Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Biochemistry ; 60(37): 2773-2780, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34469142

ABSTRACT

The prevailing opinion is that prefibrillar ß-amyloid (Aß) species, rather than end-stage amyloid fibrils, cause neuronal dysfunction in Alzheimer's disease, although the mechanisms behind Aß neurotoxicity remain to be elucidated. Luminescent conjugated oligothiophenes (LCOs) exhibit spectral properties upon binding to amyloid proteins and have previously been reported to change the toxicity of Aß1-42 and prion protein. In a previous study, we showed that an LCO, pentamer formyl thiophene acetic acid (p-FTAA), changed the toxicity of Aß1-42. Here we investigated whether an LCO, heptamer formyl thiophene acetic acid (h-FTAA), could change the toxicity of Aß1-42 by comparing its behavior with that of p-FTAA. Moreover, we investigated the effects on toxicity when Aß with the Arctic mutation (AßArc) was aggregated with both LCOs. Cell viability assays on SH-SY5Y neuroblastoma cells demonstrated that h-FTAA has a stronger impact on Aß1-42 toxicity than does p-FTAA. Interestingly, h-FTAA, but not p-FTAA, rescued the AßArc-mediated toxicity. Aggregation kinetics and binding assay experiments with Aß1-42 and AßArc when aggregated with both LCOs showed that h-FTAA and p-FTAA either interact with different species or affect the aggregation in different ways. In conclusion, h-FTAA protects against Aß1-42 and AßArc toxicity, thus showing h-FTAA to be a useful tool for improving our understanding of the process of Aß aggregation linked to cytotoxicity.


Subject(s)
Acetates/chemistry , Amyloid beta-Protein Precursor/metabolism , Thiophenes/chemistry , Acetates/metabolism , Amyloid/chemistry , Amyloid beta-Peptides/chemistry , Amyloid beta-Protein Precursor/physiology , Amyloid beta-Protein Precursor/toxicity , Amyloidogenic Proteins/chemistry , Fluorescent Dyes/chemistry , Humans , Kinetics , Luminescence , Peptide Fragments/metabolism , Protein Aggregates/drug effects , Protein Aggregates/physiology , Staining and Labeling/methods , Thiophenes/metabolism
2.
FEBS J ; 283(19): 3508-3522, 2016 10.
Article in English | MEDLINE | ID: mdl-27562772

ABSTRACT

Genetic polymorphisms of immune genes that associate with higher risk to develop Alzheimer's disease (AD) have led to an increased research interest on the involvement of the immune system in AD pathogenesis. A link between amyloid pathology and immune gene expression was suggested in a genome-wide gene expression study of transgenic amyloid mouse models. In this study, the gene expression of lysozyme, a major player in the innate immune system, was found to be increased in a comparable pattern as the amyloid pathology developed in transgenic mouse models of AD. A similar pattern was seen at protein levels of lysozyme in human AD brain and CSF, but this lysozyme pattern was not seen in a tau transgenic mouse model. Lysozyme was demonstrated to be beneficial for different Drosophila melanogaster models of AD. In flies that expressed Aß1-42 or AßPP together with BACE1 in the eyes, the rough eye phenotype indicative of toxicity was completely rescued by coexpression of lysozyme. In Drosophila flies bearing the Aß1-42 variant with the Arctic gene mutation, lysozyme increased the fly survival and decreased locomotor dysfunction dose dependently. An interaction between lysozyme and Aß1-42 in the Drosophila eye was discovered. We propose that the increased levels of lysozyme, seen in mouse models of AD and in human AD cases, were triggered by Aß1-42 and caused a beneficial effect by binding of lysozyme to toxic species of Aß1-42 , which prevented these from exerting their toxic effects. These results emphasize the possibility of lysozyme as biomarker and therapeutic target for AD.


Subject(s)
Alzheimer Disease/enzymology , Muramidase/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Brain/enzymology , Brain/pathology , Disease Models, Animal , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/ultrastructure , Eye/metabolism , Eye/ultrastructure , Female , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Muramidase/genetics , Mutation , Peptide Fragments/metabolism , RNA, Messenger/metabolism
3.
Biol Open ; 5(8): 1030-9, 2016 Aug 15.
Article in English | MEDLINE | ID: mdl-27387531

ABSTRACT

The aggregation of the amyloid-ß (Aß) peptide into fibrillar deposits has long been considered the key neuropathological hallmark of Alzheimer's disease (AD). Aß peptides are generated from proteolytic processing of the transmembrane Aß precursor protein (AßPP) via sequential proteolysis through the ß-secretase activity of ß-site AßPP-cleaving enzyme (BACE1) and by the intramembranous enzyme γ-secretase. For over a decade, Drosophila melanogaster has been used as a model organism to study AD, and two different approaches have been developed to investigate the toxicity caused by AD-associated gene products in vivo In one model, the Aß peptide is directly over-expressed fused to a signal peptide, allowing secretion of the peptide into the extracellular space. In the other model, human AßPP is co-expressed with human BACE1, resulting in production of the Aß peptide through the processing of AßPP by BACE1 and by endogenous fly γ-secretase. Here, we performed a parallel study of flies that expressed the Aß1-42 peptide alone or that co-expressed AßPP and BACE1. Toxic effects (assessed by eye phenotype, longevity and locomotor assays) and levels of the Aß1-42, Aß1-40 and Aß1-38 peptides were examined. Our data reveal that the toxic effect per amount of detected Aß1-42 peptide was higher in the flies co-expressing AßPP and BACE1 than in the Aß1-42-expressing flies, and that the co-existence of Aß1-42 and Aß1-40 in the flies co-expressing AßPP and BACE1 could be of significant importance to the neurotoxic effect detected in these flies. Thus, the toxicity detected in these two fly models seems to have different modes of action and is highly dependent on how and where the peptide is generated rather than on the actual level of the Aß1-42 peptide in the flies. This is important knowledge that needs to be taken into consideration when using Drosophila models to investigate disease mechanisms or therapeutic strategies in AD research.

4.
J Biol Chem ; 291(17): 9233-43, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-26907684

ABSTRACT

Aggregation of the amyloid-ß peptide (Aß) in the brain leads to the formation of extracellular amyloid plaques, which is one of the pathological hallmarks of Alzheimer disease (AD). It is a general hypothesis that soluble prefibrillar assemblies of the Aß peptide, rather than mature amyloid fibrils, cause neuronal dysfunction and memory impairment in AD. Thus, reducing the level of these prefibrillar species by using molecules that can interfere with the Aß fibrillation pathway may be a valid approach to reduce Aß cytotoxicity. Luminescent-conjugated oligothiophenes (LCOs) have amyloid binding properties and spectral properties that differ when they bind to protein aggregates with different morphologies and can therefore be used to visualize protein aggregates. In this study, cell toxicity experiments and biophysical studies demonstrated that the LCO p-FTAA was able to reduce the pool of soluble toxic Aß species in favor of the formation of larger insoluble nontoxic amyloid fibrils, there by counteracting Aß-mediated cytotoxicity. Moreover, p-FTAA bound to early formed Aß species and induced a rapid formation of ß-sheet structures. These p-FTAA generated amyloid fibrils were less hydrophobic and more resistant to proteolysis by proteinase K. In summary, our data show that p-FTAA promoted the formation of insoluble and stable Aß species that were nontoxic which indicates that p-FTAA might have therapeutic potential.


Subject(s)
Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Protein Aggregation, Pathological/metabolism , Thiophenes/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cell Line, Tumor , Humans , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/pathology , Protein Stability/drug effects , Protein Structure, Secondary
5.
Neurobiol Dis ; 83: 122-33, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26334479

ABSTRACT

The hallmarks of Alzheimer disease are amyloid-ß plaques and neurofibrillary tangles accompanied by signs of neuroinflammation. Lysozyme is a major player in the innate immune system and has recently been shown to prevent the aggregation of amyloid-ß1-40 in vitro. In this study we found that patients with Alzheimer disease have increased lysozyme levels in the cerebrospinal fluid and lysozyme co-localized with amyloid-ß in plaques. In Drosophila neuronal co-expression of lysozyme and amyloid-ß1-42 reduced the formation of soluble and insoluble amyloid-ß species, prolonged survival and improved the activity of amyloid-ß1-42 transgenic flies. This suggests that lysozyme levels rise in Alzheimer disease as a compensatory response to amyloid-ß increases and aggregation. In support of this, in vitro aggregation assays revealed that lysozyme associates with amyloid-ß1-42 and alters its aggregation pathway to counteract the formation of toxic amyloid-ß species. Overall, these studies establish a protective role for lysozyme against amyloid-ß associated toxicities and identify increased lysozyme in patients with Alzheimer disease. Therefore, lysozyme has potential as a new biomarker as well as a therapeutic target for Alzheimer disease.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/metabolism , Muramidase/metabolism , Peptide Fragments/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/metabolism , Amyloid beta-Peptides/ultrastructure , Animals , Brain/pathology , Cell Death , Drosophila melanogaster , Female , Humans , Insect Proteins/metabolism , Locomotion , Male , Middle Aged , Muramidase/blood , Muramidase/cerebrospinal fluid , Muramidase/pharmacology , Peptide Fragments/ultrastructure , Plaque, Amyloid/metabolism , Plaque, Amyloid/ultrastructure , Tumor Cells, Cultured , tau Proteins/metabolism
6.
Neuromolecular Med ; 16(1): 150-60, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24101586

ABSTRACT

The success of future intervention strategies for Alzheimer's disease (AD) will likely rely on the development of treatments starting early in the disease course, before irreversible brain damage occurs. The pre-symptomatic stage of AD occurs at least one decade before the clinical onset, highlighting the need for validated biomarkers that reflect this early period. Reliable biomarkers for AD are also needed in research and clinics for diagnosis, patient stratification, clinical trials, monitoring of disease progression and the development of new treatments. Changes in the lysosomal network, i.e., the endosomal, lysosomal and autophagy systems, are among the first alterations observed in an AD brain. In this study, we performed a targeted search for lysosomal network proteins in human cerebrospinal fluid (CSF). Thirty-four proteins were investigated, and six of them, early endosomal antigen 1 (EEA1), lysosomal-associated membrane proteins 1 and 2 (LAMP-1, LAMP-2), microtubule-associated protein 1 light chain 3 (LC3), Rab3 and Rab7, were significantly increased in the CSF from AD patients compared with neurological controls. These results were confirmed in a validation cohort of CSF samples, and patients with no neurochemical evidence of AD, apart from increased total-tau, were found to have EEA1 levels corresponding to the increased total-tau levels. These findings indicate that increased levels of LAMP-1, LAMP-2, LC3, Rab3 and Rab7 in the CSF might be specific for AD, and increased EEA1 levels may be a sign of general neurodegeneration. These six lysosomal network proteins are potential AD biomarkers and may be used to investigate lysosomal involvement in AD pathogenesis.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Cerebrospinal Fluid Proteins/cerebrospinal fluid , Lysosomal Membrane Proteins/cerebrospinal fluid , Lysosomal-Associated Membrane Protein 2/cerebrospinal fluid , Lysosomes/chemistry , Microtubule-Associated Proteins/cerebrospinal fluid , Vesicular Transport Proteins/cerebrospinal fluid , rab GTP-Binding Proteins/cerebrospinal fluid , rab3 GTP-Binding Proteins/cerebrospinal fluid , Adult , Aged , Aged, 80 and over , Albumins/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Autophagy , Biomarkers/cerebrospinal fluid , Endosomes/chemistry , Female , Humans , Male , Middle Aged , Nerve Tissue Proteins/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , Phagosomes/chemistry , rab7 GTP-Binding Proteins , tau Proteins/cerebrospinal fluid
7.
PLoS One ; 7(11): e50262, 2012.
Article in English | MEDLINE | ID: mdl-23166840

ABSTRACT

Alterations in lipid homeostasis are implicated in several neurodegenerative diseases, although the mechanisms responsible are poorly understood. We evaluated the impact of cholesterol accumulation, induced by U18666A, quinacrine or mutations in the cholesterol transporting Niemann-Pick disease type C1 (NPC1) protein, on lysosomal stability and sensitivity to lysosome-mediated cell death. We found that neurons with lysosomal cholesterol accumulation were protected from oxidative stress-induced apoptosis. In addition, human fibroblasts with cholesterol-loaded lysosomes showed higher lysosomal membrane stability than controls. Previous studies have shown that cholesterol accumulation is accompanied by the storage of lipids such as sphingomyelin, glycosphingolipids and sphingosine and an up regulation of lysosomal associated membrane protein-2 (LAMP-2), which may also influence lysosomal stability. However, in this study the use of myriocin and LAMP deficient fibroblasts excluded these factors as responsible for the rescuing effect and instead suggested that primarily lysosomal cholesterol content determineD the cellular sensitivity to toxic insults. Further strengthening this concept, depletion of cholesterol using methyl-ß-cyclodextrin or 25-hydroxycholesterol decreased the stability of lysosomes and cells became more prone to undergo apoptosis. In conclusion, cholesterol content regulated lysosomal membrane permeabilization and thereby influenced cell death sensitivity. Our data suggests that lysosomal cholesterol modulation might be used as a therapeutic strategy for conditions associated with accelerated or repressed apoptosis.


Subject(s)
Cell Death/physiology , Cholesterol/metabolism , Lysosomes/metabolism , Neurons/metabolism , Androstenes , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Fibroblasts/metabolism , Flow Cytometry , Humans , Hydroxycholesterols , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Lysosomal Membrane Proteins/metabolism , Lysosomes/physiology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microscopy, Phase-Contrast , Mutation/genetics , Niemann-Pick C1 Protein , Quinacrine , Statistics, Nonparametric , beta-Cyclodextrins
SELECTION OF CITATIONS
SEARCH DETAIL
...