Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Behav Brain Res ; 378: 112303, 2020 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-31622640

RESUMO

Although one of the defining characteristics of Alzheimer's disease is the presence of amyloid-beta (Aß) plaques, the early accumulation of soluble Aß oligomers (AßOs) may disrupt synaptic function and trigger cognitive impairments long before the appearance of plaques. Furthermore, murine models aimed at understanding how AßOs alter formation and retrieval of associative memories are conducted using human Aß species, which are more neurotoxic in the mouse brain than the native murine species. Unfortunately, there is currently a lack of attention in the literature as to what the murine version of the peptide (mAß) does to synaptic function and how it impacts the consolidation and retrieval of associative memories. In the current study, adult mice were infused with mAß 0, 2, 6, or 46 h after contextual-fear conditioning, and were tested 2-48 h later. Interestingly, only mAß infusions within 2 h of training reduced freezing behavior at test, indicating that mAß disrupted the consolidation, but not retrieval of fear memory. This consolidation deficit coincided with increased IL-1ß and reduced synaptophysin mRNA levels, without disrupting other synaptic signaling-related genes here examined. Despite differences between murine and human Aß, the deleterious functional outcomes of early-stage synaptic oligomer presence are similar. Thus, models utilizing or inducing the production of mAß in non-transgenic animals are useful in exploring the role of dysregulated synaptic plasticity and resultant learning deficits induced by Aß oligomers.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Comportamento Animal/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Medo/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Inflamação/induzido quimicamente , Consolidação da Memória/efeitos dos fármacos , Rememoração Mental/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/administração & dosagem , Animais , Modelos Animais de Doenças , Hipocampo/imunologia , Hipocampo/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Infusões Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
Behav Brain Res ; 379: 112366, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31743728

RESUMO

Research has demonstrated that stress can exacerbate AD pathology in transgenic mouse models of AD. The purpose of the present studies was to extend this work by determining whether a social stressor, isolation stress, would increase the number of Aß plaques in 5xFAD + transgenic mice in comparison to group-housed controls, and accelerate the onset of cognitive deficits in contextual fear-conditioning. Additionally, we aimed to determine whether the pathological impact of isolation stress could be prevented through exposure to exercise alone or to exercise and an enriched environment throughout the isolation period. Two-month-old 5xFAD + and 5xFAD- animals were isolated or group-housed for two and three months. An additional subset of 5xFAD + mice were housed in isolation, housed in isolation with an exercise wheel, or housed in isolation with an exercise wheel and an enriched environment. Both two and three months of isolation stress significantly increased the number of plaques in the hippocampus of 5xFAD + mice, and three months of isolation increased hippocampal BACE1 expression. Isolated animals also displayed a significant cognitive deficit in contextual fear-conditioning, independent of genotype. Furthermore, neither exercise nor an enriched environment were able to prevent these isolation-induced effects. Understanding how stress impacts the onset and progression of AD is critical, as many individuals endure significant stress over their lifespan, including prolonged social isolation, a societal trend likely to worsen with time.


Assuntos
Doença de Alzheimer , Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Disfunção Cognitiva , Hipocampo/metabolismo , Condicionamento Físico Animal/fisiologia , Placa Amiloide/metabolismo , Isolamento Social , Estresse Psicológico , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/metabolismo , Animais , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/prevenção & controle , Modelos Animais de Doenças , Meio Ambiente , Abrigo para Animais , Masculino , Camundongos , Camundongos Transgênicos , Corrida/fisiologia , Estresse Psicológico/complicações , Estresse Psicológico/metabolismo , Estresse Psicológico/prevenção & controle
3.
Behav Brain Res ; 359: 871-877, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30031883

RESUMO

Alzheimer's disease is marked by the presence of amyloid-beta (Aß) plaques, elevated central cytokine levels, dysregulation of BDNF-related gene expression, and cognitive decline. Previously, our laboratory has demonstrated that repeated administration of peripheral LPS is sufficient to significantly increase the presence of central Aß in the hippocampus, and that this upregulation corresponds with deficits in learning and memory. We have also previously demonstrated that the inverse benzodiazepine agonist MRK-016 (MRK) can protect against memory acquisition and consolidation errors in mice. To extend these findings, the current study explored the protective effects of MRK in the context of LPS-induced hippocampal Aß accumulation. Hippocampal Aß was significantly elevated, relative to saline-treated animals, following seven days of peripheral LPS injections. Animals were then trained in a contextual fear conditioning paradigm and were immediately treated with MRK or saline once training was complete. Behavioral testing occurred the day after training. Results from this study demonstrate that repeated injections of LPS significantly elevate hippocampal Aß, and inhibit acquisition of contextual fear. Post-training treatment with MRK restored behavioral expression of fear in LPS-treated animals, despite elevated hippocampal Aß, an effect that may be attributed to increased BDNF mRNA expression. Therefore, our data indicate that MRK can prevent LPS- induced cognitive deficits associated with elevated Aß, and restore hippocampal BDNF expression.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Transtornos Cognitivos/prevenção & controle , Agonistas GABAérgicos/uso terapêutico , Hipocampo/metabolismo , Isoxazóis/uso terapêutico , Triazinas/uso terapêutico , Peptídeos beta-Amiloides/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/genética , Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/patologia , Condicionamento Psicológico/efeitos dos fármacos , Medo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Memória/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , RNA Mensageiro/metabolismo
4.
Behav Brain Res ; 313: 219-225, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27449203

RESUMO

Alzheimer's disease is marked by the accumulation of the amyloid-beta (Aß) peptide, and increases in phosphorylation of the microtubule associated protein, tau. Changes in these proteins are considered responsible, in part, for the progressive neuronal degeneration and cognitive deficits seen in AD. We examined the effect of repeated consecutive peripheral poly I:C injections on cognitive deficits, central Aß, and phosphorylated tau accumulation, following three treatment durations: 7, 14, and 21 days. Forty-eight hours after the final injection, animals were trained in a contextual fear-conditioning paradigm, and tested 24h later. Immediately after testing, the hippocampus was collected to quantify Aß and phosphorylated tau accumulation. Results showed that, although poly I:C-induced Aß was significantly elevated at all time points examined, poly I:C only disrupted cognition after 14 and 21 days of administration. Moreover, elevations in phosphorylated tau were not seen until the 14-day time point. Interestingly, phosphorylated tau expression then declined at the 21-day time point. Finally, we demonstrated that Aß levels are a stronger predictor of cognitive dysfunction, explaining 37% of the variance, whereas phosphorylated tau levels only accounted for 0.2%. Taken together, these results support the hypothesis that inflammation-induced elevation in Aß disrupts cognition, independently of phosphorylated tau, and suggest that long-term administration of poly I:C may provide a model to investigate the contribution of long-term inflammation toward the development of Alzheimer's-like pathology.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Transtornos Cognitivos/metabolismo , Disfunção Cognitiva/metabolismo , Hipocampo/metabolismo , Poli C/farmacologia , Proteínas tau/metabolismo , Animais , Cognição/efeitos dos fármacos , Cognição/fisiologia , Transtornos Cognitivos/diagnóstico , Transtornos Cognitivos/tratamento farmacológico , Disfunção Cognitiva/diagnóstico , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Fosforilação , Poli C/administração & dosagem
5.
Neuroscience ; 331: 72-7, 2016 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-27320209

RESUMO

For years, the prevailing hypothesis for Alzheimer's Disease (AD) has proposed a mechanism by which deposition of amyloid-beta (Aß) in the brain is independent of tau-pathologies and cognitive decline. However, despite extensive research on the disease, the mechanisms underlying the etiology of tau-pathology remain unknown. Previous research in our lab has shown that imatinib methanesulfonate (IM) blocks the peripheral production of Aß in response to LPS, thereby preventing the buildup of Aß in the hippocampus, and rescuing the cognitive dysfunction that normally follows. The present study aimed to examine the link between Aß and tau following inflammation, and to expand our understanding of how IM affects AD pathology. Specifically, we hypothesized that the IM-mediated inhibition of Aß production following inflammation would successfully protect against the hyperphosphorylation of tau (ptau). Here we show that 7days of LPS treatment in male C57BL/6J mice, which normally produces elevations in peripheral and central Aß, also produces hyperphosphorylation of tau. However, just as pre-treatment and concurrent treatment with IM blocks Aß production, it also blocks the phosphorylation of tau. In addition, 7days of LPS-induced inflammation and Aß production also leads to elevated total tau protein expression. Our results may provide support for the hypothesis that enhanced expression of tau following LPS administration is a protective measure by hippocampal neurons to compensate for the loss of the microtubule-stabilizing protein due to phosphorylation. More importantly, our results support the hypothesis that blocking the production of Aß that follows inflammation also leads to reduced tau phosphorylation, lending credence to a model in which Aß initiates tau phosphorylation.


Assuntos
Hipocampo/efeitos dos fármacos , Hipocampo/imunologia , Mesilato de Imatinib/farmacologia , Proteínas tau/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Western Blotting , Peso Corporal/efeitos dos fármacos , Modelos Animais de Doenças , Lipopolissacarídeos , Masculino , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia
6.
Behav Brain Res ; 302: 171-4, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26778788

RESUMO

Peripheral administration of lipopolysaccharide (LPS) elevates production of pro-inflammatory cytokines, and motivates the expression of sickness behaviors. In this study, we tested the ability of an LPS-derived adjuvant, monophosphoryl lipid A (MPLA), to prevent LPS-induced sickness behaviors in a burrowing paradigm. Testing occurred over a three-day period. Animals received a single injection of either MPLA or saline the first two days of testing. On day three, animals received either LPS or saline. Tissue from the dorsal hippocampus was collected for qRT-PCR to assess expression of IL-1ß and IL-4. Results indicate that, during the pre-treatment phase, administration of MPLA induces an immune response sufficient to trigger sickness behaviors. However, we observed that animals pre-treated with MPLA for two days were resistant to LPS-induced sickness behaviors on day three. Results from the qRT-PCR analysis indicated that LPS-treated animals pre-treated with MPLA expressed significantly less IL-1ß compared to LPS-treated animals pre-treated with saline. However, we did not observe a significant difference in IL-4 expression between groups. Therefore, results indicate that under the given parameters of the study, MPLA pre-treatment protects against LPS-induced sickness behaviors, at least in part, by decreasing expression of the pro-inflammatory cytokine IL-1ß.


Assuntos
Adjuvantes Imunológicos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Comportamento de Doença/efeitos dos fármacos , Interleucina-1beta/metabolismo , Lipídeo A/análogos & derivados , Adjuvantes Imunológicos/administração & dosagem , Animais , Modelos Animais de Doenças , Esquema de Medicação , Interleucina-1beta/genética , Interleucina-4/genética , Interleucina-4/metabolismo , Lipídeo A/administração & dosagem , Lipídeo A/farmacologia , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo
7.
Behav Brain Res ; 288: 50-3, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25823763

RESUMO

Recent evidence suggests that inflammation-induced decrements in cognitive function can be mitigated via manipulation of excitatory or inhibitory transmission. We tested the ability of the inverse benzodiazepine agonist, MRK-016 (MRK) to protect against LPS-induced deficits in memory acquisition and consolidation, using a contextual fear conditioning (CFC) paradigm. In Experiment One, mice received lipopolysaccharide (LPS) and/or MRK injections prior to CFC training, and were then tested 24h after training. In Experiment Two, animals received similar treatment injections immediately after training, and were tested 24h later. Additionally, hippocampal samples were collected 4h after LPS injections and immediately after testing, to evaluate brain-derived neurotrophic factor (BDNF) and insulin-like growth factor 1 (IGF-1) mRNA expression. Results indicate that MRK can protect against LPS-induced learning/memory decrements in both paradigms. We also found, in both paradigms, that animals treated with LPS/Saline expressed significantly less BDNF mRNA when compared to Saline/Saline-treated animals 4h after LPS administration, but that MRK did not restore BDNF expression levels. Further, treatment administrations had no effect on IGF-1 mRNA expression at any collection time-point. In summary, MRK-016 can protect against LPS-induced deficits in memory acquisition and consolidation, in this hippocampus-dependent paradigm, though this protection occurs independently of recovery of BDNF expression.


Assuntos
Inflamação/tratamento farmacológico , Isoxazóis/farmacologia , Aprendizagem/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Memória/efeitos dos fármacos , Nootrópicos/farmacologia , Triazinas/farmacologia , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Medo/efeitos dos fármacos , Medo/fisiologia , Reação de Congelamento Cataléptica/efeitos dos fármacos , Reação de Congelamento Cataléptica/fisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Inflamação/metabolismo , Inflamação/psicologia , Fator de Crescimento Insulin-Like I/metabolismo , Aprendizagem/fisiologia , Masculino , Memória/fisiologia , Camundongos Endogâmicos C57BL , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/fisiologia , RNA Mensageiro/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA