Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell Physiol Biochem ; 55(6): 773-783, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34907696

RESUMEN

The importance of Alzheime's Disease (AD) research has never been greater from a worldwide perspective with the disease becoming increasingly prevalent with life expectancy on the rise. One emerging factor that has presented as a serious risk that still requires more research and understanding is the role and effects of Apolipoprotein E4 (ApoE4). When present, individuals are three times more likely to develop AD in their lifetime. This is due to ApoE4's ability to not only increase amyloid beta plaque aggregation ApoE4 also increases hyperphosphorylation of tau causing neurofibrillary tangles. These two factors are the well-known hallmarks for AD, which increase the importance for ApoE4 research as it affects both major aspects. Treatment for AD has always been an issue due to a variety of factors with only a few approved for use today. These approved treatments are only to ease and supress symptoms rather than treating the disease. Dementia symptoms such as memory loss, language problems, motor skills, irritability and paranoia are all symptoms that destroy patient's ability to function in their communities. Inhibiting ApoE4 and reducing its toxic effects is a promising theory that has the ability to extend AD patients' lifespan and prolong capable brain function limiting brain tissue degradation.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/antagonistas & inhibidores , Apolipoproteína E4/antagonistas & inhibidores , Inhibidores de la Colinesterasa/uso terapéutico , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Terapia Molecular Dirigida/métodos , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Apolipoproteína E4/metabolismo , COVID-19/metabolismo , COVID-19/prevención & control , COVID-19/virología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/virología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología
2.
Ann Neurol ; 89(5): 952-966, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33550655

RESUMEN

OBJECTIVE: Apolipoprotein E (ApoE) genotype is the strongest genetic risk factor for late-onset Alzheimer's disease, with the ε4 allele increasing risk in a dose-dependent fashion. In addition to ApoE4 playing a crucial role in amyloid-ß deposition, recent evidence suggests that it also plays an important role in tau pathology and tau-mediated neurodegeneration. It is not known, however, whether therapeutic reduction of ApoE4 would exert protective effects on tau-mediated neurodegeneration. METHODS: Herein, we used antisense oligonucleotides (ASOs) against human APOE to reduce ApoE4 levels in the P301S/ApoE4 mouse model of tauopathy. We treated P301S/ApoE4 mice with ApoE or control ASOs via intracerebroventricular injection at 6 and 7.5 months of age and performed brain pathological assessments at 9 months of age. RESULTS: Our results indicate that treatment with ApoE ASOs reduced ApoE4 protein levels by ~50%, significantly protected against tau pathology and associated neurodegeneration, decreased neuroinflammation, and preserved synaptic density. These data were also corroborated by a significant reduction in levels of neurofilament light chain (NfL) protein in plasma of ASO-treated mice. INTERPRETATION: We conclude that reducing ApoE4 levels should be explored further as a therapeutic approach for APOE4 carriers with tauopathy including Alzheimer's disease. ANN NEUROL 2021;89:952-966.


Asunto(s)
Apolipoproteína E4/antagonistas & inhibidores , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/etiología , Oligonucleótidos Antisentido/uso terapéutico , Tauopatías/complicaciones , Tauopatías/tratamiento farmacológico , Animales , Apolipoproteína E4/sangre , Apolipoproteína E4/genética , Colesterol/metabolismo , Giro Dentado/patología , Encefalitis/prevención & control , Técnicas de Sustitución del Gen , Inyecciones Intraventriculares , Ratones , Ratones Endogámicos C57BL , Proteínas de Neurofilamentos/metabolismo , Oligonucleótidos Antisentido/administración & dosificación , Sinapsis/efectos de los fármacos , Sinapsis/patología , Proteínas tau/metabolismo
3.
Oxid Med Cell Longev ; 2020: 5086250, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32509144

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease that causes chronic cognitive dysfunction. Most of the AD cases are late onset, and the apolipoprotein E (APOE) isoform is a key genetic risk factor. The APOE gene has 3 key alleles in humans including APOE2, APOE3, and APOE4. Among them, APOE4 is the most potent genetic risk factor for late-onset AD (LOAD), while APOE2 has a defensive effect. Research data suggest that APOE4 leads to the pathogenesis of AD through various processes such as accelerated beta-amyloid aggregations that raised neurofibrillary tangle formation, cerebrovascular diseases, aggravated neuroinflammation, and synaptic loss. However, the precise mode of actions regarding in what way APOE4 leads to AD pathology remains unclear. Since APOE contributes to several pathological pathways of AD, targeting APOE4 might serve as a promising strategy for the development of novel drugs to combat AD. In this review, we focus on the recent studies about APOE4-targeted therapeutic strategies that have been advanced in animal models and are being prepared for use in humans for the management of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Apolipoproteína E4/metabolismo , Ovillos Neurofibrilares/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Animales , Apolipoproteína E4/antagonistas & inhibidores , Apolipoproteína E4/genética , Predisposición Genética a la Enfermedad , Humanos , Terapia Molecular Dirigida , Polimorfismo Genético , Pliegue de Proteína
4.
Neurobiol Dis ; 138: 104784, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32027932

RESUMEN

Alzheimer's disease (AD) is a devastating neurodegenerative disorder that is growing in prevalence globally. It is the only major cause of death without any effective pharmacological means to treat or slow progression. Inheritance of the ε4 allele of the Apolipoprotein (APO) E gene is the strongest genetic risk factor for late-onset AD. The interaction between APOE and amyloid ß (Aß) plays a key role in AD pathogenesis. The APOE-Aß interaction regulates Aß aggregation and clearance and therefore directly influences the development of amyloid plaques, congophilic amyloid angiopathy and subsequent tau related pathology. Relatively few AD therapeutic approaches have directly targeted the APOE-Aß interaction thus far. Here we review the critical role of APOE in the pathogenesis of AD and some of the most promising therapeutic approaches that focus on the APOE-Aß interaction.


Asunto(s)
Apolipoproteínas E/metabolismo , Placa Amiloide/metabolismo , Alelos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteína E4/antagonistas & inhibidores , Apolipoproteína E4/metabolismo , Apolipoproteínas E/antagonistas & inhibidores , Encéfalo/metabolismo , Sistemas de Liberación de Medicamentos , Humanos , Ratones
5.
J Alzheimers Dis ; 73(2): 517-528, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31796678

RESUMEN

The cell surface level of apolipoprotein E receptor 2 (ApoER2) increases by cyclic transport of ApoER2 and then activates Reelin signaling pathway to exert neuroprotective function in AD. ApoER2 ligand Apolipoprotein E4 (ApoE4) inhibits the recycling of ApoER2 to the cell surface rendering neurons unresponsive to Reelin. Carnosic acid (CA) is proven to possess neuroprotective and neurotrophic functions in Alzheimer's disease (AD) mouse model. However, there are few reports about how ApoE4 impairs the recycling of ApoER2 and if CA can affect the cyclic transport of ApoER2. In this study, we demonstrated that ApoE4 attenuates the binding of sorting nexin 17 (SNX17) to ApoER2 and inhibits the recycling of ApoER2, resulting in decreased cell surface level of ApoER2. Further, we found that CA enhances the binding of SNX17 to ApoER2, counteracts the negative effects of ApoE4 on the cell surface level of ApoER2 to reverse the ApoE4-induced reduction in Reelin signaling activation by increasing the phosphorylation of the N-methyl-D-aspartate receptor (NMDAR) and cAMP-response element-binding protein (CREB) and the expression of Gria2. Thus, CA promotes neurite growth inhibited by ApoE4. Our work suggests that CA may be a potential approach to attenuate the risk of ApoE4-associated AD.


Asunto(s)
Abietanos/farmacología , Apolipoproteína E4/antagonistas & inhibidores , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Fármacos Neuroprotectores/farmacología , Serina Endopeptidasas/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Femenino , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Neuritas/efectos de los fármacos , Células PC12 , Embarazo , Ratas , Receptores AMPA/biosíntesis , Receptores AMPA/genética , Receptores de Superficie Celular/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Proteína Reelina , Nexinas de Clasificación/metabolismo
6.
Neurosci Lett ; 709: 134366, 2019 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-31336138

RESUMEN

The ε4 allele of the apolipoprotein E gene represents the most widely reproduced and robust susceptibility loci for the most common late onset and sporadic forms of Alzheimer's disease. While the discovery of this now widely replicated association was reported more than 25 years ago, few therapeutic interventions that specifically target the apolipoprotein pathway in brain have emerged. Here we discuss our current understanding of apolipoprotein E biology in brain, its relationship to the pathogenesis of Alzheimer's disease and present potential future avenues for exploration that may be amenable to drug development.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/terapia , Apolipoproteína E4/antagonistas & inhibidores , Apolipoproteína E4/metabolismo , Desarrollo de Medicamentos/tendencias , Terapia Genética/tendencias , Enfermedad de Alzheimer/genética , Apolipoproteína E4/genética , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Ensayos Clínicos como Asunto/métodos , Desarrollo de Medicamentos/métodos , Terapia Genética/métodos , Humanos , Fragmentos de Péptidos/administración & dosificación , Vacunas/administración & dosificación
7.
Curr Comput Aided Drug Des ; 15(1): 97-103, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30306878

RESUMEN

INTRODUCTION: Apolipoprotein E4 (ApoE) is a major genetic factor for developing Alzheimer's disease (AD). It plays a vital role in brain to maintain a constant supply of neuronal lipids for rapid and dynamic membrane synthesis. Aggregation of beta amyloid plaques (Aß) and neurofibrillary tangles in brain are responsible for onset of AD. The current study is designed to predict a drug against over activity of apoE4. 22 natural compounds (marine, microorganism and plant derivative) were used in current study. METHODS: These compounds were used as inhibitors to target apoE4 protein activity. Moreover, six synthetic compounds were docked with target protein to compare and analyze the docking results with natural compounds. S-Allyl-L-Cysteine, Epicatechin Gallate and Fulvic acid showed highest binding affinity (-7.1, - 7 and -7 kcal /mol respectively). Analysis of the docked complex showed that Epicatechin Gallate bonded with Gln156 and Asp35. Furthermore, Fulvic Acid showed hydrogen bonding with Glu27. Among synthetic compound, Tideglusib had highest binding affinity with target protein but did not show hydrogen bonding with any amino acid residue. Moreover, a natural compound S-Allyl-LCysteine also showed highest binding affinity but did not show hydrogen bonding with any amino acid residue. RESULTS AND CONCLUSION: Our study highlighted Epicatechin Gallate as a potential lead compound on the basis of binding affinity and hydrogen bonding to inhibit the progression of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Apolipoproteína E4/antagonistas & inhibidores , Productos Biológicos/farmacología , Simulación por Computador , Benzopiranos/química , Benzopiranos/farmacología , Productos Biológicos/química , Catequina/análogos & derivados , Catequina/química , Catequina/farmacología , Cisteína/análogos & derivados , Cisteína/química , Cisteína/farmacología , Humanos , Simulación del Acoplamiento Molecular , Dominios y Motivos de Interacción de Proteínas
8.
J Clin Invest ; 128(5): 2144-2155, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29600961

RESUMEN

The apolipoprotein E E4 allele of the APOE gene is the strongest genetic factor for late-onset Alzheimer disease (LOAD). There is compelling evidence that apoE influences Alzheimer disease (AD) in large part by affecting amyloid ß (Aß) aggregation and clearance; however, the molecular mechanism underlying these findings remains largely unknown. Herein, we tested whether anti-human apoE antibodies can decrease Aß pathology in mice producing both human Aß and apoE4, and investigated the mechanism underlying these effects. We utilized APPPS1-21 mice crossed to apoE4-knockin mice expressing human apoE4 (APPPS1-21/APOE4). We discovered an anti-human apoE antibody, anti-human apoE 4 (HAE-4), that specifically recognizes human apoE4 and apoE3 and preferentially binds nonlipidated, aggregated apoE over the lipidated apoE found in circulation. HAE-4 also binds to apoE in amyloid plaques in unfixed brain sections and in living APPPS1-21/APOE4 mice. When delivered centrally or by peripheral injection, HAE-4 reduced Aß deposition in APPPS1-21/APOE4 mice. Using adeno-associated virus to express 2 different full-length anti-apoE antibodies in the brain, we found that HAE antibodies decreased amyloid accumulation, which was dependent on Fcγ receptor function. These data support the hypothesis that a primary mechanism for apoE-mediated plaque formation may be a result of apoE aggregation, as preferentially targeting apoE aggregates with therapeutic antibodies reduces Aß pathology and may represent a selective approach to treat AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Anticuerpos Monoclonales de Origen Murino/farmacología , Apolipoproteína E4/antagonistas & inhibidores , Placa Amiloide/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Apolipoproteína E3/antagonistas & inhibidores , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Humanos , Ratones , Ratones Noqueados , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patología
9.
J Alzheimers Dis ; 54(3): 1219-1233, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27567858

RESUMEN

The allele ɛ4 of apolipoprotein E (apoE4) is the most prevalent genetic risk factor for Alzheimer's disease (AD) and is therefore a promising therapeutic target. Human and animal model studies suggest that apoE4 is hypolipidated; accordingly, we have previously shown that the retinoid X receptor (RXR) agonist bexarotene upregulates ABCA1, the main apoE-lipidating protein, resulting in increased lipidation of apoE4, and the subsequent reversal of the pathological effects of apoE4, namely: accumulation of Aß42 and hyperphosphorylated tau, as well as reduction in the levels of synaptic markers and cognitive deficits. Since the RXR system has numerous other targets, it is important to devise the means of activating ABCA1 selectively. We presently utilized CS-6253, a peptide shown to directly activate ABCA1 in vitro, and examined the extent to which it can affect the degree of lipidation of apoE4 in vivo and counteract the associated brain and behavioral pathologies. This revealed that treatment of young apoE4-targeted replacement mice with CS-6253 increases the lipidation of apoE4. This was associated with a reversal of the apoE4-driven Aß42 accumulation and tau hyperphosphorylation in hippocampal neurons, as well as of the synaptic impairments and cognitive deficits. These findings suggest that the pathological effects of apoE4 in vivo are associated with decreased activation of ABCA1 and impaired lipidation of apoE4 and that the downstream brain-related pathology and cognitive deficits can be counteracted by treatment with the ABCA1 agonist CS-6253. These findings have important clinical ramifications and put forward ABCA1 as a promising target for apoE4-related treatment of AD.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/agonistas , Apolipoproteína E4/antagonistas & inhibidores , Encéfalo/patología , Trastornos del Conocimiento/patología , Péptidos/farmacología , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Apolipoproteína E4/metabolismo , Encéfalo/efectos de los fármacos , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Péptidos/uso terapéutico
10.
Biochemistry ; 55(18): 2613-21, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27065061

RESUMEN

Apolipoprotein E4 (apoE4), one of three isoforms of apoE, is the major risk factor for developing late onset Alzheimer's disease. The only differences among these isoforms (apoE2, apoE3, and apoE4) are single amino acid changes. Yet these proteins are functionally very different. One approach to ameliorating the effect of apoE4 with respect to Alzheimer's disease would be to find small molecular weight compounds that affect the behavior of apoE4. Few studies of this approach have been carried out in part because there was no complete structure of any full-length apoE isoform until 2011. Here, we focus on one small molecular weight compound, EZ-482, and explore the effects of its binding to apoE. Using hydrogen-deuterium exchange, we determined that EZ-482 binds to the C-terminal domains of both apoE3 and apoE4. The binding to apoE4, however, is accompanied by a unique N-terminal allosteric effect. Using fluorescence methods, we determined an apparent dissociation constant of approximately 8 µM. Although EZ-482 binds to the C-terminal domain, it blocks heparin binding to the N-terminal domain. The residues of apoE that bind heparin are the same as those involved in apoE binding to LDL and LRP-1 receptors. The methods and the data presented here may serve as a template for future studies using small molecular weight compounds to modulate the behavior of apoE.


Asunto(s)
Apolipoproteína E4/química , Heparina/química , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Apolipoproteína E4/antagonistas & inhibidores , Apolipoproteína E4/metabolismo , Medición de Intercambio de Deuterio , Humanos , Lipoproteínas LDL/química , Lipoproteínas LDL/metabolismo , Dominios Proteicos , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Receptores de LDL/química , Receptores de LDL/metabolismo
11.
Biomed Res Int ; 2014: 452625, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24967370

RESUMEN

Apolipoprotein E4 (Apo E4) is the major genetic risk factor in the causation of Alzheimer's disease (AD). In this study we utilize virtual screening of the world's largest traditional Chinese medicine (TCM) database and investigate potential compounds for the inhibition of ApoE4. We present the top three TCM candidates: Solapalmitine, Isodesacetyluvaricin, and Budmunchiamine L5 for further investigation. Dynamics analysis and molecular dynamics (MD) simulation were used to simulate protein-ligand complexes for observing the interactions and protein variations. Budmunchiamine L5 did not have the highest score from virtual screening; however, the dynamics pose is similar to the initial docking pose after MD simulation. Trajectory analysis reveals that Budmunchiamine L5 was stable over all simulation times. The migration distance of Budmunchiamine L5 illustrates that docked ligands are not variable from the initial docked site. Interestingly, Arg158 was observed to form H-bonds with Budmunchiamine L5 in the docking pose and MD snapshot, which indicates that the TCM compounds could stably bind to ApoE4. Our results show that Budmunchiamine L5 has good absorption, blood brain barrier (BBB) penetration, and less toxicity according to absorption, distribution, metabolism, excretion, and toxicity (ADMET) prediction and could, therefore, be safely used for developing novel ApoE4 inhibitors.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Apolipoproteína E4 , Diseño de Fármacos , Medicina Tradicional China , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Apolipoproteína E4/antagonistas & inhibidores , Apolipoproteína E4/química , Bases de Datos Factuales , Humanos
12.
J Biol Chem ; 287(8): 5253-66, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22158868

RESUMEN

Apolipoprotein E4 (apoE4), the major genetic risk factor for late onset Alzheimer disease, assumes a pathological conformation, intramolecular domain interaction. ApoE4 domain interaction mediates the detrimental effects of apoE4, including decreased mitochondrial cytochrome c oxidase subunit 1 levels, reduced mitochondrial motility, and reduced neurite outgrowth in vitro. Mutant apoE4 (apoE4-R61T) lacks domain interaction, behaves like apoE3, and does not cause detrimental effects. To identify small molecules that inhibit domain interaction (i.e. structure correctors) and reverse the apoE4 detrimental effects, we established a high throughput cell-based FRET primary assay that determines apoE4 domain interaction and secondary cell- and function-based assays. Screening a ChemBridge library with the FRET assay identified CB9032258 (a phthalazinone derivative), which inhibits domain interaction in neuronal cells. In secondary functional assays, CB9032258 restored mitochondrial cytochrome c oxidase subunit 1 levels and rescued impairments of mitochondrial motility and neurite outgrowth in apoE4-expressing neuronal cells. These benefits were apoE4-specific and dose-dependent. Modifying CB9032258 yielded well defined structure-activity relationships and more active compounds with enhanced potencies in the FRET assay (IC(50) of 23 and 116 nm, respectively). These compounds efficiently restored functional activities of apoE4-expressing cells in secondary assays. An EPR binding assay showed that the apoE4 structure correction resulted from direct interaction of a phthalazinone. With these data, a six-feature pharmacophore model was constructed for future drug design. Our results serve as a proof of concept that pharmacological intervention with apoE4 structure correctors negates apoE4 detrimental effects in neuronal cells and could be further developed as an Alzheimer disease therapeutic.


Asunto(s)
Apolipoproteína E4/antagonistas & inhibidores , Apolipoproteína E4/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Apolipoproteína E4/química , Línea Celular , Evaluación Preclínica de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos , Modelos Moleculares , Neuronas/metabolismo , Ftalazinas/química , Ftalazinas/farmacología , Estructura Terciaria de Proteína , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
13.
CNS Neurol Disord Drug Targets ; 9(2): 140-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20205646

RESUMEN

Alzheimer's disease (AD) is accompanied by an activation of the innate immune system, and many epidemiological studies have shown reduced risk for dementia or AD associated with chronic consumption of non-steroidal anti-inflammatory drugs (NSAIDs). These observations led to animal model studies to test the hypothesis that NSAIDs can be disease-modifying for some aspects of AD pathogenesis. NSAIDs cannot only suppress inflammatory targets, which could contribute to neuroprotection, they also slow amyloid deposition by mechanisms that remain unclear. Several large clinical trials with NSAID therapies with AD subjects have failed, and cyclooxygenase-2 does not appear to be a useful target for disease modifying therapy. However, there may be apolipoprotein E E4 pharmacogenomic effects and a real but delayed positive signal in a large primary prevention trial with naproxen. This encourages researchers to re-address possible mechanisms for a stage-dependent NSAID efficacy, the subject of this review.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antiinflamatorios no Esteroideos/farmacología , Química Encefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encefalitis/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Apolipoproteína E4/antagonistas & inhibidores , Apolipoproteína E4/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiopatología , Química Encefálica/fisiología , Ensayos Clínicos como Asunto , Inhibidores de la Ciclooxigenasa 2/farmacología , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Modelos Animales de Enfermedad , Encefalitis/fisiopatología , Encefalitis/prevención & control , Humanos , Fármacos Neuroprotectores/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA