Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 14(6): 387, 2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37386014

RESUMO

Wolfram syndrome (WS) is a rare neurodegenerative disorder encompassing diabetes mellitus, diabetes insipidus, optic atrophy, hearing loss (HL) as well as neurological disorders. None of the animal models of the pathology are presenting with an early onset HL, impeding the understanding of the role of Wolframin (WFS1), the protein responsible for WS, in the auditory pathway. We generated a knock-in mouse, the Wfs1E864K line, presenting a human mutation leading to severe deafness in affected individuals. The homozygous mice showed a profound post-natal HL and vestibular syndrome, a collapse of the endocochlear potential (EP) and a devastating alteration of the stria vascularis and neurosensory epithelium. The mutant protein prevented the localization to the cell surface of the Na+/K+ATPase ß1 subunit, a key protein for the maintenance of the EP. Overall, our data support a key role of WFS1 in the maintenance of the EP and the stria vascularis, via its binding partner, the Na+/K+ATPase ß1 subunit.


Assuntos
Surdez , Síndrome de Wolfram , Animais , Humanos , Camundongos , Adenosina Trifosfatases , Membrana Celular , Epitélio , Síndrome de Wolfram/genética
2.
Toxicology ; 480: 153319, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36100137

RESUMO

Fluoronitrile gas (C4F7N, CAS number 42532-60-5) is one of the most promising candidates as insulating and/or breaking medium in high and medium voltage electrical equipment. Besides its promising properties, C4F7N gas is however not devoid of acute toxicity when used pure or in gas mixtures. The toxicity was not extensively analyzed and reported. The aim of the present study was to analyze in mice the consequences of a single exposure to C4F7N gas, at different concentrations and different timepoints after exposure. Male and female Swiss mice were exposed to breathable air or C4F7N gas, at 800 ppmv or 1500 ppmv, for 4 h on day 0. Behavioral tests (spontaneous alternation in the Y-maze and object recognition) were performed on days 1, 7 and 14 to assess memory alterations. The animals were then sacrificed and their brains dissected for biochemical analyses or fixed with paraformaldehyde for histology and immunohistochemistry. Results showed behavioral impairments and memory deficits, with impairments of alternation at days 1 and 7 and object recognition at day 14. Histological alterations of pyramidal neuronal layer in the hippocampus, neuroinflammatory astroglial reaction, and microglial alterations were observed, more marked in female than male mice. Moreover, the biochemical analyses done in the brain of 1500 ppmv exposed female mice showed a reductive stress with decreased lipid peroxidation and release of cytochrome c, leading to apoptosis with increases in caspase-9 cleavage and γ-H2AX/H2AX ratio. Finally, electrophysiological analyses using a multi-electrode array allowed the measure of the extracellular activity of pyramidal neurons in the CA2 area and revealed that exposure to the gas not only prevented the induction of long-term potentiation but even provoked an epileptoid-like activity in some neurons suggesting major alterations of synaptic plasticity. This study therefore showed that an acute exposure of mice to C4F7N gas provoked, particularly in female animals, memory alterations and brain toxicity characterized by a reductive stress, microglial toxicity, loss of synaptic plasticity and apoptosis. Its use in industrial installations must be done with extreme caution.


Assuntos
Citocromos c , Síndromes Neurotóxicas , Animais , Encéfalo/patologia , Caspase 9 , Feminino , Hipocampo/patologia , Masculino , Transtornos da Memória/patologia , Camundongos , Plasticidade Neuronal/fisiologia , Síndromes Neurotóxicas/patologia
3.
Chemistry ; 28(39): e202200786, 2022 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-35621167

RESUMO

Flavonoids are polyphenolic natural products and have shown significant potential as disease-modifying agents against neurodegenerative disorders like Alzheimer's disease (AD), with activities even in vivo. Hybridization of the natural products taxifolin and silibinin with cinnamic acid led to an overadditive effect of these compounds in several phenotypic screening assays related to neurodegeneration and AD. Therefore, we have exchanged the flavonoid part of the hybrids with different flavonoids, which show higher efficacy than taxifolin or silibinin, to improve the activity of the respective hybrids. Chemical connection between the flavonoid and cinnamic acid was realized by an amide instead of a labile ester bond to improve stability towards hydrolysis. To investigate the influence of a double bond at the C-ring of the flavonoid, the dehydro analogues of the respective hybrids were also synthesized. All compounds obtained show neuroprotection against oxytosis, ferroptosis and ATP-depletion, respectively, in the murine hippocampal cell line HT22. Interestingly, the taxifolin and the quercetin derivatives are the most active compounds, whereby the quercetin derivate shows even more pronounced activity than the taxifolin one in all assays applied. As aimed for, no hydrolysis product was found in cellular uptake experiments after 4 h whereas different metabolites were detected. Furthermore, the quercetin-cinnamic acid amide showed pronounced activity in an in vivo AD mouse model at a remarkably low dose of 0.3 mg/kg.


Assuntos
Doença de Alzheimer , Produtos Biológicos , Doença de Alzheimer/tratamento farmacológico , Amidas , Animais , Cinamatos , Flavonoides/química , Flavonoides/farmacologia , Camundongos , Quercetina , Silibina
4.
Eur J Med Chem ; 228: 114038, 2022 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-34902734

RESUMO

The sigma-1 (σ1) receptor plays a significant role in many normal physiological functions and pathological disease states, and as such represents an attractive therapeutic target for both agonists and antagonists. Here, we describe a novel series of phenoxyalkylpiperidines based on the lead compound 1-[ω-(4-chlorophenoxy)ethyl]-4-methylpiperidine (1a) in which the degree of methylation at the carbon atoms alpha to the piperidine nitrogen was systematically varied. The affinity at σ1 and σ2 receptors and at Δ8-Δ7 sterol isomerase (SI) ranged from subnanomolar to micromolar Ki values. While the highest-affinity was displayed at the σ1, the increase of the degree of methylation in the piperidine ring progressively decreased the affinity. The subnanomolar affinity 1a and 1-[ω-(4-methoxyphenoxy)ethyl]-4-methylpiperidine (1b) displayed potent anti-amnesic effects associated with σ1 receptor agonism, in two memory tests. Automated receptor-small-molecule ligand docking provided a molecular structure-based rationale for the agonistic effects of 1a and 1b. Overall, the class of the phenoxyalkylpiperidines holds potential for the development of high affinity σ1 receptor agonists, and compound 1a, that appears as the best in class (exceeding by far the activity of the reference compound PRE-084) deserves further investigation.


Assuntos
Amnésia/tratamento farmacológico , Desenvolvimento de Medicamentos , Piperidinas/farmacologia , Receptores sigma/antagonistas & inibidores , Amnésia/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Cobaias , Humanos , Ligantes , Masculino , Camundongos , Modelos Moleculares , Estrutura Molecular , Piperidinas/síntese química , Piperidinas/química , Ratos , Ratos Wistar , Receptores sigma/metabolismo , Relação Estrutura-Atividade , Células Tumorais Cultivadas , Receptor Sigma-1
5.
Int J Neuropsychopharmacol ; 24(2): 142-157, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-32977336

RESUMO

BACKGROUND: Current therapies in Alzheimer's disease (AD), including Memantine, have proven to be only symptomatic but not curative or disease modifying. Fluoroethylnormemantine (FENM) is a structural analogue of Memantine, functionalized with a fluorine group that allowed its use as a positron emission tomography tracer. We here analyzed FENM neuroprotective potential in a pharmacological model of AD compared with Memantine. METHODS: Swiss mice were treated intracerebroventricularly with aggregated Aß 25-35 peptide and examined after 1 week in a battery of memory tests (spontaneous alternation, passive avoidance, object recognition, place learning in the water-maze, topographic memory in the Hamlet). Toxicity induced in the mouse hippocampus or cortex was analyzed biochemically or morphologically. RESULTS: Both Memantine and FENM showed symptomatic anti-amnesic effects in Aß 25-35-treated mice. Interestingly, FENM was not amnesic when tested alone at 10 mg/kg, contrarily to Memantine. Drugs injected once per day prevented Aß 25-35-induced memory deficits, oxidative stress (lipid peroxidation, cytochrome c release), inflammation (interleukin-6, tumor necrosis factor-α increases; glial fibrillary acidic protein and Iba1 immunoreactivity in the hippocampus and cortex), and apoptosis and cell loss (Bcl-2-associated X/B-cell lymphoma 2 ratio; cell loss in the hippocampus CA1 area). However, FENM effects were more robust than observed with Memantine, with significant attenuations vs the Aß 25-35-treated group. CONCLUSIONS: FENM therefore appeared as a potent neuroprotective drug in an AD model, with a superior efficacy compared with Memantine and an absence of direct amnesic effect at higher doses. These results open the possibility to use the compound at more relevant dosages than those actually proposed in Memantine treatment for AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Amnésia/tratamento farmacológico , Memantina/análogos & derivados , Memantina/farmacologia , Transtornos da Memória/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/prevenção & controle , Amnésia/induzido quimicamente , Amnésia/prevenção & controle , Peptídeos beta-Amiloides/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Masculino , Memantina/administração & dosagem , Transtornos da Memória/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/administração & dosagem , Fragmentos de Peptídeos/farmacologia
7.
Hum Mol Genet ; 29(4): 529-540, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-31696229

RESUMO

Sigma-1 receptor (S1R) is an endoplasmic reticulum (ER) chaperone that not only regulates mitochondrial respiration but also controls cellular defense against ER and oxidative stress. This makes S1R a potential therapeutic target in amyotrophic lateral sclerosis (ALS). Especially, as a missense mutation E102Q in S1R has been reported in few familial ALS cases. However, the pathogenicity of S1RE102Q and the beneficial impact of S1R in the ALS context remain to be demonstrated in vivo. To address this, we generated transgenic Drosophila that expresses human wild-type S1R or S1RE102Q. Expression of mutant S1R in fly neurons induces abnormal eye morphology and locomotor defects in a dose-dependent manner. This was accompanied by abnormal mitochondrial fragmentation, reduced adenosine triphosphate (ATP) levels and a higher fatigability at the neuromuscular junction during high energy demand. Overexpressing IP3 receptor or glucose transporter mitigates the S1RE102Q-induced eye phenotype, further highlighting the role of calcium and energy metabolism in its toxicity. More importantly, we showed that wild-type S1R rescues locomotor activity and ATP levels of flies expressing the key ALS protein, TDP43. Moreover, overexpressing wild-type S1R enhances resistance of flies to oxidative stress. Therefore, our data provide the first genetic evidence that mutant S1R recapitulates ALS pathology in vivo while increasing S1R confers neuroprotection against TDP43 toxicity.


Assuntos
Esclerose Lateral Amiotrófica/genética , Receptores sigma/genética , Receptores sigma/metabolismo , Animais , Animais Geneticamente Modificados/genética , Modelos Animais de Doenças , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Retículo Endoplasmático/metabolismo , Locomoção/efeitos dos fármacos , Mitocôndrias/metabolismo , Neurônios Motores/metabolismo , Mutação/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Receptor Sigma-1
8.
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
9.
J Psychopharmacol ; 27(11): 1044-57, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23813967

RESUMO

Erythropoietin (EPO) promotes neurogenesis and neuroprotection. We here compared the protection induced by two EPO formulations in a rodent model of Alzheimer's disease (AD): rHu-EPO and a low sialic form, Neuro-EPO. We used the intracerebroventricular administration of aggregated Aß25₋35 peptide, a non-transgenic AD model. rHu-EPO was tested at 125-500 µg/kg intraperitoneally and Neuro-EPO at 62-250 µg/kg intranasally (IN). Behavioural procedures included spontaneous alternation, passive avoidance, water-maze and object recognition, to address spatial and non-spatial, short- and long-term memories. Biochemical markers of Aß25₋35 toxicity in the mouse hippocampus were examined and cell loss in the CA1 layer was determined. rHu-EPO and Neuro-EPO led to a significant prevention of Aß25₋35-induced learning deficits. Both EPO formulations prevented the induction of lipid peroxidation in the hippocampus, showing an antioxidant activity. rHu-EPO (250 µg/kg) or Neuro-EPO (125 µg/kg) prevented the Aß25₋35-induced increase in Bax level, TNFα and IL-1ß production and decrease in Akt activation. A significant prevention of the Aß25₋35-induced cell loss in CA1 was also observed. EPO is neuroprotective in the Aß25₋35 AD model, confirming its potential as an endogenous neuroprotection system that could be boosted for therapeutic efficacy. We here identified a new IN formulation of EPO showing high neuroprotective activity. Considering its efficacy, ease and safety, IN Neuro-EPO is a new promising therapeutic agent in AD.


Assuntos
Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/antagonistas & inibidores , Química Farmacêutica , Modelos Animais de Doenças , Eritropoetina/administração & dosagem , Eritropoetina/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Fragmentos de Peptídeos/antagonistas & inibidores , Administração Intranasal , Peptídeos beta-Amiloides/toxicidade , Animais , Contagem de Células , Relação Dose-Resposta a Droga , Eritropoetina/uso terapêutico , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Interleucina-1beta/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/toxicidade , Receptores da Eritropoetina/metabolismo , Reconhecimento Psicológico/efeitos dos fármacos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Retenção Psicológica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
10.
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
11.
Antioxid Redox Signal ; 16(3): 263-74, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21923553

RESUMO

AIMS: In our aging society, age-related hearing loss (ARHL) or presbycusis is increasingly important. Here, we study the mechanism of ARHL using the senescence-accelerated mouse prone 8 (SAMP8) which is a useful model to probe the effects of aging on biological processes. RESULTS: We found that the SAMP8 strain displays premature hearing loss and cochlear degeneration recapitulating the processes observed in human presbycusis (i.e., strial, sensory, and neural degeneration). The molecular mechanisms associated with premature ARHL in SAMP8 mice involve oxidative stress, altered levels of antioxidant enzymes, and decreased activity of Complexes I, II, and IV, which in turn lead to chronic inflammation and triggering of apoptotic cell death pathways. In addition, spiral ganglion neurons (SGNs) also undergo autophagic stress and accumulated lipofuscin. INNOVATION AND CONCLUSION: Our results provide evidence that targeting oxidative stress, chronic inflammation, or apoptotic pathways may have therapeutic potential. Modulation of autophagy may be another strategy. The fact that autophagic stress and protein aggregation occurred specifically in SGNs also offers promising perspectives for the prevention of neural presbycusis.


Assuntos
Autofagia , Cóclea/patologia , Inflamação/metabolismo , Presbiacusia/metabolismo , Potenciais de Ação , Fatores Etários , Animais , Apoptose , Cóclea/imunologia , Cóclea/inervação , Cóclea/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/metabolismo , Feminino , Fibroblastos/patologia , Células Ciliadas Auditivas Externas/patologia , Células Ciliadas Auditivas Externas/fisiologia , Lipofuscina/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Malondialdeído/metabolismo , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Órgão Espiral/metabolismo , Órgão Espiral/patologia , Presbiacusia/imunologia , Presbiacusia/patologia , Gânglio Espiral da Cóclea/metabolismo , Gânglio Espiral da Cóclea/patologia , Estria Vascular/patologia
12.
J Psychopharmacol ; 25(7): 960-75, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21555330

RESUMO

The sigma-1 (σ1) protein regulates calcium homeostasis and acts as an endoplasmic reticulum chaperone. It can be activated by ligands which impact memory, depression, anxiety or addiction processes. We here characterized the behavioural phenotype of knockout (KO) mice for the σ1 protein. Two-month old male σ1⁻/⁻ mice showed signs of anxiety in the open-field, passive avoidance or elevated plus-maze test, but other activity or memory responses were unchanged. Female σ1⁻/⁻ mice showed deficits in spontaneous alternation or water-maze learning. Twelve-month old σ1⁺/⁻ female mice showed deficits in alternation and σ1⁻/⁻ mice in avoidance escape latency. Two- and 14-month old female σ1⁻/⁻ mice showed decreased plasma 17ß-estradiol levels. Treatment with 17ß-estradiol (0.1, 0.2 mg/kg i.p.) reversed the spatial memory deficits in young and aged mice. Male σ1 KO mice showed enhanced response in the forced swimming test. Igmesine, a σ1 agonist, failed to decrease immobility in σ1 KO mice. Fluoxetine and sertraline were more efficient in σ1 KO mice, an effect likely related to their σ1 antagonist activity. Imipramine, desipramine and amitriptyline were equally active. σ1 protein invalidation therefore affected stress or anxiety response but not memory in males. Changes in steroid tonus in female animals led, however, to memory impairments that increased with age.


Assuntos
Ansiedade/metabolismo , Comportamento Animal , Depressão/metabolismo , Transtornos da Memória/metabolismo , Memória , Receptores sigma/deficiência , Fatores Etários , Envelhecimento , Animais , Antidepressivos/farmacologia , Ansiedade/genética , Ansiedade/psicologia , Comportamento Animal/efeitos dos fármacos , Cinamatos/farmacologia , Ciclopropanos/farmacologia , Depressão/tratamento farmacológico , Depressão/genética , Depressão/psicologia , Reação de Fuga , Estradiol/administração & dosagem , Estradiol/sangue , Feminino , Genótipo , Masculino , Aprendizagem em Labirinto , Memória/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/genética , Transtornos da Memória/psicologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora , Fenótipo , Tempo de Reação , Receptores sigma/efeitos dos fármacos , Receptores sigma/genética , Fatores Sexuais , Fatores de Tempo , Receptor Sigma-1
13.
Psychopharmacology (Berl) ; 214(2): 455-63, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20981412

RESUMO

RATIONALE: Trilostane is a competitive inhibitor of 3ß-hydroxysteroid dehydrogenase (3ß-HSD), which notably converts pregnenolone into progesterone or dehydroepiandrosterone into androstenedione. Trilostane shows antidepressant-like properties in the forced swimming test (FST). The compound, however, induced only moderate effects on neuroactive steroid levels that could be related to its behavioral efficacy. METHODS: We compared the behavioral effect of trilostane with the other 3ß-HSD inhibitor, cyanoketone, and analyzed the putative involvement of the ß-type estrogen receptor (ERß) in its antidepressant effect. RESULTS: Trilostane reduced immobility in the FST significantly at 12.5 and 25 mg/kg subcutaneously (s.c.), whereas cyanoketone (0-100 mg/kg s.c.) was ineffective. The negative ER modulator fulvestrant (ICI 182780) dose-dependently blocked the effect of trilostane (25 mg/kg). Trilostane increased circulating estradiol levels in the 12.5-50 mg/kg dose-range, and this effect was unaffected by stress and not shared by cyanoketone (25 mg/kg). The trilostane (25 mg/kg) treatment increased the ERß mRNA expression in adrenals (+100%) and centrally, in the hippocampus (+330%). Stress and cyanoketone failed to affect ERß mRNA levels in periphery or in the brain. CONCLUSIONS: These data demonstrate that the antidepressant-like potential of trilostane is not due to its 3ß-HSD inhibiting activity, since it is not shared by cyanoketone, but rather to its estrogenic activity. The compound, which releases estradiol and up-regulates ERß receptors, could be used as a therapeutic tool to allow an estrogenic facilitation of antidepressant efficacy.


Assuntos
3-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Antidepressivos/farmacologia , Comportamento Animal/efeitos dos fármacos , Di-Hidrotestosterona/análogos & derivados , Inibidores Enzimáticos/farmacologia , Receptor beta de Estrogênio/efeitos dos fármacos , 3-Hidroxiesteroide Desidrogenases/metabolismo , Glândulas Suprarrenais/efeitos dos fármacos , Glândulas Suprarrenais/metabolismo , Análise de Variância , Animais , Cianocetona/farmacologia , Di-Hidrotestosterona/farmacologia , Relação Dose-Resposta a Droga , Estradiol/análogos & derivados , Estradiol/sangue , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Fulvestranto , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , RNA Mensageiro/metabolismo , Natação , Fatores de Tempo , Regulação para Cima
14.
Pharmacol Ther ; 124(2): 195-206, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19619582

RESUMO

Originally considered an enigmatic protein, the sigma-1 receptor has recently been identified as a unique ligand-regulated molecular chaperone in the endoplasmic reticulum of cells. This discovery causes us to look back at the many proposed roles of this receptor, even before its molecular function was identified, in many diseases such as methamphetamine or cocaine addiction, amnesia, pain, depression, Alzheimer's disease, stroke, retinal neuroprotection, HIV infection, and cancer. In this review, we examine the reports that have clearly shown an agonist-antagonist relationship regarding sigma-1 receptors in models of those diseases and also review the relatively known mechanisms of action of sigma-1 receptors in an attempt to spur the speculation of readers on how the sigma-1 receptor at the endoplasmic reticulum might relate to so many diseases. We found that the most prominent action of sigma-1 receptors in biological systems including cell lines, primary cultures, and animals is the regulation and modulation of voltage-regulated and ligand-gated ion channels, including Ca(2+)-, K(+)-, Na(+), Cl(-), and SK channels, and NMDA and IP3 receptors. We found that the final output of the action of sigma-1 receptor agonists is to inhibit all above-mentioned voltage-gated ion channels, while they potentiate ligand-gated channels. The inhibition or potentiation induced by agonists is blocked by sigma-1 receptor antagonists. Other mechanisms of action of sigma-1 receptors, and to some extent those of sigma-2 receptors, were also considered. We conclude that the sigma-1 and sigma-2 receptors represent potential fruitful targets for therapeutic developments in combating many human diseases.


Assuntos
Receptores sigma/agonistas , Receptores sigma/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Analgésicos Opioides/uso terapêutico , Animais , Comportamento Aditivo/tratamento farmacológico , Comportamento Aditivo/metabolismo , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/uso terapêutico , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/metabolismo , Receptores sigma/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Receptor Sigma-1
15.
Neurobiol Aging ; 30(6): 987-1000, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17977621

RESUMO

Cystinosis is a lysosomal storage disorder characterised by progressive cystine accumulation. The causative gene, CTNS, encodes cystinosin, the lysosomal cystine transporter. Neurological deterioration is one of the last symptoms to appear and the least well characterised. Visuospatial memory deficits have been documented in patients. To determine whether the cystinosis mouse model presents similar anomalies, we studied the learning and memory abilities of young and middle-aged Ctns(-/-) mice. We did not detect deficits in young Ctns(-/-) mice. In contrast, spatial reference and working memory deficits were detected in middle-aged Ctns(-/-) mice. Elevated cystine levels were detected in the hippocampus, cerebellum, forebrain and brainstem of all Ctns(-/-) mice, which increased with age and were consistent with the appearance of impairments. Our results strongly suggest that the cystinosis-associated CNS anomalies are due to progressive cystine accumulation. Furthermore, the Ctns(-/-) mice serve as a model to investigate the evolution of these anomalies and test the efficiency of existing and novel treatments to cross the blood-brain barrier and reduce lysosomal cystine levels.


Assuntos
Envelhecimento/metabolismo , Encéfalo/metabolismo , Cistina/metabolismo , Transtornos da Memória/metabolismo , Memória , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Tecidual
16.
Chem Biol Interact ; 175(1-3): 131-4, 2008 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-18533140

RESUMO

We examined the sensitivity of AChE(+/-) mice to the amnesic effects of scopolamine and amyloid beta peptide. AChE(+/-) and AChE(+/+) littermates, tested at 5-9 weeks of age, failed to show any difference in locomotion, exploration and anxiety in the open-field test, or in-place learning in the water-maze. However, when treated with the muscarinic receptor antagonist scopolamine (0.5, 5mg/kg s.c.) 20 min before each water-maze training session, learning impairments were observed at both doses in AChE(+/+) mice, but only at the highest dose in AChE(+/-) mice. The central injection of Abeta(25-35) peptide (9 nmol) induced learning deficits only in AChE(+/+) but not in AChE(+/-) mice. Therefore, the hyper-activity of cholinergic systems in AChE(+/-) mice did not result in increased memory abilities, but prevented the deleterious effects of muscarinic blockade or amyloid toxicity.


Assuntos
Acetilcolinesterase/metabolismo , Amnésia/induzido quimicamente , Peptídeos beta-Amiloides/farmacologia , Heterozigoto , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Escopolamina/farmacologia , Acetilcolinesterase/genética , Peptídeos beta-Amiloides/química , Animais , Relação Dose-Resposta a Droga , Aprendizagem em Labirinto , Camundongos , Camundongos Knockout , Fragmentos de Peptídeos/química
17.
Hippocampus ; 18(6): 602-9, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18306297

RESUMO

Prenatal infection is a major stressful experience leading to enhanced susceptibility for mental illnesses in humans. We recently reported in rats, that oxidative stress and glutathione (GSH) shortage occurred in fetal male brain after lipopolysaccharide (LPS) to the dams and that these responses might be involved in the neurodevelopmental deficits observed in adolescent offspring. Furthermore, pretreatment with N-acetylcysteine (NAC) before LPS avoided both delayed synaptic plasticity and mnesic performance deficits. Since NAC is one of the few medications permitted in pregnant women, this study evaluated the ability of NAC to serve as a protective therapy even after the LPS challenge. Pregnant rats received a single ip injection of E. coli LPS, two days before delivery, and were given NAC in their tap water after the LPS. GSH was evaluated at the time of its expected drop in the hippocampus of male fetuses, whereas long-term potentiation (LTP) in the CA1 area of the hippocampus and spatial memory in the water-maze were recorded in 28-day-old male offspring. Post-treatment with NAC, four hours after the LPS challenge fully prevented the drop in the GSH hippocampal content. LTP, as well as spatial learning were completely protected. NAC administration at delivery also partially restored the LTP whereas post-treatment two days later was inefficient. Another set of dams were supplemented with alpha-tocopherol prior to LPS exposure, enhancing the alpha-tocopherol levels in fetal hippocampus. This treatment did not prevent the LPS-induced synaptic plasticity impairment. These results point to fetal hippocampal GSH as a major target of the detrimental effects of in utero LPS challenge. The therapeutic window of NAC extends up to birth, suggesting that this drug might be clinically useful even after an immuno-inflammatory episode.


Assuntos
Acetilcisteína/administração & dosagem , Endotoxemia/tratamento farmacológico , Potenciação de Longa Duração , Exposição Materna , Transtornos da Memória/prevenção & controle , Fármacos Neuroprotetores/administração & dosagem , Complicações na Gravidez/tratamento farmacológico , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , Acetilcisteína/farmacologia , Acetilcisteína/uso terapêutico , Animais , Antioxidantes/administração & dosagem , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Esquema de Medicação , Avaliação Pré-Clínica de Medicamentos , Endotoxemia/imunologia , Endotoxemia/fisiopatologia , Feminino , Glutationa/análise , Glutationa/deficiência , Hipocampo/química , Hipocampo/embriologia , Hipocampo/patologia , Lipopolissacarídeos/toxicidade , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/etiologia , Transtornos da Memória/fisiopatologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Gravidez , Complicações na Gravidez/imunologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Ratos , Ratos Sprague-Dawley , alfa-Tocoferol/administração & dosagem , alfa-Tocoferol/análise , alfa-Tocoferol/uso terapêutico
18.
Mol Cancer Res ; 5(11): 1147-57, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18025260

RESUMO

Chronic alcohol consumption is associated with increased risk of gastrointestinal cancer. High concentrations of ethanol trigger mucosal hyperregeneration, disrupt cell adhesion, and increase the sensitivity to carcinogens. Most of these effects are thought to be mediated by acetaldehyde, a genotoxic metabolite produced from ethanol by alcohol dehydrogenases. Here, we studied the role of low ethanol concentrations, more likely to mimic those found in the intestine in vivo, and used intestinal cells lacking alcohol dehydrogenase to identify the acetaldehyde-independent biological effects of ethanol. Under these conditions, ethanol did not stimulate the proliferation of nonconfluent cells, but significantly increased maximal cell density. Incorporation of phosphatidylethanol, produced from ethanol by phospholipase D, was instrumental to this effect. Phosphatidylethanol accumulation induced claudin-1 endocytosis and disrupted the claudin-1/ZO-1 association. The resulting nuclear translocation of ZONAB was shown to mediate the cell density increase in ethanol-treated cells. In vivo, incorporation of phosphatidylethanol and nuclear translocation of ZONAB correlated with increased proliferation in the colonic epithelium of ethanol-fed mice and in adenomas of chronic alcoholics. Our results show that phosphatidylethanol accumulation after chronic ethanol exposure disrupts signals that normally restrict proliferation in highly confluent intestinal cells, thus facilitating abnormal intestinal cell proliferation.


Assuntos
Adenocarcinoma/induzido quimicamente , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Neoplasias do Colo/induzido quimicamente , Etanol/toxicidade , Glicerofosfolipídeos/metabolismo , Proteínas de Choque Térmico/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/patologia , Adenocarcinoma/metabolismo , Álcool Desidrogenase/deficiência , Álcool Desidrogenase/genética , Animais , Proteínas Estimuladoras de Ligação a CCAAT/genética , Células CACO-2 , Contagem de Células , Claudina-1 , Neoplasias do Colo/metabolismo , Endocitose , Proteínas de Choque Térmico/genética , Humanos , Hiperplasia/induzido quimicamente , Proteínas de Membrana/metabolismo , Camundongos , Fosfolipase D/metabolismo , Fosfoproteínas/metabolismo , Proteína da Zônula de Oclusão-1
19.
Pediatr Res ; 62(2): 156-62, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17597652

RESUMO

Cystinosis is a lysosomal storage disorder characterized by abnormal accumulation of cystine, which forms crystals at high concentrations. The causative gene CTNS encodes cystinosin, the lysosomal cystine transporter. The eye is one of the first organs affected (corneal lesions and photophobia in the first and visual impairment in the second decade of life). We characterized the ocular anomalies of Ctns-/- mice to determine whether they mimic those of patients. The most dramatic cystine accumulation was seen in the iris, ciliary body, and cornea of Ctns-/- mice. Consistently, Ctns-/- mice had a low intraocular pressure (IOP) and seemed mildly photophobic. Retinal cystine levels were elevated but increased less dramatically with age. Consistently, the retina was intact and electroretinogram (ERG) profiles were normal in mice younger than 19 mo; beyond this age, retinal crystals and lesions appeared. Finally, the lens contained the lowest cystine levels and crystals were not seen. The temporospatial pattern of cystine accumulation in Ctns-/- mice parallels that of patients and validates the mice as a model for the ocular anomalies of cystinosis. This work is a prerequisite step to the testing of novel ocular cystine-depleting therapies.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Cisteína/metabolismo , Cistinose/patologia , Oftalmopatias/patologia , Olho/patologia , Sistemas de Transporte de Aminoácidos Neutros/deficiência , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Corioide/metabolismo , Corioide/patologia , Corpo Ciliar/metabolismo , Corpo Ciliar/patologia , Córnea/metabolismo , Córnea/patologia , Cistinose/complicações , Cistinose/genética , Cistinose/metabolismo , Cistinose/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Eletrorretinografia , Olho/metabolismo , Olho/fisiopatologia , Oftalmopatias/complicações , Oftalmopatias/genética , Oftalmopatias/metabolismo , Oftalmopatias/fisiopatologia , Pressão Intraocular , Iris/metabolismo , Iris/patologia , Cristalino/metabolismo , Cristalino/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fotofobia/etiologia , Fotofobia/metabolismo , Fotofobia/patologia , Reprodutibilidade dos Testes , Retina/metabolismo , Retina/patologia , Esclera/metabolismo , Esclera/patologia , Fatores de Tempo
20.
Free Radic Biol Med ; 42(8): 1231-45, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17382204

RESUMO

Prenatal infection is a major risk responsible for the occurrence of psychiatric conditions in infants. Mimicking maternal infection by exposing pregnant rodents to bacterial endotoxin lipopolysaccharide (LPS) also leads to major brain disorders in the offspring. The mechanisms of LPS action remain, however, unknown. Here, we show that LPS injection during pregnancy in rats, 2 days before delivery, triggered an oxidative stress in the hippocampus of male fetuses, evidenced by a rapid rise in protein carbonylation and by decreases in alpha-tocopherol levels and in the ratio of reduced/oxidized forms of glutathione (GSH/GSSG). Neither protein carbonylation increase nor decreases in alpha-tocopherol levels and GSH/GSSG ratio were observed in female fetuses. NMDA synaptic currents and long-term potentiation in CA1, as well as spatial recognition in the water maze, were also impaired in male but not in female 28-day-old offspring. Pretreatment with the antioxidant N-acetylcysteine prevented the LPS-induced changes in the biochemical markers of oxidative stress in male fetuses, and the delayed detrimental effects in male 28-day-old offspring, completely restoring both long-term potentiation in the hippocampus and spatial recognition performance. Oxidative stress in the hippocampus of male fetuses may thus participate in the neurodevelopmental damage induced by a prenatal LPS challenge.


Assuntos
Encéfalo/embriologia , Infecções/embriologia , Estresse Oxidativo , Animais , Encefalopatias/induzido quimicamente , Encefalopatias/embriologia , Encefalopatias/etiologia , Cromatografia Líquida de Alta Pressão , Feminino , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Hipocampo/embriologia , Hipocampo/fisiopatologia , Lipopolissacarídeos/toxicidade , Técnicas de Patch-Clamp , Gravidez , Ratos , Ratos Sprague-Dawley , alfa-Tocoferol/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA