Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4695, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824138

ABSTRACT

Which isoforms of apolipoprotein E (apoE) we inherit determine our risk of developing late-onset Alzheimer's Disease (AD), but the mechanism underlying this link is poorly understood. In particular, the relevance of direct interactions between apoE and amyloid-ß (Aß) remains controversial. Here, single-molecule imaging shows that all isoforms of apoE associate with Aß in the early stages of aggregation and then fall away as fibrillation happens. ApoE-Aß co-aggregates account for ~50% of the mass of diffusible Aß aggregates detected in the frontal cortices of homozygotes with the higher-risk APOE4 gene. We show how dynamic interactions between apoE and Aß tune disease-related functions of Aß aggregates throughout the course of aggregation. Our results connect inherited APOE genotype with the risk of developing AD by demonstrating how, in an isoform- and lipidation-specific way, apoE modulates the aggregation, clearance and toxicity of Aß. Selectively removing non-lipidated apoE4-Aß co-aggregates enhances clearance of toxic Aß by glial cells, and reduces secretion of inflammatory markers and membrane damage, demonstrating a clear path to AD therapeutics.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Apolipoprotein E4 , Apolipoproteins E , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Humans , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Animals , Apolipoprotein E4/metabolism , Apolipoprotein E4/genetics , Protein Isoforms/metabolism , Protein Isoforms/genetics , Mice , Female , Protein Aggregates , Male , Protein Aggregation, Pathological/metabolism , Mice, Transgenic , Neuroglia/metabolism
2.
Adv Drug Deliv Rev ; 140: 129-135, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30253201

ABSTRACT

Microphysiological systems (MPS) may be able to provide the pharmaceutical industry models that can reflect human physiological responses to improve drug discovery and translational outcomes. With lack of efficacy being the primary cause for drug attrition, developing MPS disease models would help researchers identify novel targets, study mechanisms in more physiologically-relevant depth, screen for novel biomarkers and test/optimize various therapeutics (small molecules, nanoparticles and biologics). Furthermore, with advances in inducible pluripotent stem cell technology (iPSC), pharmaceutical companies can access cells from patients to help recreate specific disease phenotypes in MPS platforms. Combining iPSC and MPS technologies will contribute to our understanding of the complexities of neurodegenerative diseases and of the blood brain barrier (BBB) leading to development of enhanced therapeutics.


Subject(s)
Blood-Brain Barrier , Drug Discovery , Induced Pluripotent Stem Cells , Animals , Humans , Models, Biological , Translational Research, Biomedical
3.
J Biol Chem ; 293(34): 13090-13099, 2018 08 24.
Article in English | MEDLINE | ID: mdl-29887525

ABSTRACT

The cellular prion protein (PrPC) can act as a cell-surface receptor for ß-amyloid (Aß) peptide; however, a role for PrPC in the pathogenesis of Alzheimer's disease (AD) is contested. Here, we expressed a range of Aß isoforms and PrPC in the Drosophila brain. We found that co-expression of Aß and PrPC significantly reduces the lifespan, disrupts circadian rhythms, and increases Aß deposition in the fly brain. In contrast, under the same conditions, expression of Aß or PrPC individually did not lead to these phenotypic changes. In vitro studies revealed that substoichiometric amounts of PrPC trap Aß as oligomeric assemblies and fragment-preformed Aß fibers. The ability of membrane-anchored PrPC to trap Aß as cytotoxic oligomers at the membrane surface and fragment inert Aß fibers suggests a mechanism by which PrPC exacerbates Aß deposition and pathogenic phenotypes in the fly, supporting a role for PrPC in AD. This study provides a second animal model linking PrPC expression with Aß toxicity and supports a role for PrPC in AD pathogenesis. Blocking the interaction of Aß and PrPC represents a potential therapeutic strategy.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloid/chemistry , Drosophila melanogaster/metabolism , Neurotoxicity Syndromes/etiology , Prion Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Circadian Rhythm , Disease Models, Animal , Drosophila melanogaster/growth & development , Longevity , Mesocricetus , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Protein Binding , Protein Multimerization
4.
Dis Model Mech ; 10(10): 1187-1199, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28993311

ABSTRACT

Molecular and cellular oscillations constitute an internal clock that tracks the time of day and permits organisms to optimize their behaviour and metabolism to suit the daily demands they face. The workings of this internal clock become impaired with age. In this review, we discuss whether such age-related impairments in the circadian clock interact with age-related neurodegenerative disorders, such as Alzheimer's disease. Findings from mouse and fly models of Alzheimer's disease have accelerated our understanding of the interaction between neurodegeneration and circadian biology. These models show that neurodegeneration likely impairs circadian rhythms either by damaging the central clock or by blocking its communication with other brain areas and with peripheral tissues. The consequent sleep and metabolic deficits could enhance the susceptibility of the brain to further degenerative processes. Thus, circadian dysfunction might be both a cause and an effect of neurodegeneration. We also discuss the primary role of light in the entrainment of the central clock and describe important, alternative time signals, such as food, that play a role in entraining central and peripheral circadian clocks. Finally, we propose how these recent insights could inform efforts to develop novel therapeutic approaches to re-entrain arrhythmic individuals with neurodegenerative disease.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Circadian Clocks , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Circadian Rhythm , Age Factors , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Animals , Brain/physiopathology , Circadian Clocks/genetics , Circadian Rhythm/genetics , Circadian Rhythm Signaling Peptides and Proteins/genetics , Disease Models, Animal , Energy Metabolism , Humans , Nerve Degeneration , Plaque, Amyloid , Signal Transduction , Sleep
5.
EMBO Mol Med ; 9(10): 1366-1378, 2017 10.
Article in English | MEDLINE | ID: mdl-28855301

ABSTRACT

We have characterised the proteolytic cleavage events responsible for the shedding of triggering receptor expressed on myeloid cells 2 (TREM2) from primary cultures of human macrophages, murine microglia and TREM2-expressing human embryonic kidney (HEK293) cells. In all cell types, a soluble 17 kDa N-terminal cleavage fragment was shed into the conditioned media in a constitutive process that is inhibited by G1254023X and metalloprotease inhibitors and siRNA targeting ADAM10. Inhibitors of serine proteases and matrix metalloproteinases 2/9, and ADAM17 siRNA did not block TREM2 shedding. Peptidomimetic protease inhibitors highlighted a possible cleavage site, and mass spectrometry confirmed that shedding occurred predominantly at the H157-S158 peptide bond for both wild-type and H157Y human TREM2 and for the wild-type murine orthologue. Crucially, we also show that the Alzheimer's disease-associated H157Y TREM2 variant was shed more rapidly than wild type from HEK293 cells, possibly by a novel, batimastat- and ADAM10-siRNA-independent, sheddase activity. These insights offer new therapeutic targets for modulating the innate immune response in Alzheimer's and other neurological diseases.


Subject(s)
Alzheimer Disease/genetics , Membrane Glycoproteins/metabolism , Proteolysis , Receptors, Immunologic/metabolism , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Animals, Newborn , Culture Media, Conditioned , HEK293 Cells , Humans , Ketocholesterols/pharmacology , Macrophages/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Immunologic/genetics
6.
Nat Commun ; 7: 13821, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27929117

ABSTRACT

Aberrant protein aggregation is controlled by various chaperones, including CCT (chaperonin containing TCP-1)/TCP-1/TRiC. Mutated CCT4/5 subunits cause sensory neuropathy and CCT5 expression is decreased in Alzheimer's disease. Here, we show that CCT integrity is essential for autophagosome degradation in cells or Drosophila and this phenomenon is orchestrated by the actin cytoskeleton. When autophagic flux is reduced by compromise of individual CCT subunits, various disease-relevant autophagy substrates accumulate and aggregate. The aggregation of proteins like mutant huntingtin, ATXN3 or p62 after CCT2/5/7 depletion is predominantly autophagy dependent, and does not further increase with CCT knockdown in autophagy-defective cells/organisms, implying surprisingly that the effect of loss-of-CCT activity on mutant ATXN3 or huntingtin oligomerization/aggregation is primarily a consequence of autophagy inhibition rather than loss of physiological anti-aggregation activity for these proteins. Thus, our findings reveal an essential partnership between two key components of the proteostasis network and implicate autophagy defects in diseases with compromised CCT complex activity.


Subject(s)
Autophagosomes/metabolism , Autophagy , Chaperonin Containing TCP-1/metabolism , Huntingtin Protein/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Ataxin-3/metabolism , Drosophila , Female , HeLa Cells , Humans , Lysosomes/metabolism , Male , Mice, Transgenic , RNA-Binding Proteins/metabolism
7.
Epileptic Disord ; 18(S2): 103-110, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27618835

ABSTRACT

Familial encephalopathy with neuroserpin inclusion bodies (FENIB) is a conformational proteinopathy characterised by neuronal inclusion bodies composed of the serine protease inhibitor (SERPIN), neuroserpin. Presenting clinically as a familial dementia-epilepsy syndrome, the molecular mechanism of the pathogenic abnormalities in neuroserpin has been characterised at atomic resolution. There is a remarkable genotype-phenotype correlation between the degree of molecular destabilisation of the several variants of the neuroserpin protein, their propensity to self-associate and the age of onset of the dementia-epilepsy complex. As with other serpinopathies there appears to be a mix of cell-autonomous toxicity, due to neuronal accumulation of neuroserpin, and non-cell autonomous toxicity, caused by loss of protease inhibition, in this case the dysregulated protease is likely to be tissue plasminogen activator (tPA). FENIB should be considered in cases of progressive myoclonic epilepsy and dementia particularly where there is family history of neuropsychiatric disease.


Subject(s)
Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/physiopathology , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/physiopathology , Humans
8.
Sci Rep ; 6: 33759, 2016 Sep 23.
Article in English | MEDLINE | ID: mdl-27658441

ABSTRACT

Circadian rhythms are essential for health and are frequently disturbed in disease. A full understanding of the causal relationships between behavioural and molecular circadian rhythms requires simultaneous longitudinal observations over time in individual organisms. Current experimental paradigms require the measurement of each rhythm separately across distinct populations of experimental organisms, rendering the comparability of the resulting datasets uncertain. We therefore developed FLYGLOW, an assay using clock gene controlled luciferase expression detected by exquisitely sensitive EM-CCD imaging, to enable simultaneous quantification of parameters including locomotor, sleep consolidation and molecular rhythms in single flies over days/weeks. FLYGLOW combines all the strengths of existing techniques, and also allows powerful multiparametric paired statistics. We found the age-related transition from rhythmicity to arrhythmicity for each parameter occurs unpredictably, with some flies showing loss of one or more rhythms during middle-age. Using single-fly correlation analysis of rhythm robustness and period we demonstrated the independence of the peripheral clock from circadian behaviours in wild type flies as well as in an Alzheimer's model. FLYGLOW is a useful tool for investigating the deterioration of behavioural and molecular rhythms in ageing and neurodegeneration. This approach may be applied more broadly within behavioural neurogenetics research.

9.
Neurobiol Aging ; 41: 39-52, 2016 May.
Article in English | MEDLINE | ID: mdl-27103517

ABSTRACT

Amyloid beta (Aß) peptide aggregation is linked to the initiation of Alzheimer's disease; accordingly, aggregation-prone isoforms of Aß, expressed in the brain, shorten the lifespan of Drosophila melanogaster. However, the lethal effects of Aß are not apparent until after day 15. We used shibire(TS) flies that exhibit a temperature-sensitive paralysis phenotype as a reporter of proteostatic robustness. In this model, we found that increasing age but not Aß expression lowered the flies' permissive temperature, suggesting that Aß did not exert its lethal effects by proteostatic disruption. Instead, we observed that chemical challenges, in particular oxidative stressors, discriminated clearly between young (robust) and old (sensitive) flies. Using nuclear magnetic resonance spectroscopy in combination with multivariate analysis, we compared water-soluble metabolite profiles at various ages in flies expressing Aß in their brains. We observed 2 genotype-linked metabolomic signals, the first reported the presence of any Aß isoform and the second the effects of the lethal Arctic Aß. Lethality was specifically associated with signs of oxidative respiration dysfunction and oxidative stress.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Brain/metabolism , Disease Models, Animal , Drosophila melanogaster , Proteostasis Deficiencies/etiology , Proteostasis Deficiencies/metabolism , Aging/metabolism , Alzheimer Disease/etiology , Animals , Oxidative Stress , Protein Isoforms/metabolism , Protein Isoforms/toxicity , Temperature
10.
Free Radic Biol Med ; 96: 57-66, 2016 07.
Article in English | MEDLINE | ID: mdl-27094492

ABSTRACT

Mitochondrial dysfunction has been widely associated with the pathology of Alzheimer's disease, but there is no consensus on whether it is a cause or consequence of disease, nor on the precise mechanism(s). We addressed these issues by testing the effects of expressing the alternative oxidase AOX from Ciona intestinalis, in different models of AD pathology. AOX can restore respiratory electron flow when the cytochrome segment of the mitochondrial respiratory chain is inhibited, supporting ATP synthesis, maintaining cellular redox homeostasis and mitigating excess superoxide production at respiratory complexes I and III. In human HEK293-derived cells, AOX expression decreased the production of beta-amyloid peptide resulting from antimycin inhibition of respiratory complex III. Because hydrogen peroxide was neither a direct product nor substrate of AOX, the ability of AOX to mimic antioxidants in this assay must be indirect. In addition, AOX expression was able to partially alleviate the short lifespan of Drosophila models neuronally expressing human beta-amyloid peptides, whilst abrogating the induction of markers of oxidative stress. Our findings support the idea of respiratory chain dysfunction and excess ROS production as both an early step and as a pathologically meaningful target in Alzheimer's disease pathogenesis, supporting the concept of a mitochondrial vicious cycle underlying the disease.


Subject(s)
Alzheimer Disease/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Oxidative Stress/drug effects , Oxidoreductases/genetics , Plant Proteins/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/metabolism , Animals , Antimycin A/administration & dosage , Antimycin A/analogs & derivatives , Antioxidants/metabolism , Ciona intestinalis/genetics , Ciona intestinalis/metabolism , Disease Models, Animal , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex IV/antagonists & inhibitors , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Oxidoreductases/metabolism , Plant Proteins/metabolism , Reactive Oxygen Species/metabolism , Superoxides/metabolism
11.
Methods Mol Biol ; 1303: 227-39, 2016.
Article in English | MEDLINE | ID: mdl-26235070

ABSTRACT

Fruit flies (Drosophila melanogaster) have been widely used to study the cellular and molecular basis of human neurodegenerative disease. The biological similarities between the human and the fly have been explored successfully to further investigate the pathological basis of Alzheimer's disease (AD). Here, we discuss transgenic Drosophila models systems and the methodologies that have been employed in the study of AD.


Subject(s)
Alzheimer Disease , Drosophila melanogaster , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/physiology , Female , Hybridization, Genetic , Immunoblotting , Immunohistochemistry , Longevity , Male , Motor Activity , Solubility
12.
Dis Model Mech ; 8(7): 657-67, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26035384

ABSTRACT

Metals, including iron, are present at high concentrations in amyloid plaques in individuals with Alzheimer's disease, where they are also thought to be cofactors in generating oxidative stress and modulating amyloid formation. In this study, we present data from several Drosophila models of neurodegenerative proteinopathies indicating that the interaction between iron and amyloid beta peptide (Aß) is specific and is not seen for other aggregation-prone polypeptides. The interaction with iron is likely to be important in the dimerisation of Aß and is mediated by three N-terminal histidines. Transgenic fly lines systematically expressing all combinations of His>Ala substitutions in Aß were generated and used to study the pathological role of these residues. Developmental eye phenotypes, longevity and histological examinations indicate that the N-terminal histidines have distinct position-dependent and -independent mechanisms. The former mediate the toxic effects of metals and Aß aggregation under non-oxidising conditions and the latter are relevant under oxidising conditions. Understanding how Aß mediates neurotoxic effects in vivo will help to better target pathological pathways using aggregation blockers and metal-modifying agents.


Subject(s)
Amyloid beta-Peptides/metabolism , Drosophila/metabolism , Iron/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Substitution , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila/genetics , Female , Ferritins/metabolism , Histidine/chemistry , Humans , In Vitro Techniques , Oxidation-Reduction , Phenotype , Protein Aggregates , Protein Aggregation, Pathological/etiology , Protein Aggregation, Pathological/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
13.
Hum Mol Genet ; 24(14): 3929-38, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25954034

ABSTRACT

Brain-derived neurotrophic factor (BDNF) has a crucial role in learning and memory by promoting neuronal survival and modulating synaptic connectivity. BDNF levels are lower in the brains of individuals with Alzheimer's disease (AD), suggesting a pathogenic involvement. The Drosophila orthologue of BDNF is the highly conserved Neurotrophin 1 (DNT1). BDNF and DNT1 have the same overall protein structure and can be cleaved, resulting in the conversion of a full-length polypeptide into separate pro- and mature-domains. While the BDNF mature-domain is neuroprotective, the role of the pro-domain is less clear. In flies and mammalian cells, we have identified a synergistic toxic interaction between the amyloid-ß peptide (Aß1-42) and the pro-domains of both DNT1 and BDNF. Specifically, we show that DNT1 pro-domain acquires a neurotoxic activity in the presence of Aß1-42. In contrast, DNT1 mature-domain is protective against Aß1-42 toxicity. Likewise, in SH-SY5Y cell culture, BDNF pro-domain is toxic only in the presence of Aß1-42. Western blots indicate that this synergistic interaction likely results from the Aß1-42-induced upregulation of the BDNF pro-domain receptor p75(NTR). The clinical relevance of these findings is underlined by a greater than thirty fold increase in the ratio of BDNF pro- to mature-domains in the brains of individuals with AD. This unbalanced BDNF pro:mature-domain ratio in patients represents a possible biomarker of AD and may offer a target for therapeutic intervention.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Drosophila Proteins/metabolism , Drosophila/genetics , Nerve Growth Factors/metabolism , Peptide Fragments/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/genetics , Animals , Brain/metabolism , Brain-Derived Neurotrophic Factor/genetics , Cell Line, Tumor , Cell Survival , Drosophila Proteins/genetics , Female , Genetic Markers , Humans , Male , Middle Aged , Nerve Growth Factors/genetics , Peptide Fragments/genetics , Protein Interaction Maps , Receptor, trkB/genetics , Receptor, trkB/metabolism , Up-Regulation
14.
Essays Biochem ; 56: 69-83, 2014.
Article in English | MEDLINE | ID: mdl-25131587

ABSTRACT

The formation of amyloid aggregates is a feature of most, if not all, polypeptide chains. In vivo modelling of this process has been undertaken in the fruitfly Drosophila melanogaster with remarkable success. Models of both neurological and systemic amyloid diseases have been generated and have informed our understanding of disease pathogenesis in two main ways. First, the toxic amyloid species have been at least partially characterized, for example in the case of the Aß (amyloid ß-peptide) associated with Alzheimer's disease. Secondly, the genetic underpinning of model disease-linked phenotypes has been characterized for a number of neurodegenerative disorders. The current challenge is to integrate our understanding of disease-linked processes in the fly with our growing knowledge of human disease, for the benefit of patients.


Subject(s)
Amyloidosis/metabolism , Amyloid beta-Peptides/metabolism , Amyloidosis/pathology , Animals , Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila melanogaster , Humans , tau Proteins/metabolism
15.
PLoS One ; 9(7): e102985, 2014.
Article in English | MEDLINE | ID: mdl-25080104

ABSTRACT

The human Aß peptide causes progressive paralysis when expressed in the muscles of the nematode worm, C. elegans. We have exploited this model of Aß toxicity by carrying out an RNAi screen to identify genes whose reduced expression modifies the severity of this locomotor phenotype. Our initial finding was that none of the human orthologues of these worm genes is identical with the genome-wide significant GWAS genes reported to date (the "white zone"); moreover there was no identity between worm screen hits and the longer list of GWAS genes which included those with borderline levels of significance (the "grey zone"). This indicates that Aß toxicity should not be considered as equivalent to sporadic AD. To increase the sensitivity of our analysis, we then considered the physical interactors (+1 interactome) of the products of the genes in both the worm and the white+grey zone lists. When we consider these worm and GWAS gene lists we find that 4 of the 60 worm genes have a +1 interactome overlap that is larger than expected by chance. Two of these genes form a chaperonin complex, the third is closely associated with this complex and the fourth gene codes for actin, the major substrate of the same chaperonin.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Caenorhabditis elegans/genetics , Genome-Wide Association Study , Molecular Chaperones/physiology , RNA Interference , Animals , Humans , Phenotype
16.
Acta Neuropathol Commun ; 2: 43, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24725347

ABSTRACT

INTRODUCTION: The self-assembly of Aß peptides into a range of conformationally heterogeneous amyloid states represents a fundamental event in Alzheimer's disease. Within these structures oligomeric intermediates are considered to be particularly pathogenic. To test this hypothesis we have used a conformational targeting approach where particular conformational states, such as oligomers or fibrils, are recognized in vivo by state-specific antibody fragments. RESULTS: We show that oligomer targeting with the KW1 antibody fragment, but not fibril targeting with the B10 antibody fragment, affects toxicity in Aß-expressing Drosophila melanogaster. The effect of KW1 is observed to occur selectively with flies expressing Aß(1-40) and not with those expressing Aß(1-42) or the arctic variant of Aß(1-42) This finding is consistent with the binding preference of KW1 for Aß(1-40) oligomers that has been established in vitro. Strikingly, and in contrast to the previously demonstrated in vitro ability of this antibody fragment to block oligomeric toxicity in long-term potentiation measurements, KW1 promotes toxicity in the flies rather than preventing it. This result shows the crucial importance of the environment in determining the influence of antibody binding on the nature and consequences of the protein misfolding and aggregation. CONCLUSIONS: While our data support to the pathological relevance of oligomers, they highlight the issues to be addressed when developing inhibitory strategies that aim to neutralize these states by means of antagonistic binding agents.


Subject(s)
Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Antibodies/therapeutic use , Peptide Fragments/immunology , Peptide Fragments/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/pharmacology , Animals , Animals, Genetically Modified , Antibodies/chemistry , Antibodies/genetics , Antibodies/pharmacology , Cell Line, Tumor , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster , Eye/metabolism , Eye/ultrastructure , Hippocampus/drug effects , Hippocampus/physiology , Humans , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Mice , Mice, Inbred C57BL , Neuroblastoma/pathology , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/physiopathology , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Protein Aggregation, Pathological , Protein Binding/drug effects , Protein Conformation
17.
Biochemistry ; 53(13): 2101-11, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24601543

ABSTRACT

The aggregation of ß-amyloid (Aß) into toxic oligomers is a hallmark of Alzheimer's disease pathology. Here we present a novel approach for the development of peptides capable of preventing amyloid aggregation based upon the previous selection of natural all-l peptides that bind Aß1-42. Using an intracellular selection system, successful library members were further screened via competition selection to identify the most effective peptides capable of reducing amyloid levels. To circumvent potential issues arising from stability and protease action for these structures, we have replaced all l residues with d residues and inverted the sequence. These retro-inverso (RI) peptide analogues therefore encompass reversed sequences that maintain the overall topological order of the native peptides. Our results demonstrate that efficacy in blocking and reversing amyloid formation is maintained while introducing desirable properties to the peptides. Thioflavin-T assays, circular dichroism, and oblique angle fluorescence microscopy collectively indicate that RI peptides can reduce amyloid load, while 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays demonstrate modest reductions in cell toxicity. These conclusions are reinforced using Drosophila melanogaster studies to monitor pupal hatching rates and fly locomotor activity in the presence of RI peptides delivered via RI-trans-activating transcriptional activator peptide fusions. We demonstrate that the RI-protein fragment complementation assay approach can be used as a generalized method for deriving Aß-interacting peptides. This approach has subsequently led to several peptide candidates being further explored as potential treatments for Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Peptides/pharmacology , Peptides/therapeutic use , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/toxicity , Animals , Circular Dichroism , Disease Models, Animal , Drosophila melanogaster/drug effects , Drosophila melanogaster/metabolism , Motor Activity/drug effects , PC12 Cells , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Peptide Library , Peptides/chemistry , Peptides/isolation & purification , Plaque, Amyloid/drug therapy , Protein Structure, Secondary , Rats
18.
Dis Model Mech ; 7(4): 445-58, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24574361

ABSTRACT

Circadian behavioural deficits, including sleep irregularity and restlessness in the evening, are a distressing early feature of Alzheimer's disease (AD). We have investigated these phenomena by studying the circadian behaviour of transgenic Drosophila expressing the amyloid beta peptide (Aß). We find that Aß expression results in an age-related loss of circadian behavioural rhythms despite ongoing normal molecular oscillations in the central clock neurons. Even in the absence of any behavioural correlate, the synchronised activity of the central clock remains protective, prolonging lifespan, in Aß flies just as it does in control flies. Confocal microscopy and bioluminescence measurements point to processes downstream of the molecular clock as the main site of Aß toxicity. In addition, there seems to be significant non-cell-autonomous Aß toxicity resulting in morphological and probably functional signalling deficits in central clock neurons.


Subject(s)
Alzheimer Disease/physiopathology , Behavior , Circadian Clocks , Circadian Rhythm , Disease Models, Animal , Drosophila melanogaster/physiology , Amyloid beta-Peptides/toxicity , Animals , Behavior/drug effects , Cell Death/drug effects , Circadian Clocks/drug effects , Circadian Rhythm/drug effects , Darkness , Drosophila melanogaster/drug effects , Motor Activity/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology
19.
Biochim Biophys Acta ; 1832(12): 2115-26, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23911349

ABSTRACT

The accumulation of ß-amyloid (Aß) peptide in the brain is one of the pathological hallmarks of Alzheimer's disease and is thought to be of primary aetiological significance. In an unbiased genetic screen, we identified puromycin-sensitive aminopeptidase (PSA) as a potent suppressor of Aß toxicity in a Drosophila model system. We established that coexpression of Drosophila PSA (dPSA) in the flies' brains improved their lifespan, protected against locomotor deficits, and reduced brain Aß levels by clearing the Aß plaque-like deposits. However, confocal microscopy and subcellular fractionation of amyloid-expressing 7PA2 cells demonstrated that PSA localizes to the cytoplasm. Therefore, PSA and Aß are unlikely to be in the same cellular compartment; moreover, when we artificially placed them in the same compartment in flies, we could not detect a direct epistatic interaction. The consequent hypothesis that PSA's suppression of Aß toxicity is indirect was supported by the finding that Aß is not a proteolytic substrate for PSA in vitro. Furthermore, we showed that the enzymatic activity of PSA is not required for rescuing Aß toxicity in neuronal SH-SY5Y cells. We investigated whether the stimulation of autophagy by PSA was responsible for these protective effects. However PSA's promotion of autophagosome fusion with lysosomes required proteolytic activity and so its effect on autophagy is not identical to its protection against Aß toxicity.


Subject(s)
Alzheimer Disease/prevention & control , Aminopeptidases/pharmacology , Amyloid beta-Peptides/adverse effects , Brain/metabolism , Drosophila melanogaster/metabolism , Neuroblastoma/prevention & control , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Animals, Genetically Modified , Autophagy , Blotting, Western , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Proteolysis , Puromycin/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
20.
Hum Mol Genet ; 22(22): 4616-26, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23814041

ABSTRACT

Mutants of neuroserpin are retained as polymers within the endoplasmic reticulum (ER) of neurones to cause the autosomal dominant dementia familial encephalopathy with neuroserpin inclusion bodies or FENIB. The cellular consequences are unusual in that the ordered polymers activate the ER overload response (EOR) in the absence of the canonical unfolded protein response. We use both cell lines and Drosophila models to show that the G392E mutant of neuroserpin that forms polymers is degraded by UBE2j1 E2 ligase and Hrd1 E3 ligase while truncated neuroserpin, a protein that lacks 132 amino acids, is degraded by UBE2g2 (E2) and gp78 (E3) ligases. The degradation of G392E neuroserpin results from SREBP-dependent activation of the cholesterol biosynthetic pathway in cells that express polymers of neuroserpin (G392E). Inhibition of HMGCoA reductase, the limiting enzyme of the cholesterol biosynthetic pathway, reduced the ubiquitination of G392E neuroserpin in our cell lines and increased the retention of neuroserpin polymers in both HeLa cells and primary neurones. Our data reveal a reciprocal relationship between cholesterol biosynthesis and the clearance of mutant neuroserpin. This represents the first description of a link between sterol metabolism and modulation of the proteotoxicity mediated by the EOR.


Subject(s)
Cholesterol/biosynthesis , Drosophila melanogaster/metabolism , Epilepsies, Myoclonic/metabolism , Heredodegenerative Disorders, Nervous System/metabolism , Neuropeptides/metabolism , Polymers/metabolism , Serpins/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Drosophila melanogaster/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/pathology , HeLa Cells , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/pathology , Humans , Mice , Mutant Proteins/metabolism , Neurons/metabolism , Neuropeptides/genetics , Protein Unfolding , Serpins/genetics , Signal Transduction , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Unfolded Protein Response , Neuroserpin
SELECTION OF CITATIONS
SEARCH DETAIL
...