Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Brain Commun ; 4(4): fcac192, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35928052

RESUMO

In Alzheimer's disease, synapse loss causes memory and cognitive impairment. However, the mechanisms underlying synaptic degeneration in Alzheimer's disease are not well understood. In the hippocampus, alterations in the level of cysteine string protein alpha, a molecular co-chaperone at the pre-synaptic terminal, occur prior to reductions in synaptophysin, suggesting that it is a very sensitive marker of synapse degeneration in Alzheimer's. Here, we identify putative extracellular accumulations of cysteine string alpha protein, which are proximal to beta-amyloid deposits in post-mortem human Alzheimer's brain and in the brain of a transgenic mouse model of Alzheimer's disease. Cysteine string protein alpha, at least some of which is phosphorylated at serine 10, accumulates near the core of beta-amyloid deposits and does not co-localize with hyperphosphorylated tau, dystrophic neurites or glial cells. Using super-resolution microscopy and array tomography, cysteine string protein alpha was found to accumulate to a greater extent than other pre-synaptic proteins and at a comparatively great distance from the plaque core. This indicates that cysteine string protein alpha is most sensitive to being released from pre-synapses at low concentrations of beta-amyloid oligomers. Cysteine string protein alpha accumulations were also evident in other neurodegenerative diseases, including some fronto-temporal lobar dementias and Lewy body diseases, but only in the presence of amyloid plaques. Our findings are consistent with suggestions that pre-synapses are affected early in Alzheimer's disease, and they demonstrate that cysteine string protein alpha is a more sensitive marker for early pre-synaptic dysfunction than traditional synaptic markers. We suggest that cysteine string protein alpha should be used as a pathological marker for early synaptic disruption caused by beta-amyloid.

3.
Cereb Cortex ; 32(16): 3457-3471, 2022 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34937090

RESUMO

Extensive research has uncovered diverse forms of synaptic plasticity and an array of molecular signaling mechanisms that act as positive or negative regulators. Specifically, cyclic 3',5'-cyclic adenosine monophosphate (cAMP)-dependent signaling pathways are crucially implicated in long-lasting synaptic plasticity. In this study, we examine the role of Popeye domain-containing protein 1 (POPDC1) (or blood vessel epicardial substance (BVES)), a cAMP effector protein, in modulating hippocampal synaptic plasticity. Unlike other cAMP effectors, such as protein kinase A (PKA) and exchange factor directly activated by cAMP, POPDC1 is membrane-bound and the sequence of the cAMP-binding cassette differs from canonical cAMP-binding domains, suggesting that POPDC1 may have an unique role in cAMP-mediated signaling. Our results show that Popdc1 is widely expressed in various brain regions including the hippocampus. Acute hippocampal slices from Popdc1 knockout (KO) mice exhibit PKA-dependent enhancement in CA1 long-term potentiation (LTP) in response to weaker stimulation paradigms, which in slices from wild-type mice induce only transient LTP. Loss of POPDC1, while not affecting basal transmission or input-specificity of LTP, results in altered response during high-frequency stimulation. Popdc1 KO mice also show enhanced forskolin-induced potentiation. Overall, these findings reveal POPDC1 as a novel negative regulator of hippocampal synaptic plasticity and, together with recent evidence for its interaction with phosphodiesterases (PDEs), suggest that POPDC1 is involved in modulating activity-dependent local cAMP-PKA-PDE signaling.


Assuntos
Moléculas de Adesão Celular , Hipocampo , Potenciação de Longa Duração , Proteínas Musculares , Plasticidade Neuronal , Animais , Moléculas de Adesão Celular/genética , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Hipocampo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Musculares/genética , Transmissão Sináptica
4.
Neuropharmacology ; 193: 108616, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34051268

RESUMO

Potentiation of glutamatergic synaptic transmission is thought to underlie memory. The induction of this synaptic potentiation relies on activation of NMDA receptors which allows for calcium influx into the post-synapse. A key mechanistic question for the understanding of synaptic potentiation is what signaling is activated by the calcium influx. Here, I review evidences that at mature synapses the elevated calcium levels activate primarily calcium/calmodulin-dependent kinase II (CaMKII) and cause its autophophorylation. CaMKII autophosphorylation leads to calcium-independent activity of the kinase, so that kinase signaling can outlast NMDA receptor-dependent calcium influx. Prolonged CaMKII signaling induces downstream signaling for AMPA receptor trafficking into the post-synaptic density and causes structural enlargement of the synapse. Interestingly, however, CaMKII autophosphorylation does not have such an essential role in NMDA receptor-dependent synaptic potentiation in early postnatal development and in adult dentate gyrus, where neurogenesis occurs. Additionally, in old age memory-relevant NMDA receptor-dependent synaptic plasticity appears to be due to generation of multi-innervated dendritic spines, which does not require CaMKII autophosphorylation. In conclusion, CaMKII autophosphorylation has a conditional role in the induction of NMDA receptor-dependent synaptic potentiation.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/fisiologia , Potenciais Sinápticos , Animais , Potenciação de Longa Duração , Fosforilação , Ratos , Ratos Long-Evans
5.
JCI Insight ; 5(5)2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32069266

RESUMO

Long-term memory depends on the control of activity-dependent neuronal gene expression, which is regulated by epigenetic modifications. The epigenetic modification of histones is orchestrated by the opposing activities of 2 classes of regulatory complexes: permissive coactivators and silencing corepressors. Much work has focused on coactivator complexes, but little is known about the corepressor complexes that suppress the expression of plasticity-related genes. Here, we define a critical role for the corepressor SIN3A in memory and synaptic plasticity, showing that postnatal neuronal deletion of Sin3a enhances hippocampal long-term potentiation and long-term contextual fear memory. SIN3A regulates the expression of genes encoding proteins in the postsynaptic density. Loss of SIN3A increases expression of the synaptic scaffold Homer1, alters the metabotropic glutamate receptor 1α (mGluR1α) and mGluR5 dependence of long-term potentiation, and increases activation of ERK in the hippocampus after learning. Our studies define a critical role for corepressors in modulating neural plasticity and memory consolidation and reveal that Homer1/mGluR signaling pathways may be central molecular mechanisms for memory enhancement.


Assuntos
Hipocampo/fisiologia , Proteínas de Arcabouço Homer/metabolismo , Plasticidade Neuronal/fisiologia , Receptor de Glutamato Metabotrópico 5/metabolismo , Transdução de Sinais/fisiologia , Complexo Correpressor Histona Desacetilase e Sin3/fisiologia , Animais , Hipocampo/metabolismo , Camundongos , Camundongos Mutantes , Neurônios/metabolismo , Complexo Correpressor Histona Desacetilase e Sin3/genética
6.
Mol Brain ; 11(1): 76, 2018 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-30593282

RESUMO

Shortly before he died in October 2017, John Lisman submitted an invited review to Molecular Brain on 'Criteria for identifying the molecular basis of the engram (CaMKII, PKMζ)'. John had no opportunity to read the referees' comments, and as a mark of the regard in which he was held by the neuroscience community the Editors decided to publish his review as submitted. This obituary takes the form of a series of commentaries on Lisman's review. At the same time we are publishing as a separate article a longer response by Todd Sacktor and André Fenton entitled 'What does LTP tell us about the roles of CaMKII and PKMζ in memory?' which presents the case for a rival memory molecule, PKMζ.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Memória , Animais , Hipocampo/metabolismo , Humanos , Potenciação de Longa Duração , Plasticidade Neuronal , Proteína Quinase C/metabolismo
7.
J Neurochem ; 147(1): 12-23, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29704430

RESUMO

In this review, we discuss the poorly explored role of calcium/calmodulin-dependent protein kinase II (CaMKII) in memory maintenance, and its influence on memory destabilization. After a brief review on CaMKII and memory destabilization, we present critical pieces of evidence suggesting that CaMKII activity increases retrieval-induced memory destabilization. We then proceed to propose two potential molecular pathways to explain the association between CaMKII activation and increased memory destabilization. This review will pinpoint gaps in our knowledge and discuss some 'controversial' observations, establishing the basis for new experiments on the role of CaMKII in memory reconsolidation. The role of CaMKII in memory destabilization is of great clinical relevance. Still, because of the lack of scientific literature on the subject, more basic science research is necessary to pursue this pathway as a clinical tool.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/fisiologia , Transtornos da Memória/enzimologia , Transtornos da Memória/genética , Memória/fisiologia , Animais , Humanos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
8.
Sci Rep ; 7(1): 4040, 2017 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-28642476

RESUMO

CaMK2N1 and CaMK2N2 are endogenous inhibitors of calcium/calmodulin-dependent protein kinase II (CaMKII), a key synaptic signaling molecule for learning and memory. Here, we investigated the learning and memory function of CaMK2N1 by knocking-down its expression in dorsal hippocampus of mice. We found that reduced CaMK2N1 expression does not affect contextual fear long-term memory (LTM) formation. However, we show that it impairs maintenance of established LTM, but only if retrieval occurs. CaMK2N1 knockdown prevents a decrease of threonine-286 (T286) autophosphorylation of αCaMKII and increases GluA1 levels in hippocampal synapses after retrieval of contextual fear LTM. CaMK2N1 knockdown can also increase CaMK2N2 expression, but we show that such increased expression does not affect LTM after retrieval. We also found that substantial overexpression of CaMK2N2 in dorsal hippocampus impairs LTM formation, but not LTM maintenance, suggesting that CaMKII activity is not required for LTM storage. Taken together, we propose a specific function for CaMK2N1; enabling LTM maintenance after retrieval by inhibiting T286 autophosphorylation of αCaMKII.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Memória de Longo Prazo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Medo , Expressão Gênica , Técnicas de Silenciamento de Genes , Hipocampo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Fosforilação , Interferência de RNA , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Receptores de AMPA/metabolismo
9.
Brain ; 139(Pt 10): 2751-2765, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27524794

RESUMO

Characteristic features of Alzheimer's disease are memory loss, plaques resulting from abnormal processing of amyloid precursor protein (APP), and presence of neurofibrillary tangles and dystrophic neurites containing hyperphosphorylated tau. Currently, it is not known what links these abnormalities together. Cytoplasmic FMR1 interacting protein 2 (CYFIP2) has been suggested to regulate mRNA translation at synapses and this may include local synthesis of APP and alpha-calcium/calmodulin-dependent kinase II, a kinase that can phosphorylate tau. Further, CYFIP2 is part of the Wiskott-Aldrich syndrome protein-family verprolin-homologous protein complex, which has been implicated in actin polymerization at synapses, a process thought to be required for memory formation. Our previous studies on p25 dysregulation put forward the hypothesis that CYFIP2 expression is reduced in Alzheimer's disease and that this contributes to memory impairment, abnormal APP processing and tau hyperphosphorylation. Here, we tested this hypothesis. First, in post-mortem tissue CYFIP2 expression was reduced by ∼50% in severe Alzheimer's hippocampus and superior temporal gyrus when normalized to expression of a neuronal or synaptic marker protein. Interestingly, there was also a trend for decreased expression in mild Alzheimer's disease hippocampus. Second, CYFIP2 expression was reduced in old but not in young Tg2576 mice, a model of familial Alzheimer's disease. Finally, we tested the direct impact of reduced CYFIP2 expression in heterozygous null mutant mice. We found that in hippocampus this reduced expression causes an increase in APP and ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) protein, but not mRNA expression, and elevates production of amyloid-ß42 Reduced CYFIP2 expression also increases alpha-calcium/calmodulin-dependent kinase II protein expression, and this is associated with hyperphosphorylation of tau at serine-214. The reduced expression also impairs spine maturity without affecting spine density in apical dendrites of CA1 pyramidal neurons. Furthermore, the reduced expression prevents retention of spatial memory in the water maze. Taken together, our findings indicate that reduced CYFIP2 expression triggers a cascade of change towards Alzheimer's disease, including amyloid production, tau hyperphosphorylation and memory loss. We therefore suggest that CYFIP2 could be a potential hub for targeting treatment of the disease.

10.
Mol Brain ; 8(1): 78, 2015 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-26603284

RESUMO

CaMKII is a remarkably complex protein kinase, known to have a fundamental role in synaptic plasticity and memory formation. Further, CaMKII has also been suggested to be a tau kinase. CaMKII dysregulation may therefore be a modulator of toxicity in Alzheimer's disease, a dementia characterised by aberrant calcium signalling, synapse and neuronal loss, and impaired memory. Here, we first examine the evidence for CaMKII dysregulation in Alzheimer's patients and draw parallels to findings in disease models which recapitulate key aspects of the disease. We then put forward the hypothesis that these changes critically contribute to neurodegeneration and memory impairment in Alzheimer's disease.


Assuntos
Doença de Alzheimer/enzimologia , Doença de Alzheimer/patologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Modelos Biológicos
11.
PLoS One ; 10(10): e0140821, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26469698

RESUMO

p35 is an activation subunit of the cyclin-dependent kinase 5 (CDK5), which is a Ser/Thr kinase that is expressed predominantly in neurons. Disruption of the CDK5 or p35 (CDK5R1) genes induces abnormal neuronal layering in various regions of the mouse brain via impaired neuronal migration, which may be relevant for mental retardation in humans. Accordingly, mutations in the p35 gene were reported in patients with nonsyndromic mental retardation; however, their effect on the biochemical function of p35 has not been examined. Here, we studied the biochemical effect of mutant p35 on its known properties, i.e., stability, CDK5 activation, and cellular localization, using heterologous expression in cultured cells. We also examined the effect of the mutations on axon elongation in cultured primary neurons and migration of newborn neurons in embryonic brains. However, we did not detect any significant differences in the effects of the mutant forms of p35 compared with wild-type p35. Therefore, we conclude that these p35 mutations are unlikely to cause mental retardation.


Assuntos
Deficiência Intelectual/genética , Mutação , Proteínas do Tecido Nervoso/genética , Neurônios/fisiologia , Animais , Animais Recém-Nascidos , Axônios/fisiologia , Células COS , Células Cultivadas , Chlorocebus aethiops , Embrião de Mamíferos , Humanos , Camundongos , Camundongos Endogâmicos ICR , Mutação/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo
12.
Neurobiol Learn Mem ; 124: 48-51, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25933505

RESUMO

NMDA receptor-dependent long-term potentiation (LTP) at hippocampal CA1 synapses is a well-accepted mechanism underlying long-term memory (LTM) formation. However, studies with mice that lack threonine-286 autophosphorylation of αCaMKII have shown that hippocampal LTM can be formed despite absence of NMDA receptor-dependent CA1 LTP. After multiple training trials, LTM formation in these mutants is linked to the generation of multi-innervated dendritic spines (MIS), a spine that receives typically two presynaptic inputs. PSD-95 overexpression is sufficient for MIS generation and depends on mTOR signaling. LTM that involves MIS generation appears less modifiable upon retrieval in comparison to LTM without MIS generation. Taken together, MIS generation appears to be a novel LTM mechanism after multiple training trials, which may occur in diseases with impaired LTP or conditions affecting negative feedback CaMKII signaling at the synapse.


Assuntos
Encéfalo/fisiologia , Espinhas Dendríticas/fisiologia , Potenciação de Longa Duração , Memória de Longo Prazo/fisiologia , Animais , Encéfalo/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Espinhas Dendríticas/metabolismo , Camundongos
13.
Learn Mem ; 21(11): 616-26, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25322797

RESUMO

α-calcium/calmodulin-dependent protein kinase (αCaMKII) T286-autophosphorylation provides a short-term molecular memory that was thought to be required for LTP and for learning and memory. However, it has been shown that learning can occur in αCaMKII-T286A mutant mice after a massed training protocol. This raises the question of whether there might be a form of LTP in these mice that can occur without T286 autophosphorylation. In this study, we confirmed that in CA1 pyramidal cells, LTP induced in acute hippocampal slices, after a recovery period in an interface chamber, is strictly dependent on postsynaptic αCaMKII autophosphorylation. However, we demonstrated that αCaMKII-autophosphorylation-independent plasticity can occur in the hippocampus but at the expense of synaptic specificity. This nonspecific LTP was observed in mutant and wild-type mice after a recovery period in a submersion chamber and was independent of NMDA receptors. Moreover, when slices prepared from mutant mice were preincubated during 2 h with rapamycin, high-frequency trains induced a synapse-specific LTP which was added to the nonspecific LTP. This specific LTP was related to an increase in the duration and the amplitude of NMDA receptor-mediated response induced by rapamycin.


Assuntos
Região CA1 Hipocampal/fisiologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Potenciação de Longa Duração , Células Piramidais/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Animais , Região CA1 Hipocampal/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Células Piramidais/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores
14.
Learn Mem ; 20(10): 540-52, 2013 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-24042850

RESUMO

In the adult mammalian brain, more than 250 protein kinases are expressed, but only a few of these kinases are currently known to enable learning and memory. Based on this information it appears that learning and memory-related kinases either impact on synaptic transmission by altering ion channel properties or ion channel density, or regulate gene expression and protein synthesis causing structural changes at existing synapses as well as synaptogenesis. Here, we review the roles of these kinases in short-term memory formation, memory consolidation, memory storage, retrieval, reconsolidation, and extinction. Specifically, we discuss the roles of calcium/calmodulin-dependent kinase II (CaMKII), the calcium/calmodulin kinase cascade, extracellular signal regulated kinase 1 and 2 (ERK1/2), cAMP-dependent protein kinase A (PKA), cGMP-dependent protein kinase G (PKG), the phosphatidylinositol 3-kinase (PI3K) pathway, and protein kinase M ζ (PKMζ). Although these kinases are important for learning and memory processes, much remains to be learned as to how they act. Therefore, it will be important to identify and characterize the critical phosphorylation substrates so that a sophisticated understanding of learning and memory processes will be achieved. This will also allow for a systematic analysis of dysfunctional kinase activity in mental disorders.


Assuntos
Encéfalo/enzimologia , Aprendizagem/fisiologia , Memória/fisiologia , Proteínas Quinases/metabolismo , Transdução de Sinais/fisiologia , Animais , Humanos
15.
Front Behav Neurosci ; 5: 78, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22164138

RESUMO

Ras-GRF1 is a neuronal specific guanine exchange factor that, once activated by both ionotropic and metabotropic neurotransmitter receptors, can stimulate Ras proteins, leading to long-term phosphorylation of downstream signaling. The two available reports on the behavior of two independently generated Ras-GRF1 deficient mouse lines provide contrasting evidence on the role of Ras-GRF1 in spatial memory and contextual fear conditioning. These discrepancies may be due to the distinct alterations introduced in the mouse genome by gene targeting in the two lines that could differentially affect expression of nearby genes located in the imprinted region containing the Ras-grf1 locus. In order to determine the real contribution of Ras-GRF1 to spatial memory we compared in Morris Water Maze learning Brambilla's mice with a third mouse line (GENA53) in which a non-sense mutation was introduced in the Ras-GRF1 coding region without additional changes in the genome and we found that memory in this task is normal. Also, we measured both contextual and cued fear conditioning, which were previously reported to be affected in Brambilla's mice, and we confirmed that contextual learning but not cued conditioning is impaired in both mouse lines. In addition, we also tested both lines for the first time in conditioned place aversion in the Intellicage, an ecological and remotely controlled behavioral test, and we observed normal learning. Finally, based on previous reports of other mutant lines suggesting that Ras-GRF1 may control body weight, we also measured this non-cognitive phenotype and we confirmed that both Ras-GRF1 deficient mutants are smaller than their control littermates. In conclusion, we demonstrate that Ras-GRF1 has no unique role in spatial memory while its function in contextual fear conditioning is likely to be due not only to its involvement in amygdala functions but possibly to some distinct hippocampal connections specific to contextual learning.

16.
Brain ; 134(Pt 8): 2408-21, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21772061

RESUMO

Cyclin-dependent kinase 5 is activated by small subunits, of which p35 is the most abundant. The functions of cyclin-dependent kinase 5 signalling in cognition and cognitive disorders remains unclear. Here, we show that in schizophrenia, a disorder associated with impaired cognition, p35 expression is reduced in relevant brain regions. Additionally, the expression of septin 7 and OPA1, proteins downstream of truncated p35, is decreased in schizophrenia. Mimicking a reduction of p35 in heterozygous knockout mice is associated with cognitive endophenotypes. Furthermore, a reduction of p35 in mice results in protein changes similar to schizophrenia post-mortem brain. Hence, heterozygous p35 knockout mice model both cognitive endophenotypes and molecular changes reminiscent of schizophrenia. These changes correlate with reduced acetylation of the histone deacetylase 1 target site H3K18 in mice. This site has previously been shown to be affected by truncated p35. By restoring H3K18 acetylation with the clinically used specific histone deacetylase 1 inhibitor MS-275 both cognitive and molecular endophenotypes of schizophrenia can be rescued in p35 heterozygous knockout mice. In summary, we suggest that reduced p35 expression in schizophrenia has an impact on synaptic protein expression and cognition and that these deficits can be rescued, at least in part, by the inhibition of histone deacetylase 1.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Transtornos Cognitivos/etiologia , GTP Fosfo-Hidrolases/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Esquizofrenia/metabolismo , Septinas/metabolismo , Estimulação Acústica/métodos , Análise de Variância , Animais , Benzamidas/farmacologia , Encéfalo/metabolismo , Encéfalo/patologia , Transtornos Cognitivos/genética , Endofenótipos , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Inibidores de Histona Desacetilases/farmacologia , Humanos , Inibição Psicológica , Relações Interpessoais , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Mucoproteínas/genética , Mutação/genética , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Fosfotransferases , Mudanças Depois da Morte , Piridinas/farmacologia , Recompensa , Esquizofrenia/complicações , Esquizofrenia/genética , Esquizofrenia/patologia , Fatores Sexuais , Sinapses/metabolismo
17.
Learn Mem ; 18(6): 375-83, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21597043

RESUMO

Insulin has been shown to impact on learning and memory in both humans and animals, but the downstream signaling mechanisms involved are poorly characterized. Insulin receptor substrate-2 (Irs2) is an adaptor protein that couples activation of insulin- and insulin-like growth factor-1 receptors to downstream signaling pathways. Here, we have deleted Irs2, either in the whole brain or selectively in the forebrain, using the nestin Cre- or D6 Cre-deleter mouse lines, respectively. We show that brain- and forebrain-specific Irs2 knockout mice have enhanced hippocampal spatial reference memory. Furthermore, NesCreIrs2KO mice have enhanced spatial working memory and contextual- and cued-fear memory. Deletion of Irs2 in the brain also increases PSD-95 expression and the density of dendritic spines in hippocampal area CA1, possibly reflecting an increase in the number of excitatory synapses per neuron in the hippocampus that can become activated during memory formation. This increase in activated excitatory synapses might underlie the improved hippocampal memory formation observed in NesCreIrs2KO mice. Overall, these results suggest that Irs2 acts as a negative regulator on memory formation by restricting dendritic spine generation.


Assuntos
Proteínas Substratos do Receptor de Insulina/metabolismo , Memória/fisiologia , Análise de Variância , Animais , Condicionamento Psicológico/fisiologia , Espinhas Dendríticas/ultraestrutura , Proteína 4 Homóloga a Disks-Large , Comportamento Exploratório/fisiologia , Medo , Guanilato Quinases/metabolismo , Hipocampo/citologia , Proteínas Substratos do Receptor de Insulina/deficiência , Aprendizagem em Labirinto/fisiologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/genética , Neurônios/metabolismo , Neurônios/ultraestrutura , Teste de Desempenho do Rota-Rod/métodos , Deleção de Sequência/genética
18.
Biol Psychiatry ; 70(2): 159-68, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21616478

RESUMO

BACKGROUND: The cyclin-dependent kinase 5 activator p35 can be cleaved into p25. Formation of p25 has been suggested to contribute to neurodegeneration in Alzheimer's disease (AD). However, overexpression of low levels of p25 in mice enhances memory formation. Therefore, it has been suggested that p25 formation might be an event early in AD to compensate for impairments in synaptic plasticity. Ongoing p25 formation has been hypothesized to contribute to neurodegeneration at the later stages of AD. METHODS: Here, we tested the early compensation hypothesis by analyzing the levels of p25 and its precursor p35 in AD postmortem samples from different brain regions at different stages of tau pathology, using quantitative Western blots. Furthermore, we studied p35 and p25 during spatial memory formation. By employing quantitative mass spectrometry, we identified proteins downstream of p25, which were then studied in AD samples. RESULTS: We found that p25 is generated during spatial memory formation. Furthermore, we demonstrate that overexpression of p25 in the physiological range increases the expression of two proteins implicated in spine formation, septin 7 and optic atrophy 1. We show that the expression of p35 and p25 is reduced as an early event in AD. Moreover, expression of the p25-regulated protein optic atrophy 1 was reduced in a time course similar to p25 expression. CONCLUSIONS: Our findings suggest that p25 generation is a mechanism underlying hippocampal memory formation that is impaired in the early stages of AD. Our findings argue against the previously raised early compensation hypothesis and they propose that p25-mediated neurotoxicity does not occur in AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Regulação para Baixo/genética , Memória/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/ultraestrutura , Ciclina D1/metabolismo , Modelos Animais de Doenças , GTP Fosfo-Hidrolases/metabolismo , Humanos , Espectrometria de Massas/métodos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Mutação/genética , Proteínas do Tecido Nervoso/genética , Fosfopiruvato Hidratase/metabolismo , Septinas/metabolismo , Sinapses/metabolismo , Sinapses/ultraestrutura , Sinaptossomos/metabolismo , Sinaptossomos/ultraestrutura , Proteínas tau/metabolismo
20.
Brain Res Bull ; 85(1-2): 2-8, 2011 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-21070840

RESUMO

Ca(2+)/calmodulin-dependent kinase II (CaMKII) is an abundant synaptic signalling molecule that is essential for memory formation and the induction of synaptic potentiation. Additionally, CaMKII plays a prominent role in synaptic tagging and metaplasticity. These abilities are mediated by kinase activity as well as binding to a wide variety of synaptic proteins, including NMDA receptor subunits, modulating CaMKII location and activity. A characteristic feature is that autophosphorylation of CaMKII switches the kinase into autonomous activity. Since CaMKII can be autonomously active and because CaMKII is required for the formation of memory it is important that the kinase activity is adequately switched off. However, the exact time window of increased activity and how this is terminated, it is still matter of debate. After training in a memory task CaMKII activity is increased for at least 30 min. This CaMKII activity and further activation of CaMKII may be regulated by changes in the expression of two endogenous CaMKII inhibitor proteins, CaMKII inhibitor Alpha and Beta, as they are up-regulated early after training. These endogenous inhibitors specifically block CaMKII activity and they inhibit the association with NMDA receptor subunits. Thus, regulation of the expression of endogenous CaMKII inhibitors may constitute a novel negative feedback on CaMKII signalling during memory formation.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Isoenzimas/metabolismo , Memória/fisiologia , Transdução de Sinais/fisiologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/química , Inibidores Enzimáticos/metabolismo , Humanos , Isoenzimas/química , Plasticidade Neuronal/fisiologia , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Ligação Proteica , Proteínas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA