Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 158: 105486, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34450329

RESUMO

Non-convulsive epileptiform activity is a common and under-studied comorbidity of Alzheimer's disease that may significantly contribute to onset of clinical symptoms independently of other neuropathological features such as ß-amyloid deposition. We used repeated treatment with low dose kainic acid (KA) to trigger sub-threshold epileptiform activity in young (less than 6 months) wild-type (WT) and APP/PSEN1 mice to test the role of disruption to the glutamatergic system in epileptiform activity changes and the development of memory deficits. Short-term repeated low-dose KA (five daily treatments with 5 mg/kg, IP) impaired long-term potentiation in hippocampus of APP/PSEN1 but not WT mice. Long-term repeated low-dose KA (fourteen weeks of bi-weekly treatment with 7.5-10 mg/kg) led to high mortality in APP/PSEN1 mice. KA treatment also impaired memory retention in the APP/PSEN1 mice in a Morris water maze task under cognitively challenging reversal learning conditions where the platform was moved to a new location. Four weeks of bi-weekly treatment with 5 mg/kg KA also increased abnormal spike activity in APP/PSEN1 and not WT mice but did not impact sleep/wake behavioral states. These findings suggest that hyperexcitability in Alzheimer's disease may indeed be an early contributor to cognitive decline that is independent of heavy ß-amyloid-plaque load, which is absent in APP/PSEN1 mice under 6 months of age.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Ácido Glutâmico/metabolismo , Homeostase/fisiologia , Presenilina-1/genética , Animais , Eletroencefalografia , Epilepsia/induzido quimicamente , Epilepsia/genética , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Ácido Caínico , Potenciação de Longa Duração , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/psicologia , Camundongos , Camundongos Endogâmicos C57BL , Placa Amiloide/patologia
2.
Genes Brain Behav ; 16(6): 619-626, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28220990

RESUMO

Smith-Lemli-Opitz syndrome is an inherited monogenic disorder in which mutations to the 7-dehydrocholesterol (7-DHC) reductase (Dhcr7) gene lead to deficits in cholesterol synthesis. As a result, many patients suffer from gross physiological and neurological deficits. The purpose of this study was to identify a potential abnormal behavioral phenotype in a compound mutant mouse model for Smith-Lemli-Opitz disease (Dhcr7 Δ3-5/T93M ) to further validate the model and to provide potential targets for future therapeutic interventions. We also sought to identify some of the underlying changes in brain function that may be responsible for behavioral differences among groups. The Dhcr7 compound mutant mice were smaller than their single mutant littermates. Both single and compound heterozygous mice made fewer ultrasonic vocalizations when separated from the dam, which may suggest a communication deficit in these animals. Striking increases of the highly oxidizable 7-DHC were observed in the compound mutant mice. 7-Dehydrocholesterol is the precursor to cholesterol and builds up because of decreased function of the mutated Dhcr7 enzyme. Additionally, several differences were noted in the serotonergic system including increased expression of the serotonin transporter and increased uptake of serotonin by isolated synaptosomes. We propose that changes to the oxidative environment during development can have a significant impact on the development of serotonergic function and that this contributes to behavioral differences observed in the mutant mice.


Assuntos
Estresse Oxidativo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Serotonina/metabolismo , Síndrome de Smith-Lemli-Opitz/genética , Vocalização Animal , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Desidrocolesteróis/metabolismo , Feminino , Heterozigoto , Masculino , Camundongos , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Síndrome de Smith-Lemli-Opitz/metabolismo , Síndrome de Smith-Lemli-Opitz/fisiopatologia , Ondas Ultrassônicas
3.
Pharmacol Biochem Behav ; 106: 101-8, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23541496

RESUMO

Smith-Lemli-Opitz syndrome (SLOS) is a developmental disorder resulting from mutations to the Dhcr7 gene, which is required for cholesterol synthesis. Patients with SLOS typically exhibit a number of severe behavioral deficits and many are diagnosed with autistic spectrum disorder. Although the molecular pathophysiology underlying behavioral changes in SLOS and autism spectrum disorders is poorly understood, there is evidence for the involvement of the serotonergic system in SLOS and autism in general. Behavioral testing was undertaken to ascertain the basal behavioral differences between Dhcr7-heterozygous (HET) and wild-type control mice and explore the utility of a Dhcr7-HET mouse line in the development of new treatments for this disorder. Dhcr7-HET mice did not differ from wild-type control mice on basic measures of locomotor activity, anxiety and neuromuscular ability. However, female Dhcr7-HET mice at 6 months of age or older were significantly more likely to win on the social dominance tube test against an unfamiliar mouse. Pharmacological testing, using the 5-HT2A agonist 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI), showed increased head-twitch response in Dhcr7-HET mice, which was apparent from 6 months of age. No differences were found between the genotypes in testing for 5-HT1A agonist 8-OH-DPAT-induced hypothermia. These data indicate an underlying dysfunction of the 5-HT2A receptors in Dhcr7-HET mice that warrants further investigation to establish how this may relate to behavioral disturbances in human patients carrying Dhcr7 mutations.


Assuntos
Comportamento Animal , Modelos Animais de Doenças , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Serotonina/fisiologia , Síndrome de Smith-Lemli-Opitz/fisiopatologia , Animais , Feminino , Masculino , Camundongos
4.
Free Radic Biol Med ; 49(5): 821-9, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20541602

RESUMO

The sodium-dependent vitamin C transporter (SVCT2) is responsible for the transport of vitamin C into cells in multiple organs, from either the blood or the cerebrospinal fluid. Mice null for SVCT2 (SVCT2(-/-)) do not survive past birth but the cause of death has not yet been ascertained. After mating of SVCT2(+/-) males and SVCT2(+/-) females, fewer SVCT2(-/-) and SVCT2(+/-) progeny were observed than would be expected according to Mendelian ratios. Vitamin C levels in SVCT2(-/-), SVCT2(+/-), and SVCT2(+/+) were genotype-dependent. SVCT2(-/-) fetuses had significantly lower vitamin C levels than littermates in placenta, cortex, and lung, but not in liver (the site of vitamin C synthesis). Low vitamin C levels in placenta and cortex were associated with elevations in several markers of oxidative stress: malondialdehyde, isoketals, F(2)-isoprostanes, and F(4)-neuroprostanes. Oxidative stress was not elevated in fetal SVCT2(-/-) lung tissue despite low vitamin C levels. In addition to the expected severe hemorrhage in cortex, we also found hemorrhage in the brain stem, which was accompanied by cell loss. We found evidence of increased apoptosis in SVCT2(-/-) mice and disruption of the basement membrane in fetal brain. Together these data show that SVCT2 is critical for maintaining vitamin C levels in fetal and placental tissues and that the lack of SVCT2, and the resulting low vitamin C levels, results in fetal death and, in SVCT2(-/-) mice that survive the gestation period, in oxidative stress and cell death.


Assuntos
Ácido Ascórbico/sangue , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Estresse Oxidativo/genética , Simportadores/genética , Animais , Encéfalo/embriologia , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular/genética , Regulação para Baixo/genética , Feminino , Genótipo , Masculino , Malondialdeído/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/fisiologia , Placenta/metabolismo , Placenta/patologia , Gravidez , Transportadores de Sódio Acoplados à Vitamina C , Simportadores/metabolismo , Simportadores/fisiologia , Distribuição Tecidual , Regulação para Cima/genética
5.
Pharmacol Biochem Behav ; 94(4): 543-52, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19941887

RESUMO

Vitamin C is a powerful antioxidant and its levels are decreased in Alzheimer's patients. Even sub-clinical vitamin C deficiency could impact disease development. To investigate this principle we crossed APP/PSEN1 transgenic mice with Gulo knockout mice unable to synthesize their own vitamin C. Experimental mice were maintained from 6 weeks of age on standard (0.33 g/L) or reduced (0.099 g/L) levels of vitamin C and then assessed for changes in behavior and neuropathology. APP/PSEN1 mice showed impaired spatial learning in the Barnes maze and water maze that was not further impacted by vitamin C level. However, long-term decreased vitamin C levels led to hyperactivity in transgenic mice, with altered locomotor habituation and increased omission errors in the Barnes maze. Decreased vitamin C also led to increased oxidative stress. Transgenic mice were more susceptible to the activity-enhancing effects of scopolamine and low vitamin C attenuated these effects in both genotypes. These data indicate an interaction between the cholinergic system and vitamin C that could be important given the cholinergic degeneration associated with Alzheimer's disease.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Deficiência de Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/psicologia , Presenilina-1/genética , Amiloide/metabolismo , Animais , Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/mortalidade , Encéfalo/metabolismo , Encéfalo/patologia , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Feminino , L-Gulonolactona Oxidase/genética , Peroxidação de Lipídeos , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Estresse Oxidativo/fisiologia , Taxa de Sobrevida
6.
Nutr Neurosci ; 12(5): 203-18, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19761651

RESUMO

The present study investigated the relationships among oxidative stress, beta-amyloid and cognitive abilities in the APP/PSEN1 double-transgenic mouse model of Alzheimer's disease. In two experiments, long-term dietary supplements were given to aged APP/PSEN1 mice containing vitamin C alone (1 g/kg diet; Experiment 1) or in combination with a high (750 IU/kg diet, Experiments 1 and 2) or lower (400 IU/kg diet, Experiment 2) dose of vitamin E. Oxidative stress, measured by F(4)-neuroprostanes or malondialdehyde, was elevated in cortex of control-fed APP/PSEN1 mice and reduced to wild-type levels by vitamin supplementation. High-dose vitamin E with C was less effective at reducing oxidative stress than vitamin C alone or the low vitamin E+C diet combination. The high-dose combination also impaired water maze performance in mice of both genotypes. In Experiment 2, the lower vitamin E+C treatment attenuated spatial memory deficits in APP/PSEN1 mice and improved performance in wild-type mice in the water maze. Amyloid deposition was not reduced by antioxidant supplementation in either experiment.


Assuntos
Doença de Alzheimer/metabolismo , Antioxidantes/administração & dosagem , Memória , Estresse Oxidativo , Envelhecimento , Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Antioxidantes/efeitos adversos , Antioxidantes/análise , Ácido Ascórbico/administração & dosagem , Ácido Ascórbico/efeitos adversos , Ácido Ascórbico/análise , Comportamento Animal/fisiologia , Química Encefálica , Cognição , Dieta , Modelos Animais de Doenças , Feminino , Fígado/química , Masculino , Camundongos , Camundongos Transgênicos , Placa Amiloide/metabolismo , Presenilina-1/deficiência , Presenilina-1/genética , Presenilina-1/fisiologia , Vitamina E/administração & dosagem , Vitamina E/efeitos adversos , Vitamina E/análise
7.
Behav Brain Res ; 205(2): 550-8, 2009 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-19703495

RESUMO

Vitamin C (ascorbate) has important antioxidant functions that can help protect against oxidative stress in the brain and damage associated with neurodegenerative disorders such as Alzheimer's disease. When administered parenterally ascorbate can bypass saturable uptake mechanisms in the gut and thus higher tissue concentrations can be achieved than by oral administration. In the present study we show that ascorbate (125 mg/kg) administered intraperitoneally (i.p.) 1-h before testing, partially attenuated scopolamine-induced (1 mg/kg i.p.) cognitive deficits in Morris water maze performance in young mice. Cumulative search error, but not escape latency nor path length, was significantly improved during acquisition in ascorbate plus scopolamine-treated mice although performance did not equal that of control mice. During the probe trial, scopolamine led to increased search error and chance level of time spent in the platform quadrant, whereas mice pre-treated with ascorbate prior to scopolamine did not differ from control mice on these measures. Ascorbate had no effect on unimpaired, control mice and neither did it reduce the peripheral, activity-increasing effects of scopolamine. Ascorbate alone increased acetylcholinesterase activity in the medial forebrain area but had no effect in cortex or striatum. This change, and its action against the amnestic effects of the muscarinic antagonist scopolamine, suggest that ascorbate may be acting in part via altered cholinergic signaling. However, further investigation is necessary to isolate the cognition-enhancing effects of ascorbate.


Assuntos
Ácido Ascórbico/farmacologia , Deficiências da Aprendizagem/tratamento farmacológico , Aprendizagem em Labirinto/efeitos dos fármacos , Nootrópicos/farmacologia , Percepção Espacial/efeitos dos fármacos , Acetilcolinesterase/metabolismo , Animais , Ácido Ascórbico/sangue , Ácido Ascórbico/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/enzimologia , Córtex Cerebral/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/enzimologia , Corpo Estriado/metabolismo , Reação de Fuga/efeitos dos fármacos , Feminino , Deficiências da Aprendizagem/induzido quimicamente , Fígado/efeitos dos fármacos , Fígado/metabolismo , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/enzimologia , Prosencéfalo/metabolismo , Escopolamina , Fatores de Tempo
8.
Pharmacol Biochem Behav ; 93(4): 443-50, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19539642

RESUMO

Alzheimer's disease is a progressive and fatal neurodegenerative disease characterized by a build up of amyloid beta (Abeta) deposits, elevated oxidative stress, and deterioration of the cholinergic system. The present study investigated short-term cognitive-enhancing effects of acute intraperitoneal (i.p.) Vitamin C (ascorbate) treatment in APP/PSEN1 mice, a mouse model of Alzheimer's disease. Middle-aged (12 months) and very old (24 months) APP/PSEN1 bigenic and wild-type mice were treated with ascorbate (125 mg/kg i.p.) or the vehicle 1 h before testing on Y-maze spontaneous alternation and Morris water maze tasks. Very old mice performed more poorly on cognitive tasks than middle-aged mice. Ascorbate treatment improved Y-maze alternation rates and swim accuracy in the water maze in both wild-type and APP/PSEN1 mice. Abeta deposits and oxidative stress both increased with age, and acetylcholinesterase (AChE) activity was significantly reduced in APP/PSEN1 compared to wild-type mice. However, the short course of acute ascorbate treatment did not alter Alzheimer-like neuropathological features of plaque deposition, oxidative stress, or AChE activity. These data suggest that ascorbate may have noötropic functions when administered parenterally in high doses and that the mode of action is via an acute, pharmacological-like mechanism that likely modulates neurotransmitter function.


Assuntos
Envelhecimento/psicologia , Peptídeos beta-Amiloides/genética , Antioxidantes/farmacologia , Ácido Ascórbico/farmacologia , Aprendizagem em Labirinto/efeitos dos fármacos , Presenilina-1/genética , Acetilcolinesterase/metabolismo , Peptídeos beta-Amiloides/fisiologia , Animais , Química Encefálica/efeitos dos fármacos , Malondialdeído/metabolismo , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Presenilina-1/fisiologia
9.
Behav Brain Res ; 198(1): 247-51, 2009 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-18996418

RESUMO

The effects of abnormally high or low stress on learning are well established. The Barnes maze and Morris water maze are two commonly used tests of spatial memory, of which the water maze is considered more stressful; however, until now this has not been demonstrated empirically. In the present study, mice matched for performance on commonly used anxiety tasks were trained on either the Barnes maze or water maze or received no cognitive testing. Water-maze training induced greater increases in plasma corticosterone than did Barnes maze training, assessed 30 min after the final session. Importantly, spatial learning was inversely correlated with corticosterone levels in the water maze but not the Barnes maze, suggesting that performance on the water maze may be more affected by test-induced stress even within wild-type subjects of the same age and gender. These findings are important when considering the appropriate cognitive tasks for any experiment in which stress responses may differ systematically across groups.


Assuntos
Ansiedade/etiologia , Corticosterona/fisiologia , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Estresse Psicológico/etiologia , Análise de Variância , Animais , Ansiedade/fisiopatologia , Ansiedade/psicologia , Cognição/fisiologia , Corticosterona/sangue , Comportamento Exploratório/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Desempenho Psicomotor/fisiologia , Radioimunoensaio , Comportamento Social , Percepção Espacial/fisiologia , Comportamento Espacial/fisiologia , Estresse Psicológico/fisiopatologia , Estresse Psicológico/psicologia , Natação
10.
Genes Brain Behav ; 6(1): 54-65, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17233641

RESUMO

Mice co-expressing the Swedish amyloid precursor protein mutation (APP(Swe)) and exon 9 deletion (DeltaE9) of the PSEN1 gene begin to develop amyloid plaques at 6-7 months of age. We demonstrate here a spatial learning deficit in 7-month-old APP(Swe) + PSEN1DeltaE9 bigenic mice using an adaptation of the Barnes maze. Mice were first trained on a cued target followed by a hidden-target condition. Although bigenic mice quickly learned the cued-target version of the task, they were significantly impaired when switched to the hidden-target version. In contrast, a separate group of double-transgenic mice trained first on the spatial hidden-target version of the task were unimpaired relative to wild-type controls. We propose that processes such as general rule learning, context learning and exploratory habituation exert a greater influence when the testing environment is novel and overshadow the spatial memory deficit in naive bigenic mice. However, when cued-target training is conducted first, these processes habituate and the spatial learning deficit is unmasked. Seven-month-old APP(Swe) + PSEN1DeltaE9 mice were unimpaired on tests of memory that did not involve learning the rules governing spatial associations.


Assuntos
Doença de Alzheimer/metabolismo , Ansiedade/metabolismo , Aprendizagem em Labirinto/fisiologia , Presenilina-1/metabolismo , Comportamento Espacial/fisiologia , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Ansiedade/genética , Encéfalo/metabolismo , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Feminino , Deleção de Genes , Masculino , Camundongos , Camundongos Mutantes Neurológicos , Camundongos Transgênicos , Presenilina-1/genética , Percepção Espacial/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA