Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Molecules ; 28(2)2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36677925

RESUMO

Radioiodinated imaging agents for Aß amyloid plaque imaging in Alzheimer's disease (AD) patients have not been actively pursued. Our previous studies employed the "diaza" derivatives [11C]TAZA and [18F]flotaza in order to develop successful positron emission tomography (PET) imaging agents for Aß plaques. There is a need for radioiodinated imaging agents for Aß plaques for single photon emission computed tomography (SPECT) and PET imaging. We report our findings on the preparation of [124/125I]IAZA, a "diaza" analog of [11C]TAZA and [18F]flotaza, and the evaluation of binding to Aß plaques in the postmortem human AD brain. The binding affinity of IAZA for Aß plaques was Ki = 10.9 nM with weak binding affinity for neurofibrillary tangles (Ki = 3.71 µM). Both [125I]IAZA and [124I]IAZA were produced in >25% radiochemical yield and >90% radiochemical purity. In vitro binding of [125I]IAZA and [124I]IAZA in postmortem human AD brains was higher in gray matter containing Aß plaques compared to white matter (ratio of gray to white matter was >7). Anti-Aß immunostaining strongly correlated with [124/125I]IAZA in postmortem AD human brains. The binding of [124/125I]IAZA in postmortem human AD brains was displaced by the known Aß plaque imaging agents. Thus, radiolabeled [124/123I]IAZA may potentially be a useful PET or SPECT radioligand for Aß plaques in brain imaging studies.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/metabolismo , Radioisótopos do Iodo/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Placa Amiloide/diagnóstico por imagem , Placa Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo
2.
Mar Drugs ; 20(4)2022 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-35447926

RESUMO

This paper examined the toxins naturally produced by marine dinoflagellates and their effects on increases in ß-amyloid plaques along with tau protein hyperphosphorylation, both major drivers of Alzheimer's disease (AD). This approach is in line with the demand for certain natural compounds, namely those produced by marine invertebrates that have the potential to be used in the treatment of AD. Current advances in AD treatment are discussed as well as the main factors that potentially affect the puzzling global AD pattern. This study focused on yessotoxins (YTXs), gymnodimine (GYM), spirolides (SPXs), and gambierol, all toxins that have been shown to reduce ß-amyloid plaques and tau hyperphosphorylation, thus preventing the neuronal or synaptic dysfunction that ultimately causes the cell death associated with AD (or other neurodegenerative diseases). Another group of toxins described, okadaic acid (OA) and its derivatives, inhibit protein phosphatase activity, which facilitates the presence of phosphorylated tau proteins. A few studies have used OA to trigger AD in zebrafish, providing an opportunity to test in vivo the effectiveness of new drugs in treating or attenuating AD. Constraints on the production of marine toxins for use in these tests have been considered. Different lines of research are anticipated regarding the action of the two groups of toxins.


Assuntos
Doença de Alzheimer , Dinoflagellida , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides , Animais , Dinoflagellida/metabolismo , Toxinas Marinhas/farmacologia , Ácido Okadáico/farmacologia , Placa Amiloide , Peixe-Zebra/metabolismo , Proteínas tau/metabolismo
3.
Molecules ; 27(14)2022 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-35889425

RESUMO

Several fluorine-18-labeled PET ß-amyloid (Aß) plaque radiotracers for Alzheimer's disease (AD) are in clinical use. However, no radioiodinated imaging agent for Aß plaques has been successfully moved forward for either single-photon emission computed tomography (SPECT) or positron emission tomography (PET) imaging. Radioiodinated pyridyl benzofuran derivatives for the SPECT imaging of Aß plaques using iodine-123 and iodine-125 are being pursued. In this study, we assess the iodine-124 radioiodinated pyridyl benzofuran derivative 5-(5-[124I]iodobenzofuran-2-yl)-N,N-dimethylpyridin-2-amine ([124I]IBETA) (Ki = 2.36 nM) for utilization in PET imaging for Aß plaques. We report our findings on the radioiododestannylation reaction used to prepare [124/125I]IBETA and evaluate its binding to Aß plaques in a 5 × FAD mouse model and postmortem human AD brain. Both [125I]IBETA and [124I]IBETA are produced in >25% radiochemical yield and >85% radiochemical purity. The in vitro binding of [125I]IBETA and [124I]IBETA in transgenic 5 × FAD mouse model for Aß plaques was high in the frontal cortex, anterior cingulate, thalamus, and hippocampus, which are regions of high Aß accumulation, with very little binding in the cerebellum (ratio of brain regions to cerebellum was >5). The in vitro binding of [125I]IBETA and [124I]IBETA in postmortem human AD brains was higher in gray matter containing Aß plaques compared to white matter (ratio of gray to white matter was >5). Anti-Aß immunostaining strongly correlated with [124/125I]IBETA regional binding in both the 5 × FAD mouse and postmortem AD human brains. The binding of [124/125I]IBETA in 5 × FAD mouse and postmortem human AD brains was displaced by the known Aß plaque imaging agent, Flotaza. Preliminary PET/CT studies of [124I]IBETA in the 5 × FAD mouse model suggested [124I]IBETA was relatively stable in vivo with a greater localization of [124I]IBETA in the brain regions with a high concentration of Aß plaques. Some deiodination was observed at later time points. Therefore, [124I]IBETA may potentially be a useful PET radioligand for Aß plaques in brain studies.


Assuntos
Doença de Alzheimer , Benzofuranos , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Benzofuranos/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Modelos Animais de Doenças , Humanos , Radioisótopos do Iodo , Camundongos , Camundongos Transgênicos , Placa Amiloide/diagnóstico por imagem , Placa Amiloide/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/metabolismo
4.
Bioorg Med Chem ; 50: 116462, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34695709

RESUMO

Alzheimers disease (AD) is the most prominent neurodegenerative disorder with high medical need. Protein-protein-interactions (PPI) interactions have a critical role in AD where ß-amyloid structures (Aß) build toxic oligomers. Design of disease modifying multi target directed ligand (MTDL) has been performed, which disable PPI on the one hand and on the other hand, act as procognitive antagonists at the histamine H3 receptor (H3R). The synthetized compounds are structurally based on peptidomimetic amino acid-like structures mainly as keto, diketo-, or acyl variations of a piperazine moiety connected to an H3R pharmacophore. Most of them showed low nanomolar affinities at H3R and some with promising affinity to Aß-monomers. The structure-activity relationships (SAR) described offer new possibilities for MTDL with an optimized profile combining symptomatic and potential causal therapeutic approaches in AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/antagonistas & inibidores , Antagonistas dos Receptores Histamínicos H3/farmacologia , Peptidomiméticos/farmacologia , Piperazina/farmacologia , Receptores Histamínicos H3/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Relação Dose-Resposta a Droga , Antagonistas dos Receptores Histamínicos H3/síntese química , Antagonistas dos Receptores Histamínicos H3/química , Humanos , Estrutura Molecular , Peptidomiméticos/síntese química , Peptidomiméticos/química , Piperazina/síntese química , Piperazina/química , Relação Estrutura-Atividade
5.
Molecules ; 26(5)2021 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-33652938

RESUMO

PET of ß-Amyloid plaques (Aß) using [18F]florbetaben ([18F]FBB) and [18F]fluorodeoxyglucose ([18F]FDG) increasingly aid clinicians in early diagnosis of dementia, including Alzheimer's disease (AD), frontotemporal disease, dementia with Lewy bodies, and vascular dementia. The aim of this retrospective analysis was to evaluate clinical relevance of [18F]FBB, [18F]FDG PET and complimentary CSF measurements in patients with suspected dementia. In this study, 40 patients with clinically suspected or history of dementia underwent (1) measurement of Aß peptides, total tau, and p-tau protein levels in the cerebrospinal fluid (CSF) compared with healthy controls (HC); (2) clinical and neuropsychological assessment, which included Consortium to Establish a Registry for Alzheimer's Disease neuropsychological assessment battery (CERAD-NAB); (3) [18F]FBB and [18F]FDG PET imaging within an average of 3 weeks. The subjects were within 15 days stratified using PET, CSF measurements as HC, mild cognitive impaired (MCI) and dementia including Alzheimer´s disease. The predictive dementia-related cognitive decline values were supporting the measurements. PET images were evaluated visually and quantitatively using standard uptake value ratios (SUVR). Twenty-one (52.5%) subjects were amyloid-positive (Aß+), with a median neocortical SUVR of 1.80 for AD versus 1.20 relative to the respective 19 (47.5 %) amyloid-negative (Aß-) subjects. Moreover, the [18F]FDG and [18F]FBB confirmed within a sub-group of 10 patients a good complimentary role by correlation between amyloid pathology and brain glucose metabolism in 8 out of 10 subjects. The results suggest the clinical relevance for [18F]FBB combined with [18F]FDG PET retention and CFS measurements serving the management of our patients with dementia. Therefore, [18F]FBB combined with [18F]FDG PET is a helpful tool for differential diagnosis, and supports the patients' management as well as treatment.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Disfunção Cognitiva/diagnóstico por imagem , Demência/diagnóstico por imagem , Fluordesoxiglucose F18/administração & dosagem , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/isolamento & purificação , Encéfalo/diagnóstico por imagem , Encéfalo/fisiopatologia , Disfunção Cognitiva/diagnóstico , Disfunção Cognitiva/fisiopatologia , Demência/diagnóstico , Demência/fisiopatologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Proteínas tau/genética , Proteínas tau/isolamento & purificação
6.
Molecules ; 25(4)2020 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-32098280

RESUMO

Ironically, population aging which is considered a public health success has been accompanied by a myriad of new health challenges, which include neurodegenerative disorders (NDDs), the incidence of which increases proportionally to age. Among them, Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common, with the misfolding and the aggregation of proteins being common and causal in the pathogenesis of both diseases. AD is characterized by the presence of hyperphosphorylated τ protein (tau), which is the main component of neurofibrillary tangles (NFTs), and senile plaques the main component of which is ß-amyloid peptide aggregates (Aß). The neuropathological hallmark of PD is α-synuclein aggregates (α-syn), which are present as insoluble fibrils, the primary structural component of Lewy body (LB) and neurites (LN). An increasing number of non-invasive PET examinations have been used for AD, to monitor the pathological progress (hallmarks) of disease. Notwithstanding, still the need for the development of novel detection tools for other proteinopathies still remains. This review, although not exhaustively, looks at the timeline of the development of existing tracers used in the imaging of Aß and important moments that led to the development of these tracers.


Assuntos
Doença de Alzheimer/diagnóstico , Encéfalo/diagnóstico por imagem , Doença de Parkinson/diagnóstico , Compostos Radiofarmacêuticos/uso terapêutico , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/isolamento & purificação , Encéfalo/patologia , Humanos , Doença de Parkinson/diagnóstico por imagem , Doença de Parkinson/genética , Doença de Parkinson/patologia , Placa Amiloide/diagnóstico por imagem , Placa Amiloide/patologia , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/química , alfa-Sinucleína/genética , alfa-Sinucleína/isolamento & purificação , Proteínas tau/genética , Proteínas tau/isolamento & purificação
7.
Bioorg Med Chem ; 26(12): 3352-3358, 2018 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-29751990

RESUMO

In the amyloid cascade hypothesis, ß-amyloid (Aß) plaques is one of the major pathological biomarkers in the Alzheimer's disease (AD) brain. We report the synthesis and evaluation of novel radiofluorinated chalcones, [18F]4-dimethylamino-4'-fluoro-chalcone ([18F]DMFC) and [18F]4'-fluoro-4-methylamino-chalcone ([18F]FMC), as Aß imaging probes. The conversion of iodine directly introduced to the chalcone backbone into fluorine was successfully carried out by 18F-labeling via the corresponding boronate precursors, achieving the direct introduction of fluorine-18 into the chalcone backbone to prepare [18F]DMFC and [18F]FMC. In a biodistribution study using normal mice, [18F]DMFC and [18F]FMC showed a higher initial uptake (4.43 and 5.47% ID/g at 2 min postinjection, respectively) into and more rapid clearance (0.52 and 0.66% ID/g at 30 min postinjection, respectively) from the brain than a Food and Drug Administration (FDA)-approved Aß imaging agent ([18F]Florbetapir), meaning the improvement of the probability of detecting Aß plaques and the reduction of non-specific binding in the brain. In the in vitro binding studies using aggregates of recombinant Aß peptides, [18F]DMFC and [18F]FMC showed high binding affinity to recombinant Aß aggregates at the Kd values of 4.47 and 6.50 nM, respectively. In the in vitro autoradiography (ARG) experiment with AD brain sections, [18F]DMFC and [18F]FMC markedly accumulated only in a region with abundant Aß plaques, indicating that they clearly recognized human Aß plaques in vitro. These encouraging results suggest that [18F]DMFC and [18F]FMC may be promising PET probes for the detection of an amyloid pathology and the early diagnosis of AD with marked accuracy.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Chalconas/química , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/diagnóstico por imagem , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/genética , Animais , Encéfalo/diagnóstico por imagem , Chalconas/metabolismo , Chalconas/farmacocinética , Radioisótopos de Flúor/química , Iodo/química , Masculino , Camundongos , Camundongos Transgênicos , Tomografia por Emissão de Pósitrons , Ligação Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Distribuição Tecidual
8.
J Nanobiotechnology ; 16(1): 32, 2018 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-29587747

RESUMO

BACKGROUND: Memantine, drug approved for moderate to severe Alzheimer's disease, has not shown to be fully effective. In order to solve this issue, polylactic-co-glycolic (PLGA) nanoparticles could be a suitable solution to increase drug's action on the target site as well as decrease adverse effects. For these reason, Memantine was loaded in biodegradable PLGA nanoparticles, produced by double emulsion method and surface-coated with polyethylene glycol. MEM-PEG-PLGA nanoparticles (NPs) were aimed to target the blood-brain barrier (BBB) upon oral administration for the treatment of Alzheimer's disease. RESULTS: The production parameters were optimized by design of experiments. MEM-PEG-PLGA NPs showed a mean particle size below 200 nm (152.6 ± 0.5 nm), monomodal size distribution (polydispersity index, PI < 0.1) and negative surface charge (- 22.4 mV). Physicochemical characterization of NPs confirmed that the crystalline drug was dispersed inside the PLGA matrix. MEM-PEG-PLGA NPs were found to be non-cytotoxic on brain cell lines (bEnd.3 and astrocytes). Memantine followed a slower release profile from the NPs against the free drug solution, allowing to reduce drug administration frequency in vivo. Nanoparticles were able to cross BBB both in vitro and in vivo. Behavioral tests carried out on transgenic APPswe/PS1dE9 mice demonstrated to enhance the benefit of decreasing memory impairment when using MEM-PEG-PLGA NPs in comparison to the free drug solution. Histological studies confirmed that MEM-PEG-PLGA NPs reduced ß-amyloid plaques and the associated inflammation characteristic of Alzheimer's disease. CONCLUSIONS: Memantine NPs were suitable for Alzheimer's disease and more effective than the free drug.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Antiparkinsonianos/farmacocinética , Disfunção Cognitiva/tratamento farmacológico , Portadores de Fármacos , Memantina/farmacocinética , Nanopartículas/química , Placa Amiloide/tratamento farmacológico , Administração Oral , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Antiparkinsonianos/química , Antiparkinsonianos/farmacologia , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/fisiopatologia , Modelos Animais de Doenças , Composição de Medicamentos/métodos , Emulsões , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Memantina/química , Memantina/farmacologia , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Tamanho da Partícula , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Poliésteres/química , Polietilenoglicóis/química
9.
BMC Anesthesiol ; 18(1): 47, 2018 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-29699479

RESUMO

BACKGROUND: There is increasing interest in whether anesthetic agents affect the risk or progression of Alzheimer's disease (AD). To mitigate many of the methodological issues encountered in human retrospective cohort studies we have used a transgenic model of AD to investigate the effect of propofol on AD pathology. METHODS: Six month-old amyloid precursor protein/presenilin 1 (APP/PS1) transgenic AD mice and control mice were exposed to 3 doses of propofol (200 mg/kg) or vehicle, delivered at monthly intervals. RESULTS: There was no difference in the extent of ß-amyloid (Aß) immunolabeled plaque deposition in APP/PS1 mice in vehicle versus propofol treatment groups. We also detected no difference in plaque-associated synapse loss in APP/PS1 mice following repeat propofol exposure relative to vehicle. Western blotting indicated that there was no difference in post-synaptic density protein 95, synaptophysin or glutamic acid decarboxylase 65/67 expression in control or APP/PS1 mice subjected to repeat propofol treatment relative to vehicle. CONCLUSIONS: These data suggest that repeat propofol anesthesia may not exacerbate plaque deposition or associated synapse loss in AD. Interestingly, this data also provides some of the first evidence suggesting that repeat propofol exposure in adult wild-type mice does not result in robust long-term alterations in the levels of key excitatory and inhibitory synaptic markers.


Assuntos
Doença de Alzheimer/patologia , Anestésicos Intravenosos/farmacologia , Encéfalo/efeitos dos fármacos , Placa Amiloide/patologia , Propofol/farmacologia , Sinapses/efeitos dos fármacos , Doença de Alzheimer/induzido quimicamente , Anestésicos Intravenosos/administração & dosagem , Animais , Western Blotting , Encéfalo/patologia , Encéfalo/ultraestrutura , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Transgênicos , Placa Amiloide/induzido quimicamente , Propofol/administração & dosagem , Sinapses/patologia
10.
Zhonghua Yi Xue Za Zhi ; 97(41): 3258-3262, 2017 Nov 07.
Artigo em Chinês | MEDLINE | ID: mdl-29141366

RESUMO

Objective: To construct magnetic nanoparticles targeting ß-amyloid (Aß) plaques, the pathological biomarker of Alzheimer's disease (AD) and to study their binding capability in vitro. Methods: Superparamagnetic nanoparticles Mn(0.6)Zn(0.4)Fe(2)O(4) (MZF) were coated with amphiphilic star-block copolymeric micelles and modified with Aß-specific probe Pittsburgh compound B (PiB) to construct a novel magnetic nanoparticle MZF-PiB, which specifically targeted amyloid plaques. Transmission electron microscope was used to study the morphological features of MZF-PiB. Superparamagnetism of MZF-PiB was assessed by its r(2) relaxation rate by using 3.0 T MRI scanner. Cytotoxic test was applied to determine biosafety of MZF-PiB nanoparticles in differentiated human neuroblastoma cells (SH-SY5Y) and Madin-Darby canine kidney (MDCK). In vitro binding tests were conducted via immunohistochemistry on 6-month old AD mice brain sections. Differences of cell viability between groups were compared with one-way analysis of variance. Results: MZF-PiB nanoparticles were successfully constructed. Transmission electron microscope images showed that the nanoparticles were about 100 nm in size. The r(2) relaxation rate was 163.11 mMS(-1). No differences were found in cell viability of SH-SY5Y and MDCK incubated with MZF-PiB suspension for 24 h or 48 h when compared with those of untreated cells (F=2.336, 2.539, 0.293, 1.493, all P>0.05). In vitro binding tests indicated that the MZF-PiB were specifically bound to amyloid plaques. The smallest size of detected plaques was 27 µm. Conclusion: PiB-modified nanoparticles targeting Aß are biologically safe and highly superparamagnetic, possessing the capability to detect amyloid plaques early in vitro and the potential for early diagnosis of AD.


Assuntos
Doença de Alzheimer/complicações , Compostos de Anilina , Nanopartículas de Magnetita , Placa Amiloide/diagnóstico , Tiazóis , Peptídeos beta-Amiloides , Animais , Encéfalo/patologia , Cães , Humanos , Camundongos , Tomografia por Emissão de Pósitrons
11.
J Neurosci Res ; 93(8): 1258-66, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25711385

RESUMO

Inappropriate activation of cyclin-dependent kinase 5 (CDK5) resulting from proteolytic release of the activator fragment p25 from the membrane contributes to the formation of neurofibrillary tangles, ß-amyloid (ßA) aggregation, and chronic neurodegeneration. At 18 months of age, 3× Tg-AD mice were sacrificed after either 3 weeks (short term) or 1 year (long term) of CDK5 knockdown. In short-term-treated animals, CDK5 knockdown reversed ßA aggregation in the hippocampi via inhibitory phosphorylation of glycogen synthase kinase 3ß Ser9 and activation of phosphatase PP2A. In long-term-treated animals, CDK5 knockdown induced a persistent reduction in CDK5 and prevented ßA aggregation, but the effect on amyloid precursor protein processing was reduced, suggesting that yearly booster therapy would be required. These findings further validate CDK5 as a target for preventing or blocking amyloidosis in older transgenic mice.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Quinase 5 Dependente de Ciclina/antagonistas & inibidores , Marcação de Genes/métodos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Agregação Patológica de Proteínas/prevenção & controle , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Quinase 5 Dependente de Ciclina/genética , Quinase 5 Dependente de Ciclina/metabolismo , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Camundongos , Camundongos Transgênicos , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo
12.
Bratisl Lek Listy ; 116(10): 580-1, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26531866

RESUMO

Pathologic characteristics of Alzheimer disease (AD) are ß-amyloid (Aß) plaques, neurofibrillary tangles (NFT) and neurodegeneration. Currently, there is no cure for AD. Cilostazol, a selective inhibitor of type 3 phosphodiesterase, is likely to be a promising agent for AD. In the brain of the experimental animals it significantly reduced the Aß amyloid plaques. Initial clinical reports on the effect of Cilostazol in AD patients are promising. In mice, stem cells favourably influence the pathogenetic process critical in AD, by reducing deposits of Aß plaques. Clinical trials of the drug, called Betablock, are already underway in Britain. Successful management and resolution of AD in man will still require further intensive research (Fig. 4, Ref. 11).


Assuntos
Doença de Alzheimer , Gerenciamento Clínico , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/epidemiologia , Doença de Alzheimer/terapia , Animais , Saúde Global , Humanos , Morbidade
13.
Curr Med Chem ; 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39206477

RESUMO

INTRODUCTION: Alzheimer's Disease (AD) is a complicated and advanced neurodegenerative condition accompanied by gradual cholinergic neuronal death and higher levels of monoamine oxidase-B (MAO-B) enzyme. In this study, a series of novel hybrid compounds combining 1,3,4-oxadiazole and quinoline moieties were synthesized and evaluated for their potential as inhibitors of acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), and MAO enzymes. METHODS: The chemical structures of the synthesized compounds were confirmed using various analytical techniques, such as mass spectrometry, infrared spectroscopy (IR), proton nuclear magnetic resonance (1H-NMR), and carbon and nuclear magnetic resonance (13C-NMR). The final products were evaluated for anticholinesterase potential by applying modified Ellman's spectrometric method, whereas a fluorometric method was used to assess MAO inhibition properties. In-silico studies using molecular docking and molecular dynamics simulation (MDS) methods has been also conducted. RESULTS: Among the synthesized compounds, 5a, 5c, and 6a demonstrated substantial activity against AChE, with IC50 values of 0.033 µM, 0.096 µM, and 0.177 µM, respectively. A molecular docking study was performed to elucidate the binding modes and establish the structure-activity relationship (SAR) of the most active compounds (5a, 5c, and 6a). Molecular dynamics simulation (MDS) of the most potent compound, 5a, was also conducted to examine the stability of the interactions with the receptor. Moreover, the physicochemical properties of the active products were also studied. CONCLUSION: Overall, this research contributes to the development of 1,3,4-oxadiazole- quinoline hybrids as potential AChE inhibitors for the treatment of Alzheimer's disease.

14.
Int Immunopharmacol ; 113(Pt A): 109368, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36330912

RESUMO

As the most common neurodegenerative disease, Alzheimer's disease (AD) exhibits an incomprehensible pathogenesis, which has led to the continuous failure of drug development in recent years. Although neuronal damage is considered a pathological feature of AD, treatment strategies targeting ß-amyloid (Aß) have not achieved beneficial effects. In-depth research on glial cells has revealed the strong importance and application prospects of astrocytes in the recovery of cognitive functions. This review summarizes the role of astrocytes in AD and the possibility of therapeutic strategies targeting astrocytes. Astrocytes are involved in brain lipid metabolism and can regulate the synthesis and degradation of Aß to affect the pathology of AD. The tau protein is phosphorylated by astrocytes, and this phosphorylation leads to the formation of neurofibrillary tangles (NFTs). Astrocytes can express a variety of receptors and inflammatory factors and participate in the neuroinflammatory process and the release of proinflammatory mediators. When the glutamate produced by the neurons is not cleared by astrocytes, neurons undergo apoptosis due to blocked cell metabolism. Therapies for astrocytes are highly efficient, and these include stem cell therapy, gene editing technology, astrocyte transformation and chemical drugs. Here, we discuss the advantages and disadvantages of animal and cell models applied to the study of targeted astrocyte therapies. This study helps elucidate the mechanism of astrocytes in AD and promotes the clinical application of potential therapeutic strategies targeting astrocytes.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Animais , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Astrócitos , Doenças Neurodegenerativas/metabolismo , Peptídeos beta-Amiloides/metabolismo , Neurônios
15.
Curr Neuropharmacol ; 20(4): 675-692, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33998995

RESUMO

Alzheimer's disease (AD) is one of the major reasons for 60-80% cases of senile dementia occurring as a result of the accumulation of plaques and tangles in the hippocampal and cortical neurons of the brain leading to neurodegeneration and cell death. The other pathological features of AD comprise abnormal microvasculature, network abnormalities, interneuronal dysfunction, increased ß-amyloid production and reduced clearance, increased inflammatory response, elevated production of reactive oxygen species, impaired brain metabolism, hyperphosphorylation of tau, and disruption of acetylcholine signaling. Among all these pathologies, Mitochondrial Dysfunction (MD), regardless of it being an inciting insult or a consequence of the alterations, is related to all the associated AD pathologies. Observed altered mitochondrial morphology, distribution and movement, increased oxidative stress, dysregulation of enzymes involved in mitochondrial functioning, impaired brain metabolism, and impaired mitochondrial biogenesis in AD subjects suggest the involvement of mitochondrial malfunction in the progression of AD. Here, various pre-clinical and clinical evidence establishing MD as a key mediator in the progression of neurodegeneration in AD are reviewed and discussed with an aim to foster future MD based drug development research for the management of AD.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Desenvolvimento de Medicamentos , Humanos , Mitocôndrias/metabolismo , Placa Amiloide/metabolismo
16.
Neurol Res ; 44(12): 1053-1065, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35981107

RESUMO

OBJECTIVE: Numerous pathological variations and complex interactions are involved in the long period prior to cognitive decline in brains with Alzheimer's disease (AD). Thus, elucidation of the pathological disorders can facilitate early AD diagnosis. The aim of this study was to investigate the age-specific pathological changes of ß-amyloid plaques in brain tissues of AD mice at different ages. METHODS: We arranged the most widely available APP/PS1 transgenic AD models into six age groups: 3, 4 and 6 months (these three groups mimicked early-clinical stage AD), 9, 12 and 15 months (these three groups mimicked late-clinical stage AD). Cell morphology and arrangement in the cortex and hippocampus were observed by hematoxylin and eosin (HE) staining. Congo red staining and immunohistochemical staining were performed to exhibit the distribution of ß-amyloid plaques in the cortex and hippocampus of AD brains. RESULTS: Our results found that as age increased, the nuclei of cortical and hippocampal cells in AD mice were severely damaged. The number and area of ß-amyloid plaques increased in AD mice in correspondence with age revealed by histological experiments. Importantly, ß-amyloid plaques were detected in the cortex and hippocampus of 6-month-old AD mice shown by Congo red staining while detected in the cortex and hippocampus of 4-month-old AD mice shown by immunohistochemical staining. CONCLUSIONS: The current study revealed the age-related pathological changes of ß-amyloid plaques in the cortex and hippocampus of AD mice and displayed a higher specificity of immunohistochemical staining than Congo red staining when detecting pathological changes of brain tissues.


Assuntos
Fatores Etários , Doença de Alzheimer , Placa Amiloide , Animais , Camundongos , Peptídeos beta-Amiloides , Camundongos Transgênicos
17.
Biomed Pharmacother ; 151: 113188, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35676788

RESUMO

Protein tyrosine phosphatases (PTPs) are important regulator of neuronal signal transduction and a growing number of PTPs have been implicated in Alzheimer's disease (AD). In the brains of patients with AD, there are a variety of abnormally phosphorylated proteins, which are closely related to the abnormal expression and activity of PTPs. ß-Amyloid plaques (Aß) and hyperphosphorylated tau protein are two pathological hallmarks of AD, and their accumulation ultimately leads to neurodegeneration. Studies have shown that protein phosphorylation signaling pathways mediates intracellular accumulation of Aß and tau during AD development and are involved in synaptic plasticity and other stress responses. Here, we summarized the roles of PTPs related to the pathogenesis of AD and analyzed their therapeutic potential in AD.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Humanos , Fosforilação , Placa Amiloide , Proteínas Tirosina Fosfatases , Proteínas tau/metabolismo
18.
Basic Clin Neurosci ; 12(3): 383-394, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34917297

RESUMO

INTRODUCTION: The increasing cases of Alzheimer Disease (AD) has caused numerous problems. The risk of developing AD increases in menopausal women, too. Apigenin and ß-estradiol are effective antioxidant and neuroprotective agents. We conducted the present study to explore their combined effects on ß-amyloid plaque formation, memory, and learning in ovariectomized rats. METHODS: Forty-two Wistar rats were randomly assigned into 6 groups: 1) ovariectomized (OVX), 2) OVX + apigenin, 3) OVX + ß-estradiol, 4) OVX + apigenin + ß-estradiol, 5 &6) vehicle shams for E2 and API, and 7) surgical sham. Treatment was done with apigenin and ß-estradiol. Then, we studied the formation of ß-amyloid plaques, neuronal density in the hippocampus area, apoptosis, memory, and learning. RESULTS: Findings showed the significant formation of ß-amyloid plaques in the hippocampus of OVX animals and their memory impairment. Apigenin and ß-estradiol significantly reduced the number of ß-amyloid plaques, as well as the symptoms of memory impairment and learning, and decreased the expression of caspase-3 in treated animals. CONCLUSION: Accordingly, ß-estradiol and apigenin could have more potent therapeutic effects on AD.

19.
Curr Alzheimer Res ; 18(6): 470-481, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34455970

RESUMO

BACKGROUND: In Alzheimer's disease (AD), and amyloid models such as the 5XFAD mouse, butyrylcholinesterase (BChE) is associated with ß-amyloid (Aß) plaques and has unique biochemical features which distinguish it from that found in neurons. It has been suggested that BChE associated with Aß plaques may be involved in the maturation of this structure and thus disease progression. OBJECTIVE: Currently, it is unknown whether BChE bound to Aß plaques has altered biochemical properties due to a different primary structure or because of the association of this enzyme with Aß plaques. Also, the source and binding mechanism of this BChE remains unknown. METHODS: Brain tissue sections from the 5XFAD/BChE-KO mouse were incubated with exogenous sources of BChE and stained for this enzyme's activity. Efforts were made to determine what region of BChE or Aß may be involved in this association. RESULTS: We found that incubation of 5XFAD/BChE-KO brain tissues with exogenous BChE led to this enzyme becoming associated with Aß plaques and neurons. In contrast to neuronal BChE, the BChE bound to Aß plaques had similar biochemical properties to those seen in AD. Mutations to BChE and efforts to block Aß epitomes failed to prevent this association. CONCLUSION: The association of BChE with Aß plaques, and the resultant biochemical changes, suggests that BChE may undergo a conformational change when bound to Aß plaques but not neurons. The 5XFAD/BChE-KO model is ideally suited to explore the binding mechanism of BChE to Aß plaques as well as the involvement of BChE in AD pathogenesis.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Butirilcolinesterase/metabolismo , Placa Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos
20.
Front Endocrinol (Lausanne) ; 11: 560375, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33224105

RESUMO

Insulin plays a major neuroprotective and trophic function for cerebral cell population, thus countering apoptosis, beta-amyloid toxicity, and oxidative stress; favoring neuronal survival; and enhancing memory and learning processes. Insulin resistance and impaired cerebral glucose metabolism are invariantly reported in Alzheimer's disease (AD) and other neurodegenerative processes. AD is a fatal neurodegenerative disorder in which progressive glucose hypometabolism parallels to cognitive impairment. Although AD may appear and progress in virtue of multifactorial nosogenic ingredients, multiple interperpetuative and interconnected vicious circles appear to drive disease pathophysiology. The disease is primarily a metabolic/energetic disorder in which amyloid accumulation may appear as a by-product of more proximal events, especially in the late-onset form. As a bridge between AD and type 2 diabetes, activation of c-Jun N-terminal kinase (JNK) pathway with the ensued serine phosphorylation of the insulin response substrate (IRS)-1/2 may be at the crossroads of insulin resistance and its subsequent dysmetabolic consequences. Central insulin axis bankruptcy translates in neuronal vulnerability and demise. As a link in the chain of pathogenic vicious circles, mitochondrial dysfunction, oxidative stress, and peripheral/central immune-inflammation are increasingly advocated as major pathology drivers. Pharmacological interventions addressed to preserve insulin axis physiology, mitochondrial biogenesis-integral functionality, and mitophagy of diseased organelles may attenuate the adjacent spillover of free radicals that further perpetuate mitochondrial damages and catalyze inflammation. Central and/or peripheral inflammation may account for a local flood of proinflammatory cytokines that along with astrogliosis amplify insulin resistance, mitochondrial dysfunction, and oxidative stress. All these elements are endogenous stressor, pro-senescent factors that contribute to JNK activation. Taken together, these evidences incite to identify novel multi-mechanistic approaches to succeed in ameliorating this pandemic affliction.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Metabolismo Energético/fisiologia , Resistência à Insulina/fisiologia , Peptídeos beta-Amiloides/metabolismo , Animais , Humanos , Estresse Oxidativo/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa