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











Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 18(11): e0294280, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37948406

RESUMO

Chemotherapy-induced memory loss ("chemobrain") can occur following treatment with the widely used chemotherapeutic agent doxorubicin (DOX). However, the mechanisms through which DOX induces cognitive dysfunction are not clear, and there are no commercially available therapies for its treatment or prevention. Therefore, the aim of this study was to determine the therapeutic potential of phenyl-2-aminoethyl selenide (PAESe), an antioxidant drug previously demonstrated to reduce cardiotoxicity associated with DOX treatment, against DOX-induced chemobrain. Four groups of male athymic NCr nude (nu/nu) mice received five weekly tail-vein injections of saline (Control group), 5 mg/kg of DOX (DOX group), 10 mg/kg PAESe (PAESe group), or 5 mg/kg DOX and 10 mg/kg PAESe (DOX+PAESe group). Spatial memory was evaluated using Y-maze and novel object location tasks, while synaptic plasticity was assessed through the measurement of field excitatory postsynaptic potentials from the Schaffer collateral circuit. Western blot analyses were performed to assess hippocampal protein and phosphorylation levels. In this model, DOX impaired synaptic plasticity and memory, and increased phosphorylation of protein kinase B (Akt) and extracellular-regulated kinase (ERK). Co-administration of PAESe reduced Akt and ERK phosphorylation and ameliorated the synaptic and memory deficits associated with DOX treatment.


Assuntos
Disfunção Cognitiva , Potenciação de Longa Duração , Camundongos , Animais , Masculino , Proteínas Proto-Oncogênicas c-akt/metabolismo , Doxorrubicina/farmacologia , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Hipocampo/metabolismo , Cognição
2.
Cells ; 12(21)2023 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-37947603

RESUMO

Cannabis is now one of the most commonly used illicit substances among pregnant women. This is particularly concerning since developmental exposure to cannabinoids can elicit enduring neurofunctional and cognitive alterations. This study investigates the mechanisms of learning and memory deficits resulting from prenatal cannabinoid exposure (PCE) in adolescent offspring. The synthetic cannabinoid agonist WIN55,212-2 was administered to pregnant rats, and a series of behavioral, electrophysiological, and immunochemical studies were performed to identify potential mechanisms of memory deficits in the adolescent offspring. Hippocampal-dependent memory deficits in adolescent PCE animals were associated with decreased long-term potentiation (LTP) and enhanced long-term depression (LTD) at hippocampal Schaffer collateral-CA1 synapses, as well as an imbalance between GluN2A- and GluN2B-mediated signaling. Moreover, PCE reduced gene and protein expression of neural cell adhesion molecule (NCAM) and polysialylated-NCAM (PSA-NCAM), which are critical for GluN2A and GluN2B signaling balance. Administration of exogenous PSA abrogated the LTP deficits observed in PCE animals, suggesting PSA mediated alterations in GluN2A- and GluN2B- signaling pathways may be responsible for the impaired hippocampal synaptic plasticity resulting from PCE. These findings enhance our current understanding of how PCE affects memory and how this process can be manipulated for future therapeutic purposes.


Assuntos
Canabinoides , Moléculas de Adesão de Célula Nervosa , Humanos , Ratos , Feminino , Animais , Gravidez , Adolescente , Moléculas de Adesão de Célula Nervosa/metabolismo , Canabinoides/farmacologia , Canabinoides/metabolismo , Plasticidade Neuronal/fisiologia , Hipocampo/metabolismo , Transtornos da Memória/metabolismo
3.
ACS Pharmacol Transl Sci ; 4(1): 179-192, 2021 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-33615171

RESUMO

Since the first discovery of its ibuprofen-like anti-inflammatory activity in 2005, the olive phenolic (-)-oleocanthal gained great scientific interest and popularity due to its reported health benefits. (-)-Oleocanthal is a monophenolic secoiridoid exclusively occurring in extra-virgin olive oil (EVOO). While several groups have investigated oleocanthal pharmacokinetics (PK) and disposition, none was able to detect oleocanthal in biological fluids or identify its PK profile that is essential for translational research studies. Besides, oleocanthal could not be detected following its addition to any fluid containing amino acids or proteins such as plasma or culture media, which could be attributed to its unique structure with two highly reactive aldehyde groups. Here, we demonstrate that oleocanthal spontaneously reacts with amino acids, with high preferential reactivity to glycine compared to other amino acids or proteins, affording two products: an unusual glycine derivative with a tetrahydropyridinium skeleton that is named oleoglycine, and our collective data supported the plausible formation of tyrosol acetate as the second product. Extensive studies were performed to validate and confirm oleocanthal reactivity, which were followed by PK disposition studies in mice, as well as cell culture transport studies to determine the ability of the formed derivatives to cross physiological barriers such as the blood-brain barrier. To the best of our knowledge, we are showing for the first time that (-)-oleocanthal is biochemically transformed to novel products in amino acids/glycine-containing fluids, which were successfully monitored in vitro and in vivo, creating a completely new perspective to understand the well-documented bioactivities of oleocanthal in humans.

4.
Aging (Albany NY) ; 13(5): 6634-6661, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33591941

RESUMO

As a major pathological hallmark of Alzheimer's disease (AD), amyloid-ß (Aß) is regarded as a causative factor for cognitive impairment. Extensive studies have found Aß induces a series of pathophysiological responses, finally leading to memory loss in AD. Our previous results demonstrated that cysteinyl leukotrienes receptor 1 (CysLT1R) antagonists improved exogenous Aß-induced memory impairment. But the role of CysLT1R in AD and its underlying mechanisms still remain elusive. In this study, we investigated CysLT1R levels in AD patients and APP/PS1 mice. We also generated APP/PS1-CysLT1R-/- mice by clustered regulatory interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9)-mediated CysLT1R deletion in APP/PS1 mice and studied the effect of CysLT1R knockout on amyloidogenesis, synapse structure and plasticity, cognition, neuroinflammation, and kynurenine pathway. These attributes were also studied after lentivirus-mediated knockdown of CysLT1R gene in APP/PS1 mice. We found that CysLT1R knockout or knockdown could conserve synaptic structure and plasticity, and improve cognition in APP/PS1 mice. These effects were associated with concurrent decreases in amyloid processing, reduced neuroinflammation and suppression of the kynurenine pathway. Our study demonstrates that CysLT1R deficiency can mediate several beneficial effects against AD pathogenesis, and genetic/pharmacological ablation of this protein could be a potential therapeutic option for AD.


Assuntos
Amiloidose/prevenção & controle , Sistemas CRISPR-Cas , Disfunção Cognitiva/prevenção & controle , Deleção de Genes , Receptores de Leucotrienos/genética , Transmissão Sináptica , Precursor de Proteína beta-Amiloide , Animais , Modelos Animais de Doenças , Camundongos Knockout , Camundongos Transgênicos , Plasticidade Neuronal
5.
Neuropharmacology ; 131: 143-153, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29248482

RESUMO

AIMS: To investigate restorative effects of the receptor for advanced glycation end products (RAGE)-specific inhibitor FPS-ZM1 on abnormal amyloid ß (Aß) influx across the blood brain-barrier (BBB) and cognitive deficits in db/db mice. METHODS: Aß influx across the BBB was determined by intra-arterial infusion of 125I-Aß1-40. Receptor for advanced glycation end products (RAGE), Aß, NF-κB p65, caspase-3, Bax, Bcl-2, PSD-95 and synaptophysin were assayed by Western blot, immunohistochemistry or RT-PCR. Apoptosis was quantified by TUNEL assay. In vivo hippocampal long term potentiation (LTP) recording, Golgi Staining, Morris water maze (MWM) task and Y-maze test were performed. RESULTS: FPS-ZM1 (1.0 mg/kg i.p.) inhibited Aß influx across the BBB and expression of RAGE participating in Aß influx, consequently decreased hippocampal Aß1-40 and Aß1-42 in db/db mice. After FPS-ZM1 treatment, NF-κB signaling was inhibited, and neuronal apoptosis was reduced, which revealed by less TUNEL + cells, reduced caspase-3 activity and higher ratio of Bcl-2/Bax. In addition, FPS-ZM1 improved hippocampal plasticity evidenced by enhanced in vivo LTP and the restoration of spine deficit and increased PSD-95 expression in hippocampal neuron. Further studies found that FPS-ZM1 treatment alleviated cognitive deficits shown by better performance in behavioral tests, without significant metabolic effects on blood glucose, insulin and cerebral AGEs. CONCLUSION: Downregulation of abnormal Aß influx across the BBB by FPS-ZM1 at higher dosage contributes to reduced neuronal apoptosis, improved hippocampal plasticity and cognitive impairment in db/db mice.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Benzamidas/uso terapêutico , Barreira Hematoencefálica/fisiologia , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Diabetes Mellitus Tipo 2/complicações , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Peptídeos beta-Amiloides/farmacocinética , Animais , Benzamidas/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/ultraestrutura , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/ultraestrutura , Caspase 3/metabolismo , Diabetes Mellitus Tipo 2/genética , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Fragmentos de Peptídeos/farmacocinética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Receptor para Produtos Finais de Glicação Avançada/genética , Receptores para Leptina/deficiência , Receptores para Leptina/genética , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética , Proteína X Associada a bcl-2/metabolismo
6.
J Affect Disord ; 227: 672-680, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29174741

RESUMO

BACKGROUND: Recent studies demonstrated beneficial effects of zileuton, a 5-lipoxygenase (5LO) inhibitor, on some brain diseases in animal models, but the role of zileuton in the depression remains unknown. METHODS: We investigated the effects of zileuton on depressive behaviors using tail suspension test (TST), forced swimming test (FST) and novelty-suppressed feeding test (NSFT) in mice injected with lipopolysaccharide (LPS). The 5LO level, activation of microglia, NF-κB p65, TNF-α, IL-1ß, brain-derived neurotrophic factor (BDNF), and c-AMP response element-binding protein (CREB) were determined in the mouse hippocampus. RESULTS: We firstly found that the expression of hippocampal 5LO was gradually increased over LPS exposure and was reversed by fluoxetine administration. Zileuton significantly suppressed LPS-induced depressive behaviors, evidenced by the decreases in immobility time in TST and FST, as well as the latency to feed in NSFT. This treatment pronouncedly alleviated LPS-induced neuroinflammatory response, characterized by decreased 5LO, suppressed activation of microglia, decreased NF-κB p65, TNF-α and IL-1ß, and significantly increased the ratio of p-CREB/CREB or mBDNF/proBDNF in the hippocampus of the LPS-challenged mice. CONCLUSIONS: Zileuton abrogates LPS-induced depressive-like behaviors and neuroinflammation, and enhances CREB/BDNF signaling in the hippocampus, suggesting that zileuton could have potential therapeutic value for depression.


Assuntos
Antidepressivos/farmacologia , Hipocampo/efeitos dos fármacos , Hidroxiureia/análogos & derivados , Animais , Antidepressivos/uso terapêutico , Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , AMP Cíclico/metabolismo , Depressão/tratamento farmacológico , Depressão/metabolismo , Transtorno Depressivo/tratamento farmacológico , Modelos Animais de Doenças , Fluoxetina/farmacologia , Hipocampo/metabolismo , Hidroxiureia/farmacologia , Hidroxiureia/uso terapêutico , Interleucina-1beta/metabolismo , Lipopolissacarídeos , Masculino , Camundongos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/efeitos dos fármacos
7.
Mol Cell Endocrinol ; 389(1-2): 40-7, 2014 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-24424441

RESUMO

While the conflict between basic science evidence for estrogen neuroprotection and the lack of effectiveness in clinical trials is only now being resolved, it is clear that strategies for estrogen neuroprotection that avoid activation of ERs have the potential for clinical application. Herein we review the evidence from both in vitro and in vivo studies that describe high potency neuroprotection with non-feminizing estrogens. We have characterized many of the essential chemical features of non-feminizing estrogens that eliminate or reduce ER binding while maintaining or enhancing neuroprotection. Additionally, we provide evidence that these non-feminizing estrogens have efficacy in protecting the brain from AD neuropathology and traumatic brain injury. In conclusion, it appears that the non-feminizing estrogen strategy for neuroprotection is a viable option to achieve the beneficial neuroprotective effects of estrogens while eliminating the toxic off-target effects of chronic estrogen administration.


Assuntos
Estrogênios/metabolismo , Estrogênios/farmacologia , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ensaios Clínicos como Assunto , Feminino , Humanos , Masculino , Receptores de Estrogênio/metabolismo
8.
Neurotoxicology ; 27(5): 710-20, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16824603

RESUMO

Fish contain methylmercury and the potentially protective element, selenium. Blood and brain concentrations of these elements were determined in female rats after consuming AIN-93-based diets containing 0.06 or 0.6 ppm of selenium (Se) and drinking water containing 0, 0.5, or 5 ppm of mercury as methylmercury (MeHg) for 6 or 18 months. Brain and blood concentrations of mercury and selenium were also evaluated in neonates after gestational exposure. For adult rats in the high-Se, high-Hg condition, brain selenium content was 0.35 ppm and 1.8 ppm after 6 and 18 months, respectively, but for every other adult-onset condition, it was 0.1 ppm. Blood selenium varied less than two-fold despite a 10-fold difference in diet. After 6 months, mercury content in the brain showed a greater than 10-fold difference between the mercury groups, and interacted somewhat with dietary selenium. After 18 months, no mercury was detected in the brains of the 0.5 ppm groups, and their blood mercury also fell. For the 5.0 ppm groups, brain mercury increased slightly (low Se diet) or several-fold (high Se diet) over that seen at 6 months, and blood mercury also increased. Neonatal selenium concentrations were more labile than adults, and mercury in neonates was generally higher. All animals exposed to 5 ppm of mercury experienced a molar excess of mercury over selenium. Animals exposed to 0.5 ppm mercury showed a balance between mercury and selenium or a selenium excess, depending on the condition.


Assuntos
Encéfalo , Compostos de Metilmercúrio/administração & dosagem , Compostos de Metilmercúrio/sangue , Efeitos Tardios da Exposição Pré-Natal , Selênio/administração & dosagem , Selênio/sangue , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Química Encefálica/efeitos dos fármacos , Dieta , Relação Dose-Resposta a Droga , Feminino , Masculino , Gravidez , Ratos , Ratos Long-Evans , Fatores de Tempo , Distribuição Tecidual
9.
Neurotoxicol Teratol ; 27(4): 629-41, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16024222

RESUMO

It has been suggested that docosahexaenoic acid (DHA) or other n-3 polyunsaturated fatty acids (PUFAs) may prevent or ameliorate methyl mercury's neurotoxicity. To examine interactions between PUFAs and methyl mercury exposure, sixty-six female Long-Evans rats were exposed to methyl mercury continuously via drinking water from fifteen weeks of age. Water included methyl mercury concentrations of 0, 0.5, and 5.0 ppm, creating estimated intakes of about 0, 40, and 400 microg/kg/day across exposure groups. An additional fifty-eight female offspring were exposed to methyl mercury only during gestation. Rats consumed one of two diets, each based on AIN-93 formulation, providing a 2 (generation) X 2 (diet) X 3 (methyl mercury exposure) factorial experimental design. A "coconut oil" diet (1/3 of fats were provided by coconut oil) was marginally adequate in n-3 PUFAs and contained no DHA. A "fish oil" diet was rich in n-3 fatty acids, including DHA. The diets were approximately equal in n-6 fatty acids. Forelimb grip strength declined with age for all groups, but the decline was greatest for those exposed chronically to 400 microg/kg/day of methyl mercury. This high-dose group also displayed hind limb crossing, gait disorders, and diminished running wheel activity. Dietary n-3 fatty acids did not influence these effects. Chronic exposure to 400 microg/kg/day of methyl mercury resulted in blood and brain concentrations of about 70 and 10 ppm, respectively, approximately 50-fold higher than concentrations seen in rats exposed to 40 microg/kg/day. Rats that became ill and died before the experiment ended had higher concentrations of mercury than their cohorts who survived to the end. Organic mercury was highly correlated with total mercury in these rats but inorganic mercury remained approximately constant. Some deaths were due to urolithiasis (kidney or bladder stones) associated with a dietary contaminant and that was eventually fatal to 22% of the females in the colony. Neurobehavioral effects are reported on rats that did not become ill.


Assuntos
Óleos de Peixe/administração & dosagem , Mercúrio/sangue , Compostos de Metilmercúrio/toxicidade , Transtornos das Habilidades Motoras/dietoterapia , Efeitos Tardios da Exposição Pré-Natal , Fatores Etários , Análise de Variância , Animais , Comportamento Animal , Química Encefálica/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Atividade Motora/efeitos dos fármacos , Transtornos das Habilidades Motoras/induzido quimicamente , Transtornos das Habilidades Motoras/fisiopatologia , Gravidez , Desempenho Psicomotor/efeitos dos fármacos , Ratos , Ratos Long-Evans , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA