Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EMBO Mol Med ; 15(11): e15984, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37792911

RESUMO

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


Assuntos
Sistema de Sinalização das MAP Quinases , Neuroproteção , Animais , Humanos , Camundongos , alfa Carioferinas/farmacologia , Cognição , Fosforilação , Transdução de Sinais
2.
Biol Psychiatry ; 81(3): 179-192, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27587266

RESUMO

BACKGROUND: Dysregulation of Ras-extracellular signal-related kinase (ERK) signaling gives rise to RASopathies, a class of neurodevelopmental syndromes associated with intellectual disability. Recently, much attention has been directed at models bearing mild forms of RASopathies whose behavioral impairments can be attenuated by inhibiting the Ras-ERK cascade in the adult. Little is known about the brain mechanisms in severe forms of these disorders. METHODS: We performed an extensive characterization of a new brain-specific model of severe forms of RASopathies, the KRAS12V mutant mouse. RESULTS: The KRAS12V mutation results in a severe form of intellectual disability, which parallels mental deficits found in patients bearing mutations in this gene. KRAS12V mice show a severe impairment of both short- and long-term memory in a number of behavioral tasks. At the cellular level, an upregulation of ERK signaling during early phases of postnatal development, but not in the adult state, results in a selective enhancement of synaptogenesis in gamma-aminobutyric acidergic interneurons. The enhancement of ERK activity in interneurons at this critical postnatal time leads to a permanent increase in the inhibitory tone throughout the brain, manifesting in reduced synaptic transmission and long-term plasticity in the hippocampus. In the adult, the behavioral and electrophysiological phenotypes in KRAS12V mice can be temporarily reverted by inhibiting gamma-aminobutyric acid signaling but not by a Ras-ERK blockade. Importantly, the synaptogenesis phenotype can be rescued by a treatment at the developmental stage with Ras-ERK inhibitors. CONCLUSIONS: These data demonstrate a novel mechanism underlying inhibitory synaptogenesis and provide new insights in understanding mental dysfunctions associated to RASopathies.


Assuntos
Encéfalo/fisiologia , Neurônios GABAérgicos/fisiologia , Deficiência Intelectual/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Sinapses/fisiologia , Animais , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Neurônios GABAérgicos/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiologia , Potenciais Pós-Sinápticos Inibidores , Potenciação de Longa Duração , Transtornos da Memória/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Receptores de GABA/metabolismo , Comportamento Social , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
3.
Elife ; 52016 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-27557444

RESUMO

Ras-ERK signalling in the brain plays a central role in drug addiction. However, to date, no clinically relevant inhibitor of this cascade has been tested in experimental models of addiction, a necessary step toward clinical trials. We designed two new cell-penetrating peptides - RB1 and RB3 - that penetrate the brain and, in the micromolar range, inhibit phosphorylation of ERK, histone H3 and S6 ribosomal protein in striatal slices. Furthermore, a screening of small therapeutics currently in clinical trials for cancer therapy revealed PD325901 as a brain-penetrating drug that blocks ERK signalling in the nanomolar range. All three compounds have an inhibitory effect on cocaine-induced ERK activation and reward in mice. In particular, PD325901 persistently blocks cocaine-induced place preference and accelerates extinction following cocaine self-administration. Thus, clinically relevant, systemically administered drugs that attenuate Ras-ERK signalling in the brain may be valuable tools for the treatment of cocaine addiction.


Assuntos
Comportamento Animal/efeitos dos fármacos , Cocaína/administração & dosagem , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Proteínas ras/antagonistas & inibidores , Animais , Benzamidas/metabolismo , Peptídeos Penetradores de Células/metabolismo , Corpo Estriado/efeitos dos fármacos , Difenilamina/análogos & derivados , Difenilamina/metabolismo , Camundongos
4.
Psychopharmacology (Berl) ; 233(15-16): 2943-54, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27245230

RESUMO

RATIONALE: Despite the critical role attributed to phosphorylated extracellular signal regulated kinase (pERK1/2) in the nucleus accumbens (Acb) in the actions of addictive drugs, the effects of morphine on ERK1/2 phosphorylation in this area are still controversial. OBJECTIVES: In order to investigate further this issue, we studied (1) the ability of morphine to affect ERK1/2 phosphorylation in the shell (AcbSh) and core (AcbC) of Sprague-Dawley and Wistar rats and of CD-1 and C57BL/6J mice and (2) the role of dopamine D1 and µ-opioid receptors in Sprague-Dawley rats and CD-1 mice. METHODS: The pERK1/2 expression was assessed by immunohistochemistry. RESULTS: In rats, morphine decreased AcbSh and AcbC pERK1/2 expression, whereas in mice, increased it preferentially in the AcbSh compared with the AcbC. Systemic SCH 39166 decreased pERK1/2 expression on its own in the AcbSh and AcbC of Sprague-Dawley rats and CD-1 mice; furthermore, in rats, SCH 39166 disclosed the ability of morphine to stimulate pERK1/2 expression. Systemic (rats and mice) and intra-Acb (rats) naltrexone prevented both decreases, in rats, and increases, in mice. CONCLUSIONS: These findings confirm the differential effects of morphine in rats and mice Acb and that D1 receptors exert a facilitatory role on ERK1/2 phosphorylation; furthermore, they indicate that, in rats, removal of the D1-dependent pERK1/2 expression discloses the stimulatory influence of morphine on ERK1/2 phosphorylation and that the morphine's ability to decrease pERK1/2 expression is mediated by Acb µ-opioid receptors. Future experiments may disentangle the psychopharmacological significance of the effects of morphine on pERK1/2 in the Acb.


Assuntos
Analgésicos Opioides/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/efeitos dos fármacos , Morfina/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Receptores de Dopamina D1/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Animais , Benzazepinas/farmacologia , Antagonistas de Dopamina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Núcleo Accumbens/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores de Dopamina D1/metabolismo , Receptores Opioides mu/metabolismo
5.
Biol Psychiatry ; 77(2): 106-15, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24844602

RESUMO

BACKGROUND: Bidirectional long-term plasticity at the corticostriatal synapse has been proposed as a central cellular mechanism governing dopamine-mediated behavioral adaptations in the basal ganglia system. Balanced activity of medium spiny neurons (MSNs) in the direct and the indirect pathways is essential for normal striatal function. This balance is disrupted in Parkinson's disease and in l-3,4-dihydroxyphenylalanine (l-DOPA)-induced dyskinesia (LID), a common motor complication of current pharmacotherapy of Parkinson's disease. METHODS: Electrophysiological recordings were performed in mouse cortico-striatal slice preparation. Synaptic plasticity, such as long-term potentiation (LTP) and depotentiation, was investigated. Specific pharmacological inhibitors or genetic manipulations were used to modulate the Ras-extracellular signal-regulated kinase (Ras-ERK) pathway, a signal transduction cascade implicated in behavioral plasticity, and synaptic activity in different subpopulations of striatal neurons was measured. RESULTS: We found that the Ras-ERK pathway, is not only essential for long-term potentiation induced with a high frequency stimulation protocol (HFS-LTP) in the dorsal striatum, but also for its reversal, synaptic depotentiation. Ablation of Ras-guanine nucleotide-releasing factor 1 (Ras-GRF1), a neuronal activator of Ras proteins, causes a specific loss of HFS-LTP in the medium spiny neurons in the direct pathway without affecting LTP in the indirect pathway. Analysis of LTP in animals with unilateral 6-hydroxydopamine lesions (6-OHDA) rendered dyskinetic with chronic L-DOPA treatment reveals a complex, Ras-GRF1 and pathway-independent, apparently stochastic involvement of ERK. CONCLUSIONS: These data not only demonstrate a central role for Ras-ERK signaling in striatal LTP, depotentiation, and LTP restored after L-DOPA treatment but also disclose multifaceted synaptic adaptations occurring in response to dopaminergic denervation and pulsatile administration of L-DOPA.


Assuntos
Corpo Estriado/fisiopatologia , Discinesia Induzida por Medicamentos/fisiopatologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Plasticidade Neuronal/fisiologia , ras-GRF1/metabolismo , Animais , Antiparkinsonianos/toxicidade , Butadienos/farmacologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiopatologia , Corpo Estriado/efeitos dos fármacos , Dopamina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Levodopa/toxicidade , Camundongos Knockout , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Nitrilas/farmacologia , Oxidopamina , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/fisiopatologia , Inibidores de Proteínas Quinases/farmacologia , Técnicas de Cultura de Tecidos , ras-GRF1/genética
6.
PLoS One ; 6(4): e19419, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21559369

RESUMO

Radial Glia (RG) cells constitute the major population of neural progenitors of the mouse developing brain. These cells are located in the ventricular zone (VZ) of the cerebral cortex and during neurogenesis they support the generation of cortical neurons. Later on, during brain maturation, RG cells give raise to glial cells and supply the adult mouse brain of Neural Stem Cells (NSC). Here we used a novel transgenic mouse line expressing the CreER(T2) under the control of AspM promoter to monitor the progeny of an early cohort of RG cells during neurogenesis and in the post natal brain. Long term fate mapping experiments demonstrated that AspM-expressing RG cells are multi-potent, as they can generate neurons, astrocytes and oligodendrocytes of the adult mouse brain. Furthermore, AspM descendants give also rise to proliferating progenitors in germinal niches of both developing and post natal brains. In the latter--i.e. the Sub Ventricular Zone--AspM descendants acquired several feature of neural stem cells, including the capability to generate neurospheres in vitro. We also performed the selective killing of these early progenitors by using a Nestin-GFP(flox)-TK allele. The forebrain specific loss of early AspM expressing cells caused the elimination of most of the proliferating cells of brain, a severe derangement of the ventricular zone architecture, and the impairment of the cortical lamination. We further demonstrated that AspM is expressed by proliferating cells of the adult mouse SVZ that can generate neuroblasts fated to become olfactory bulb neurons.


Assuntos
Células-Tronco Embrionárias/citologia , Proteínas do Tecido Nervoso/fisiologia , Células-Tronco Neurais/citologia , Neuroglia/citologia , Alelos , Animais , Proteínas de Ligação a Calmodulina , Técnicas de Cultura de Células , Linhagem da Célula , Proliferação de Células , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Prosencéfalo/patologia , Fatores de Tempo
7.
Methods Mol Biol ; 661: 205-20, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20811985

RESUMO

Accumulating evidence indicates that p44(ERK1) and p42(ERK2) mitogen-activated protein kinases (MAPKs) have distinct quantitative roles in cell signaling. In our recently proposed model of regulation of ERK1 and ERK2, p42 plays a major role in delivering signals from the cell membrane to the nucleus, while p44 acts as a partial agonist of ERK2 toward effectors and downstream activators, thus providing a fine tuning system of the global signaling output. Here, we describe systems to modulate MAPK signaling in vitro and in vivo via lentiviral vector (LV)-mediated gene transfer, using three systems: RNAi with small hairpin RNAs, microRNA-mediated gene knockdown, and expression of signaling-interfering mutants of MEK1. We show, by using proliferation assays in mouse embryo fibroblasts (MEF) and NIH 3T3 cells, that gene knockdown of ERK1 promotes cell proliferation in a manner indistinguishable from a constitutively active MEK1 construct, while ERK2 RNAi causes a significant growth arrest, similar to that observed with the ectopic expression of a dominant negative MEK1 mutant.


Assuntos
Ensaios Enzimáticos/métodos , Vetores Genéticos/genética , Lentivirus/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Animais , Fosfatos de Cálcio/metabolismo , Proliferação de Células , Técnicas de Silenciamento de Genes , Humanos , Camundongos , MicroRNAs/genética , Proteína Quinase 1 Ativada por Mitógeno/deficiência , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/deficiência , Proteína Quinase 3 Ativada por Mitógeno/genética , Células NIH 3T3 , RNA Interferente Pequeno/genética , Transfecção
8.
J Neurosci Methods ; 185(1): 1-14, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19699233

RESUMO

The use of viral vectors as gene transfer tools for the central nervous system has seen a significant growth in the last decade. Improvements in the safety, efficiency and specificity of vectors for clinical applications have proven to be beneficial also for basic neuroscience research. This review will discuss the viral systems currently available to neuroscientists and some of the recent achievements in the study of synaptic function, memory and drug addiction.


Assuntos
Encéfalo/metabolismo , Técnicas de Transferência de Genes/tendências , Vetores Genéticos/genética , Vírus/genética , Animais , Química Encefálica/genética , Regulação da Expressão Gênica , Humanos , Aprendizagem/fisiologia , Plasticidade Neuronal/genética , Transtornos Relacionados ao Uso de Substâncias/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...