Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neurochem Int ; 165: 105510, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36893915

RESUMO

Clinical and experimental studies have shown that the sharp reduction of estrogen is one of the important reasons for the high incidence of Alzheimer's disease (AD) in elderly women, but there is currently no such drug for treatment of AD. Our group first designed and synthesized a novel compound R-9-(4fluorophenyl)-3-methyl-10,10,-Hydrogen-6-hydrogen-benzopyran named FMDB. In this study, our aim is to investigate the neuroprotective effects and mechanism of FMDB in APP/PS1 transgenic mice. 6 months old APP/PS1 transgenic mice were intragastrical administered with FMDB (1.25, 2.5 and 5 mg/kg) every other day for 8 weeks. LV-ERß-shRNA was injected bilaterally into the hippocampus of APP/PS1 mice to knockdown estrogen receptor ß (ERß). We found that FMDB ameliorated cognitive impairment in the Morris water maze and novel object recognition tests, increased hippocampal neurogenesis and prevented hippocampal apoptotic responses in APP/PS1 mice. Importantly, FMDB activated nuclear ERß mediated CBP/p300, CREB and brain-derived neurotrophic factor (BDNF) signaling, and membrane ERß mediated PI3K/Akt, CREB and BDNF signaling in the hippocampus. Our study demonstrated the contributions and mechanism of FMDB to cognition, neurogenesis and apoptosis in APP/PS1 mice. These lay the experimental foundation for the development of new anti-AD drugs.


Assuntos
Doença de Alzheimer , Fármacos Neuroprotetores , Camundongos , Animais , Feminino , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Camundongos Transgênicos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Fosfatidilinositol 3-Quinases , Receptor beta de Estrogênio , Cognição , Hipocampo/metabolismo , Modelos Animais de Doenças , Neurogênese , Apoptose , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Presenilina-1/genética
2.
Neurobiol Dis ; 175: 105922, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36371059

RESUMO

Our previous study suggests that hippocampal cysteinyl leukotriene receptor 1 (CysLT1R) could be involved in depression. Herein we hypothesize that CysLT1R may regulate depression by affecting synaptic glutamate cycling based on existence of CysLT1R in the astrocytes that participate in occurrence of depression. We found that CysLT1R expression was significantly increased in the astrocyte of chronic unpredictable mild stress (CUMS)-induced depression-like mice, CysLT1R astrocyte-specific conditional knockout (AcKO) significantly improved depression-like behaviors, as indicated by decreased immobility time in the forced swimming test and tail suspension test and increased sucrose preference in the sucrose preference test, and knockdown of CysLT1R in the astrocyte of dentate gyrus (DG), the region with the most significant increase of CysLT1R in the astrocyte of depression-like mice, produced similar effects. Correspondingly, overexpression of CysLT1R in the astrocyte of DG induced depression-like behaviors in mice. The further study showed that CysLT1R AcKO ameliorated synaptic plasticity impairment, as reflected by increased synapse, LTP and PSD95, and promoted glutamate transporter 1 (GLT-1) expression by inhibiting NF-κB p65 nuclear translocation mediated by ß-arestin2 and clatrhin, subsequently decreased glutamate in synaptic cleft and GluN2B on postsynaptic membrane in depression-like mice. The present study also showed that GLT-1 agonist or NF-κB inhibitor ameliorated depressive-like behaviors induced by overexpression of the astrocyte CysLT1R of DG. Our study demonstrated that astrocyte CysLT1R regulated depression by modulating glutamate synaptic transmission, suggesting that CysLT1R could be a potential target for developing novel drugs of anti-depression.


Assuntos
Astrócitos , Depressão , Ácido Glutâmico , Receptores de Leucotrienos , Transmissão Sináptica , Animais , Camundongos , Astrócitos/metabolismo , Ácido Glutâmico/metabolismo , Hipocampo/metabolismo , NF-kappa B/metabolismo , Estresse Psicológico , Sacarose/metabolismo , Sacarose/farmacologia , Receptores de Leucotrienos/metabolismo , Depressão/metabolismo , Depressão/patologia
3.
Ann Transl Med ; 9(20): 1531, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34790737

RESUMO

BACKGROUND: Our previous studies demonstrated that cysteinyl leukotrienes receptor 1 (CysLT1R) knockout, pharmacological blockade, or hippocampus knockdown produced beneficial effects against Alzheimer's disease (AD); however, whether CysLT1R upregulation has deleterious effects on AD remains elusive. METHODS: In this study, we investigated the changes in behaviors, hippocampal amyloidogenesis, and synapse plasticity after CysLT1R overexpression by microinfusion of the lentiviral vector, containing its coding sequence of mouse (LV-CysLT1R), into the bilateral dentate gyri (DG) of the hippocampus or CysLT1R activation by repeated systemic administration of its agonist YM-17690 (0.1 mg/kg, once a day, i.p., for 28 d). RESULTS: The behavior data showed that overexpression of CysLT1R in hippocampal DG or administration of YM-17690 deteriorated behavioral performance in Morris water maze (MWM), Y-maze tests, and novel object recognition (NOR) in young APP/PS1 mice. The further studies showed that these treatments significantly destroyed synaptic function, as evidenced by impaired hippocampal long-term potentiation (LTP), decreased spine density, low number of synapses, and decreased postsynaptic protein (PSD95), and promoted the generation of amyloid ß (Aß) through increased expression of BACE1 and PS1 in the hippocampus of young APP/PS1 mice. CONCLUSIONS: Together, our results indicate that CysLT1R upregulation accelerates memory impairment in young APP/PS1 mice, which is associated with promoting synaptic dysfunction and amyloidogenesis in the hippocampus.

4.
Ann Transl Med ; 9(18): 1471, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34734023

RESUMO

BACKGROUND: Cognitive impairment is a serious complication of diabetes that manifests as an impairment of spatial memory and learning ability. Its pathogenesis is unclear, and effective therapeutic drugs are very limited. Our group designed and synthesized a novel compound named 3-p-tolyl-9H-xanthen-9-one (Tozan). In this study, we sought to investigate the effects and mechanism of Tozan on diabetic cognitive impairment. METHODS: Methylglyoxal (MG)-induced SH-SY5Y cells and streptozotocin (STZ)-induced type 1 diabetic mice were treated with Tozan. Methyl thiazolul tetrazolium (MTT) and lactate dehydrogenase (LDH) were used to test cytotoxicity. Morris water maze (MWM) and Y-maze tests were used to evaluate cognitive function. Immunofluorescence and western blot analyses were used to evaluate neurogenesis, apoptosis, and signal transduction pathway-related proteins. In addition, Lentivirus (LV)-estrogen receptor beta (ERß)-ribonucleic acid interference (RNAi) was used to knockdown the ERß gene in SH-SY5Y cells. RESULTS: We found that Tozan ameliorated MG-induced cytotoxicity in SH-SY5Y cells, improved cognitive dysfunction in STZ-induced type 1 diabetic mice, increased neurogenesis, and prevented apoptotic responses in vitro and in vivo. Importantly, Tozan (2, 4, and 8 mg/kg) mediated phosphatidylinositol-3-kinase and protein kinase B cAMP-response element binding protein (PI3K/Akt-CREB) signaling by activating membrane ERß, and a high dose of Tozan (8 mg/kg) mediated CREB signaling by activating nuclear ERß in the hippocampus. Notably, Tozan did not have an anti-apoptosis and regeneration protective role in ERß gene knockdown cells. CONCLUSIONS: Our study demonstrates Tozan's contributions to and role in cognition, neurogenesis, and apoptosis in diabetes, and lays an experimental foundation for the development of new anti-diabetic cognitive impairment drugs.

5.
Int Immunopharmacol ; 78: 105947, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31796384

RESUMO

Our previous study has found that zileuton, a selective 5-lipoxygenase (5LO) inhibitor, abrogated lipopolysaccharide-induced depressive-like behaviors and hippocampal neuroinflammation. Herein, we further extended our curiosity to investigate effects of zileuton on stress-induced depressive-like behaviors. Our data indicated that zileuton significantly ameliorated depressive-like behaviors in mice subjected to chronic mild stress (CMS), as shown in the tail suspension test, forced swimming test and novelty-suppressed feeding test. The further studies indicated that zileuton suppressed hippocampal neuroinflammation, evidenced by lower levels of TNF-α, IL-1ß and nuclear NF-κB p65 as well as decreased number of Iba1-positive cells. It also significantly ameliorated hippocampal apoptosis, indicated by deceased number of TUNEL-positive cells, deceased ratio of cleaved caspase-3/procaspase-3 and increased ratio of Bcl-2/Bax. More importantly, zileuton increased the level of synaptic proteins PSD-95 and SYN and the number of NeuN+/BrdU+ cells in the hippocampus. Over all, zileuton alleviated CMS-induced depressive-like behaviors, neuroinflammatory and apoptotic responses, abnormalities of synapse and neurogenesis in the hippocampus, suggesting that it might has beneficial effects on depression.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Encefalite/tratamento farmacológico , Hidroxiureia/análogos & derivados , Fármacos Neuroprotetores/uso terapêutico , Estresse Psicológico/tratamento farmacológico , Animais , Anti-Inflamatórios/farmacologia , Antidepressivos/farmacologia , Apoptose/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Depressão/metabolismo , Depressão/patologia , Encefalite/metabolismo , Encefalite/patologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Hidroxiureia/farmacologia , Hidroxiureia/uso terapêutico , Interleucina-1beta/metabolismo , Masculino , Camundongos Endogâmicos ICR , Neurogênese/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Estresse Psicológico/metabolismo , Estresse Psicológico/patologia , Sinapses/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
6.
J Asian Nat Prod Res ; 22(9): 864-878, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31347387

RESUMO

This study aimed to evaluate whether mogrol, a main bioactive ingredient of Siraitia grosvenorii, could attenuate LPS-induced memory impairment in mice. The behavioral tests and immunohistochemical analysis and Western blot were performed. The present results showed that oral administration of mogrol (20, 40, 80 mg/kg) significantly improved LPS-induced memory impairment in mice. The results also indicated that mogrol treatment significantly reduced the number of Iba1-positive cells, the nuclear NF-κB p65 and levels of TNF-α, IL-1ß and IL-6 both in the hippocampus and frontal cortex of LPS-challenged mice. [Formula: see text].


Assuntos
Inflamação , Lipopolissacarídeos , Animais , Hipocampo , Camundongos , Estrutura Molecular , NF-kappa B , Fator de Necrose Tumoral alfa
7.
Int Immunopharmacol ; 77: 105918, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31639616

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disease that affects cognition and behavior. The neuroinflammatory response in the brain is an important pathological characteristic in AD. In this study, we investigated the neuroprotective effects of 1-Methylnicotinamide (MNA), known as the main metabolite of nicotinamide, on reducing lipopolysaccharide (LPS)-induced cognitive deficits via targeting neuroinflammation and neuronal apoptosis. We found that the mice treated with LPS exhibited cognitive deficits in the novel object recognition, Morris water maze and Y-maze avoidance tests. However, intragastric administration of MNA (100 or 200 mg/kg) for 3 weeks significantly attenuated LPS-induced cognitive deficits in mice. Importantly, MNA treatment suppressed the protein expression of nuclear factor-kappa B p65 (NF-κB p65), pro-inflammatory cytokines (TNF-α, IL-6) and decreased the activation of microglia and astrocytes in the hippocampus and frontal cortex of LPS-induced mice. In addition, MNA treatment suppressed neuronal apoptosis by reducing the number of TUNEL-positive cells, caspase-3 activation and increasing the level of Bcl-2/Bax ratio in the hippocampus and frontal cortex. These findings indicate that MNA could be a potential neuroprotective drug in neurodegenerative diseases such as AD.


Assuntos
Anti-Inflamatórios/uso terapêutico , Transtornos Cognitivos/tratamento farmacológico , Transtornos da Memória/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Niacinamida/análogos & derivados , Animais , Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/imunologia , Lobo Frontal/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Interleucina-6/imunologia , Lipopolissacarídeos , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/imunologia , Camundongos Endogâmicos ICR , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Niacinamida/farmacologia , Niacinamida/uso terapêutico , Fator de Transcrição RelA/imunologia , Fator de Necrose Tumoral alfa/imunologia
8.
Exp Neurol ; 311: 33-43, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30201537

RESUMO

Estrogen receptors (ERs) are thought to be associated with the onset and progression of neurodegenerative injuries and diseases, but the relationship and mechanisms underlying between ERs and cognition in type 2 diabetes remain elusive. In the current study, we investigated the effects of ERα and ERß on the cognition, neurogenesis and apoptosis in high-fat diet and streptozocin-induced diabetic mice. We found that ERα and/or ERß activation using their agonists (0.5 mg/kg E2, PPT or DPN) ameliorate memory impairment in the Morris water maze and Y-maze tests, increase hippocampal neurogenesis and prevent hippocampal apoptotic responses. Importantly, treatment with the pharmacologic ERs agonists caused significant increases in the membrane ERα and ERß expression and subsequent PI3K/Akt, CREB and BDNF activation in the hippocampus of type 2 diabetes mellitus mice. Our data indicate that ERα and ERß are involved in the cognitive impairment in type 2 diabetes, and that activated ERs, such as application of ERs agonists, could be a novel and promising strategy for the treatment of diabetic cognitive impairment.


Assuntos
Apoptose/fisiologia , Disfunção Cognitiva/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Neurogênese/fisiologia , Animais , Apoptose/efeitos dos fármacos , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/psicologia , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/psicologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/psicologia , Estradiol/farmacologia , Estradiol/uso terapêutico , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Feminino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos ICR , Neurogênese/efeitos dos fármacos , Distribuição Aleatória
9.
Horm Behav ; 105: 95-103, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30096284

RESUMO

Estrogen receptors (ERs) are thought to be associated with the onset and progression of neurodegenerative injuries and diseases, but the relationship and mechanisms underlying between ERs and cognition in type 1 diabetes remain elusive. In the current study, we investigated the effects of ERα and ERß on the memory impairment and apoptosis in streptozotocin-induced diabetic mice. We found that ERα and/or ERß activation using their agonists (0.5 mg/kg E2, PPT or DPN) ameliorate memory impairment in the Morris water maze (MWM) and Y-maze tests and suppress apoptosis as evidenced by decreased caspase-3 activity and increased ratio of Bcl-2/Bax. Importantly, treatment with the pharmacologic ERs agonists caused significant increases in the membrane ERα and ERß expression and subsequent PI3K/Akt, CREB and BDNF activation in the hippocampus of diabetic mice. Our data indicate that ERα and ERß are involved in the cognitive impairment of type 1 diabetes and that activation of ERs via administration of ERs agonists could be a novel and promising strategy for the treatment of diabetic cognitive impairment.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Diabetes Mellitus Experimental/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Disfunção Cognitiva/etiologia , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/psicologia , Estradiol/farmacologia , Estradiol/uso terapêutico , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/metabolismo , Feminino , Hipocampo/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Fármacos Neuroprotetores/farmacologia , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Ovariectomia , Fenóis/farmacologia , Fenóis/uso terapêutico , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Estreptozocina
10.
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
11.
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
12.
Acta Pharmacol Sin ; 38(4): 477-487, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28112182

RESUMO

Evidence suggests that neuroinflammation is involved in depression and that the cysteinyl leukotriene receptor 1 (CysLT1R) plays a potential pathophysiological role in several types of CNS disorders. Our previous study has shown that knockdown of hippocampal CysLT1R in mice prevents the depressive-like phenotype and neuroinflammation induced by chronic mild stress (CMS). Here, we examined the effects of hippocampal CysLT1R knockdown and CysLT1R blockade on LPS-induced depressive-like behavior in mice. We found that injection of LPS (0.5 mg/kg, ip) caused marked increase in hippocampal CysLT1R expression, which was reversed by pretreatment with fluoxetine (20 mg·kg-1·d-1 for 7 d, ig). Knockdown of hippocampal CysLT1R or blockade of CysLT1R by pretreatment with pranlukast (0.5 mg/kg, ip) significantly suppressed LPS-induced depressive behaviors, as evidenced by decreases in mouse immobility time in the forced swimming test (FST) and tail suspension test (TST) and latency to feed in the novelty-suppressed feeding (NSF) test. Moreover, both CysLT1R knockdown and CysLT1R blockade markedly prevented LPS-induced neuroinflammation, as shown by the suppressed activation of microglia and NF-κB signaling as well as the hippocampal levels of TNF-α and IL-1ß in mice. Our results suggest that CysLT1R may be involved in LPS-induced depressive-like behaviors and neuroinflammation, and that downregulation of CysLT1R could be a novel and potential therapeutic strategy for the treatment of depression, at least partially due to its role in neuroinflammation.


Assuntos
Depressão/tratamento farmacológico , Hipocampo/metabolismo , Lipopolissacarídeos/farmacologia , Receptores de Leucotrienos/genética , Animais , Cromonas/uso terapêutico , Depressão/metabolismo , Depressão/psicologia , Fluoxetina/uso terapêutico , Técnicas de Silenciamento de Genes , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Antagonistas de Leucotrienos/uso terapêutico , Masculino , Camundongos Endogâmicos ICR , Receptores de Leucotrienos/metabolismo , Transdução de Sinais
13.
Artigo em Inglês | MEDLINE | ID: mdl-27720931

RESUMO

Our previous studies showed that cysteinyl leukotrienes receptor 1 (CysLT1R) is upregulated in amyloid-ß (Aß)-induced neurotoxicity and that administration of CysLT1R antagonists such as pranlukast or montelukast can ameliorate memory impairment in mice. In the current study, we sought to explore the role of CysLT1R in intracerebroventricular streptozotocin (STZ-ICV)-induced mouse model of memory impairment and neuroinflammation through shRNA-mediated knockdown of CysLT1R and also its pharmacological blockade by pranlukast. ICR mice were infused with STZ (3.0mg/kg) by a single bilateral stereotaxic ICV microinjection followed by administration of CysLT1R-shRNA (intra-hippocampal) or pranlukast (intragastric, IG). After 21days, a set of behavioral and biochemical tests were performed in order to assess the degree of memory impairment and neuroinflammation in mice. STZ-infused mice spent less time in the target quadrant of Morris water maze test and took more time to find the shock-free arm in modified Y-maze test, which were rescued in the CysLT1R-knockdowned or pranlukast-treated mice. STZ-induced memory impairment was also accompanied by an elevated level of hippocampal CysLT1R, microglial activation, increased IL-1ß, and TNF-α. Such elevation of these factors was found to be mediated through the classical NF-κB pathway and administration of CysLT1R-shRNA or pranlukast for 21days reversed all these parameters, suggesting a role of CysLT1R in STZ-induced memory deficit and neuroinflammation.


Assuntos
Regulação para Baixo/fisiologia , Encefalite/etiologia , Transtornos da Memória/complicações , Receptores de Leucotrienos/metabolismo , Animais , Antiasmáticos/uso terapêutico , Antibióticos Antineoplásicos/toxicidade , Proteínas de Ligação ao Cálcio/metabolismo , Cromonas/uso terapêutico , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Histonas/metabolismo , Humanos , Injeções Intraventriculares , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/induzido quimicamente , Camundongos , Camundongos Endogâmicos ICR , Proteínas dos Microfilamentos/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/uso terapêutico , Receptores de Leucotrienos/genética , Transdução de Sinais/efeitos dos fármacos , Estreptozocina/toxicidade , Fatores de Tempo , Transdução Genética , Transfecção
14.
Neurotoxicology ; 57: 214-222, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27702591

RESUMO

Extensive studies have demonstrated that neuroinflammation is associated with Alzheimer's disease (AD) and cysteinyl leukotriene receptor 1 (CysLT1R) was involved in neuroinflammation. Montelukast, a highly selective CysLT1R antagonist, has been reported to attenuate learning and memory impairments in the amyloid-ß-induced mouse model of AD. However, whether montelukast also exerts beneficial effects on streptozotocin (STZ)-induced memory deficits in mice is not well known. In the present study, we aimed to investigate the effects of montelukast on STZ-induced cognitive impairment, neuroinflammation and apoptosis in mice. Our data showed that intra-hippocampal microinfusion of STZ resulted in learning and memory impairments, including increased escape latency during acquisition trials and decreased exploratory activities in the probe trial in Morris watermaze (MWM) task, and decreased number of correct choices and increased latency to enter the shock-free compartment in Y-maze test, and caused neuroinflammatory and apoptotic responses, evidenced by increments of nuclear NF-κB p65, TNF-α, IL-1ß, cleaved caspase-3, Bax as well as decreased expression of Bcl-2 in hippocampus. Interestingly, STZ treatment led to up-regulation of protein and mRNA of CysLT1R in hippocampus. Of note, consecutive oral administration of montelukast (1 or 2mg/kg, 3 weeks) remarkably attenuated these effects induced by STZ. However, montelukast had no effect on normal mice. These results suggest that montelukast improves memory impairment and inhibits neuroinflammation and apoptosis in mice exposed to STZ. Montelukast may provide a novel strategy for treating or preventing AD.


Assuntos
Acetatos/uso terapêutico , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/etiologia , Quinolinas/uso terapêutico , Estreptozocina/toxicidade , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Ciclopropanos , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Interleucina-1beta/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/metabolismo , Receptores de Leucotrienos/metabolismo , Sulfetos , Fator de Necrose Tumoral alfa/metabolismo , Proteína X Associada a bcl-2
15.
Metab Brain Dis ; 31(3): 663-71, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26864581

RESUMO

Amyloid-ß deposition is thought to be associated with memory deficits, neuroinflammation, apoptotic responses, and progressive neuronal death manifested in Alzheimer's disease. Peroxisome proliferator-activated receptor δ (PPARδ) is a transcription factor with potent anti-inflammatory effect. In the current study, the effect of GW0742, a selective PPARδ agonist, on Aß1-42-induced neurotoxicity was investigated in the hippocampus of mice. Intra-hippocampal infusion of aggregated Aß1-42 oligomer (410pmol/mouse) remarkably damaged learning and memory in the Morris water maze (MWM) and Y-maze tests, accompanied by decreased expression of PPARδ in the hippocampus as confirmed by Western blot. Intra-hippocampal infusion of GW0742 (1.06 mM/mouse) significantly improved Aß1-42-induced memory deficits in mice, reversed Aß1-42-induced hippocampal PPARδ down-regulation and repressed Aß1-42-triggered neuroinflammatory and apoptotic responses, indicated by decreased nuclear NF-κB p65, TNF-α, IL-1ß as well as a decrease in cleaved caspase-3 and increased ratio of Bcl-2/Bax in the hippocampus. These results suggest that PPARδ activation ameliorates Aß1-42-induced hippocampal neurotoxicity, and it might play a crucial role in Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Hipocampo/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , PPAR delta/agonistas , Fragmentos de Peptídeos/toxicidade , Tiazóis/farmacologia , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Hipocampo/metabolismo , Interleucina-1beta/metabolismo , Masculino , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/metabolismo , Camundongos , Camundongos Endogâmicos ICR , NF-kappa B/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , PPAR delta/metabolismo , Tiazóis/uso terapêutico , Fator de Necrose Tumoral alfa/metabolismo
16.
Psychopharmacology (Berl) ; 233(9): 1739-49, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26546369

RESUMO

RATIONALE: Numerous studies have demonstrated that neuroinflammation is associated with depression-like symptoms and neuropsychological disturbances, and cysteinyl leukotriene receptor 1 (CysLT1R) was reported to be involved in neuroinflammation. The pathophysiological role of CysLT1R has been reported in several types of brain damage. However, the role of CysLT1R in depression remains to be elucidated. OBJECTIVES: We aimed to investigate the effect of hippocampal CysLT1R downregulation on depressive behaviors and neuroinflammatory responses in mice exposed to chronic mild stress (CMS). RESULTS: We firstly found that expression of hippocampal CysLT1R was gradually increased over CMS exposure, while 3 weeks treatment with fluoxetine reversed the increment of hippocampal CysLT1R expression. Hippocampal CysLT1R knockdown suppressed CMS-induced depressive-like behaviors as evidenced by decreases in immobility time in tail suspension test (TST), decreased latency to feed in novelty-suppressed feeding (NSF) test, and by increase in the number of entries and decrease in time spent in the open arm in elevated plus maze (EPM) test. Increments of hippocampal NF-κB p65, IL-1ß, and TNF-α induced by CMS were also prevented by hippocampal CysLT1R knockdown beforehand. CONCLUSIONS: Hippocampal CysLT1R participates in depression, and knockdown of hippocampal CysLT1R prevents CMS-induced depressive-like behaviors and neuroinflammation, suggesting that suppression of CysLT1R could prevent the development of depression.


Assuntos
Depressão/etiologia , Depressão/genética , Hipocampo/metabolismo , Inflamação/complicações , Inflamação/genética , Receptores de Leucotrienos/genética , Estresse Psicológico/metabolismo , Estresse Psicológico/psicologia , Animais , Antidepressivos de Segunda Geração/farmacologia , Ansiedade/psicologia , Citocinas/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Fluoxetina/farmacologia , Técnicas de Silenciamento de Genes , Elevação dos Membros Posteriores/psicologia , Hipocampo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR
17.
Br J Pharmacol ; 173(2): 372-85, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26507867

RESUMO

BACKGROUND AND PURPOSE: There is emerging evidence suggesting that abnormal transport of amyloid-ß (Aß) across the blood-brain barrier (BBB) is involved in diabetes-associated cognitive decline. We investigated whether PPARγ agonists restore Aß transport across the BBB and hippocampal plasticity in db/db mice. EXPERIMENTAL APPROACH: Efflux and influx of Aß across the BBB were determined by stereotaxic intra-cerebral or i.a. infusion of [(125) I]-Aß1-40 respectively. Receptor for advanced glycation end products (RAGE) and low-density lipoprotein receptor-related protein 1 (LRP1), which are involved in Aß influx and efflux, PPARγ and NF-κB p65 at the BBB, as well as hippocampal Aß, caspase-3, Bax and Bcl-2 were assayed by Western blot, immunohistochemistry and RT-PCR. In vivo, hippocampal LTP was recorded, and Morris water maze and Y-maze tasks were performed. KEY RESULTS: Treatment with PPARγ agonists, rosiglitazone (0.8 mg·kg(-1) ) and pioglitazone (9.0 mg·kg(-1) ), for 6 weeks significantly increased Aß efflux and decreased Aß influx across the BBB in db/db mice. Concomitantly, they decreased hippocampal Aß1-40 and Aß1-42 , suppressed neuronal apoptosis, as indicated by decreased caspase-3 activity and increased ratio of Bcl-2/Bax, and increased hippocampal plasticity, characterized by an enhanced in vivo LTP and better performance in behavioural tests. Furthermore, the PPARγ agonists induced the expression of LRP1 gene by activation of PPARγ and suppressed RAGE gene expression by inactivation of NF-κB signalling at the BBB of db/db mice. CONCLUSIONS AND IMPLICATIONS: PPARγ agonists modify abnormal Aß transport across the BBB and this is accompanied by amelioration of ß-amyloidosis and an improvement in hippocampal plasticity in diabetic mice.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/metabolismo , Hipocampo/metabolismo , Plasticidade Neuronal/fisiologia , PPAR gama/agonistas , PPAR gama/metabolismo , Fragmentos de Peptídeos/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Pioglitazona , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Rosiglitazona , Tiazolidinedionas/farmacologia
18.
Int J Neuropsychopharmacol ; 17(4): 581-92, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24229499

RESUMO

Deposition of extracellular amyloid-ß (Aß) peptide is one of the pathological hallmarks of Alzheimer's disease (AD). Accumulation of Aß is thought to associate with cognition deficits, neuroinflammation and apoptosis observed in AD. However, effective neuroprotective approaches against Aß neurotoxicity are unavailable. In the present study, we analysed the effects of pranlukast, a selective cysteinyl leukotriene receptor 1 (CysLT1R) antagonist, on the impairment of learning and memory formation induced by Aß and the probable underlying electrophysiological and molecular mechanisms. We found that bilateral intrahippocampal injection of Aß1₋42 resulted in a significant decline of spatial learning and memory of mice in the Morris water maze (MWM) and Y-maze tests, together with a serious depression of in vivo hippocampal long-term potentiation (LTP) in the CA1 region of the mice. Importantly, this treatment caused significant increases in CysLT1R expression and subsequent NF-κB signaling, caspase-3 activation and Bcl-2 downregulation in the hippocampus or prefrontal cortex. Oral administration of pranlukast at 0.4 or 0.8 mg/kg for 4 wk significantly reversed Aß1₋42-induced impairments of cognitive function and hippocampal LTP in mice. Furthermore, pranlukast reversed Aß1₋42-induced CysLT1R upregulation, and markedly suppressed the Aß1₋42-triggered NF-κB pathway, caspase-3 activation and Bcl-2 downregulation in the hippocampus and prefrontal cortex in mice. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay confirmed its presence in the brain after oral administration of pranlukast in mice. These data disclose novel findings about the therapeutic potential of pranlukast, revealing a previously unknown therapeutic possibility to treat memory deficits associated with AD.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Cromonas/farmacologia , Transtornos Cognitivos/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Antagonistas de Leucotrienos/farmacologia , Fragmentos de Peptídeos/farmacologia , Peptídeos beta-Amiloides/administração & dosagem , Animais , Comportamento Animal/efeitos dos fármacos , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/fisiopatologia , Cromonas/administração & dosagem , Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Hipocampo/fisiopatologia , Aprendizagem/efeitos dos fármacos , Antagonistas de Leucotrienos/administração & dosagem , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Memória/efeitos dos fármacos , Camundongos Endogâmicos ICR , Fragmentos de Peptídeos/administração & dosagem , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiopatologia , Receptores de Leucotrienos/efeitos dos fármacos
19.
Neurobiol Aging ; 35(3): 590-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24269024

RESUMO

Accumulation of amyloid-ß (Aß) is thought to be associated with the progressive neuronal death observed in Alzheimer's disease, but the mechanisms underlying neurotoxicity triggered by Aß remain elusive. In the current study, we investigated the roles of cysteinyl leukotriene receptor 1 (CysLT1R) in Aß1-42-induced neurotoxicity in vitro or in vivo. In vitro exposure of mouse primary neurons to Aß1-42 caused a gradual increases in CysLT1R expression. In vivo bilateral intrahippocampal injection of Aß1-42 also elicited time-dependent increases of CysLT1R expression in the hippocampus and cortex of mice. The CysLT1R antagonist pranlukast not only reversed Aß1-42-induced upregulation of CysLT1R, but also suppressed Aß1-42-triggered neurotoxicity evidenced by enhanced nuclear factor-kappa B p65, activated caspase-3, decreased B-cell lymphoma-2 and cell viability and impaired memory. Furthermore, chronic treatment with pranlukast produced similar beneficial effects on memory behavior and hippocampal long-term potentiation to memantine or donepezil in intrahippocampal Aß1-42-injected mice. Our data indicate that CysLT1R is involved in Aß1-42-induced neurotoxicity, and that blockade of CysLT1R, such as application of CysLT1R antagonist, could be a novel and promising strategy for the treatment of Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Córtex Cerebral/metabolismo , Expressão Gênica/efeitos dos fármacos , Hipocampo/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fragmentos de Peptídeos/toxicidade , Receptores de Leucotrienos/genética , Receptores de Leucotrienos/fisiologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Cromonas/farmacologia , Cromonas/uso terapêutico , Hipocampo/citologia , Antagonistas de Leucotrienos/farmacologia , Antagonistas de Leucotrienos/uso terapêutico , Masculino , Memória , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Terapia de Alvo Molecular , NF-kappa B/metabolismo , Neurônios/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Leucotrienos/metabolismo , Regulação para Cima/efeitos dos fármacos
20.
Neurochem Int ; 62(3): 340-7, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23318673

RESUMO

Although the pathogenesis of sporadic Alzheimer's disease (AD) is not clearly understood, neuroinflammation has been known to play a role in the pathogenesis of AD. To investigate a functional link between the neuroinflammation and AD, the effect of leukotriene D4 (LTD4), an inflammatory lipid mediator, was studied on amyloid-ß generation in vitro. Application of LTD4 to cell monolayers at concentrations up to 40 nM LTD4 caused increases in the Aß releases. Concentrations ≥ 40 nM LTD4 decreased neuronal viability. Application of 20 nM LTD4 caused a significant increase in Aß generation, as assessed by ELISA or Western blotting, without significant cytotoxicity. At this concentration, exposure of neurons to LTD4 for 24h produced maximal effect in the Aß generation, and significant increases in the expressions of cysteinyl leukotriene 1 receptor (CysLT(1)R) and activity of ß- or γ-secretase with complete abrogation by the selective CysLT(1)R antagonist pranlukast. Exposure of neurons to LTD4 for 1h showed activation of NF-κB pathway, by assessing the levels of p65 or phospho-p65 in the nucleus, and either CysLT(1)R antagonist pranlukast or NF-κB inhibitor PDTC prevented the nuclear translocation of p65 and the consequent phosphorylation. PDTC also inhibited LTD4-induced elevations of ß- or γ-secretase activity and Aß generation in vitro. Overall, our data show for the first time that LTD4 causes Aß production by enhancement of ß- or γ-secretase resulting from activation of CysLT(1)R-mediated NF-κB signaling pathway. These findings provide a novel pathologic link between neuroinflammation and AD.


Assuntos
Peptídeos beta-Amiloides/biossíntese , Leucotrieno D4/farmacologia , NF-kappa B/metabolismo , Neurônios/metabolismo , Receptores de Leucotrienos/fisiologia , Animais , Sequência de Bases , Western Blotting , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Camundongos , Camundongos Endogâmicos ICR , Reação em Cadeia da Polimerase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA