Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mar Drugs ; 22(3)2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38535440

RESUMO

The microalgae Phaeodactylum tricornutum (PT) is distinguished by its rich nutrient profile, characterized by well-documented neuroprotective activities, including fucoxanthin (FX), a major carotenoid and polyunsaturated omega-3 fatty acids (n-3 PUFA). The current study aims to evaluate the protective effects of a standardized extract of PT (Mi136) containing 2% FX on cognitive function, oxidative stress, and inflammation parameters in a mouse model of accelerated aging. Seventy-two (72) male mice were randomly assigned to the blank control group (BC), negative control group (NC), and four similar microalgae extract of PT groups (branded as BrainPhyt™) with different human equivalent doses to evaluate potential dose-response effects. From day 01 to day 51, mice in the BC group were injected with a 0.9% normal saline solution, while mice in all other groups were subcutaneously injected with D-galactose (D-Gal) at a dose of 150 mg/kg once per day, five days per week. Results indicated that, for the three higher microalgae extract of PT dose groups, spatial cognitive function, swim latency, and step-through latency impairments induced by chronic D-Gal intoxication were significantly and fully inhibited, with mean values similar to those in the BC group during each day of testing. Similar benefits were observed in biochemical analysis, specifically regarding brain and plasma levels of lipid peroxidation, TNF-α, and IL-6 markers. These data underscore the positive effects of a standardized extract of PT containing 2% FX on cognitive function parameters such as spatial working memory, long-term memory, and short-term memory through the regulation of oxidative stress and inflammation pathways.


Assuntos
Disfunção Cognitiva , Ácidos Graxos Ômega-3 , Microalgas , Humanos , Masculino , Animais , Camundongos , Galactose , Cognição , Inflamação
2.
Front Neurosci ; 14: 563, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32612499

RESUMO

Donepezil (DPZ) is an acetylcholinesterase inhibitor used in Alzheimer's disease to restore cognitive functions but is endowed with limited efficacy. Recent studies pointed out the implication of astroglial networks in cognitive processes, notably via astrocyte connexins (Cxs), proteins involved in gap junction intercellular communications. Hence, we investigated the impact on cognition of pharmacological or genetic modulations of those astrocyte Cxs during DPZ challenge in two rodent models of Alzheimer's disease-like memory deficits. We demonstrated that the Cx modulator mefloquine (MEF) significantly enhanced the procognitive effect of DPZ in both models. In parallel, we determined that MEF potentiated DPZ-induced release of acetylcholine in hippocampus. Finally, local genetic silencing of astrocyte Cxs in the hippocampus was also found to enhance the procognitive effect of DPZ, pointing out the importance of Cx-dependent astrocyte networks in memory processes.

3.
CNS Neurol Disord Drug Targets ; 19(3): 227-240, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32479250

RESUMO

AIM: The aim of this study is to examine the effect of etifoxine on ß-amyloid-induced toxicity models. BACKGROUND: Etifoxine is an anxiolytic compound with a dual mechanism of action; it is a positive allosteric modulator of GABAergic receptors as well as a ligand for the 18 kDa mitochondrial Translocator Protein (TSPO). TSPO has recently raised interest in Alzheimer's Disease (AD), and experimental studies have shown that some TSPO ligands could induce neuroprotective effects in animal models. OBJECTIVE: In this study, we examined the potential protective effect of etifoxine in an in vitro and an in vivo model of amyloid beta (Aß)-induced toxicity in its oligomeric form, which is a crucial factor in AD pathologic mechanisms. METHODS: Neuronal cultures were intoxicated with Aß1-42, and the effects of etifoxine on oxidative stress, Tau-hyperphosphorylation and synaptic loss were quantified. In a mice model, behavioral deficits induced by intracerebroventricular administration of Aß25-35 were measured in a spatial memory test, the spontaneous alternation and in a contextual memory test, the passive avoidance test. RESULTS: In neuronal cultures intoxicated with Aß1-42, etifoxine dose-dependently decreased oxidative stress (methionine sulfoxide positive neurons), tau-hyperphosphorylation and synaptic loss (ratio PSD95/synaptophysin). In a mice model, memory impairments were fully alleviated by etifoxine administered at anxiolytic doses (12.5-50mg/kg). In addition, markers of oxidative stress and apoptosis were decreased in the hippocampus of these animals. CONCLUSION: Our results have shown that in these two models, etifoxine could fully prevent neurotoxicity and pathological changes induced by Aß. These results confirm that TSPO ligands could offer an interesting therapeutic approach to Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Ansiolíticos/uso terapêutico , Oxazinas/uso terapêutico , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/toxicidade , Animais , Apoptose/efeitos dos fármacos , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Masculino , Camundongos , Neurônios/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos
4.
J Alzheimers Dis ; 44(2): 507-24, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25298201

RESUMO

Aftins (amyloid forty-two inducers) represent a novel class of tri-substituted purines derived from roscovitine, able to promote the generation of amyloid-ß (Aß)1-42 from amyloid-ß protein precursor through γ-secretase activation in cell cultures. We here examined whether aftin-4 could provoke an amyloid-like toxicity in vivo in mice. The intracerebroventricular administration of aftin-4 (3-20 nmol) increased Aß1-42, but not Aß1-40, content in the mouse hippocampus, between 5 and 14 days after injection. Aftin-4 injection increased lipid peroxidation levels in the hippocampus, an index of oxidative stress. It increased brain contents in pro-inflammatory cytokines, IL-1ß, IL-6, and TNFα, and GFAP immunolabeling, showing astrocytic reaction. Expression of the synaptic marker synaptophysin was decreased by aftin-4. Finally, the treatment provoked marked learning deficits, observed using different memory procedures: Spontaneous alternation in the Y-maze, place learning in the water-maze, and passive avoidance response. The systemic intraperitoneal injection of aftin-4 in the 3-30 mg/kg dose range also induced oxidative stress and learning deficits. All these alterations could be blocked by pre-treatment with the γ-secretase inhibitor BMS-299,897, confirming that the mechanism of action of aftin-4 involves secretase activity. Furthermore, we examined if the cholinesterase inhibitor donepezil and the non-steroidal anti-inflammatory drug ibuprofen could prevent aftin-4-induced memory impairments, cytokine release, and lipid peroxidation. Donepezil prevented all alterations, whereas ibuprofen prevented the increases in cytokine release and lipid peroxidation, but only marginally the memory impairments. As a whole, this study showed that in vivo injection of aftin-4 results in a rapid, acute Alzheimer's disease-like toxicity in the rodent brain.


Assuntos
Adenina/análogos & derivados , Encefalite/induzido quimicamente , Encefalite/fisiopatologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Adenina/toxicidade , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Butiratos/farmacologia , Inibidores da Colinesterase/farmacologia , Donepezila , Relação Dose-Resposta a Droga , Encefalite/prevenção & controle , Inibidores Enzimáticos/farmacologia , Hidrocarbonetos Halogenados/farmacologia , Ibuprofeno/farmacologia , Indanos/farmacologia , Deficiências da Aprendizagem/induzido quimicamente , Deficiências da Aprendizagem/fisiopatologia , Deficiências da Aprendizagem/prevenção & controle , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/fisiopatologia , Transtornos da Memória/prevenção & controle , Camundongos , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Fragmentos de Peptídeos/metabolismo , Piperidinas/farmacologia
5.
Psychopharmacology (Berl) ; 231(17): 3293-3312, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24481566

RESUMO

RATIONALE: Pregnenolone sulfate (PREGS) and dehydroepiandrosterone sulphate (DHEAS) are pro-amnesic, anti-amnesic and neuroprotective steroids in rodents. In Alzheimer's disease (AD) patient's brains, their low concentrations are correlated with high levels of Aß and tau proteins. The unnatural enantiomer ent-PREGS enhanced memory in rodents. We investigated here whether ent-PREGS and ent-DHEAS could be neuroprotective in AD models. OBJECTIVE: The effects of PREGS, ent-PREGS, DHEAS and ent-DHEAS against Aß25-35 peptide-induced toxicity were examined in vitro on B104 neuroblastoma cells and in vivo in mice. METHODS: B104 cells pretreated with the steroids before Aß25-35 were analysed by flow cytometry measuring cell viability and death processes. Mice injected intracerebroventricularly with Aß25-35 and the steroids were analysed for their memory abilities. Additionally, lipid peroxidation levels in the hippocampus were measured. RESULTS: ent-PREGS and PREGS significantly attenuated the Aß25-35-induced decrease in cell viability. Both steroids prevented the Aß25-35-induced increase in late apoptotic cells. PREGS further attenuated the ratio of necrotic cells. ent-DHEAS and DHEAS significantly reduced the Aß25-35-induced toxicity and prevented the cells from entering late apoptosis and necrosis. All steroids stimulated neurite outgrowth per se and prevented the Aß25-35-induced decrease. In vivo, ent-PREGS and ent-DHEAS significantly attenuated the Aß25-35-induced decrease in memory (spontaneous alternation and passive avoidance) and an increase in lipid peroxidation levels. In contrast to the natural steroids, both enantiomers prevented amnesia when injected 6 h before Aß25-35 in contrast to the natural steroids. CONCLUSION: The unnatural steroids ent-PREGS and ent-DHEAS are potent neuroprotective agents and could be effective therapeutical tools in AD.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/toxicidade , Sulfato de Desidroepiandrosterona/farmacologia , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/toxicidade , Pregnenolona/farmacologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Técnicas In Vitro , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Neuritos/efeitos dos fármacos , Ratos
6.
Front Cell Neurosci ; 8: 463, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25653589

RESUMO

Alzheimer's disease (AD), the most prevalent dementia in the elderly, is characterized by progressive synaptic and neuronal loss. Mitochondrial dysfunctions have been consistently reported as an early event in AD and appear before Aß deposition and memory decline. In order to define a new neuroprotectant strategy in AD targeting mitochondrial alterations, we develop tetrahydro-N,N-dimethyl-2,2-diphenyl-3-furanmethanamine (ANAVEX2-73, AE37), a mixed muscarinic receptor ligand and a sigma-1 receptor (σ1R) agonist. We previously reported that ANAVEX2-73 shows anti-amnesic and neuroprotective activities in mice injected intracerebroventricular (ICV) with oligomeric amyloid-ß25-35 peptide (Aß25-35). The σ1R is present at mitochondria-associated endoplasmic reticulum (ER) membranes, where it acts as a sensor/modulator of ER stress responses and local Ca(2+) exchanges with the mitochondria. We therefore evaluated the effect of ANAVEX2-73 and PRE-084, a reference σ1R agonist, on preservation of mitochondrial integrity in Aß25-35-injected mice. In isolated mitochondria from hippocampus preparations of Aß25-35 injected animals, we measured respiration rates, complex activities, lipid peroxidation, Bax/Bcl-2 ratios and cytochrome c release into the cytosol. Five days after Aß25-35 injection, mitochondrial respiration in mouse hippocampus was altered. ANAVEX2-73 (0.01-1 mg/kg IP) restored normal respiration and PRE-084 (0.5-1 mg/kg IP) increased respiration rates. Both compounds prevented Aß25-35-induced increases in lipid peroxidation levels, Bax/Bcl-2 ratio and cytochrome c release into the cytosol, all indicators of increased toxicity. ANAVEX2-73 and PRE-084 efficiently prevented the mitochondrial respiratory dysfunction and resulting oxidative stress and apoptosis. The σ1R, targeted selectively or non-selectively, therefore appears as a valuable target for protection against mitochondrial damages in AD.

7.
Neuropsychopharmacology ; 38(9): 1706-23, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23493042

RESUMO

The main objective of the present study was to establish whether the mixed σ1/muscarinic ligand ANAVEX2-73, shown to be neuroprotective in Alzheimer's disease (AD) models in vivo and currently in clinical phase I/IIa, could have the ability to reduce the appearance of hyperphosphorylated Tau and amyloid-ß1₋42 (Aß1₋42 in the Aß25₋35 mouse model of AD. We therefore first confirmed that Aß25₋35 injection induced hyperphosphorylation of Tau protein, by showing that it rapidly decreased Akt activity and activated glycogen synthase kinase-3ß (GSK-3ß) in the mouse hippocampus. Second, we showed that the kinase activation, and resulting Tau alteration, directly contributed to the amyloid toxicity, as co-administration of the selective GSK-3ß inhibitor 2-thio(3-iodobenzyl)-5-(1-pyridyl)-[1,3,4]-oxidiazole blocked both Tau phosphorylation and Aß25₋35-induced memory impairments. Third, we analyzed the ANAVEX2-73 effect on Tau phosphorylation and activation of the related kinase pathways (Akt and GSK-3ß). And fourth, we also addressed the impact of the drug on Aß25₋35-induced Aß1₋42 seeding and observed that the compound significantly blocked the increase in Aß1₋42 and C99 levels in the hippocampus, suggesting that it may alleviate amyloid load in AD models. The comparison with PRE-084, a selective and reference σ1 receptor agonist, and xanomeline, a muscarinic ligand presenting similar profile as ANAVEX2-73 on M1 and M2 subtypes, confirmed that both muscarinic and σ1 targets are involved in the ANAVEX2-73 effects. The drug, acting synergistically on both targets, but with moderate affinity, presents a promising pharmacological profile.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/biossíntese , Furanos/farmacologia , Agonistas Muscarínicos/farmacologia , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/biossíntese , Receptores sigma/agonistas , Proteínas tau/metabolismo , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/toxicidade , Animais , Relação Dose-Resposta a Droga , Furanos/uso terapêutico , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Infusões Intraventriculares , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/tratamento farmacológico , Camundongos , Morfolinas/farmacologia , Morfolinas/uso terapêutico , Agonistas Muscarínicos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Oxidiazóis/administração & dosagem , Oxidiazóis/farmacologia , Oxidiazóis/uso terapêutico , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/toxicidade , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/administração & dosagem , Piridinas/farmacologia , Piridinas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Tiadiazóis/farmacologia , Tiadiazóis/uso terapêutico
8.
Neuropsychopharmacology ; 38(5): 817-25, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23303044

RESUMO

Oxidative stress is recognized as one of the earliest and most intense pathological processes in Alzheimer's disease (AD), and the antioxidant vitamin E has been shown to efficiently prevent amyloid plaque formation and neurodegeneration. Plasma phospholipid transfer protein (PLTP) has a major role in vitamin E transfers in vivo, and PLTP deficiency in mice is associated with reduced brain vitamin E levels. To determine the impact of PLTP on amyloid pathology in vivo, we analyzed the vulnerability of PLTP-deficient (PLTP-KO) mice to the toxic effects induced by intracerebroventricular injection of oligomeric amyloid-ß 25-35 (Aß 25-35) peptide, a non-transgenic model of AD. Under basal conditions, PLTP-KO mice showed increased cerebral oxidative stress, increased brain Aß 1-42 levels, and a lower expression of the synaptic function marker synaptophysin, as compared with wild-type mice. This PLTP-KO phenotype was associated with increased memory impairment 1 week after Aß25-35 peptide injection. Restoration of brain vitamin E levels in PLTP-KO mice through a chronic dietary supplementation prevented Aß 25-35-induced memory deficits and reduced cerebral oxidative stress and toxicity. We conclude that PLTP, through its ability to deliver vitamin E to the brain, constitutes an endogenous neuroprotective agent. Increasing PLTP activity may offer a new way to develop neuroprotective therapies.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/genética , Fragmentos de Peptídeos/toxicidade , Proteínas de Transferência de Fosfolipídeos/deficiência , Análise de Variância , Animais , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Fosfatidilcolinas/metabolismo , Reconhecimento Psicológico/efeitos dos fármacos , Esfingomielinas/metabolismo , alfa-Tocoferol/metabolismo
9.
Eur J Pharmacol ; 698(1-3): 193-9, 2013 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-23123349

RESUMO

Alzheimer's disease pathomimetic toxicity could be induced in mice within one week after the intracerebroventricular (i.c.v.) injection of an aggregated preparation of the highly toxic and endogenous amyloid-ß fragment Aß(25-35). It was recently reported that Aß(25-35) also provokes a modification of APP processing with accumulation of endogenous Aß(1-42). We here analyzed whether a γ-secretase inhibitor, BMS-299897, attenuated this Aß(25-35)-induced Aß(1-42) seeding and toxicity. The compound was administered at 0.1-1 nmol/mouse, concomittantly with Aß(25-35) (9 nmol) in male Swiss mice. After one week, the contents in Aß(1-42) and Aß(1-40), and the levels in lipid peroxidation were analyzed in the mouse hippocampus. Mice were submitted to spontaneous alternation, passive avoidance and object recognition to analyze their short- and long-term memory abilities. Aß(25-35) increased Aß(1-42) content (+240%) but failed to affect Aß(1-40). BMS-299897 blocked the increase in Aß(1-42) content and decreased Aß(1-40) levels significantly. The compound did not affect Aß(25-35)-induced increase in hippocampal lipid peroxidation. Behaviorally, BMS-299897 blocked the Aß(25-35)-induced deficits in spontaneous alternation or novel object recognition, using a 1h intertrial time interval. BMS-299896 failed to affect the passive avoidance impairments or novel object recognition, using a 24h intertrial time interval. These results confirmed that Aß(25-35) injection provoked an accumulation in endogenous Aß(1-42), an effect blocked by γ-secretase inhibition. This Aß(1-42) accumulation marginally contributed to the toxicity or long-term memory deficits. However, since the seeded Aß(1-42) affected short-term memory, the rapid Aß(25-35) injection Alzheimer's disease model could be used to screen the activity of new secretase inhibitors.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Peptídeos beta-Amiloides/biossíntese , Peptídeos beta-Amiloides/farmacologia , Butiratos/farmacologia , Hidrocarbonetos Halogenados/farmacologia , Transtornos da Memória/tratamento farmacológico , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/farmacologia , Inibidores de Proteases/farmacologia , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Butiratos/uso terapêutico , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Hidrocarbonetos Halogenados/uso terapêutico , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/metabolismo , Transtornos da Memória/fisiopatologia , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Inibidores de Proteases/uso terapêutico , Fatores de Tempo
10.
J Pharmacol Sci ; 115(3): 279-92, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21427517

RESUMO

Selective agonists of the sigma-1 (σ(1)) ligand-operated chaperone protein, like igmesine or PRE-084, are antidepressants in preclinical depression models. σ(1)-Protein activation may contribute to the antidepressant efficacy of drugs known to act as selective serotonin-reuptake inhibitors (SSRI) or noradrenaline reuptake inhibitors through direct or indirect involvement of the σ(1)-receptor in the drug effect. We here compared antidepressant effects in two behavioral procedures, the forced swimming test (FST) and conditioned fear stress (CFS). The involvement of the σ(1)-receptor was examined using a co-treatment with the σ(1)-antagonist BD1047 or using σ(1)-knockout (KO) mice. Igmesine but not PRE-084 decreased FST immobility. The SSRI fluoxetine and sertraline, but not fluvoxamine, and the tricyclic antidepressants imipramine, desipramine, and amitriptyline were also effective. Only the effect of igmesine was blocked by BD1047 or in σ(1)-KO mice. Igmesine, PRE-084, fluvoxamine, and sertraline decreased the CFS immobility in a BD1047- and σ(1)-KO-sensitive manner. Among tricyclics, only amitriptyline was effective and its effect was unaffected by BD1047 or in σ(1)-KO mice. The behavioral effects induced by mixed σ(1)-receptor/SSRI antidepressants, like fluvoxamine or sertraline, may therefore involve a non-selective action at both targets. Moreover, the CFS appears to more reliably uncover a σ(1) pharmacological component in antidepressant screening.


Assuntos
Inibidores da Captação Adrenérgica/farmacologia , Antidepressivos Tricíclicos/farmacologia , Antidepressivos/farmacologia , Comportamento Animal/efeitos dos fármacos , Depressão/tratamento farmacológico , Receptores sigma/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Animais , Etilenodiaminas/farmacologia , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Receptores sigma/agonistas , Receptores sigma/antagonistas & inibidores , Estresse Psicológico , Natação
11.
J Psychopharmacol ; 25(8): 1101-17, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20829307

RESUMO

Tetrahydro-N, N-dimethyl-2, 2-diphenyl-3-furanmethanamine hydrochloride (ANAVEX2-73) binds to muscarinic acetylcholine and sigma(1) (σ(1)) receptors with affinities in the low micromolar range. We characterized its anti-amnesic and neuroprotective potentials in pharmacological and pathological amnesia models. Spatial working memory was evaluated using spontaneous alternation in the Y-maze and non-spatial memory using passive avoidance procedures. ANAVEX2-73 (0.01-3.0 mg/kg i.p.) alleviated the scopolamine- and dizocilpine-induced learning impairments. ANAVEX2-73 (300 µg/kg) also reversed the learning deficits in mice injected with Aß(25-35) peptide, a non-transgenic Alzheimer's disease model. When the drug was injected simultaneously with Aß(25-35), 7 days before the tests, it blocked the appearance of learning impairments. This protective activity was confirmed since ANAVEX2-73 blocked the Aß(25-35)-induced oxidative stress in the hippocampus. This effect was differentially sensitive to the muscarinic receptor antagonist scopolamine or the σ(1) protein antagonist BD1047, confirming the mixed muscarinic/σ(1) pharmacological action. Finally, its unique demethyl metabolite, ANAVEX19-144, was also effective and ANAVEX2-73 presented a longer duration of action, effective 12 h before Aß(25-35), than its related compound ANAVEX1-41. The neuroprotective activity of ANAVEX2-73, its mixed cholinergic/σ(1) activity, its low active dose range and its long duration of action together reinforce its therapeutic potential in Alzheimer's disease.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Preparações de Ação Retardada/farmacologia , Receptores Muscarínicos/metabolismo , Receptores sigma/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/toxicidade , Animais , Aprendizagem da Esquiva , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Furanos/metabolismo , Furanos/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Ligantes , Masculino , Memória de Curto Prazo/efeitos dos fármacos , Camundongos , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Ligação Proteica
12.
Neuropsychopharmacology ; 34(6): 1552-66, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19052542

RESUMO

The antiamnesic and neuroprotective activities of the new aminotetrahydrofuran derivative tetrahydro-N,N-dimethyl-5,5-diphenyl-3-furanmethanamine hydrochloride (ANAVEX1-41), a nonselective muscarinic receptor ligand and sigma1 protein activator, were examined in mice injected intracerebroventricularly with amyloid beta(25-35) (Abeta(25-35)) peptide (9 nmol). Abeta(25-35) impaired significantly spontaneous alternation performance, a spatial working memory, and passive avoidance response. When ANAVEX1-41 (1-1000 microg/kg i.p.) was administered 7 days after Abeta(25-35), ie, 20 min before the behavioral tests, it significantly reversed the Abeta(25-35)-induced deficits, the most active doses being in the 3-100 microg/kg range. When the compound was preadministered 20 min before Abeta(25-35), ie, 7 days before the tests, it prevented the learning impairments at 30-100 microg/kg. Morphological analysis of corticolimbic structures showed that Abeta(25-35) induced a significant cell loss in the CA1 pyramidal cell layer of the hippocampus that was prevented by ANAVEX1-41 (100 microg/kg). Increased number of glial fibrillary acidic protein immunopositive cells in the retrosplenial cortex or throughout the hippocampus revealed an Abeta(25-35)-induced inflammation that was prevented by ANAVEX1-41. The drug also prevented the parameters of Abeta(25-35)-induced oxidative stress measured in hippocampus extracts, ie, the increases in lipid peroxidation and protein nitration. ANAVEX1-41, however, failed to prevent Abeta(25-35)-induced caspase-9 expression. The compound also blocked the Abeta(25-35)-induced caspase-3 expression, a marker of apoptosis. Both the muscarinic antagonist scopolamine and the sigma1 protein inactivator BD1047 prevented the beneficial effects of ANAVEX1-41 (30 or 100 microg/kg) against Abeta(25-35)-induced learning impairments, suggesting that muscarinic and sigma1 targets are involved in the drug effect. A synergic effect could indeed account for the very low active doses measured in vivo. These data outline the therapeutic potential of ANAVEX1-41 as a neuroprotective agent in Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Aprendizagem da Esquiva/efeitos dos fármacos , Furanos/uso terapêutico , Deficiências da Aprendizagem/tratamento farmacológico , Transtornos da Memória/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Fragmentos de Peptídeos/toxicidade , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Caspase 3/metabolismo , Caspase 9/metabolismo , Morte Celular/efeitos dos fármacos , Etilenodiaminas/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Deficiências da Aprendizagem/induzido quimicamente , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Transtornos da Memória/induzido quimicamente , Camundongos , Antagonistas Muscarínicos/farmacologia , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Escopolamina/farmacologia , Percepção Espacial/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...