Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Exp Gerontol ; 157: 111645, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34843902

RESUMO

BACKGROUND: Oxidative stress implicates in Alzheimer's disease (AD) pathophysiology, and associates with the creation of end products of free radical reactions, are known as lipophilic fluorescent products (LFPs). This study aimed to evaluate the probable parallel alterations in the spectral properties of the LFPs in the hippocampus tissues, cerebrospinal fluid (CSF), plasma, and erythrocytes during AD model induction by intra-cerebroventricular (ICV) amyloid ß-protein fragment 25-35 (Aß) injection. METHODS: Male rats received an intra-ICV injection of Aß. Hippocampus, CSF, plasma, and erythrocytes were harvested at 5, 14, and 21 days after Aß injection. The fluorescent intensity of LFPs was assessed by spectrofluorimetry using synchronous fluorescence spectra 25 (SYN 25) and 50 (SYN 50) in the range of 250-500 nm. Hippocampal tissue malondialdehyde (MDA) and superoxide dismutase (SOD) were also measured. Cognitive alterations were evaluated using Morris water maze (MWM) test. RESULTS: The parallel significant rise in the fluorescence intensity of LFPs was detected in the hippocampus, CSF, plasma, and erythrocytes, 14, and 21 days after ICV-Aß injection. These alterations were found in both types of synchronous spectra 25, and 50, and were coincided with hippocampal cognitive decline, the MDA rise, and decrease of SOD activity. There was a positive correlation between hippocampus homogenate, and plasma or CSF rise in fluorescence intensity. CONCLUSION: Data showed that the Aß increased hippocampal MDA, and decreased SOD activity, led to a higher rate of oxidative products and subsequently resulted in an increase in LFPs fluorescence intensity during the development of cognitive decline. LFPs' alterations reflect a comprehensive view of tissue redox status. The fluorescence properties of LFPs indicate their composition, which may pave the way to trace the different pathological states.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Peptídeos beta-Amiloides/toxicidade , Animais , Modelos Animais de Doenças , Eritrócitos , Hipocampo , Masculino , Aprendizagem em Labirinto , Estresse Oxidativo , Fragmentos de Peptídeos , Ratos
2.
Cells ; 10(9)2021 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-34572130

RESUMO

BACKGROUND: Whole transgenic or non-transgenic organism model systems allow the screening of pharmacological compounds for protective actions in Alzheimer's disease (AD). AIM: In this study, a plant parasitic nematode, Globodera pallida, which assimilates intact peptides from the external environment, was investigated as a new potential non-transgenic model system of AD. Methods: Fresh second-stage juveniles of G. pallida were used to measure their chemosensory, perform immunocytochemistry on their neurological structures, evaluate their survival rate, measure reactive oxygen species, and determine total oxidized glutathione to reduced glutathione ratio (GSSG/GSH) levels, before and after treatment with 100 µM of various amyloid beta (Aß) peptides (1-40, 1-42, 17-42, 17-40, 1-28, or 1-16). Wild-type N2 C. elegans (strain N2) was cultured on Nematode Growth Medium and directly used, as control, for chemosensory assays. RESULTS: We demonstrated that: (i) G. pallida (unlike Caenorhabditis elegans) assimilates amyloid-ß (Aß) peptides which co-localise with its neurological structures; (ii) pre-treatment with various Aß isoforms (1-40, 1-42, 17-42, 17-40, 1-28, or 1-16) impairs G. pallida's chemotaxis to differing extents; (iii) Aß peptides reduced survival, increased the production of ROS, and increased GSSG/GSH levels in this model; (iv) this unique model can distinguish differences between different treatment concentrations, durations, and modalities, displaying good sensitivity; (v) clinically approved neuroprotective agents were effective in protecting G. pallida from Aß (1-42) exposure. Taken together, the data indicate that G. pallida is an interesting in vivo model with strong potential for discovery of novel bioactive compounds with anti-AD activity.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/toxicidade , Animais Geneticamente Modificados/fisiologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Tylenchoidea/fisiologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/fisiologia , Quimiotaxia , Tylenchoidea/efeitos dos fármacos
3.
J Biophotonics ; 13(7): e202000067, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32306519

RESUMO

In this study, a neurotoxicity model of zebrafish induced by amyloid beta (Aß) protein was developed and evaluated in vivo by optical coherence tomography (OCT). Aß protein and phosphate buffer saline (PBS) were separately injected into the head of two groups of adult zebrafish (n = 6 per group). Congo-red staining results confirmed that Aß protein had penetrated into brain tissue. All zebrafish were imaged with OCT on the 0th, 5th, 10th, 15th and 20th day postinjection. OCT images showed that PBS is not toxic to brain tissue. However, significant brain atrophy could be seen in the OCT images of zebrafish injected with Aß-protein that was verified by histological consequences. In addition, zebrafish in the model group showed memory decline in behavioral tests. This study verified the feasibility of in vivo long-term assessment of Aß protein-induced brain atrophy in adult zebrafish by OCT that has great potential to be applied in the neurological diseases research.


Assuntos
Tomografia de Coerência Óptica , Peixe-Zebra , Peptídeos beta-Amiloides/toxicidade , Animais , Atrofia , Encéfalo/diagnóstico por imagem
4.
Metab Brain Dis ; 35(2): 263-274, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31853828

RESUMO

Regulatory role of vitamin D (VitD) in cognitive memory and learning has been proposed. Here, we examine the behavioral and biochemical effects of VitD in Alzheimer's disease (AD), as the most common form of dementia, in male Wistar rats. Animals (n = 48) were randomly divided into six groups: control, sham solvent, sham surgery, VitD (by intraperitoneal injection), AD (receiving intrahippocampal injection of amyloid-beta peptide, Aß), and combination of VitD and Aß. Learning and memory functions were investigated through the passive avoidance and the Morris water maze (MWM) tasks. Moreover, oxidative stress biomarkers including total antioxidant capacity (TAC), total thiol groups (TTG), lipid peroxidation (LPO), and DNA damage were assessed in hippocampus and serum. In passive avoidance task, Aß significantly impaired the step-through latency and time in dark compartment. It also increased escape latency and time spent in the target quadrant in the MWM. VitD administration attenuated the Aß-induced memory impairment in passive avoidance and MWM tests. Furthermore, VitD reduced deleterious biochemical effect of Aß by enhancing the levels of TAC and TTG in addition to decreasing LPO and DNA damage levels in both hippocampus and serum. We showed, for the first time, that VitD administration improves the impaired Aß-induced memory and that, by acting as a strong antioxidant, it can attenuate the stress oxidative biomarkers in hippocampus and serum of rats with AD. Altogether, our results provide evidence for further application of VitD in neurodegenerative disorders such as AD to enlighten the involved mechanisms.


Assuntos
Doença de Alzheimer/sangue , Doença de Alzheimer/tratamento farmacológico , Aprendizagem da Esquiva/efeitos dos fármacos , Modelos Animais de Doenças , Hipocampo/metabolismo , Vitamina D/uso terapêutico , Doença de Alzheimer/induzido quimicamente , Peptídeos beta-Amiloides/toxicidade , Animais , Aprendizagem da Esquiva/fisiologia , Hipocampo/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/fisiologia , Masculino , Distribuição Aleatória , Ratos , Ratos Wistar , Resultado do Tratamento , Vitamina D/farmacologia
5.
Drug Deliv Transl Res ; 8(5): 1406-1420, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30117120

RESUMO

Curcumin (CUR), a natural polyphenolic compound, is considered as one of the most potential candidates against Alzheimer disease (AD) by targeting multiple pathologies such as amyloid-beta, tau phosphorylation, and oxidative stress. Poor physicochemical profile and oral bioavailability (BA) are the major contributors to its failure in clinical trials. Lack of success in numerous drug clinical trials for the treatment of AD urges the need of repositioning of CUR. To overcome its limitation and enhance oral BA, Novel CUR Formulation (NCF) was developed using self-nanomicellizing solid dispersion strategy which displayed 117-fold enhancement in oral BA of CUR. NCF was tested using SH-SY5Y695 APP human neuroblastoma cell line against the cytotoxicity induced by copper metal ion, H2O2, and Aß42 oligomer and compared with CUR control. The safety and efficacy of NCF on mice AD-like behavioral deficits (open field, novel objective recognition, Y-maze, and Morris water maze tests) were assessed in transgenic AD (APPSwe/PS1deE9) mice model. In SH-SY5Y695 APP human neuroblastoma cell line, NCF showed better safety and efficacy against the cytotoxicity due to the significantly enhancement of cellular uptake. It not only prevents the deterioration of cognitive functions of the aged APPSwe/PS1deE9 mice during aging but also reverses the cognitive functions to their much younger age which is also better than the currently available approved options. Moreover, NCF was proved as well tolerated with no appearance of any significant toxicity via oral administration. The results of the study demonstrated the potential of NCF to improve the efficacy of CUR without compromising its safety profile, and pave the way for clinical development for AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Disfunção Cognitiva/prevenção & controle , Curcumina/administração & dosagem , Nanoestruturas/química , Administração Oral , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/toxicidade , Animais , Linhagem Celular , Cobre/toxicidade , Curcumina/efeitos adversos , Curcumina/química , Modelos Animais de Doenças , Composição de Medicamentos , Humanos , Peróxido de Hidrogênio/toxicidade , Masculino , Camundongos , Camundongos Transgênicos , Micelas , Fragmentos de Peptídeos/toxicidade
6.
J Biophotonics ; 10(2): 311-319, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27090065

RESUMO

Manipulating neural activity is crucial for studying the neural connectivity and the pathophysiology of neurodegenerative disease. Among various techniques for neural activation, direct optical stimulation method with femtosecond-pulsed laser is simple and can be specifically applied on a single neuron. Brief irradiation of femtosecond laser pulses on a neuron elevates intracellular calcium, and it propagates to adjacent neurons. However, the mechanisms of laser-induced neural activation are still unclear. In this report, we have elucidated the mechanism of laser-induced neural activation which could be mediated by superoxide, specifically blocked by diphenyleneiodonium chloride, and depletion in intracellular calcium storage. Furthermore, we also showed that the propagation of calcium initiated by laser stimulation is dependent on the presence of extracellular calcium as well as electrical and chemical synapses. We verified the applicability of such mechanism for the assessment of neuronal functionality, by measuring calcium elevation, intracellular calcium propagation, ROS increase, and performing cell death assay in vehicle and Aß-treated neurons. This work suggests promising applications of the potential for implementing such laser-induced neural activation for rapid and reliable drug screening.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Lasers , Neurônios/efeitos da radiação , Animais , Cálcio/metabolismo , Morte Celular , Células Cultivadas , Hipocampo/citologia , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Sinapses
7.
J Neuroinflammation ; 13(1): 221, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27578213

RESUMO

BACKGROUND: Obesity has been identified as a risk factor for cognitive decline and Alzheimer's disease (AD). The aim of this study was to investigate the effect of obesity on neuroinflammation and cerebral glucose metabolism using PET in a mouse model of ß-amyloidosis and determine the relationship between these PET imaging biomarkers, pathogenic changes, and functional outcomes. METHODS: Three-month-old C57BL/J6 mice were fed either a standard (control group) or high-fat diet (obese group) for 3 months and intracerebroventricularly infused with vehicle or human beta amyloid 1-42 (Aß42). We assessed obesity-induced abnormalities in peripheral metabolic indices including adiposity, fasting glucose, and glucose tolerance. Brain glucose metabolism was assessed by (18)F-FDG PET, and glial activation was assessed using the translocator protein (TSPO) ligand (11)C-PBR-28. TSPO expression was confirmed by immunohistochemistry of brain sections obtained from scanned mice. The association between inflammatory state and (11)C-PBR-28 PET signals was characterized by examination of the cytokine expression profile in both the serum and hippocampus by antibody array. Learning and memory performance was assessed in the object recognition task, and anxiety-related behavior was assessed in the elevated plus maze. RESULTS: Obesity combined with Aß infusion promoted neuroinflammation and cerebral hypermetabolism, and these signals were significant predictors of learning and memory performance in the object recognition task. In vivo TSPO signals were associated with inflammatory markers including CXCL1, CXCL2, CXCL12, CCL3, CCL5, TIMP-1, G-CSF, sICAM-1, and IL-1ra. CONCLUSIONS: In vivo cerebral metabolism and TSPO signals indicate that obesity can accelerate amyloid-induced inflammation and associated cognitive decline.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Amiloidose/diagnóstico por imagem , Modelos Animais de Doenças , Obesidade/diagnóstico por imagem , Fragmentos de Peptídeos/toxicidade , Tomografia por Emissão de Pósitrons , Amiloidose/induzido quimicamente , Amiloidose/imunologia , Animais , Dieta Hiperlipídica/efeitos adversos , Inflamação/induzido quimicamente , Inflamação/diagnóstico por imagem , Inflamação/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/imunologia , Tomografia por Emissão de Pósitrons/métodos , Distribuição Aleatória
8.
J Alzheimers Dis ; 46(4): 1091-101, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26402635

RESUMO

Utilizing decision making biomarkers in drug development requires thorough assay validation. Special considerations need to be taken into account when monitoring biomarkers using immunoassays in the presence of therapeutic antibodies. We have developed robust and sensitive assays to assess target engagement and proof of mechanism to support the clinical progression of a human monoclonal antibody against the neurotoxic amyloid-ß (Aß)42 peptide. Here we present the introduction of novel pre-treatment steps to ensure drug-tolerant immunoassays and describe the validation of the complete experimental procedures to measure total Aß42 concentration (bound and unbound) in cerebrospinal fluid (CSF) and plasma, free Aß42 concentration (unbound) in CSF, and Aß40 concentration in CSF. The difference in composition of the matrices (CSF and plasma) and antigen levels therein, in combination with the hydrophobic properties of Aß protein, adds to the complexity of validation. Monitoring pharmacodynamics of an Aß42 specific monoclonal antibody in a non-human primate toxicology study using these assays, we demonstrated a 1500-fold and a 3000-fold increase in total Aß42 in plasma, a 4-fold and 8-fold increase in total Aß42 in CSF together with a 95% and 96% reduction of free Aß42 in CSF following weekly intravenous injections of 10 mg/kg and 100 mg/kg, respectively. Levels of Aß40 were unchanged. The accuracy of these data is supported by previous pre-clinical studies as well as predictive pharmacokinetic/pharmacodynamics modeling. In contrast, when analyzing the same non-human primate samples excluding the pre-treatment steps, we were not able to distinguish between free and total Aß42. Our data clearly demonstrate the importance of thorough evaluation of antibody interference and appropriate validation to monitor different types of biomarkers in the presence of a therapeutic antibody.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/toxicidade , Anticorpos Monoclonais/uso terapêutico , Imunoensaio/métodos , Fragmentos de Peptídeos/líquido cefalorraquidiano , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/toxicidade , Doença de Alzheimer/sangue , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/induzido quimicamente , Peptídeos beta-Amiloides/sangue , Animais , Modelos Animais de Doenças , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Macaca fascicularis , Masculino , Fragmentos de Peptídeos/sangue , Reprodutibilidade dos Testes , Fatores de Tempo
9.
ACS Chem Neurosci ; 4(4): 547-65, 2013 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-23379636

RESUMO

The synthesis, molecular modeling, and pharmacological analysis of phenoxyalkylamino-4-phenylnicotinates (2-7), phenoxyalkoxybenzylidenemalononitriles (12, 13), pyridonepezils (14-18), and quinolinodonepezils (19-21) are described. Pyridonepezils 15-18 were found to be selective and moderately potent regarding the inhibition of hAChE, whereas quinolinodonepezils 19-21 were found to be poor inhibitors of hAChE. The most potent and selective hAChE inhibitor was ethyl 6-(4-(1-benzylpiperidin-4-yl)butylamino)-5-cyano-2-methyl-4-phenylnicotinate (18) [IC(50) (hAChE) = 0.25 ± 0.02 µM]. Pyridonepezils 15-18 and quinolinodonepezils 20-21 are more potent selective inhibitors of EeAChE than hAChE. The most potent and selective EeAChE inhibitor was ethyl 6-(2-(1-benzylpiperidin-4-yl)ethylamino)-5-cyano-2-methyl-4-phenylnicotinate (16) [IC(50) (EeAChE) = 0.0167 ± 0.0002 µM], which exhibits the same inhibitory potency as donepezil against hAChE. Compounds 2, 7, 13, 17, 18, 35, and 36 significantly prevented the decrease in cell viability caused by Aß(1-42). All compounds were effective in preventing the enhancement of AChE activity induced by Aß(1-42). Compounds 2-7 caused a significant reduction whereas pyridonepezils 17 and 18, and compound 16 also showed some activity. The pyrazolo[3,4-b]quinolines 36 and 38 also prevented the upregulation of AChE induced by Aß(1-42). Compounds 2, 7, 12, 13, 17, 18, and 36 may act as antagonists of voltage sensitive calcium channels, since they significantly prevented the Ca(2+) influx evoked by KCl depolarization. Docking studies show that compounds 16 and 18 adopted different orientations and conformations inside the active-site gorges of hAChE and hBuChE. The structural and energetic features of the 16-AChE and 18-AChE complexes compared to the 16-BuChE and 18-BuChE complexes account for a higher affinity of the ligand toward AChE. The present data indicate that compounds 2, 7, 17, 18, and 36 may represent attractive multipotent molecules for the potential treatment of Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Butirilcolinesterase , Inibidores da Colinesterase/síntese química , Inibidores da Colinesterase/farmacologia , Modelos Moleculares , Fragmentos de Peptídeos/toxicidade , Butirilcolinesterase/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Humanos
10.
Neurobiol Dis ; 7(6 Pt B): 644-56, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11114262

RESUMO

It has been shown recently that the generation of an abnormal transmembrane form of the prion protein ((Ctm)PrP) is involved in the neurodegeneration process during inherited and infectious prion diseases but a causative relationship has never been established. We wanted to know if and how the proposed transmembrane domain of PrP could induce neuronal dysfunction. Thus, we investigated the neurotoxic properties of two peptides whose sequences are encompassed within this domain. We show that PrP peptides 118-135 and 105-132 as well as an amidated more soluble peptide 105-132 induce the death of pure cortical neurons originating from normal and PrP knockout mice. This can be correlated with the high propensity of these peptides to insert stably into and to destabilize cell membranes. Through this study, we have identified a novel mechanism of neurotoxicity for PrP, which directly involves membrane perturbation; this mechanism is independent of fibril formation and probably corresponds to the effect of the transmembrane insertion of (Ctm)PrP.


Assuntos
Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Doenças Priônicas/metabolismo , Príons/toxicidade , Peptídeos beta-Amiloides/toxicidade , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Aminoácidos Excitatórios/farmacologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica , Modelos Moleculares , Método de Monte Carlo , Neurônios/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/ultraestrutura , Doenças Priônicas/etiologia , Príons/biossíntese , Príons/síntese química , Príons/química , Príons/ultraestrutura , Estrutura Terciária de Proteína
11.
Neurobiol Aging ; 13(5): 609-12, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1461350

RESUMO

Cell culture systems for evaluating the biological effects of the beta-amyloid protein are potentially important tools in the study of the pathogenesis of Alzheimer's disease. In this report, methodological considerations in the assessment of beta-amyloid neurotoxicity are discussed. Chronic incubation of beta 1-40 in primary human cortical cultures results in progressive neuronal degeneration. The neurodegenerative process occurs in association with localized deposition of beta-amyloid on the neuronal soma ultimately resulting in the formation of compact beta-amyloid deposits. A beta 1-40 preparation from another laboratory was tested that did not form neuronal beta-amyloid deposits and was not neurotoxic. Thus, the conformational state of the beta 1-40 peptide leading to the formation of neuronal amyloid deposits is an important determinant of neurotoxicity. Variables in peptide preparation that influence this property may account for variation in neurotoxic potency.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Sistema Nervoso/patologia , Animais , Células Cultivadas , Humanos , Sistema Nervoso/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA