Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Kaohsiung J Med Sci ; 40(6): 553-560, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38623867

RESUMO

Working memory (WM) is a cognitive function important for guiding the on-going or upcoming behavior. A memory-related protein Arc (activity-regulated cytoskeleton-associated protein) is implicated in long-term memory consolidation. Recent evidence further suggests the involvement of hippocampal Arc in spatial WM. The medial prefrontal cortex (mPFC) is a key brain region mediating WM. However, the role of mPFC Arc in WM is still uncertain. To investigate whether mPFC Arc protein is involved in WM performance, delayed non-match to sample (DNMS) T-maze task was performed in rats with or without blocking new synthesis of mPFC Arc. In DNMS task, a 10-s or 30-s delay between the sample run and the choice run was given to evaluate WM performance. To block new Arc protein synthesis during the DNMS task, Arc antisense oligodeoxynucleotides (ODNs) were injected to the bilateral mPFC. The results show that, in rats without surgery for cannula implantation and subsequent intracerebral injection of ODNs, WM was functioning well during the DNMS task with a delay of 10 s but not 30 s, which was accompanied with a significantly increased level of mPFC Arc protein, indicating a possible link between enhanced Arc protein expression and the performance of WM. After preventing the enhancement of mPFC Arc protein expression with Arc antisense ODNs, rat's WM performance was impaired. These findings support enhanced mPFC Arc protein expression playing a role during WM performance.


Assuntos
Proteínas do Citoesqueleto , Memória de Curto Prazo , Proteínas do Tecido Nervoso , Córtex Pré-Frontal , Animais , Córtex Pré-Frontal/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas do Citoesqueleto/genética , Masculino , Memória de Curto Prazo/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Ratos , Aprendizagem em Labirinto/fisiologia , Ratos Sprague-Dawley
2.
Neurosci Lett ; 818: 137533, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37865186

RESUMO

Hypercholesterolemia is a risk factor for Alzheimer's disease (AD). Plasma cholesterol does not pass the blood-brain barrier whereas its metabolite 27-hydroxycholesterol (27-OHC) can enter the brain. High 27-OHC in the brain has been suggested to mediate hypercholesterolemia-induced impairments of learning and memory through promoting amyloid-ß accumulation and facilitating synaptic disruption. In AD brains, the activity of histone deacetylase (HDAC) is elevated. Treating AD animals with HDAC inhibitors decreases amyloid-ß levels and synaptic damages, which leads to memory improvement. Whether HDAC activity is involved in the actions of 27-OHC is still uncertain. In this study, 4 weekly injections of 27-OHC/vehicle were given to rats followed by 3 daily injections of HDAC inhibitor trichostatin (TSA)/vehicle. The results of Morris water maze test reveal that all rats have intact spatial learning ability during the 5-d training phase. However, the behavioral performance during the probe trial was impaired by 27-OHC treatment, which was improved by adding TSA treatments. Furthermore, 27-OHC treatments reduced the hippocampal levels of acetylated histone H3, acetylated α tubulin, insulin-degrading enzyme and postsynaptic protein PSD-95, indicating that 27-OHC treatments may induce enhanced HDAC activity, decreased amyloid-ß clearance and synaptic disruption. All reduced levels returned to the basal levels by adding TSA treatments. These findings support our hypothesis that HDAC activity is enhanced following long-term exposure to excess 27-OHC.


Assuntos
Doença de Alzheimer , Inibidores de Histona Desacetilases , Hipercolesterolemia , Animais , Ratos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Hipercolesterolemia/metabolismo , Aprendizagem Espacial
3.
J Cell Physiol ; 235(2): 1065-1075, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31264208

RESUMO

Chronic high-fat-diet (HFD) consumption can lead to the development of brain insulin resistance, which then exerts deleterious effects on learning and memory. Activity-regulated cytoskeleton-associated protein (Arc) is a memory-related protein, and its expression can be induced by insulin stimulation. In HFD-fed animals, their basal Arc protein levels in cerebral cortex and hippocampus are reduced. However, the effects of HFD on novelty-induced Arc protein expression that is important for cognitive function is still unknown. In the present study, after feeding HFD (60% kcal from fat) for 5 weeks, mice developed brain insulin resistance and had a significant reduction in the novelty-induced but not the basal Arc protein levels in their hippocampi. Further experiments were performed in primary rat hippocampal neurons. The results show that, under the condition of neuronal insulin resistance, acute insulin stimulation induced less activation of the phosphatidylinositol 3-kinase/protein kinase B/p70 ribosomal S6 kinase (PI3K/Akt/p70S6K) pathway, resulting in reduced induction of Arc protein expression. Accordingly, it is suggested that following HFD feeding, the reduction in novelty-induced Arc protein expression in animal's hippocampus is probably related to a suppressed activation of the PI3K/Akt/p70S6K pathway due to the existence of brain insulin resistance.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Dieta Hiperlipídica , Gorduras na Dieta/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Ração Animal/análise , Animais , Células Cultivadas , Proteínas do Citoesqueleto/genética , Hipocampo/citologia , Insulina/farmacologia , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo
4.
J Cell Physiol ; 231(11): 2428-38, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26895748

RESUMO

Cholinergic dysfunction in the brain is closely related to cognitive impairment including memory loss. In addition to the degeneration of basal forebrain cholinergic neurons, deficits in the cholinergic receptor signaling may also play an important role. In the present study, to examine the cholinergic signaling pathways responsible for the induction of a memory-related postsynaptic protein, a cholinergic agonist carbachol was used to induce the expression of activity-regulated cytoskeleton associated protein (Arc) in primary rat cortical neurons. After pretreating neurons with various antagonists or inhibitors, the levels of carbachol-induced Arc protein expression were detected by Western blot analysis. The results show that carbachol induces Arc protein expression mainly through activating M1 acetylcholine receptors and the downstream phospholipase C pathway, which may lead to the activation of the MAPK/ERK signaling pathway. Importantly, carbachol-mediated M2 receptor activation exerts negative effects on Arc protein expression and thus counteracts the enhanced effects of M1 activation. Furthermore, it is suggested for the first time that M1-mediated enhancement of N-methyl-D-aspartate receptor (NMDAR) responses, leading to Ca(2+) entry through NMDARs, contributes to carbachol-induced Arc protein expression. These findings reveal a more complete cholinergic signaling that is responsible for carbachol-induced Arc protein expression, and thus provide more information for developing treatments that can modulate cholinergic signaling and consequently alleviate cognitive impairment. J. Cell. Physiol. 231: 2428-2438, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Acetilcolina/metabolismo , Córtex Cerebral/citologia , Proteínas do Citoesqueleto/metabolismo , Memória , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Transdução de Sinais , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Carbacol/farmacologia , Células Cultivadas , Memória/efeitos dos fármacos , Modelos Biológicos , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ratos , Receptor Muscarínico M1/antagonistas & inibidores , Receptor Muscarínico M2/antagonistas & inibidores , Receptores Colinérgicos , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fosfolipases Tipo C/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
5.
Cell Mol Life Sci ; 71(20): 4069-80, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24705985

RESUMO

Learning and memory depend on long-term synaptic plasticity including long-term potentiation (LTP) and depression (LTD). Activity-regulated cytoskeleton-associated protein (Arc) plays versatile roles in synaptic plasticity mainly through inducing F-actin formation, underlying consolidation of LTP, and promoting AMPA receptor (AMPAR) endocytosis, underlying LTD. Insulin can also induce LTD by facilitating the internalization of AMPARs. In neuroblastoma cells, insulin induced a dramatic increase in Arc mRNA and Arc protein levels, which may underlie the memory-enhancing action of insulin. Thus, a hypothesis was made that, in response to insulin, increased AMPAR endocytosis leads to enhanced Arc expression, and vice versa. Primary cultures of neonatal Sprague-Dawley rat cortical neurons were used. Using Western-blot analysis and immunofluorescent staining, our results reveal that inhibiting AMPAR-mediated responses with AMPAR antagonists significantly enhanced whereas blocking AMPAR endocytosis with various reagents significantly prevented insulin (200 nM, 2 h)-induced Arc expression. Furthermore, via surface biotinylation assay, we demonstrate that acute blockade of new Arc synthesis after insulin stimulation using Arc antisense oligodeoxynucleotide prevented insulin-stimulated AMPAR endocytosis. These findings suggest for the first time that an interaction exists between insulin-stimulated AMPAR endocytosis and insulin-induced Arc expression.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Endocitose/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Insulina/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Receptores de AMPA/metabolismo , Animais , Células Cultivadas , Clatrina/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/citologia , Neurônios/metabolismo , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Quinoxalinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/antagonistas & inibidores , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
6.
Artigo em Inglês | MEDLINE | ID: mdl-24141945

RESUMO

Protein kinase M zeta (PKMζ) and the kidney and brain protein (KIBRA) play important roles in various forms of memories. However, whether they are involved in performing the T-maze task is still unknown. In this study, the delayed nonmatch-to-sample (DNMS) task in a T-maze was given to rats. The percentage of correct choices denoting the performance accuracy was calculated and the protein levels of PKMζ and KIBRA in rat's prefrontal cortex were measured. The results showed significantly increased performance accuracy after the training phase, which was maintained on the next day in groups with a delay of 10 s but not 30 s, indicating that 30 s is too long for rats to maintain working memory. As for the expressions of PKMζ and KIBRA, significant increases were observed 1 day after the training phase, indicating that the formation of reference memory accompanies an increase in PKMζ and KIBRA. No significant difference was found among groups with various delay intervals, indicating that the expressions of PKMζ and KIBRA exert no effects on the performance of working memory. These results provide the first evidence that KIBRA as well as PKMζ is closely related to reference memory but not working memory in rats.


Assuntos
Aprendizagem em Labirinto/fisiologia , Memória de Curto Prazo/fisiologia , Memória/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Fosfoproteínas/metabolismo , Córtex Pré-Frontal/fisiologia , Proteína Quinase C/metabolismo , Animais , Western Blotting , Masculino , Prática Psicológica , Ratos , Ratos Sprague-Dawley , Análise e Desempenho de Tarefas , Fatores de Tempo
7.
J Toxicol Sci ; 38(5): 783-91, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24067726

RESUMO

The purpose of this study was to investigate the effects of di-(2-ethylhexyl) phthalate (DEHP) treatment on MyoD and myogenin expression and myotube formation in the murine C2C12 cells. Myogenic differentiation is principally regulated by activities of myogenic regulatory factors, such as MyoD and myogenin, leading the elongation and fusion of mononucleated myoblasts into multinucleated myotubes. In the present study, myogenic differentiation of C2C12 cells was induced by serum deprivation with medium containing vehicle or DEHP (10, 100, 1,000 µg/ml) for 5 days. Using 3-(4,5-dimethylthiazol-2-yl) 2,5-diphenyltetrazolium bromide (MTT) assay clearly demonstrated cell viability was not affected by DEHP at any given dose. At the dose of 1,000 µg/ml DEHP, the elongation of multinucleated myotubes, and the percent of nuclei incorporated into myosin heavy chain (MyHC)-stained myotubes were markedly reduced. In addition, immunoblotting revealed expression of muscle specific marker MyHC, as well as myogenic regulatory factors MyoD and myogenin, were reduced in DEHP-treated myotubes during myogenic differentiation. Taken together, the results showed that DEHP may impair myogenic differentiation through repression of myogenic regulatory factors, such as MyoD and myogenin, resulting in a reduction of MyHC expression. This in vitro study suggests that DEHP may be an environmental risk factor for myogenesis.


Assuntos
Diferenciação Celular/genética , Dietilexilftalato/toxicidade , Disruptores Endócrinos/toxicidade , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Proteína MyoD/metabolismo , Mioblastos/citologia , Miogenina/metabolismo , Plastificantes/toxicidade , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Camundongos
8.
Toxicol Lett ; 200(1-2): 67-76, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21040763

RESUMO

Although many studies have demonstrated that aluminum (Al) exposure impairs learning and memory, its underlying mechanism is still uncertain. Long-lasting forms of synaptic plasticity that underlie memory are dependent on new protein synthesis. In particular, activity-regulated cytoskeleton-associated protein (Arc) has a versatile role in synaptic plasticity, and its synthesis can be induced by brain-derived neurotrophic factor (BDNF). BDNF-induced Arc expression has been suggested to play a fundamental role in the stabilization of synaptic plasticity. In the present study, the pretreatment of Al(malt)3 at nonlethal level (200 µM, 24 h) significantly reduced BDNF (10 ng/ml, 1h)-induced Arc expression in SH-SY5Y human neuroblastoma cells. BDNF-induced activation of ERK but not PI3K signaling pathway was interfered with the Al(malt)3 pretreatment, resulting in the subsequent reduction of BDNF-induced phosphorylation of 4EBP1, p70S6K, and eIF4E. Reduced phospho-4EBP1 and phospho-eIF4E hindered the initiation step of translation, which may lead to a reduction in BDNF-induced Arc expression. However, reduced phospho-p70S6K did not influence the phosphorylation of eEF2K and eEF2, indicating no significant effect on BDNF-enhanced translation elongation. Therefore, even at nonlethal level, Al(malt)3 pretreatment reduced BDNF-induced Arc expression, which was caused by interrupting the ERK signaling pathway as well as the subsequent translation initiation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Proteínas do Citoesqueleto/biossíntese , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Proteínas do Tecido Nervoso/biossíntese , Neuroblastoma/metabolismo , Compostos Organometálicos/farmacologia , Pironas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Plasticidade Neuronal/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosforilação
9.
Toxicol Sci ; 116(1): 264-72, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20395307

RESUMO

Aluminum (Al) is a neurotoxicant and is implicated in several neurodegenerative diseases, including Alzheimer's disease (AD). In AD brains, one of the pathological hallmarks is the extracellular deposition of senile plaques, which are mainly composed of aggregated amyloid-beta (Abeta). Endoproteolysis of the amyloid-beta precursor protein (AbetaPP) by the beta-secretase and the gamma-secretase generates Abeta. AbetaPP can also be cleaved by the alpha-secretase within the Abeta region, which releases a soluble fragment sAPPalpha and precludes the formation of Abeta. Al has been reported to increase the level of Abeta, promote Abeta aggregation, and increase Abeta neurotoxicity. In contrast, small G protein Rho and its effector, Rho-associated kinase (ROCK), are known to negatively regulate the amount of Abeta. Inhibition of the Rho-ROCK pathway may underlie the ability of nonsteroidal anti-inflammatory drugs and statins to reduce Abeta production. Whether the Rho-ROCK pathway is involved in Al-induced elevation and aggregation of Abeta is unknown. In the present study, cultured rat cortical neurons were treated with Al(malt)(3) in the absence or presence of ROCK inhibitor Y-27632. After the treatment of Al(malt)(3), the cell viability and the level of sAPPalpha were reduced, whereas the amyloid fibrils in the conditioned media were increased. Treatment with Y-27632 prevented these adverse effects of Al(malt)(3) and thus maintained neuronal survival. These results reveal that the activation of the Rho-ROCK signaling pathway was involved in Al-induced effects in terms of the cell viability, the production of sAPPalpha, and the formation of amyloid fibril, which provides a novel mechanism underlying Al-induced neurotoxicity.


Assuntos
Alumínio/toxicidade , Córtex Cerebral/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Córtex Cerebral/citologia , Meios de Cultivo Condicionados , Neurônios/citologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...