Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Hum Mol Genet ; 24(18): 5198-210, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26123488

RESUMO

Accumulation of amyloid-ß (Aß) in synaptic mitochondria is associated with mitochondrial and synaptic injury. The underlying mechanisms and strategies to eliminate Aß and rescue mitochondrial and synaptic defects remain elusive. Presequence protease (PreP), a mitochondrial peptidasome, is a novel mitochondrial Aß degrading enzyme. Here, we demonstrate for the first time that increased expression of active human PreP in cortical neurons attenuates Alzheimer disease's (AD)-like mitochondrial amyloid pathology and synaptic mitochondrial dysfunction, and suppresses mitochondrial oxidative stress. Notably, PreP-overexpressed AD mice show significant reduction in the production of proinflammatory mediators. Accordingly, increased neuronal PreP expression improves learning and memory and synaptic function in vivo AD mice, and alleviates Aß-mediated reduction of long-term potentiation (LTP). Our results provide in vivo evidence that PreP may play an important role in maintaining mitochondrial integrity and function by clearance and degradation of mitochondrial Aß along with the improvement in synaptic and behavioral function in AD mouse model. Thus, enhancing PreP activity/expression may be a new therapeutic avenue for treatment of AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Agregação Patológica de Proteínas/metabolismo , Serina Endopeptidases/metabolismo , Sinapses/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Comportamento Animal , Células Cultivadas , Cognição , Modelos Animais de Doenças , Expressão Gênica , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Transgênicos , Estresse Oxidativo , Proteólise , Serina Endopeptidases/genética
2.
J Chem Inf Model ; 54(3): 902-12, 2014 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-24555519

RESUMO

Cyclophilin D (CypD) is a peptidyl prolyl isomerase F that resides in the mitochondrial matrix and associates with the inner mitochondrial membrane during the mitochondrial membrane permeability transition. CypD plays a central role in opening the mitochondrial membrane permeability transition pore (mPTP) leading to cell death and has been linked to Alzheimer's disease (AD). Because CypD interacts with amyloid beta (Aß) to exacerbate mitochondrial and neuronal stress, it is a potential target for drugs to treat AD. Since appropriately designed small organic molecules might bind to CypD and block its interaction with Aß, 20 trial compounds were designed using known procedures that started with fundamental pyrimidine and sulfonamide scaffolds know to have useful therapeutic effects. Two-dimensional (2D) quantitative structure-activity relationship (QSAR) methods were applied to 40 compounds with known IC50 values. These formed a training set and were followed by a trial set of 20 designed compounds. A correlation analysis was carried out comparing the statistics of the measured IC50 with predicted values for both sets. Selectivity-determining descriptors were interpreted graphically in terms of principle component analyses. These descriptors can be very useful for predicting activity enhancement for lead compounds. A 3D pharmacophore model was also created. Molecular dynamics simulations were carried out for the 20 trial compounds with known IC50 values, and molecular descriptors were determined by 2D QSAR studies using the Lipinski rule-of-five. Fifteen of the 20 molecules satisfied all 5 Lipinski rules, and the remaining 5 satisfied 4 of the 5 Lipinski criteria and nearly satisfied the fifth. Our previous use of 2D QSAR, 3D pharmacophore models, and molecular docking experiments to successfully predict activity indicates that this can be a very powerful technique for screening large numbers of new compounds as active drug candidates. These studies will hopefully provide a basis for efficiently designing and screening large numbers of more potent and selective inhibitors for CypD treatment of AD.


Assuntos
Ciclofilinas/antagonistas & inibidores , Desenho de Fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Peptidil-Prolil Isomerase F , Ciclofilinas/metabolismo , Humanos , Simulação de Acoplamento Molecular , Relação Quantitativa Estrutura-Atividade
3.
Aging Cell ; 20(5): e13368, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33951271

RESUMO

Mitochondrial dysfunction is one of the early pathological features of Alzheimer's disease (AD). Accumulation of cerebral and mitochondrial Aß links to mitochondrial and synaptic toxicity. We have previously demonstrated the mechanism by which presequence peptidase (PITRM1)-mediated clearance of mitochondrial Aß contributes to mitochondrial and cerebral amyloid pathology and mitochondrial and synaptic stress in adult transgenic AD mice overexpressing Aß up to 12 months old. Here, we investigate the effect of PITRM1 in an advanced age AD mouse model (up to 19-24 months) to address the fundamental unexplored question of whether restoration/gain of PITRM1 function protects against mitochondrial and synaptic dysfunction associated with Aß accumulation and whether this protection is maintained even at later ages featuring profound amyloid pathology and synaptic failure. Using newly developed aged PITRM1/Aß-producing AD mice, we first uncovered reduction in PITRM1 expression in AD-affected cortex of AD mice at 19-24 months of age. Increasing neuronal PITRM1 activity/expression re-established mitochondrial respiration, suppressed reactive oxygen species, improved synaptic function, and reduced loss of synapses even at advanced ages (up to 19-24 months). Notably, loss of PITRM1 proteolytic activity resulted in Aß accumulation and failure to rescue mitochondrial and synaptic function, suggesting that PITRM1 activity is required for the degradation and clearance of mitochondrial Aß and Aß deposition. These data indicate that augmenting PITRM1 function results in persistent life-long protection against Aß toxicity in an AD mouse model. Therefore, augmenting PITRM1 function may enhance Aß clearance in mitochondria, thereby maintaining mitochondrial integrity and ultimately slowing the progression of AD.


Assuntos
Doença de Alzheimer/enzimologia , Metaloendopeptidases/metabolismo , Mitocôndrias/enzimologia , Neurônios/enzimologia , Sinapses/metabolismo , Envelhecimento , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/metabolismo , Cognição , Modelos Animais de Doenças , Feminino , Inflamação , Masculino , Metaloendopeptidases/genética , Camundongos Transgênicos , Mitocôndrias/metabolismo , Neurônios/metabolismo , Sinapses/fisiologia
4.
Eur J Med Chem ; 76: 506-16, 2014 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-24602793

RESUMO

Amyloid-ß (Aß), a neurotoxic peptide, is linked to the onset of Alzheimer's disease (AD). Increased Aß content within neuronal cell mitochondria is a pathological feature in both human and mouse models with AD. This accumulation of Aß within the mitochondrial landscape perpetuates increased free radical production and activation of the apoptotic pathway. Human Presequence Protease (hPreP) is responsible for the degradation of mitochondrial amyloid-ß peptide in human neuronal cells, and is thus an attractive target to increase the proteolysis of Aß. Therefore, it offers a potential target for Alzheimer's drug design, by identifying potential activators of hPreP. We applied structure-based drug design, combined with experimental methodologies to investigate the ability of various compounds to enhance hPreP proteolytic activity. Compounds 3c &4c enhanced hPreP-mediated proteolysis of Aß (1-42), pF1ß (2-54) and fluorogenic-substrate V. These results suggest that activation of hPreP by small benzimidazole derivatives provide a promising avenue for AD treatment.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Ativadores de Enzimas/uso terapêutico , Peptídeo Hidrolases/efeitos dos fármacos , Peptídeos beta-Amiloides/metabolismo , Desenho de Fármacos , Ativadores de Enzimas/química , Ativadores de Enzimas/farmacologia , Humanos , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Proteólise , Espectrometria de Massas por Ionização por Electrospray
5.
J Alzheimers Dis ; 42(1): 333-44, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24858403

RESUMO

A major obstacle to the development of effective treatment of Alzheimer's disease (AD) is successfully delivery of drugs to the brain. We have previously identified a series of benzothiazole phosphonate compounds that block the interaction of amyloid-ß peptide with amyloid-ß binding alcohol dehydrogenase (ABAD). A selective and sensitive method for the presence of three new benzothiazole ABAD inhibitors in mouse plasma, brain, and artificial cerebrospinal fluid has been developed and validated based on high performance liquid chromatography tandem mass spectrometry. Mass spectra were generated using Micromass Quattro Ultima "triple" quadrupole mass spectrometer equipped with an Electrospray Ionization interface. Good linearity was obtained over a concentration range of 0.05-2.5 µg/ml. The lowest limit of quantification and detection was found to be 0.05 µg/ml. All inter-day accuracies and precisions were within ± 15% of the nominal value and ± 20%, respectively, at the lower limit of quantitation. The tested compounds were stable at various conditions with recoveries >90.0% (RSD <10%). The method used for pharmacokinetic studies of compounds in mouse cerebrospinal fluid, plasma, and brain is accurate, precise, and specific with no matrix effect. Pharmacokinetic data showed that these compounds penetrate the blood-brain barrier (BBB) yielding 4-50 ng/ml peak brain concentrations and 2 µg/ml peak plasma concentrations from a 10 mg/kg dose. These results indicate that our newly synthesized small molecule ABAD inhibitors have a good drug properties with the ability to cross the blood-brain barrier, which holds a great potential for AD therapy.


Assuntos
3-Hidroxiacil-CoA Desidrogenases/antagonistas & inibidores , Barreira Hematoencefálica/metabolismo , Inibidores Enzimáticos/farmacocinética , Doença de Alzheimer/tratamento farmacológico , Animais , Encéfalo/metabolismo , Calibragem , Cromatografia Líquida de Alta Pressão , Camundongos , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa