Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Kidney Int ; 100(3): 570-584, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34051264

RESUMO

Ischemia-reperfusion injury is a major cause of acute kidney injury. Recent studies on the pathophysiology of ischemia-reperfusion-induced acute kidney injury showed that immunologic responses significantly affect kidney ischemia-reperfusion injury and repair. Nuclear factor (NF)-ĸB signaling, which controls cytokine production and cell survival, is significantly involved in ischemia-reperfusion-induced acute kidney injury, and its inhibition can ameliorate ischemic acute kidney injury. Using EXPLOR, a novel, optogenetically engineered exosome technology, we successfully delivered the exosomal super-repressor inhibitor of NF-ĸB (Exo-srIĸB) into B6 wild type mice before/after kidney ischemia-reperfusion surgery, and compared outcomes with those of a control exosome (Exo-Naïve)-injected group. Exo-srIĸB treatment resulted in lower levels of serum blood urea nitrogen, creatinine, and neutrophil gelatinase-associated lipocalin in post-ischemic mice than in the Exo-Naïve treatment group. Systemic delivery of Exo-srIĸB decreased NF-ĸB activity in post-ischemic kidneys and reduced apoptosis. Post-ischemic kidneys showed decreased gene expression of pro-inflammatory cytokines and adhesion molecules with Exo-srIĸB treatment as compared with the control. Intravital imaging confirmed the uptake of exosomes in neutrophils and macrophages. Exo-srIĸB treatment also significantly affected post-ischemic kidney immune cell populations, lowering neutrophil, monocyte/macrophage, and T cell frequencies than those in the control. Thus, modulation of NF-ĸB signaling through exosomal delivery can be used as a novel therapeutic method for ischemia-reperfusion-induced acute kidney injury.


Assuntos
Injúria Renal Aguda , Exossomos , Traumatismo por Reperfusão , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/prevenção & controle , Animais , Rim , Camundongos , Camundongos Endogâmicos C57BL , Inibidor de NF-kappaB alfa , Traumatismo por Reperfusão/prevenção & controle
2.
Am J Reprod Immunol ; 85(2): e13329, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32846024

RESUMO

The extracellular vesicles (EVs) research area has grown rapidly because of their pivotal roles in intercellular communications and maintaining homeostasis of individual organism. As a subtype of EVs, exosomes are made via unique biogenesis pathway and exhibit disparate functional and phenotypic characteristics. Functionally, exosomes transfer biological messages from donor cell to recipient cell, which makes exosomes as a novel therapeutic platform delivering therapeutic materials to the target tissue/cell. Currently, both academia and industry try to develop exosome platform-based therapeutics for disease management, some of which are already in clinical trials. In this review, we will discuss focusing on therapeutic values of exosomes, recent advances in therapeutic exosome platform development, and late development of exosome therapeutics in diverse therapeutic areas.


Assuntos
Terapia Biológica/métodos , Exossomos/metabolismo , Animais , Comunicação Celular , Sistemas de Liberação de Medicamentos , Vesículas Extracelulares , Humanos
3.
J Ethnopharmacol ; 232: 119-129, 2019 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-30576772

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Polygala tenuifolia Willdenow root extract (BT-11) has beneficial effects on central nervous system disorders in human. The safety of BT-11 should be elucidated in younger person further. AIM OF THE STUDY: To evaluate the safety of BT-11 in human aged from 9 to 19 years old. MATERIAL AND METHODS: The safety was evaluated in randomly assigned subjects who received the test products (61 subjects in BT-11 300 mg daily or 60 subjects in matching placebo) for 12 weeks. Adverse reactions were analyzed by the incidence rate, type, and severity. The clinical examination included hematology and blood chemistry tests, urinalysis, vital signs, body weight, and electrocardiogram (ECG). RESULTS: Eleven adverse reactions were observed in ten subjects receiving BT-11 while seven adverse reactions in six subjects receiving placebo. There were no statistical differences in the incidence of adverse reactions between the two groups. Serious adverse reactions such as acute appendicitis and acute viral gastroenteritis were observed in the BT-11 group4 and the placebo group, respectively. However, it was confirmed that they were not associated with the test product. All other adverse reactions observed during the test period were resolved completely without special treatment. No statistical difference was also observed in safety laboratory tests, vital signs, and ECG between two groups. CONCLUSIONS: This study demonstrates the safety of BT-11 in the adolescent by showing no apparent adverse reactions related to it.


Assuntos
Extratos Vegetais/administração & dosagem , Polygala , Adolescente , Adulto , Criança , Método Duplo-Cego , Feminino , Humanos , Masculino , Raízes de Plantas , Pós , Adulto Jovem
4.
CNS Neurol Disord Drug Targets ; 15(8): 935-944, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27577574

RESUMO

We previously demonstrated that dehydroevodiamine•HCl (DHED), which was purified from Evodia rutaecarpa Bentham (Rutaceae), has beneficial effects on memory impairment and neuronal damage in three disease models. To investigate the preventive action of DHED in Alzheimer's disease (AD), a progressive neurodegenerative disorder characterized by memory decline, amyloid-ß (Aß) protein-containing neuritic plaques and neurofibrillary tangles, in this study, we proposed that DHED may be therapeutically effective against the memory impairment and disease-related neurochemical changes that occur in Tg2576 (Tg) mice. DHED (0.5 mg/kg) was intraperitoneally administered to 7-month-old Tg and wild type mice for 4 months. In passive avoidance and water maze tests, DHED improved memory impairment of Tg mice after 4 months of administration. DHED also reduced cortical levels of soluble Aß40, soluble Aß42 and total Aß peptides in the Tg mice. Additionally, we investigated whether DHED may be a ß-secretase inhibitor that affects the production of Aß related to the formation of neuritic plaques. DHED directly inhibited ß-secretase activity in a concentrationdependent manner. The concentration required for 50 % enzyme inhibition (IC50) was 40.96 µM, and DHED may act as a competitive inhibitor of ß-secretase. Moreover, DHED interacted strongly with BACE1 (ß-secretase 2QP8), as demonstrated in the analysis of the binding mode of DHED in the active site of human BACE1. In conclusion, DHED may exhibit therapeutic effects for AD as a ß-secretase inhibitor.


Assuntos
Alcaloides/uso terapêutico , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Córtex Cerebral/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análise de Variância , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/genética , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Mutação/genética , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/genética
5.
J Neurosci Res ; 93(8): 1267-78, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25711470

RESUMO

Monoamine oxidase B (MAO-B) is well known as a therapeutic target for Parkinson's disease (PD). MAO-B inhibitors retain antiparkinsonism abilities to improve motor function and prevent neuronal loss by decreasing dopamine metabolism and oxidative stress in the brain. From the study to find novel antiparkinsonism drugs that can inhibit MAO-B activity, neuronal loss, and behavioral deficits in the mouse model of PD, we identified that 1-[2-(4-benzyloxyphenoxy)ethyl]imidazole (BPEI) or safinamide strongly and selectively inhibited MAO-B activities in a dose-dependent manner (IC50 of BPEI and safinamide for MAO-B were 0.016 and 0.0021 µM and for MAO-A were 70.0 and 370 µM, respectively). In ex vivo studies after an administration (30 mg/kg, i.p.) of BPEI or safinamide to normal mice, the MAO-B activity in the brain was reduced by up to 90.6% or 82.4% at 1.0 hr. BPEI (20 mg/kg, i.p.) or safinamide (20 mg/kg, i.p.) significantly reversed the behavioral impairments, dopamine levels in the striatum, and neuronal loss in the substantia nigra of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice compared with the MPTP-alone-treated group. In the 6-hydroxydopamine-induced PD rat model, behavioral improvement by levodopa sparing activity was observed in the BPEI- or safinamide-treated (20 mg/kg, i.p.) rats. Moreover, BPEI revealed additional curative activities for nonmotor symptoms of PD such as pain, anxiety, epilepsy, and depression in rodent disease models. Therefore, BPEI has broad therapeutic potential for treating motor symptoms via strong and selective inhibitory effects on MAO-B, with additional benefits for comorbid symptoms in PD.


Assuntos
Modelos Animais de Doenças , Imidazóis/uso terapêutico , Intoxicação por MPTP/enzimologia , Intoxicação por MPTP/prevenção & controle , Inibidores da Monoaminoxidase/uso terapêutico , Neurônios/enzimologia , Animais , Contagem de Células , Imidazóis/farmacologia , Intoxicação por MPTP/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Monoaminoxidase/metabolismo , Inibidores da Monoaminoxidase/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Doença de Parkinson/enzimologia , Doença de Parkinson/patologia , Doença de Parkinson/prevenção & controle , Ratos , Ratos Sprague-Dawley
6.
Neurosci Lett ; 557 Pt B: 95-100, 2013 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-24176882

RESUMO

Medium spiny neurons (MSNs) are the major type of neurons found in the striatum. The dendritic spines on these cells contain glutamatergic synaptic contacts between the cortex (or the thalamus) and the striatum. The complexity of the dendritic structure of MSNs may therefore reflect the functional status of the basal ganglia because the striatum is the major input structure in which signals from different regions are integrated. We examined the structural alterations in the dendrites of striatal MSNs in an 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of Parkinson's disease (PD). Acute MPTP treatment rapidly damaged dopaminergic neurons and their terminals within the striatum and caused behavioral impairments. However, mice injected with MPTP spontaneously recovered from these behavioral impairments within one week. This recovery was accompanied by the restoration of dendritic structures on MSNs, but the damage to dopaminergic neurons remained extensive. Furthermore, we demonstrated that rasagiline, a monoamine oxidase-B (MAO-B) inhibitor that has been shown to be efficacious for PD, could enhance the dendritic complexity of cultured MSNs. The effect of rasagiline on the spine-like structures of dendrites, however, appears not to require DA availability because the small protrusions of dendrites in cultured MSNs without major source of DA input was similarly changed by rasagiline. Our data suggest that the dendritic structures of striatal MSNs change dynamically, reflecting the progression of motor-related symptoms in PD, and the restoration of functional synapses in the MSNs of PD patients may constitute a clinical target for symptomatic alleviation.


Assuntos
Corpo Estriado/patologia , Dendritos/ultraestrutura , Transtornos Parkinsonianos/patologia , Animais , Comportamento Animal , Células Cultivadas , Modelos Animais de Doenças , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Recuperação de Função Fisiológica
7.
PLoS One ; 7(9): e44259, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028510

RESUMO

The p53 and NF-κB pathways play important roles in diverse cellular functions, including cell growth, apoptosis, and tumorigenesis. Mutations that inactivate the p53 gene and constitutive NF-κB pathway activation are common occurrences in human cancers. Although many drugs are being developed that selectively activate p53 or inhibit NF-κB, there are few drug candidates that can do both. Simultaneous activation of p53 and inhibition of the NF-κB pathway is therefore a prime target for new cancer drug development. This study is the first report of a high-throughput approach with mass compounds that concurrently target both pathways. Using a cell-based screening assay and a library of 200,000 synthetic compounds, we identified 9 small molecules that simultaneously inhibit NF-κB and activate p53. One of these compounds, N-2, increased the expression of p53 target genes, including p21 and GADD45a. In addition, N-2 inhibited the transcriptional activity of NF-κB, concomitantly repressing interleukin-6 and monocyte chemotactic protein-1 (MCP-1) expression. When cell lines derived from a diverse range of cancers were treated in vitro with N-2, we observed increased cell death. N-2 also significantly inhibited allograft growth in murine models of melanoma and lung carcinoma. Our findings suggest that N-2 may act as a bivalent anti-cancer agent through simultaneous modulation of NF-κB and p53 activities.


Assuntos
Antineoplásicos/farmacologia , NF-kappa B/antagonistas & inibidores , Fenantridinas/farmacologia , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/classificação , Linhagem Celular Tumoral , Quimiocina CCL2/genética , Ensaios de Seleção de Medicamentos Antitumorais , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes , Ensaios de Triagem em Larga Escala , Humanos , Interleucina-6/genética , Dose Letal Mediana , Camundongos , NF-kappa B/genética , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Fenantridinas/química , Fenantridinas/classificação , Piridinas/química , Piridinas/classificação , Bibliotecas de Moléculas Pequenas , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética , Transcriptoma , Proteína Supressora de Tumor p53/genética
8.
J Neurochem ; 109(5): 1483-96, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19457162

RESUMO

Although there is known to be a marked concentration of reactive microglia in the substantia nigra pars compacta (SNpc) of patients with Parkinson's disease (PD), a disorder in which alpha-synuclein plays a key pathogenic role, the specific roles of alpha-synuclein and microglia remains poorly understood. In this study, we investigated the effects of alpha-synuclein and the mechanisms of invasive microglial migration into the SNpc. We show that alpha-synuclein up-regulates the expressions of the cell adhesion molecule CD44 and the cell surface protease membrane-type 1 matrix metalloproteinase through the extracellular regulated kinases 1/2 pathway. These concurrent inductions increased the generation of soluble CD44 to liberate microglia from the surrounding extracellular matrix for migration. The effects of alpha-synuclein were identical in BV-2 murine microglial cells subjected to cDNA transfection and extracellular treatment. These inductions in primary microglial cultures of C57Bl/6 mice were identical to those in BV-2 cells. alpha-Synuclein-induced microglial migration into the SNpc was confirmed in vivo using a 6-hydroxydopamine mouse model of PD. Our data demonstrate a correlation between alpha-synuclein-induced phenotypic changes and microglial migration. With the recruitment of the microglial population into the SNpc during dopaminergic neurodegeneration, alpha-synuclein may play a role in accelerating the pathogenesis of PD.


Assuntos
Movimento Celular/fisiologia , Receptores de Hialuronatos/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Microglia/fisiologia , Regulação para Cima/fisiologia , alfa-Sinucleína/fisiologia , Animais , Encéfalo/citologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Transplante de Células/métodos , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Receptores de Hialuronatos/genética , Metaloproteinase 14 da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Oxidopamina/toxicidade , Doença de Parkinson/etiologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Doença de Parkinson/cirurgia , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Substância Negra/metabolismo , Substância Negra/cirurgia , Transfecção , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , alfa-Sinucleína/genética
9.
J Neurosci Res ; 87(1): 260-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18712849

RESUMO

In Oriental medicine, roots of Polygala tenuifolia Willdenow have been known to be an important herb that exhibits sedative effects in insomnia, palpitation with anxiety, restlessness, and disorientation in humans. We previously reported that BT-11, extracted from those roots, improved scopolamine-induced amnesia in rats and inhibited acetylcholinesterase activities in vitro. Therefore, we proposed that BT-11 could remedy stress-induced memory deficits in rats. In this study, the stress-induced memory impairments in rats were significantly reversed almost to the control level by BT-11 treatment. To seek an active component of BT-11 that plays an important role in antipsychotic effects, we compared BT-11 with 3,4,5-trimethoxycinnamic acid (TMCA), which is a constituent of those root extracts. However, the effects of TMCA were less or were not consistent with those of BT-11 in some of tests. In particular, BT-11 reversed the stress-induced reduction of glucose utilization by [(18)fluorodeoxyglucose]FDG-PET and the levels of neural cell adhesion molecule (NCAM) in rat brains to the control levels, whereas TMCA did not. Therefore, BT-11 improved stress-induced memory impairments through increment of glucose utilization and total NCAM levels in rat brains. In conclusion, BT-11 may be strongly effective against stress-induced amnesia in rats, through the combined effects of TMCA and other active components of BT-11.


Assuntos
Encéfalo/metabolismo , Glucose/metabolismo , Transtornos da Memória/tratamento farmacológico , Moléculas de Adesão de Célula Nervosa/metabolismo , Fitoterapia/métodos , Polygala/química , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Cicloexilaminas/uso terapêutico , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Fluordesoxiglucose F18/metabolismo , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , Transtornos da Memória/fisiopatologia , Tomografia por Emissão de Pósitrons/métodos , Ratos , Ratos Wistar , Estresse Psicológico/complicações
10.
J Neurosci Res ; 85(11): 2500-11, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17600377

RESUMO

To develop a novel and effective drug that could enhance cognitive function and neuroprotection, we newly synthesized maltolyl p-coumarate by the esterification of maltol and p-coumaric acid. In the present study, we investigated whether maltolyl p-coumarate could improve cognitive decline in scopolamine-injected rats and in amyloid beta peptide(1-42)-infused rats. Maltolyl p-coumarate was found to attenuate cognitive deficits in both rat models using passive avoidance test and to reduce apoptotic cell death observed in the hippocampus of the amyloid beta peptide(1-42)-infused rats. We also examined the neuroprotective effects of maltolyl p-coumarate in vitro using SH-SY5Y cells. Cells were pretreated with maltolyl p-coumarate, before exposed to amyloid beta peptide(1-42), glutamate or H2O2. We found that maltolyl p-coumarate significantly decreased apoptotic cell death and reduced reactive oxygen species, cytochrome c release, and caspase 3 activation. Taking these in vitro and in vivo results together, our study suggests that maltolyl p-coumarate is a potentially effective candidate against Alzheimer's disease that is characterized by wide spread neuronal death and progressive decline of cognitive function.


Assuntos
Ácidos Cumáricos/farmacologia , Demência/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Pironas/farmacologia , Peptídeos beta-Amiloides/toxicidade , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3/efeitos dos fármacos , Cognição/efeitos dos fármacos , Citocromos c/efeitos dos fármacos , Demência/induzido quimicamente , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Marcação In Situ das Extremidades Cortadas , Antagonistas Muscarínicos/toxicidade , Fármacos Neuroprotetores/química , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Escopolamina/toxicidade
11.
Neuropsychopharmacology ; 32(11): 2393-404, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17406652

RESUMO

Minocycline is a semi-synthetic tetracycline antibiotic that effectively crosses the blood-brain barrier. Minocycline has been reported to have significant neuroprotective effects in models of cerebral ischemia, traumatic brain injury, amyotrophic lateral sclerosis, and Huntington's and Parkinson's diseases. In this study, we demonstrate that minocycline has neuroprotective effects in in vitro and in vivo Alzheimer's disease models. Minocycline was found to attenuate the increases in the phosphorylation of double-stranded RNA-dependent serine/threonine protein kinase, eukaryotic translation initiation factor-2 alpha and caspase 12 activation induced by amyloid beta peptide1-42 treatment in NGF-differentiated PC 12 cells. In addition, increases in the phosphorylation of eukaryotic translation initiation factor-2 alpha were attenuated by administration of minocycline in Tg2576 mice, which harbor mutated human APP695 gene including the Swedish double mutation and amyloid beta peptide(1-42)-infused rats. We found that minocycline administration attenuated deficits in learning and memory in amyloid beta peptide(1-42)-infused rats. Increased phosphorylated state of eukaryotic translation initiation factor-2 alpha is observed in Alzheimer's disease patients' brains and may result in impairment of cognitive functions in Alzheimer's disease patients by decreasing the efficacy of de novo protein synthesis required for synaptic plasticity. On the basis of these results, minocycline may prove to be a good candidate as an effective therapeutic agent for Alzheimer's disease.


Assuntos
Doença de Alzheimer/complicações , Transtornos Cognitivos , Minociclina/uso terapêutico , Neurônios/patologia , Fármacos Neuroprotetores/uso terapêutico , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/genética , Peptídeos beta-Amiloides , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Encéfalo/patologia , Estudos de Casos e Controles , Morte Celular/efeitos dos fármacos , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/patologia , Modelos Animais de Doenças , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Minociclina/farmacologia , Fator de Crescimento Neural/farmacologia , Fármacos Neuroprotetores/farmacologia , Células PC12/efeitos dos fármacos , Fragmentos de Peptídeos , Ratos , Ratos Wistar , Transfecção
12.
Mol Cell Biol ; 26(11): 4327-38, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16705182

RESUMO

Amyloid precursor protein (APP) has eight potential phosphorylation sites in its cytoplasmic domain. Recently, it has demonstrated that the constitutive phosphorylation of APP at T668 (APP695 isoform numbering) was observed specifically in the brain. Neuron-specific phosphorylation of APP at T668 is thought to be important for neuronal functions of APP, although its exact physiological significance remains to be clarified. In this study, we show that the phosphorylation of the APP intracellular domain (AICD) at T668 is essential for its binding to Fe65 and its nuclear translocation and affects the resultant neurotoxicity, possibly mediated through the induction of glycogen synthase kinase 3beta and tau phosphorylation by enhancing the formation of a ternary complex with Fe65 and CP2 transcription factor. Taken together, these results suggest that the phosphorylation of AICD at T668 contributes to the neuronal degeneration in Alzheimer's disease (AD) by regulating its translocation into the nucleus and then affects neurodegeneration; therefore, the specific inhibitor of T668 phosphorylation might be the target of AD therapy.


Assuntos
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Núcleo Celular/metabolismo , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Treonina/metabolismo , Transporte Ativo do Núcleo Celular , Doença de Alzheimer/patologia , Animais , Encéfalo/citologia , Encéfalo/patologia , Morte Celular , Células Cultivadas , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Humanos , Camundongos , Camundongos Transgênicos , Mutação/genética , Fator de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Células PC12 , Fosforilação , Estrutura Terciária de Proteína , Transporte Proteico , Ratos , Proteínas tau/metabolismo
13.
FASEB J ; 20(6): 729-31, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16467370

RESUMO

Although chronic stress is known to be linked with memory and other neurological disorders, little is known about the relationship between chronic stress and the onset or development of Alzheimer's disease (AD). In this study, we investigated the effects of long-term stress on the onset and severity of cognitive deficits and pathological changes in APPV717I-CT100 mice overexpressing human APP-CT100 containing the London mutation (V717I) after exposure to immobilization stress. We found that chronic immobilization stress accelerated cognitive impairments, as accessed by the Passive avoidance and the Social Transfer of Food Preference (STFP) tests. Moreover, the numbers and densities of vascular and extracellular deposits containing amyloid beta peptide (Abeta) and carboxyl-terminal fragments of amyloid precursor protein (APP-CTFs), which are pathologic markers of AD, were significantly elevated in stressed animals, especially in the hippocampus. Moreover, stressed animals, also showed highly elevated levels of neurodegeneration and tau phosphorylation and increased intraneuronal Abeta and APP-CTFs immunoreactivities in the hippocampus and in the entorhinal and piriform cortex. This study provides the first evidence that chronic stress accelerates the onset and severity of cognitive deficits and that these are highly correlated with pathological changes, which thus indicates that chronic stress may be an important contributor to the onset and development of AD.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Deficiências da Aprendizagem/metabolismo , Transtornos da Memória/metabolismo , Estresse Psicológico/metabolismo , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Córtex Cerebral/metabolismo , Doença Crônica , Corticosterona/sangue , Modelos Animais de Doenças , Hipocampo/metabolismo , Camundongos , Camundongos Transgênicos , Restrição Física/efeitos adversos , Estresse Psicológico/genética
14.
Mol Pharmacol ; 69(1): 76-84, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16223958

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) exert anti-inflammatory, analgesic, and antipyretic activities and suppress prostaglandin synthesis by inhibiting cyclooxygenase, an enzyme that catalyzes the formation of prostaglandin precursors from arachidonic acid. Epidemiological observations indicate that the long-term treatment of patients suffering from rheumatoid arthritis with NSAIDs results in reduced risk and delayed onset of Alzheimer's disease. In this study, we investigated the therapeutic potential for Alzheimer's disease of mefenamic acid, a commonly used NSAID that is a cyclooxygenase-1 and 2 inhibitor with only moderate anti-inflammatory properties. We found that mefenamic acid attenuates the neurotoxicities induced by amyloid beta peptide (Abeta)(1-42) treatment and the expression of a Swedish double mutation (KM595/596NL) of amyloid precursor protein (Swe-APP) or the C-terminal fragments of APP (APP-CTs) in neuronal cells. We also show that mefenamic acid decreases the production of the free radical nitric oxide and reduces cytochrome c release from mitochondria induced by Abeta(1-42), Swe-APP, or APP-CTs in neuronal cells. In addition, mefenamic acid up-regulates expression of the antiapoptotic protein Bcl-X(L). Moreover, our study demonstrates for the first time that mefenamic acid improves learning and memory impairment in an Abeta(1-42)-infused Alzheimer's disease rat model. Taking these in vitro and in vivo results together, our study suggests that mefenamic acid could be used as a therapeutic agent in Alzheimer's disease.


Assuntos
Doença de Alzheimer/fisiopatologia , Anti-Inflamatórios não Esteroides/farmacologia , Transtornos Cognitivos/tratamento farmacológico , Ácido Mefenâmico/farmacologia , Fármacos Neuroprotetores/farmacologia , Peptídeos beta-Amiloides/metabolismo , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Caspase 3 , Caspases/metabolismo , Diferenciação Celular/efeitos dos fármacos , Ativação Enzimática , Masculino , Ácido Mefenâmico/uso terapêutico , Potenciais da Membrana/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Fator de Crescimento Neural/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Células PC12 , Fragmentos de Peptídeos/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Transfecção
15.
Int J Dev Neurosci ; 24(1): 81-5, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16289942

RESUMO

Huntington's disease (HD) is a dominant neurodegenerative disorder caused by the expansion of a CAG repeat in the gene encoding huntingtin. Moreover, the nuclear targeting of mutant huntingtin increases cellular toxicity, whereas normal huntingtin resides mainly in the cytoplasm, and is associated with membranes or microtubules. Huntingtin is enriched in neurons and its expression is increased during neural development. The inactivation of the HD gene results in embryonic lethality before nervous system development. Thus, huntingtin is critical during early embryonic development. Nevertheless, the function of huntingtin at this stage is unknown, even the distribution of the protein has not been described. The present study was undertaken to elucidate the distribution of huntingtin during the early developmental period in the mouse embryo. At the preimplantation stage, huntingtin was detected in nuclei up to 2.5 days post coitum (dpc), but disappeared from nuclei during the blastocyst stage (3.5 dpc). Following this stage, huntingtin was mainly localized in the cytoplasm and co-localized with mitotic spindles. These data suggest that the nuclear targeting of normal huntingtin is required during early embryo development in mice.


Assuntos
Blastocisto/metabolismo , Núcleo Celular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Animais , Blastocisto/citologia , Humanos , Proteína Huntingtina , Doença de Huntington , Imuno-Histoquímica , Camundongos , Oócitos/citologia , Oócitos/metabolismo
16.
FASEB J ; 18(13): 1615-7, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15289452

RESUMO

Though the etiology of Parkinson's disease (PD) remains unclear, alpha-synuclein (alpha-SN) is regarded as a major causative agent of PD. Several lines of evidence indicate that immunological abnormalities are associated with PD for unknown reasons. The present study was performed to assess whether peripheral blood mononuclear cells (PBMCs) show altered alpha-SN expression in PD patients and to identify its functions, which may be related to peripheral immune abnormalities in PD. alpha-SN was found to be expressed more in 151 idiopathic PD (IPD) patients than in 101 healthy controls, who nevertheless showed as age-dependent increases. By in vitro transfection, alpha-SN expression was shown to be correlated with glucocorticoid sensitive apoptosis, possibly caused by the enhanced expression of glucocorticoid receptor (GR), caspase activations (caspase-8, caspase-9), CD95 up-regulation, and reactive oxygen species (ROS) production. An understanding of the correlation between alpha-SN levels and apoptosis in the presence of the coordinated involvement of multiple processes would provide an insight into the molecular basis of the disease. The present study provides a clue that the alpha-SN may be one of the primary causes of the immune abnormalities observed in PD and offers new targets for pharmacotherapeutic intervention.


Assuntos
Apoptose , Regulação da Expressão Gênica , Linfócitos/imunologia , Linfócitos/patologia , Proteínas do Tecido Nervoso/metabolismo , Doença de Parkinson/imunologia , Doença de Parkinson/metabolismo , Estudos de Casos e Controles , Caspases/metabolismo , Células Cultivadas , Ativação Enzimática , Humanos , Linfócitos/enzimologia , Linfócitos/metabolismo , Proteínas do Tecido Nervoso/genética , Doença de Parkinson/genética , Doença de Parkinson/patologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Glucocorticoides/metabolismo , Sinucleínas , alfa-Sinucleína , Receptor fas/genética , Receptor fas/metabolismo
17.
J Neurosci Res ; 75(1): 117-24, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14689454

RESUMO

The AICD (APP intracellular Domain) and C31, caspase-cleaved C-terminal fragment of APP, have been found in Alzheimer's disease (AD) patients' brains and have been reported to induce apoptosis in neuronal cells. In recent, the C-terminal fragments of amyloid precursor protein (APP-CTs) have been reported to form a complex with Fe65 and the histone acetyltransferase Tip60 and are thought to be involved in gene transcription. In this study, based on the hypothesis that APP-CTs might exert neurotoxicity by inducing some gene transcription, we investigated the effects of APP-CTs on histone acetylation which indicates that transcription is actively going on and also on the relationship between histone acetylation and the cytotoxicity induced by APP-CTs in nerve growth factor (NGF)-differentiated PC12 cells and rat primary cortical neurons. Here we demonstrate that the expression of APP-CTs [C31, AICD (C59) and C99] induces increases in acetylation of histone 3 and histone 4 and that treatment with sodium butyrate, an inhibitor of histone deacetylase, significantly enhances the cytotoxicity induced by APP-CTs. The acetylation of histone plays an important role in allowing regulatory proteins to access DNA and is likely to be a major factor in the regulation of gene transcription. Taken together, our results suggest that APP-CTs exert neurotoxicity by transcription-dependent mechanisms and this might contribute to the pathogenesis of AD.


Assuntos
Precursor de Proteína beta-Amiloide/toxicidade , Histonas/metabolismo , Acetilação/efeitos dos fármacos , Precursor de Proteína beta-Amiloide/química , Animais , Apoptose/efeitos dos fármacos , Western Blotting/métodos , Butiratos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Interações Medicamentosas , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas/métodos , Indóis/metabolismo , L-Lactato Desidrogenase/metabolismo , Estrutura Molecular , Fator de Crescimento Neural/farmacologia , Células PC12 , Ratos , Ratos Sprague-Dawley , Transfecção
18.
FASEB J ; 17(13): 1951-3, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12923068

RESUMO

The AICD (amyloid precursor protein [APP] intracellular domain) and C31, the caspase-cleaved C-terminal fragment of APP, have been found in the brains of patients with Alzheimer's disease (AD). Here, we demonstrate for the first time that the C-terminal fragments of APP (AICD [C57, C59] and C31) exert neurotoxicity on differentiated PC 12 cells and rat primary cortical neurons by inducing the expression of glycogen synthase kinase 3beta, forming a ternary complex with Fe65 and CP2/LSF/LBP1 in the nucleus, whereas deletion mutants and a point mutant with Y682G of the YENPTY domain, a Fe65 binding domain, do not. Moreover, expression of APP770 and Swedish mutant form of APP increased the levels of C-terminal fragments of APP (APP-CTs) in neuronal cells and also induced the up-regulation of glycogen synthase kinase-3beta at both the mRNA and the protein levels. In addition, we show that CP2/LSF/LBP1 binding site (nt +0 to approximately +10) in human glycogen synthase kinase 3beta promoter region is essential for the induction of the gene transcription by APP-CTs. The neurotoxicities induced by APP-CTs (AICD and C31) were accompanied by an increase in the active form of glycogen synthase kinase-3beta, and by the induction of tau phosphorylation and a reduction in nuclear beta-catenin levels, and led to apoptosis.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Quinase 3 da Glicogênio Sintase/genética , Ativação Transcricional , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Quinase 3 da Glicogênio Sintase/biossíntese , Glicogênio Sintase Quinase 3 beta , Modelos Genéticos , Mutação , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Células PC12 , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/toxicidade , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA , Ratos , Fatores de Transcrição/metabolismo , Transfecção
19.
Arzneimittelforschung ; 53(6): 393-401, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12872609

RESUMO

The general pharmacological properties of indolo[2',3':3,4]pyrido[2,1-b]quinazolinium-5,7,8,13-tetrahydro-14- methyl-5-oxo-chloride (dehydroevodiamine-HCl, DHED, CAS 67909-49-3), a new potential anti-dementia agent, were studied to investigate side effects using various experimental animals. Both oral and intraperitoneal administration of DHED had no effects on the central nervous system except that they showed an analgesic activity. DHED had no significant effect on heart rate, blood pressure and coronary flow in isolated rat hearts. DHED had negligible effects on the autonomic nervous system and smooth muscle in isolated rat ileum, rat vas deferens and rat aorta. DHED did not influence the gastrointestinal system except that it inhibited the intestinal travel of a charcoal meal in mice. Neither blood coagulation mechanism nor liver function was affected by DHED. Therefore, it is concluded from these general pharmacological studies that DHED does not induce any serious side effects at the dose levels showing anticholinesterase and memory enhancing activities in the experimental animals.


Assuntos
Alcaloides/farmacologia , Demência/tratamento farmacológico , Animais , Sistema Nervoso Autônomo/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Sistema Digestório/efeitos dos fármacos , Cobaias , Hemodinâmica/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Testes de Função Hepática , Masculino , Camundongos , Camundongos Endogâmicos ICR , Músculo Liso/efeitos dos fármacos , Sistema Nervoso/efeitos dos fármacos , Coelhos , Ratos , Ratos Sprague-Dawley , Sistema Respiratório/efeitos dos fármacos
20.
FASEB J ; 17(11): 1556-8, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12824278

RESUMO

Many environmental factors during the pre- or postnatal period can affect an individual's cognitive function and neural development throughout life. Little is known, however, about the combined effects of the pre- and postnatal environments on cognitive function of adult offspring and structural alterations in the adult brain. In this study, we confirmed that pre- or postnatal stress impaired learning and memory performance of rats. Conversely, pre- or postnatal enriched housing improved behavioral performance. These experience-dependent behavioral alterations were consistent with changes in 5-bromo-2'-deoxyuridine-labeled cell number in the granule cell layer of the hippocampus and in the expression level of synaptic markers such as neuronal cell adhesion molecule and synaptophysin, and expression of a neurotrophic factor, brain-derived neurotrophic factor. Postnatal stress appeared to have no influence on cell proliferation, however. We did find that postnatal environment could attenuate prenatal effects partly via a longitudinal cross-housing study, in which pups born to mothers housed under enriched conditions were reared under stressful conditions and vice versa. These results suggest that postnatal environmental manipulations can counteract the cognitive alterations in early adulthood and the structural changes in the young adult brain induced by prenatal experience.


Assuntos
Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Cognição , Plasticidade Neuronal , Animais , Animais Recém-Nascidos , Encéfalo/fisiologia , Divisão Celular , Giro Denteado/citologia , Giro Denteado/embriologia , Giro Denteado/crescimento & desenvolvimento , Exposição Ambiental , Estudos Longitudinais , Masculino , Memória , Modelos Neurológicos , Proteínas do Tecido Nervoso/metabolismo , Ratos , Sinapses/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...