Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 24(22): 6492-504, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26358770

RESUMO

Glycosylation with O-linked ß-N-acetylglucosamine (O-GlcNAc) is one of the protein glycosylations affecting various intracellular events. However, the role of O-GlcNAcylation in neurodegenerative diseases such as Alzheimer's disease (AD) is poorly understood. Mitochondrial adenosine 5'-triphosphate (ATP) synthase is a multiprotein complex that synthesizes ATP from ADP and Pi. Here, we found that ATP synthase subunit α (ATP5A) was O-GlcNAcylated at Thr432 and ATP5A O-GlcNAcylation was decreased in the brains of AD patients and transgenic mouse model, as well as Aß-treated cells. Indeed, Aß bound to ATP synthase directly and reduced the O-GlcNAcylation of ATP5A by inhibition of direct interaction between ATP5A and mitochondrial O-GlcNAc transferase, resulting in decreased ATP production and ATPase activity. Furthermore, treatment of O-GlcNAcase inhibitor rescued the Aß-induced impairment in ATP production and ATPase activity. These results indicate that Aß-mediated reduction of ATP synthase activity in AD pathology results from direct binding between Aß and ATP synthase and inhibition of O-GlcNAcylation of Thr432 residue on ATP5A.


Assuntos
Doença de Alzheimer/metabolismo , ATPases Mitocondriais Próton-Translocadoras/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Fatores Acopladores da Fosforilação Oxidativa/metabolismo , Acetilglucosamina/metabolismo , Trifosfato de Adenosina/metabolismo , Doença de Alzheimer/enzimologia , Doença de Alzheimer/genética , Animais , Células CHO , Cricetulus , Modelos Animais de Doenças , Glicosilação , Células HeLa , Humanos , Camundongos , Camundongos Transgênicos , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , ATPases Mitocondriais Próton-Translocadoras/genética , Fatores Acopladores da Fosforilação Oxidativa/genética , Processamento de Proteína Pós-Traducional , beta-N-Acetil-Hexosaminidases/metabolismo
2.
Mol Pharmacol ; 88(2): 245-55, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25976766

RESUMO

The clinical benefit of current anticancer regimens for lung cancer therapy is still limited due to moderate efficacy, drug resistance, and recurrence. Therefore, the development of effective anticancer drugs for first-line therapy and for optimal second-line treatment is necessary. Because the 90-kDa molecular chaperone heat shock protein (Hsp90) contributes to the maturation of numerous mutated or overexpressed oncogenic proteins, targeting Hsp90 may offer an effective anticancer therapy. Here, we investigated antitumor activities and toxicity of a novel deguelin-derived C-terminal Hsp90 inhibitor, designated L80. L80 displayed significant inhibitory effects on the viability, colony formation, angiogenesis-stimulating activity, migration, and invasion of a panel of non-small cell lung cancer cell lines and their sublines with acquired resistance to paclitaxel with minimal toxicity to normal lung epithelial cells, hippocampal cells, vascular endothelial cells, and ocular cells. Biochemical analyses and molecular docking simulation revealed that L80 disrupted Hsp90 function by binding to the C-terminal ATP-binding pocket of Hsp90, leading to the disruption of the interaction between hypoxia-inducible factor (HIF)-1α and Hsp90, downregulation of HIF-1α and its target genes, including vascular endothelial growth factor (VEGF) and insulin-like growth factor 2 (IGF2), and decreased the expression of various Hsp90 client proteins. Consistent with these in vitro findings, L80 exhibited significant antitumor and antiangiogenic activities in H1299 xenograft tumors. These results suggest that L80 represents a novel C-terminal Hsp90 inhibitor with effective anticancer activities with minimal toxicities.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/síntese química , Benzopiranos/administração & dosagem , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Proteínas de Choque Térmico HSP90/química , Neoplasias Pulmonares/tratamento farmacológico , Quinolinas/administração & dosagem , Rotenona/análogos & derivados , Animais , Antineoplásicos/farmacologia , Benzopiranos/síntese química , Benzopiranos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Células Endoteliais da Veia Umbilical Humana , Humanos , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos SCID , Quinolinas/síntese química , Quinolinas/farmacologia , Rotenona/administração & dosagem , Rotenona/síntese química , Rotenona/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Bioorg Med Chem ; 23(15): 4919-4935, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26051601

RESUMO

Receptor for advanced glycation end products (RAGE) is known to be involved in the transportation of amyloid ß (Aß) peptides and causes the accumulation of Aß in the brain. Moreover, recent studies suggest that the interactions between RAGE and Aß peptides may be the culprit behind Alzheimer's disease (AD). Inhibitors of the RAGE-Aß interactions would not only prevent the accumulation of toxic Aß in the brain, and but also block the progress of AD, therefore, have the potential to provide a 'disease-modifying therapy'. In this study, we have developed a series of 6-phenoxy-2-phenylbenzoxazole analogs as novel inhibitors of RAGE. Among these derivatives, we found several effective inhibitors that block the RAGE-Aß interactions without causing significant cellular toxicity. Further testing showed that compound 48 suppressed Aß induced toxicity in mouse hippocampal neuronal cells and reduced Aß levels in the brains of a transgenic mouse model of AD after oral administration.


Assuntos
Benzoxazóis/química , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Peptídeos beta-Amiloides/análise , Peptídeos beta-Amiloides/toxicidade , Animais , Benzoxazóis/síntese química , Benzoxazóis/farmacologia , Encéfalo/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Transferência Ressonante de Energia de Fluorescência , Humanos , Camundongos , Camundongos Transgênicos , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Relação Estrutura-Atividade
4.
J Neurosci ; 32(26): 8845-54, 2012 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-22745485

RESUMO

The blood-brain barrier (BBB), which is formed by adherens and tight junctions (TJs) of endothelial cells, maintains homeostasis of the brain. Disrupted intracellular Ca²âº homeostasis and breakdown of the BBB have been implicated in the pathogenesis of Alzheimer's disease (AD). The receptor for advanced glycation end products (RAGE) is known to interact with amyloid ß-peptide (Aß) and mediate Aß transport across the BBB, contributing to the deposition of Aß in the brain. However, molecular mechanisms underlying Aß-RAGE interaction-induced alterations in the BBB have not been identified. We found that Aß1₋42 induces enhanced permeability, disruption of zonula occludin-1 (ZO-1) expression in the plasma membrane, and increased intracellular calcium and matrix metalloproteinase (MMP) secretion in cultured endothelial cells. Neutralizing antibodies against RAGE and inhibitors of calcineurin and MMPs prevented Aß1₋42-induced changes in ZO-1, suggesting that Aß-RAGE interactions alter TJ proteins through the Ca²âº-calcineurin pathway. Consistent with these in vitro findings, we found disrupted microvessels near Aß plaque-deposited areas, elevated RAGE expression, and enhanced MMP secretion in microvessels of the brains of 5XFAD mice, an animal model for AD. We have identified a potential molecular pathway underlying Aß-RAGE interaction-induced breakage of BBB integrity. This pathway might play an important role in the pathogenesis of AD.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/patologia , Calcineurina/metabolismo , Cálcio/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais/fisiologia , Junções Íntimas/metabolismo , Fatores Etários , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/genética , Animais , Transporte Biológico , Barreira Hematoencefálica/efeitos dos fármacos , Linhagem Celular Transformada , Claudinas/metabolismo , Dextranos/metabolismo , Dipeptídeos/farmacologia , Modelos Animais de Doenças , Células Endoteliais , Feminino , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Regulação da Expressão Gênica/genética , Transportador de Glucose Tipo 1/metabolismo , Humanos , Imageamento Tridimensional , Imunossupressores/farmacologia , Metaloproteinases da Matriz/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Microvasos/patologia , Mutação/genética , Fragmentos de Peptídeos/farmacologia , Fosfoproteínas/metabolismo , Presenilina-1/genética , Inibidores de Proteases/farmacologia , Receptor para Produtos Finais de Glicação Avançada , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Tacrolimo/farmacologia , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/patologia , Junções Íntimas/ultraestrutura , Transfecção , Proteína da Zônula de Oclusão-1
5.
J Neurosci ; 32(9): 3081-7, 2012 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-22378880

RESUMO

Activated microglia and reactive astrocytes are commonly found in and around the senile plaque, which is the central pathological hallmark of Alzheimer's disease. Astrocytes respond to neuronal activity through the release of gliotransmitters such as glutamate, D-serine, and ATP. However, it is largely unknown whether and how gliotransmitters affect neuronal functions. In this study, we explored the effect of a gliotransmitter, ATP, on neurons damaged by ß-amyloid peptide (Aß). We found that Aß(1-42) (Aß42) increased the release of ATP in cultures of primary astrocytes and U373 astrocyte cell line. We also found that exogenous ATP protected Aß42-mediated reduction in synaptic molecules, such as NMDA receptor 2A and PSD-95, through P2 purinergic receptors and prevented Aß42-induced spine reduction in cultured primary hippocampal neurons. Moreover, ATP prevented Aß42-induced impairment of long-term potentiation in acute hippocampal slices. Our findings suggest that Aß-induced release of gliotransmitter ATP plays a protective role against Aß42-mediated disruption of synaptic plasticity.


Assuntos
Trifosfato de Adenosina/metabolismo , Peptídeos beta-Amiloides/toxicidade , Astrócitos/metabolismo , Plasticidade Neuronal/fisiologia , Fragmentos de Peptídeos/toxicidade , Sinapses/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Linhagem Celular Tumoral , Células Cultivadas , Potenciais Pós-Sinápticos Excitadores/fisiologia , Humanos , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Fragmentos de Peptídeos/antagonistas & inibidores , Ratos Sprague-Dawley , Sinapses/efeitos dos fármacos
6.
J Neurosci ; 32(17): 5953-63, 2012 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-22539855

RESUMO

Alzheimer's disease (AD) in the early stages is characterized by memory impairment, which may be attributable to synaptic dysfunction. Oxidative stress, mitochondrial dysfunction, and Ca²âº dysregulation are key factors in the pathogenesis of AD, but the causal relationship between these factors and synaptic dysfunction is not clearly understood. We found that in the hippocampus of an AD mouse model (Tg2576), mitochondrial Ca²âº handling in dentate granule cells was impaired as early as the second postnatal month, and this Ca²âº dysregulation caused an impairment of post-tetanic potentiation in mossy fiber-CA3 synapses. The alteration of cellular Ca²âº clearance in Tg2576 mice is region-specific within hippocampus because in another region, CA1 pyramidal neuron, no significant difference in Ca²âº clearance was detected between wild-type and Tg2576 mice at this early stage. Impairment of mitochondrial Ca²âº uptake was associated with increased mitochondrial reactive oxygen species and depolarization of mitochondrial membrane potential. Mitochondrial dysfunctions in dentate granule cells and impairment of post-tetanic potentiation in mossy fiber-CA3 synapses were fully restored when brain slices obtained from Tg2576 were pretreated with antioxidant, suggesting that mitochondrial oxidative stress initiates other dysfunctions. Reversibility of early dysfunctions by antioxidants at the preclinical stage of AD highlights the importance of early diagnosis and antioxidant therapy to delay or prevent the disease processes.


Assuntos
Doença de Alzheimer/patologia , Giro Denteado/patologia , Mitocôndrias/patologia , Fibras Musgosas Hipocampais/fisiopatologia , Plasticidade Neuronal/fisiologia , Neurônios/ultraestrutura , Transmissão Sináptica/fisiologia , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Geneticamente Modificados , Antioxidantes/farmacologia , Biofísica , Cálcio/metabolismo , Cromanos/farmacologia , Giro Denteado/metabolismo , Modelos Animais de Doenças , Interações Medicamentosas , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Humanos , Técnicas In Vitro , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/genética , Camundongos , Mutação/genética , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/farmacologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Compostos de Rutênio/farmacologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética
7.
Biochem Biophys Res Commun ; 439(2): 252-7, 2013 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-23973487

RESUMO

Amyloid-ß (Aß) peptide is central to the development of brain pathology in Alzheimer disease (AD) patients. Association with receptors for advanced glycation end-products (RAGE) enables the transport of Aß peptide from circulating blood to human brain, and also causes the activation of the NF-κB signaling pathway. Here we show that two ß-strands of RAGE participate in the interaction with Aß peptide. Serial deletion analysis of the RAGE V domain indicates that the third and eighth ß-strands are required for interaction with Aß peptide. Site-directed mutagenesis of amino acids located in the third and eighth ß-strands abolish the interaction of RAGE with Aß peptide. Wild-type RAGE activates the NF-κB signaling pathway in response to Aß peptide treatment, while a RAGE mutant defective in Aß binding does not. Furthermore, use of peptide for the third ß-strand or a RAGE monoclonal antibody that targets the RAGE-Aß interaction interface inhibited transport of the Aß peptide across the blood brain barrier in a mice model. These results provide information crucial to the development of RAGE-derived therapeutic reagents for Alzheimer disease.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/metabolismo , Receptores Imunológicos/química , Receptores Imunológicos/metabolismo , Doença de Alzheimer/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Transporte Biológico , Deleção de Genes , Humanos , Masculino , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , NF-kappa B/imunologia , Mapas de Interação de Proteínas , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia
8.
Proc Natl Acad Sci U S A ; 107(31): 13872-7, 2010 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-20643929

RESUMO

By using a highly sensitive technique of atomic force microscopy-based single-cell compression, the rigidity of cultured N2a and HT22 neuronal cells was measured as a function of amyloid-beta42 (Abeta42) protein treatment. Abeta42 oligomers led to significant cellular stiffening; for example, 90-360% higher force was required to reach 80% deformation for N2a cells. Disaggregated or fibrillar forms of Abeta42 showed much less change. These observations were explained by a combination of two factors: (i) incorporation of oligomer into cellular membrane, which resulted in an increase in the Young's modulus of the membrane from 0.9+/-0.4 to 1.85+/-0.75 MPa for N2a cells and from 1.73+/-0.90 to 5.5+/-1.4 MPa for HT22 cells, and (ii) an increase in intracellular osmotic pressure (e.g., from 7 to 40 Pa for N2a cells) through unregulated ion influx. These findings and measurements provide a deeper, more characteristic, and quantitative insight into interactions between cells and Abeta42 oligomers, which have been considered the prime suspect for initiating neuronal dysfunction in Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/química , Comunicação Celular , Neurônios/química , Animais , Linhagem Celular , Camundongos , Neurônios/fisiologia , Dinâmica não Linear , Multimerização Proteica
9.
Proteins ; 79(2): 402-16, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21069870

RESUMO

The molecular pathogenesis of disorders arising from protein misfolding and aggregation is difficult to elucidate, involving a complex ensemble of intermediates, whose toxicity depends upon their state of progression along distinct processing pathways. To address the complex misfolding and aggregation that initiates the toxic cascade resulting in Alzheimer's disease (AD), we have developed a 2,2,6,6-tetramethylpiperidine-1-oxyl-4-amino-4-carboxylic acid spin-labeled amyloid-ß (Aß) peptide to observe its isoform-dependent interaction with the apoE protein. Although most individuals carry the E3 isoform of apoE, ∼15% of humans carry the E4 isoform, which is recognized as the most significant genetic determinant for Alzheimer's. ApoE is consistently associated with the amyloid plaque marker for AD. A vital question centers on the influence of the two predominant isoforms, E3 and E4, on Aß peptide processing and hence Aß toxicity. We used electron paramagnetic resonance (EPR) spectroscopy of incorporated spin labels to investigate the interaction of apoE with the toxic oligomeric species of Aß in solution. EPR spectra of the spin-labeled side chain report on side chain and backbone dynamics as well as the spatial proximity of spins in an assembly. Our results indicate oligomer binding involves the C-terminal domain of apoE, with apoE3 reporting a much greater response through this conformational marker. Coupled with SPR binding measurements, apoE3 displays a higher affinity and capacity for the toxic Aß oligomer. These findings support the hypothesis that apoE polymorphism and Alzheimer's risk can largely be attributed to the reduced ability of apoE4 to function as a clearance vehicle for the toxic form of Aß.


Assuntos
Peptídeos beta-Amiloides/química , Apolipoproteínas E/química , Animais , Espectroscopia de Ressonância de Spin Eletrônica , Humanos , Camundongos , Ligação Proteica , Isoformas de Proteínas/química , Ressonância de Plasmônio de Superfície
10.
J Neurochem ; 108(4): 1097-1108, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19141069

RESUMO

Small beta-amyloid (Abeta) 1-42 aggregates are toxic to neurons and may be the primary toxic species in Alzheimer's disease (AD). Methods to reduce the level of Abeta, prevent Abeta aggregation, and eliminate existing Abeta aggregates have been proposed for treatment of AD. A tricyclic pyrone named CP2 is found to prevent cell death associated with Abeta oligomers. We studied the possible mechanisms of neuroprotection by CP2. Surface plasmon resonance spectroscopy shows a direct binding of CP2 with Abeta42 oligomer. Circular dichroism spectroscopy reveals monomeric Abeta42 peptide remains as a random coil/alpha-helix structure in the presence of CP2 over 48 h. Atomic force microscopy studies show CP2 exhibits similar ability to inhibit Abeta42 aggregation as that of Congo red and curcumin. Atomic force microscopy closed-fluid cell study demonstrates that CP2 disaggregates Abeta42 oligomers and protofibrils. CP2 also blocks Abeta fibrillations using a protein quantification method. Treatment of 5x familial Alzheimer's disease mice, a robust Abeta42-producing animal model of AD, with a 2-week course of CP2 resulted in 40% and 50% decreases in non-fibrillar and fibrillar Abeta species, respectively. Our results suggest that CP2 might be beneficial to AD patients by preventing Abeta aggregation and disaggregating existing Abeta oligomers and protofibrils.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/antagonistas & inibidores , Encéfalo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/antagonistas & inibidores , Placa Amiloide/efeitos dos fármacos , Pironas/farmacologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Substâncias Macromoleculares/metabolismo , Espectroscopia de Ressonância Magnética , Camundongos , Camundongos Transgênicos , Estrutura Molecular , Neurofibrilas/efeitos dos fármacos , Neurofibrilas/metabolismo , Neurofibrilas/patologia , Fármacos Neuroprotetores/uso terapêutico , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/metabolismo , Polímeros/metabolismo , Estrutura Secundária de Proteína/efeitos dos fármacos , Estrutura Secundária de Proteína/fisiologia , Pironas/uso terapêutico , Resultado do Tratamento
11.
Biochem Biophys Res Commun ; 386(1): 175-80, 2009 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-19508864

RESUMO

The protease BACE1 (beta-site APP-cleaving enzyme 1) is essential for the generation of amyloid beta (Abeta) from amyloid precursor protein (APP). Although BACE1 is expressed primarily in neurons, which are a principal source of Abeta in the brain, the mechanism that underlies basal expression of BACE1 in neurons has not been studied thoroughly. In the present study, we found that endogenous BACE1 expression was mediated by constitutive JAK2/STAT1 activation in neurons. Inhibition of the JAK2/STAT1 signaling pathway, using AG490 (a JAK2 inhibitor), a dominant-negative form of STAT1, and SOCS1 and SOCS3 overexpression, reduced levels of BACE1 promoter activity, expression of endogenous BACE1, and generation of Abeta. These results were recapitulated in the SH-SY5Y neuronal cell line, primary cultured neurons, and mouse brains. Therefore, we propose that constitutive JAK2/STAT1 activation mediates endogenous BACE1 expression in neurons and that inhibition of JAK2/STAT1 signaling abrogates basal levels of BACE1 expression and Abeta generation.


Assuntos
Secretases da Proteína Precursora do Amiloide/genética , Ácido Aspártico Endopeptidases/genética , Regulação Enzimológica da Expressão Gênica , Janus Quinase 2/metabolismo , Neurônios/enzimologia , Fator de Transcrição STAT1/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Células Cultivadas , Humanos , Interferon gama/farmacologia , Camundongos , Neurônios/efeitos dos fármacos , Regiões Promotoras Genéticas
12.
Bioorg Med Chem Lett ; 19(3): 670-4, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19121940

RESUMO

The SbetaC gene is conditionally expressed a 99-residue carboxy terminal fragment, C99, of amyloid precursor protein in MC65 cells and causes cell death. Consequently, MC65 cell line was used to identify inhibitors of toxicity related to intracellular amyloid beta (Abeta) oligomers. Compounds that reduce the level of Abeta peptides, prevent Abeta aggregation, or eliminate existing Abeta aggregates may be used in the treatment of Alzheimer's disease (AD). Previously, we found that a tricyclic pyrone (TP) molecule, compound 1, prevents MC65 cell death and inhibits Abeta aggregation. Hence various TPs containing heterocycle at C7 side chain and a nitrogen at position 2 or 5 were synthesized and their MC65 cell protective activities evaluated. TPs containing N3'-adenine moiety such as compounds 1 and 11 are most active with EC(50) values of 0.31 and 0.35 microM, respectively. EC(50) values of tricyclic N5-analog, pyranoisoquinolinone 13, and N2-analog, pyranopyridinone 20, are 2.49 and 1.25 microM, respectively, despite the lack of adenine moiety. Further investigation of tricyclic N2- and N5-analogs is warranted.


Assuntos
Precursor de Proteína beta-Amiloide/química , Química Farmacêutica/métodos , Neurônios/metabolismo , Adenina/química , Doença de Alzheimer/tratamento farmacológico , Morte Celular , Linhagem Celular , Desenho de Fármacos , Humanos , Modelos Químicos , Neurônios/efeitos dos fármacos , Nitrogênio/química , Peptídeos/química , Piridonas/química , Pironas/química
13.
J Neurochem ; 104(2): 457-68, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17953662

RESUMO

Several small molecule ligands for amyloid-beta (Abeta) fibrils deposited in brain have been developed to facilitate radiological diagnosis of Alzheimer's disease (AD). Recently, the build-up of Abeta oligomers (AbetaO) in brain has been recognized as an additional hallmark of AD and may play a more significant role in early stages. Evidence suggests that quantitative assessment of AbetaO would provide a more accurate index of therapeutic effect of drug trials. Therefore, there is an urgent need to develop methods for efficient identification as well as structural analysis of AbetaO. We found that some well established amyloid ligands, analogs of Congo red and thioflavin-T (ThT), bind AbetaO with high affinity and detect AbetaO in vitro and in vivo. Binding studies revealed the presence of binding sites for Congo red- and thioflavin-T-analogs on AbetaO. Furthermore, these ligands can be used for imaging intracellular AbetaO in living cells and animals and as positive contrast agent for ultrastructural imaging of AbetaO, two applications useful for structural analysis of AbetaO in cells. We propose that by improving the binding affinity of current ligands, in vivo imaging of AbetaO is feasible by a 'signal subtraction' procedure. This approach may facilitate the identification of individuals with early AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Corantes , Vermelho Congo , Corantes Fluorescentes , Hipocampo/patologia , Tiazóis , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/ultraestrutura , Benzotiazóis , Linhagem Celular Tumoral , Corantes/química , Vermelho Congo/química , Corantes Fluorescentes/química , Humanos , Microscopia Eletrônica de Transmissão , Neuroblastoma/patologia , Ressonância de Plasmônio de Superfície/métodos , Tiazóis/química
14.
J Med Chem ; 61(1): 396-402, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29161514

RESUMO

We developed an orally active and blood-brain-barrier-permeable benzofuran analogue (8, MDR-1339) with potent antiaggregation activity. Compound 8 restored cellular viability from Aß-induced cytotoxicity but also improved the learning and memory function of AD model mice by reducing the Aß aggregates in the brains. Given the high bioavailability and brain permeability demonstrated in our pharmacokinetic studies, 8 will provide a novel scaffold for an Aß-aggregation inhibitor that may offer an alternative treatment for AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/química , Benzofuranos/química , Benzofuranos/farmacologia , Agregados Proteicos/efeitos dos fármacos , Administração Oral , Animais , Benzofuranos/farmacocinética , Benzofuranos/uso terapêutico , Disponibilidade Biológica , Linhagem Celular , Cães , Humanos , Camundongos , Ratos
15.
FASEB J ; 20(1): 157-9, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16293708

RESUMO

As an essential protease in the generation of amyloid beta, gamma-secretase is believed to play an important role in the pathogenesis of Alzheimer's disease. Although a great deal of progress has been made in identifying the components of gamma-secretase complex, the endogenous regulatory mechanism of gamma-secretase is unknown. Here we show that gamma-secretase is endogenously regulated via extracellular signal regulated MAP kinase (ERK) 1/2-dependent mitogen-activated protein kinase (MAPK) pathway. The inhibition of ERK1/2 activity, either by a treatment with a MEK inhibitor or an ERK knockdown transfection, dramatically increased gamma-secretase activity in several different cell types. JNK or p38 kinase inhibitors had little effect, indicating that the effect is specific to ERK1/2-dependent MAPK pathway. Conversely, increased ERK1/2 activity, by adding purified active ERK1/2 or EGF-induced activation of ERK1/2, significantly reduced gamma-secretase activity, demonstrating down-regulation of gamma-secretase activity by ERK1/2. Whereas gamma-secretase expression was not affected by ERK1/2, its activity was enhanced by phosphatase treatment, indicating that ERK1/2 regulates gamma-secretase activity by altering the pattern of phophorylation. Among the components of isolated gamma-secretase complex, only nicastrin was phosphorylated by ERK1/2, and it precipitated with ERK1/2 in a co-immunoprecipitation assay, which suggests binding between ERK1/2 and nicastrin. Our results show that ERK1/2 is an endogenous regulator of gamma-secretase, which raises the possibility that ERK1/2 down-regulates gamma-secretase activity by directly phosphorylating nicastrin.


Assuntos
Endopeptidases/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidases , Linhagem Celular , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação , Fosforilação , Interferência de RNA , Receptores Notch/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Neuroreport ; 18(12): 1225-9, 2007 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-17632272

RESUMO

Amyloid precursor protein generates the secreted amyloid precursor protein alpha, which protects hippocampal neurons from ischemic injury and facilitates neuronal survival and synaptogenesis in the developing nervous system. Here, we examined whether platelet-derived growth factor regulates the generation of secreted amyloid precursor protein alpha during the neuronal differentiation of hippocampal precursor cells, HiB5. We showed that platelet-derived growth factor promoted amyloid precursor protein production and secreted amyloid precursor protein alpha secretion. These effects of platelet-derived growth factor were diminished by the PI3K-specific inhibitor wortmannin and the protein kinase C-specific inhibitor GF109203X, suggesting the involvement of the PI3K and protein kinase C-signaling pathway. Furthermore, the conditioned media enriched with secreted amyloid precursor protein alpha promoted the survival of HiB5 cells during neuronal differentiation. These results suggest that the neurotrophic effect of platelet-derived growth factor is mediated in part via upregulation of the expression and release of secreted amyloid precursor protein alpha.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Diferenciação Celular/fisiologia , Hipocampo/embriologia , Neurônios/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Células-Tronco/metabolismo , Precursor de Proteína beta-Amiloide/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Meios de Cultivo Condicionados/farmacologia , Inibidores Enzimáticos/farmacologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Humanos , Neurônios/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Células-Tronco/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
17.
Brain Res ; 1130(1): 223-34, 2007 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-17157826

RESUMO

The discovery of small molecule inhibitors of cytotoxicity induced by amyloid-beta (Abeta) oligomers, either applied extracellularly or accumulated intraneuronally, is an important goal of drug development for Alzheimer's disease (AD), but has been limited by the lack of efficient screening methods. Here we describe our approach using two cell-based methods. The first method takes advantage of the unique ability of extracellularly applied Abeta oligomers to rapidly induce the exocytosis of formazan formed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). We employed a short protocol to quantify this toxicity, and quickly identified two novel inhibitors, code-named CP2 and A5, from two compound libraries. A second independent screen of the same libraries using our previously published MC65 protection assay, which identifies inhibitors of toxicity related to intracellular Abeta oligomers, also selected the same two leads, suggesting that both assays select for the same anti-Abeta oligomer properties displayed by these compounds. We further demonstrated that A5 attenuated the progressive aggregation of existing Abeta oligomers, reduced the level of intracellular Abeta oligomers, and prevented the Abeta oligomer-induced death of primary cortical neurons, effects similar to those demonstrated by CP2. Our results suggest that, when combined, the two methods would generate fewer false results and give a high likelihood of identifying leads that show promises in ameliorating Abeta oligomer-induced toxicities within both intraneuronal and extracellular sites. Both assays are simple, suitable for rapid screening of a large number of medicinal libraries, and amenable for automation.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Carbazóis/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Formazans/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Sais de Tetrazólio/metabolismo , Peptídeos beta-Amiloides/administração & dosagem , Animais , Bioensaio/métodos , Linhagem Celular , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Humanos , Camundongos , Proteínas do Tecido Nervoso/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia
18.
J Med Chem ; 60(6): 2573-2590, 2017 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-28234463

RESUMO

Glutaminyl cyclase (QC) has been implicated in the formation of toxic amyloid plaques by generating the N-terminal pyroglutamate of ß-amyloid peptides (pGlu-Aß) and thus may participate in the pathogenesis of Alzheimer's disease (AD). We designed a library of glutamyl cyclase (QC) inhibitors based on the proposed binding mode of the preferred substrate, Aß3E-42. An in vitro structure-activity relationship study identified several excellent QC inhibitors demonstrating 5- to 40-fold increases in potency compared to a known QC inhibitor. When tested in mouse models of AD, compound 212 significantly reduced the brain concentrations of pyroform Aß and total Aß and restored cognitive functions. This potent Aß-lowering effect was achieved by incorporating an additional binding region into our previously established pharmacophoric model, resulting in strong interactions with the carboxylate group of Glu327 in the QC binding site. Our study offers useful insights in designing novel QC inhibitors as a potential treatment option for AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Aminoaciltransferases/antagonistas & inibidores , Peptídeos beta-Amiloides/antagonistas & inibidores , Desenho de Fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/uso terapêutico , Doença de Alzheimer/metabolismo , Aminoaciltransferases/química , Aminoaciltransferases/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Derivados de Benzeno/química , Derivados de Benzeno/farmacologia , Derivados de Benzeno/uso terapêutico , Inibidores Enzimáticos/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Simulação de Acoplamento Molecular
19.
Mol Neurodegener ; 11: 4, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26758977

RESUMO

BACKGROUND: The receptor for advanced glycation end products (RAGE) has been found to interact with amyloid ß (Aß). Although RAGE does not have any kinase motifs in its cytosolic domain, the interaction between RAGE and Aß triggers multiple cellular signaling involved in Alzheimer's disease (AD). However, the mechanism of signal transduction by RAGE remains still unknown. Therefore, identifying binding proteins of RAGE may provide novel therapeutic targets for AD. RESULTS: In this study, we identified p38-regulated/activated protein kinase (PRAK) as a novel RAGE interacting molecule. To investigate the effect of Aß on PRAK mediated RAGE signaling pathway, we treated SH-SY5Y cells with monomeric form of Aß. We demonstrated that Aß significantly increased the phosphorylation of PRAK as well as the interaction between PRAK and RAGE. We showed that knockdown of PRAK rescued mTORC1 inactivation induced by Aß treatment and decreased the formation of Aß-induced autophagosome. CONCLUSIONS: We provide evidence that PRAK plays a critical role in AD pathology as a key interactor of RAGE. Thus, our data suggest that PRAK might be a potential therapeutic target of AD involved in RAGE-mediated cell signaling induced by Aß.


Assuntos
Doença de Alzheimer/metabolismo , Autofagia/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transdução de Sinais , Peptídeos beta-Amiloides/metabolismo , Humanos , Neurônios/metabolismo , Fosforilação , Transdução de Sinais/fisiologia
20.
J Alzheimers Dis ; 53(4): 1563-76, 2016 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-27392853

RESUMO

Development of a simple, non-invasive early diagnosis platform of Alzheimer's disease (AD) using blood is urgently required. Recently, PiB-PET imaging has been shown to be powerful to quantify amyloid-ß plaque loads leading to pathophysiological alterations in AD brains. Thus, there has been a need for serum biomarkers reflecting PiB-PET imaging data as an early diagnosis platform of AD. Here, using LC-MS/MS analysis coupled with isobaric tagging, we performed comprehensive proteome profiling of serum samples from cognitively normal controls, mild cognitive impairment (MCI), and AD patients, who were selected using PiB-PET imaging. Comparative analysis of the proteomes revealed 79 and 72 differentially expressed proteins in MCI and AD, respectively, compared to controls. Integrated analysis of these proteins with genomic and proteomic data of AD brain tissues, together with network analysis, identified three biomarker candidates representing the altered proteolysis-related process in MCI or AD: proprotein convertase subtilisin/kexin type 9 (PCSK9), coagulation factor XIII, A1 polypeptide (F13A1), and dermcidin (DCD). In independent serum samples of MCI and AD, we confirmed the elevation of the candidates using western blotting and ELISA. Our results suggest that these biomarker candidates can serve as a potential non-invasive early diagnosis platform reflecting PiB-PET imaging for MCI and AD.


Assuntos
Doença de Alzheimer/sangue , Doença de Alzheimer/diagnóstico por imagem , Proteínas Sanguíneas/metabolismo , Disfunção Cognitiva/sangue , Disfunção Cognitiva/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Pró-Proteína Convertase 9/metabolismo , Idoso , Idoso de 80 Anos ou mais , Compostos de Anilina , Cromatografia Líquida , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Fator XIII , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Pessoa de Meia-Idade , Peptídeos , Espectrometria de Massas em Tandem , Tiazóis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA