS) and the loss of tight junction proteins such as Claudin 5, ZO-1, and inflammatory signaling in in vitro brain endothelial cells (bEnd.3 cells) under Aß toxicity. Our results showed that Acrp30 (a globular form of adiponectin) reduces the expression of proinflammatory cytokines and the expression of RAGE as Aß transporters into brain. Moreover, we found that Acrp 30 attenuated the apoptosis and the tight junction disruption through AdipoR1-mediated NF-κB pathway in Aß-exposed bEnd.3 cells. Thus, we suggest that adiponectin is an attractive therapeutic target for treating BBB breakdown in AD brain.


Assuntos
Adiponectina/metabolismo , Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/metabolismo , Claudina-5/metabolismo , Receptores de Adiponectina/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo , Doença de Alzheimer/patologia , Animais , Apoptose/fisiologia , Encéfalo/citologia , Encéfalo/metabolismo , Linhagem Celular , Sobrevivência Celular/fisiologia , Células Endoteliais/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , Óxido Nítrico/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Receptor para Produtos Finais de Glicação Avançada/biossíntese , Receptores de LDL/biossíntese , Junções Íntimas/metabolismo , Proteínas Supressoras de Tumor/biossíntese
12.
Mol Brain ; 10(1): 35, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28764741

RESUMO

Alzheimer's disease (AD) is defined by the excessive accumulation of toxic peptides, such as beta amyloid (Aß) plaques and intracellular neurofibrillary tangles (NFT). The risk factors associated with AD include genetic mutations, aging, insulin resistance, and oxidative stress. To date, several studies that have demonstrated an association between AD and diabetes have revealed that the common risk factors include insulin resistance, sleep disturbances, blood brain barrier (BBB) disruption, and altered glucose homeostasis. Many researchers have discovered that there are mechanisms common to both diabetes and AD. AD that results from insulin resistance in the brain is termed "type 3 diabetes". Melatonin synthesized by the pineal gland is known to contribute to circadian rhythms, insulin resistance, protection of the BBB, and cell survival mechanisms. Here, we review the relationship between melatonin and type 3 diabetes, and suggest that melatonin might regulate the risk factors for type 3 diabetes. We suggest that melatonin is crucial for attenuating the onset of type 3 diabetes by intervening in Aß accumulation, insulin resistance, glucose metabolism, and BBB permeability.


Assuntos
Diabetes Mellitus/patologia , Progressão da Doença , Melatonina/metabolismo , Animais , Barreira Hematoencefálica/patologia , Humanos , Resistência à Insulina , Fatores de Risco
13.
Pharmacol Res ; 121: 42-51, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28416463

RESUMO

Evidence suggests that the stress hormones glucocorticoids (GCs) can cause cognitive deficits and neurodegeneration. Previous studies have found GCs facilitate physiological synapse weakening, termed long-term depression (LTD), though the precise mechanisms underlying this are poorly understood. Here we show that GCs activate glycogen synthase kinase-3 (GSK-3), a kinase crucial to synapse weakening signals. Critically, this ultimately leads to phosphorylation of the microtubule associated protein tau, specifically at the serine 396 residue, and this is a causal factor in the GC-mediated impairment of synaptic function. These findings reveal the link between GCs and synapse weakening signals, and the potential for stress-induced priming of neurodegeneration. This could have important implications for our understanding of how stress can lead to neurodegenerative disease.


Assuntos
Glucocorticoides/metabolismo , Hipocampo/fisiologia , Potenciação de Longa Duração , Sinapses/fisiologia , Proteínas tau/metabolismo , Animais , Quinase 3 da Glicogênio Sintase/metabolismo , Fosforilação , Ratos , Transdução de Sinais
14.
Neuroscientist ; 23(2): 137-151, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-26908138

RESUMO

Tauopathies encompass a broad range of neurodegenerative diseases featuring extensive neuronal death and cognitive decline. However, research over the past 30 years has failed to significantly advance our understanding of how tau causes dementia, limiting the design of rational therapeutics. It has become evident that we need to expand our understanding of tau in physiology, in order to delineate how tau may contribute to pathology. This review discusses recent evidence that has uncovered a novel aspect of tau function, based on its previously uncharacterized localization to the synapse. Here, multiple streams of evidence support a critical role for synaptic tau in the regulation of synapse physiology. In particular, long-term depression, a form of synaptic weakening, is dependent on the presence of tau in hippocampal neurons. The regulation of tau by specific phosphorylation events downstream of GSK-3ß activation appears to be integral to this signaling role. We also describe how the regulation of synapse physiology by tau and its phosphorylation may inform our understanding of tauopathies and comorbid diseases. This work should provide a platform for future tau biology research in addition to therapeutic design.


Assuntos
Demência/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Doenças Neurodegenerativas/metabolismo , Transdução de Sinais/fisiologia , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Humanos
15.
Neuropharmacology ; 112(Pt A): 221-227, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27561971

RESUMO

α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) are the primary conduits of excitatory synaptic transmission. AMPARs are predominantly Ca2+-impermeable in the matured excitatory synapse, except under certain circumstances. Growing evidence implicates the Ca2+ permeability of AMPARs in the regulation of long-term synaptic plasticity and in the pathophysiology of several neurological disorders. Therefore, the Ca2+ conductance of AMPARs may have both physiological and pathological roles at synapses. However, our understanding of the role of Ca2+ permeable AMPARs (CP-AMPARs) in Alzheimer's disease is limited. Here we discuss insights into the potential CP-AMPAR mediated pathophysiology of Alzheimer's disease, including: 1. Ca2+-mediated aberrant regulation of synapse weakening mechanisms, and 2. neuronal network dysfunction in the brain. Consideration of CP-AMPARs as primary drivers of pathophysiology could help in understanding synaptopathologies, and highlights the potential of CP-AMPARs as therapeutic targets in Alzheimer's disease. This article is part of the Special Issue entitled 'Ionotropic glutamate receptors'.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Cálcio/metabolismo , Receptores de AMPA/metabolismo , Sinapses/metabolismo , Peptídeos beta-Amiloides/administração & dosagem , Animais , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Plasticidade Neuronal , Neurônios/metabolismo , Neurônios/fisiologia , Receptores de AMPA/fisiologia , Sinapses/fisiologia , Transmissão Sináptica
16.
Sci Rep ; 6: 29152, 2016 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-27377368

RESUMO

Mouse models of Alzheimer's disease (AD) have been developed to study the pathophysiology of amyloid ß protein (Aß) toxicity, which is thought to cause severe clinical symptoms such as memory impairment in AD patients. However, inconsistencies exist between studies using these animal models, specifically in terms of the effects on synaptic plasticity, a major cellular model of learning and memory. Whereas some studies find impairments in plasticity in these models, others do not. We show that long-term potentiation (LTP), in the CA1 region of hippocampal slices from this mouse, is impared at Tg2576 adult 6-7 months old. However, LTP is inducible again in slices taken from Tg2576 aged 14-19 months old. In the aged Tg2576, we found that the percentage of parvalbumin (PV)-expressing interneurons in hippocampal CA1-3 region is significantly decreased, and LTP inhibition or reversal mediated by NRG1/ErbB signaling, which requires ErbB4 receptors in PV interneurons, is impaired. Inhibition of ErbB receptor kinase in adult Tg2576 restores LTP but impairs depotentiation as shown in aged Tg2576. Our study suggests that hippocampal LTP reemerges in aged Tg2576. However, this reemerged LTP is an insuppressible form due to impaired NRG1/ErbB signaling, possibly through the loss of PV interneurons.


Assuntos
Envelhecimento/patologia , Doença de Alzheimer/fisiopatologia , Potenciação de Longa Duração/fisiologia , Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Receptores ErbB/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Interneurônios/metabolismo , Masculino , Transtornos da Memória/complicações , Transtornos da Memória/fisiopatologia , Camundongos Transgênicos , Neuregulina-1/metabolismo , Plasticidade Neuronal , Parvalbuminas/metabolismo , Reconhecimento Psicológico
17.
Sci Rep ; 6: 26676, 2016 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-27225731

RESUMO

Synaptogenic adhesion molecules play critical roles in synapse formation. SALM5/Lrfn5, a SALM/Lrfn family adhesion molecule implicated in autism spectrum disorders (ASDs) and schizophrenia, induces presynaptic differentiation in contacting axons, but its presynaptic ligand remains unknown. We found that SALM5 interacts with the Ig domains of LAR family receptor protein tyrosine phosphatases (LAR-RPTPs; LAR, PTPδ, and PTPσ). These interactions are strongly inhibited by the splice insert B in the Ig domain region of LAR-RPTPs, and mediate SALM5-dependent presynaptic differentiation in contacting axons. In addition, SALM5 regulates AMPA receptor-mediated synaptic transmission through mechanisms involving the interaction of postsynaptic SALM5 with presynaptic LAR-RPTPs. These results suggest that postsynaptic SALM5 promotes synapse development by trans-synaptically interacting with presynaptic LAR-RPTPs and is important for the regulation of excitatory synaptic strength.


Assuntos
Processamento Alternativo/fisiologia , Axônios/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/metabolismo , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Animais , Moléculas de Adesão Celular Neuronais/genética , Camundongos , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/genética , Sinapses/genética
18.
Pharmacol Res ; 104: 97-107, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26687096

RESUMO

This study describes a fundamental functional difference between the two main polymorphisms of the pro-form of brain-derived neurotrophic factor (proBDNF), providing an explanation as to why these forms have such different age-related neurological outcomes. Healthy young carriers of the Met66 form (present in ∼30% Caucasians) have reduced hippocampal volume and impaired hippocampal-dependent memory function, yet the same polymorphic population shows enhanced cognitive recovery after traumatic brain injury, delayed cognitive dysfunction during aging, and lower risk of late-onset Alzheimer's disease (AD) compared to those with the more common Val66 polymorphism. To examine the differences between the protein polymorphisms in structure, kinetics of binding to proBDNF receptors and in vitro function, we generated purified cleavage-resistant human variants. Intriguingly, we found no statistical differences in those characteristics. As anticipated, exogenous application of proBDNF Val66 to rat hippocampal slices dysregulated synaptic plasticity, inhibiting long-term potentiation (LTP) and facilitating long-term depression (LTD). We subsequently observed that this occurred via the glycogen synthase kinase 3ß (GSK3ß) activation pathway. However, surprisingly, we found that Met66 had no such effects on either LTP or LTD. These novel findings suggest that, unlike Val66, the Met66 variant does not facilitate synapse weakening signaling, perhaps accounting for its protective effects with aging.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Precursores de Proteínas/genética , Sinapses/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Glicogênio Sintase Quinase 3 beta/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Humanos , L-Lactato Desidrogenase/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Polimorfismo Genético , Precursores de Proteínas/metabolismo , Ratos Wistar , Proteínas Recombinantes/farmacologia , Sinapses/efeitos dos fármacos , Proteínas tau/metabolismo
19.
J Neurochem ; 135(2): 207-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26451974

RESUMO

This Editorial highlights a study by Hunsberger et al. (2015) in the current issue of Journal of Neurochemistry, in which the authors explore the effects of riluzole (R) treatment on tau-P301L transgenic mice. The authors employed a comprehensive analysis of possible restorative effects of the drug by examining glutamate levels in subregions of the hippocampus, expression of tau and its hyper-phosphorylated forms, and memory function using behavioral tests. The authors report a simultaneous reduction in glutamate reuptake and an increase in glutamate release in the tau-P301L model, both of which are ameliorated with riluzole treatment. The authors' findings have implications for our understanding of synaptic transmission mechanisms also associated with Alzheimer's disease pathology.


Assuntos
Transtornos Cognitivos/prevenção & controle , Transtornos Cognitivos/psicologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/metabolismo , Fármacos Neuroprotetores/farmacologia , Riluzol/farmacologia , Tauopatias/prevenção & controle , Tauopatias/psicologia , Proteínas tau/biossíntese , Animais , Humanos
20.
Sci Rep ; 5: 12624, 2015 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-26220330

RESUMO

Cyclin Y (CCNY) is a member of the cyclin protein family, known to regulate cell division in proliferating cells. Interestingly, CCNY is expressed in neurons that do not undergo cell division. Here, we report that CCNY negatively regulates long-term potentiation (LTP) of synaptic strength through inhibition of AMPA receptor trafficking. CCNY is enriched in postsynaptic fractions from rat forebrain and is localized adjacent to postsynaptic sites in dendritic spines in rat hippocampal neurons. Using live-cell imaging of a pH-sensitive AMPA receptor, we found that during LTP-inducing stimulation, CCNY inhibits AMPA receptor exocytosis in dendritic spines. Furthermore, CCNY abolishes LTP in hippocampal slices. Taken together, our findings demonstrate that CCNY inhibits plasticity-induced AMPA receptor delivery to synapses and thereby blocks LTP, identifying a novel function for CCNY in post-mitotic cells.


Assuntos
Ciclinas/metabolismo , Exocitose/fisiologia , Potenciação de Longa Duração/fisiologia , Neurônios/fisiologia , Receptores de AMPA/metabolismo , Animais , Western Blotting , Células Cultivadas , Ciclinas/genética , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/fisiologia , Células HEK293 , Hipocampo/citologia , Hipocampo/metabolismo , Hipocampo/fisiologia , Humanos , Microscopia Confocal , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Prosencéfalo/citologia , Prosencéfalo/metabolismo , Prosencéfalo/fisiologia , Interferência de RNA , Ratos Wistar , Imagem com Lapso de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA