Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Article in English | MEDLINE | ID: mdl-33833060

ABSTRACT

Parkinson's disease is characterized by accumulation of α-synuclein (αSyn). Release of oligomeric/fibrillar αSyn from damaged neurons may potentiate neuronal death in part via microglial activation. Heretofore, it remained unknown if oligomeric/fibrillar αSyn could activate the nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome in human microglia and whether anti-αSyn antibodies could prevent this effect. Here, we show that αSyn activates the NLRP3 inflammasome in human induced pluripotent stem cell (hiPSC)-derived microglia (hiMG) via dual stimulation involving Toll-like receptor 2 (TLR2) engagement and mitochondrial damage. In vitro, hiMG can be activated by mutant (A53T) αSyn secreted from hiPSC-derived A9-dopaminergic neurons. Surprisingly, αSyn-antibody complexes enhanced rather than suppressed inflammasome-mediated interleukin-1ß (IL-1ß) secretion, indicating these complexes are neuroinflammatory in a human context. A further increase in inflammation was observed with addition of oligomerized amyloid-ß peptide (Aß) and its cognate antibody. In vivo, engraftment of hiMG with αSyn in humanized mouse brain resulted in caspase-1 activation and neurotoxicity, which was exacerbated by αSyn antibody. These findings may have important implications for antibody therapies aimed at depleting misfolded/aggregated proteins from the human brain, as they may paradoxically trigger inflammation in human microglia.


Subject(s)
Inflammasomes/metabolism , Microglia/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Parkinson Disease/immunology , alpha-Synuclein/immunology , Amyloid beta-Peptides/immunology , Antibodies/immunology , Cell Differentiation , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/cytology , Microglia/cytology , Toll-Like Receptor 2/metabolism , alpha-Synuclein/genetics
3.
J Neurosci ; 41(10): 2264-2273, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33483428

ABSTRACT

Synaptic and neuronal loss are major neuropathological characteristics of Parkinson's disease. Misfolded protein aggregates in the form of Lewy bodies, comprised mainly of α-synuclein (αSyn), are associated with disease progression, and have also been linked to other neurodegenerative diseases, including Lewy body dementia, Alzheimer's disease, and frontotemporal dementia. However, the effects of αSyn and its mechanism of synaptic damage remain incompletely understood. Here, we show that αSyn oligomers induce Ca2+-dependent release of glutamate from astrocytes obtained from male and female mice, and that mice overexpressing αSyn manifest increased tonic release of glutamate in vivo In turn, this extracellular glutamate activates glutamate receptors, including extrasynaptic NMDARs (eNMDARs), on neurons both in culture and in hippocampal slices of αSyn-overexpressing mice. Additionally, in patch-clamp recording from outside-out patches, we found that oligomerized αSyn can directly activate eNMDARs. In organotypic slices, oligomeric αSyn induces eNMDAR-mediated synaptic loss, which can be reversed by the drug NitroSynapsin. When we expose human induced pluripotent stem cell-derived cerebrocortical neurons to αSyn, we find similar effects. Importantly, the improved NMDAR antagonist NitroSynapsin, which selectively inhibits extrasynaptic over physiological synaptic NMDAR activity, protects synapses from oligomeric αSyn-induced damage in our model systems, thus meriting further study for its therapeutic potential.SIGNIFICANCE STATEMENT Loss of synaptic function and ensuing neuronal loss are associated with disease progression in Parkinson's disease (PD), Lewy body dementia (LBD), and other neurodegenerative diseases. However, the mechanism of synaptic damage remains incompletely understood. α-Synuclein (αSyn) misfolds in PD/LBD, forming Lewy bodies and contributing to disease pathogenesis. Here, we found that misfolded/oligomeric αSyn releases excessive astrocytic glutamate, in turn activating neuronal extrasynaptic NMDA receptors (eNMDARs), thereby contributing to synaptic damage. Additionally, αSyn oligomers directly activate eNMDARs, further contributing to damage. While the FDA-approved drug memantine has been reported to offer some benefit in PD/LBD (Hershey and Coleman-Jackson, 2019), we find that the improved eNMDAR antagonist NitroSynapsin ameliorates αSyn-induced synaptic spine loss, providing potential disease-modifying intervention in PD/LBD.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , alpha-Synuclein/metabolism , Animals , Astrocytes/drug effects , Astrocytes/pathology , Cells, Cultured , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synapses/metabolism , Synapses/pathology , alpha-Synuclein/pharmacology
4.
Acta Neuropathol Commun ; 8(1): 133, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32787922

ABSTRACT

Alpha-synucleinopathies are a group of progressive neurodegenerative disorders, characterized by intracellular deposits of aggregated α-synuclein (αS). The clinical heterogeneity of these diseases is thought to be attributed to conformers (or strains) of αS but the contribution of inclusions in various cell types is unclear. The aim of the present work was to study αS conformers among different transgenic (TG) mouse models of α-synucleinopathies. To this end, four different TG mouse models were studied (Prnp-h[A53T]αS; Thy1-h[A53T]αS; Thy1-h[A30P]αS; Thy1-mαS) that overexpress human or murine αS and differed in their age-of-symptom onset and subsequent disease progression. Postmortem analysis of end-stage brains revealed robust neuronal αS pathology as evidenced by accumulation of αS serine 129 (p-αS) phosphorylation in the brainstem of all four TG mouse lines. Overall appearance of the pathology was similar and only modest differences were observed among additionally affected brain regions. To study αS conformers in these mice, we used pentameric formyl thiophene acetic acid (pFTAA), a fluorescent dye with amyloid conformation-dependent spectral properties. Unexpectedly, besides the neuronal αS pathology, we also found abundant pFTAA-positive inclusions in microglia of all four TG mouse lines. These microglial inclusions were also positive for Thioflavin S and showed immunoreactivity with antibodies recognizing the N-terminus of αS, but were largely p-αS-negative. In all four lines, spectral pFTAA analysis revealed conformational differences between microglia and neuronal inclusions but not among the different mouse models. Concomitant with neuronal lesions, microglial inclusions were already present at presymptomatic stages and could also be induced by seeded αS aggregation. Although nature and significance of microglial inclusions for human α-synucleinopathies remain to be clarified, the previously overlooked abundance of microglial inclusions in TG mouse models of α-synucleinopathy bears importance for mechanistic and preclinical-translational studies.


Subject(s)
Microglia/pathology , Neurons/pathology , Synucleinopathies/pathology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Animals , Disease Models, Animal , Humans , Inclusion Bodies/pathology , Mice , Mice, Transgenic , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Protein Conformation , Synucleinopathies/genetics , alpha-Synuclein/chemistry
5.
J Nucl Med ; 61(7): 965-970, 2020 07.
Article in English | MEDLINE | ID: mdl-32482792

ABSTRACT

Transthyretin and light-chain amyloidosis are the 2 main causes of cardiac amyloidosis. Recent developments in molecular imaging have transformed our ability to diagnose transthyretin cardiac amyloidosis noninvasively and unmasked a hitherto unrecognized prevalence of the disease. This review summarizes the current and evolving imaging approaches, their molecular structural basis, and the gaps in imaging capabilities that have arisen as a result of parallel developments in pharmacotherapy delivering the first effective treatment options for this condition.


Subject(s)
Amyloidosis/diagnostic imaging , Cardiomyopathies/diagnostic imaging , Molecular Imaging/methods , Humans
6.
Circ Cardiovasc Imaging ; 13(2): e010249, 2020 02.
Article in English | MEDLINE | ID: mdl-32063053

ABSTRACT

BACKGROUND: Technetium-99 m pyrophosphate protocols for transthyretin cardiac amyloidosis diagnosis have variably used 1- and 3-hour imaging time points. We investigated whether imaging at 1 hour with superior efficiency had comparable diagnostic accuracy as 3-hour imaging. METHODS: This is a registry analysis of patients with suspected transthyretin cardiac amyloidosis referred for technetium-99 m pyrophosphate at a single tertiary center from June 2015 through January 2019. Patients underwent planar and single-photon emission computed tomography (SPECT) imaging at 1 and 3 hours. A positive Tc-99m pyrophosphate study was defined by the presence of diffuse myocardial tracer uptake on SPECT. For planar imaging, visual semiquantitative (grades 0-3, ≥2 considered positive) and quantitative heart to contralateral ratios (≥1.5 considered positive) were used. RESULTS: Two hundred thirty-three patients (69% men; median age, 77 [69-83] years) underwent the study protocol. There were 60 (25.8%) patients with diffuse myocardial uptake, 1 (0.4%) with regional uptake, and 172 (73.8%) with no myocardial uptake. Results of SPECT were identical at 1 and 3 hours. Planar imaging at 1 hour had 98% sensitivity and 96% specificity. Planar grade 0 uptake or heart to contralateral ratio ≤1.2 and planar grade 3 uptake or heart to contralateral ratio ≥2.0 were always associated with negative and positive SPECT, respectively. For planar grades 1 and 2 uptake and heart to contralateral ratio 1.3 to 1.9, SPECT was needed to make a diagnosis. No patient with light-chain cardiac amyloidosis had positive SPECT. CONCLUSIONS: An efficient 1-hour technetium-99 m pyrophosphate protocol had comparable diagnostic performance to a 3-hour protocol.


Subject(s)
Amyloid Neuropathies, Familial/diagnosis , Cardiomyopathies/diagnosis , Technetium Tc 99m Pyrophosphate/pharmacology , Tomography, Emission-Computed, Single-Photon/methods , Aged , Aged, 80 and over , Diagnosis, Differential , Female , Follow-Up Studies , Humans , Male , Radiopharmaceuticals/pharmacology , Reproducibility of Results , Retrospective Studies , Time Factors
8.
Sci Transl Med ; 9(407)2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28904227

ABSTRACT

Increasing evidence supports the hypothesis that soluble misfolded protein assemblies contribute to the degeneration of postmitotic tissue in amyloid diseases. However, there is a dearth of reliable nonantibody-based probes for selectively detecting oligomeric aggregate structures circulating in plasma or deposited in tissues, making it difficult to scrutinize this hypothesis in patients. Hence, understanding the structure-proteotoxicity relationships driving amyloid diseases remains challenging, hampering the development of early diagnostic and novel treatment strategies. We report peptide-based probes that selectively label misfolded transthyretin (TTR) oligomers circulating in the plasma of TTR hereditary amyloidosis patients exhibiting a predominant neuropathic phenotype. These probes revealed that there are much fewer misfolded TTR oligomers in healthy controls, in asymptomatic carriers of mutations linked to amyloid polyneuropathy, and in patients with TTR-associated cardiomyopathies. The absence of misfolded TTR oligomers in the plasma of cardiomyopathy patients suggests that the tissue tropism observed in the TTR amyloidoses is structure-based. Misfolded oligomers decrease in TTR amyloid polyneuropathy patients treated with disease-modifying therapies (tafamidis or liver transplant-mediated gene therapy). In a subset of TTR amyloid polyneuropathy patients, the probes also detected a circulating TTR fragment that disappeared after tafamidis treatment. Proteomic analysis of the isolated TTR oligomers revealed a specific patient-associated signature composed of proteins that likely associate with the circulating TTR oligomers. Quantification of plasma oligomer concentrations using peptide probes could become an early diagnostic strategy, a response-to-therapy biomarker, and a useful tool for understanding structure-proteotoxicity relationships in the TTR amyloidoses.


Subject(s)
Amyloidosis, Familial/blood , Molecular Probes/chemistry , Peptides/chemistry , Prealbumin/metabolism , Protein Folding , Protein Multimerization , Amyloidosis, Familial/genetics , Benzoxazoles/pharmacology , Case-Control Studies , Cross-Linking Reagents/chemistry , Diazomethane/chemistry , Genotype , Humans , Ions , Light , Molecular Weight , Prealbumin/chemistry , Protein Structure, Secondary , Proteolysis , Proteomics , Solubility
9.
J Neurosci ; 36(18): 5084-93, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27147660

ABSTRACT

UNLABELLED: The aggregation of amyloid-ß peptide (Aß) in brain is an early event and hallmark of Alzheimer's disease (AD). We combined the advantages of in vitro and in vivo approaches to study cerebral ß-amyloidosis by establishing a long-term hippocampal slice culture (HSC) model. While no Aß deposition was noted in untreated HSCs of postnatal Aß precursor protein transgenic (APP tg) mice, Aß deposition emerged in HSCs when cultures were treated once with brain extract from aged APP tg mice and the culture medium was continuously supplemented with synthetic Aß. Seeded Aß deposition was also observed under the same conditions in HSCs derived from wild-type or App-null mice but in no comparable way when HSCs were fixed before cultivation. Both the nature of the brain extract and the synthetic Aß species determined the conformational characteristics of HSC Aß deposition. HSC Aß deposits induced a microglia response, spine loss, and neuritic dystrophy but no obvious neuron loss. Remarkably, in contrast to in vitro aggregated synthetic Aß, homogenates of Aß deposits containing HSCs induced cerebral ß-amyloidosis upon intracerebral inoculation into young APP tg mice. Our results demonstrate that a living cellular environment promotes the seeded conversion of synthetic Aß into a potent in vivo seeding-active form. SIGNIFICANCE STATEMENT: In this study, we report the seeded induction of Aß aggregation and deposition in long-term hippocampal slice cultures. Remarkably, we find that the biological activities of the largely synthetic Aß aggregates in the culture are very similar to those observed in vivo This observation is the first to show that potent in vivo seeding-active Aß aggregates can be obtained by seeded conversion of synthetic Aß in a living (wild-type) cellular environment.


Subject(s)
Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/pathology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Neurites/pathology , Neurons/pathology , Organ Culture Techniques
10.
Acta Neuropathol ; 131(1): 5-25, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26715565

ABSTRACT

In brains of patients with Alzheimer's disease (AD), Aß peptides accumulate in parenchyma and, almost invariably, also in the vascular walls. Although Aß aggregation is, by definition, present in AD, its impact is only incompletely understood. It occurs in a stereotypical spatiotemporal distribution within neuronal networks in the course of the disease. This suggests a role for synaptic connections in propagating Aß pathology, and possibly of axonal transport in an antero- or retrograde way-although, there is also evidence for passive, extracellular diffusion. Striking, in AD, is the conjunction of tau and Aß pathology. Tau pathology in the cell body of neurons precedes Aß deposition in their synaptic endings in several circuits such as the entorhino-dentate, cortico-striatal or subiculo-mammillary connections. However, genetic evidence suggests that Aß accumulation is the first step in AD pathogenesis. To model the complexity and consequences of Aß aggregation in vivo, various transgenic (tg) rodents have been generated. In rodents tg for the human Aß precursor protein, focal injections of preformed Aß aggregates can induce Aß deposits in the vicinity of the injection site, and over time in more distant regions of the brain. This suggests that Aß shares with α-synuclein, tau and other proteins the property to misfold and aggregate homotypic molecules. We propose to group those proteins under the term "propagons". Propagons may lack the infectivity of prions. We review findings from neuropathological examinations of human brains in different stages of AD and from studies in rodent models of Aß aggregation and discuss putative mechanisms underlying the initiation and spread of Aß pathology.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/pathology , tau Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Humans
11.
Nat Neurosci ; 18(11): 1559-61, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26352792

ABSTRACT

Cerebral ß-amyloidosis is induced by inoculation of Aß seeds into APP transgenic mice, but not into App(-/-) (APP null) mice. We found that brain extracts from APP null mice that had been inoculated with Aß seeds up to 6 months previously still induced ß-amyloidosis in APP transgenic hosts following secondary transmission. Thus, Aß seeds can persist in the brain for months, and they regain propagative and pathogenic activity in the presence of host Aß.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/pharmacology , Brain/drug effects , Brain/metabolism , Plaque, Amyloid/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/administration & dosage , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/pathology , Animals , Brain/pathology , Disease Models, Animal , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Plaque, Amyloid/pathology
12.
Nat Rev Drug Discov ; 14(11): 759-80, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26338154

ABSTRACT

The aggregation of specific proteins is hypothesized to underlie several degenerative diseases, which are collectively known as amyloid disorders. However, the mechanistic connection between the process of protein aggregation and tissue degeneration is not yet fully understood. Here, we review current and emerging strategies to ameliorate aggregation-associated degenerative disorders, with a focus on disease-modifying strategies that prevent the formation of and/or eliminate protein aggregates. Persuasive pharmacological and genetic evidence now supports protein aggregation as the cause of postmitotic tissue dysfunction or loss. However, a more detailed understanding of the factors that trigger and sustain aggregate formation and of the structure-activity relationships underlying proteotoxicity is needed to develop future disease-modifying therapies.


Subject(s)
Neurodegenerative Diseases/drug therapy , Protein Aggregation, Pathological/drug therapy , Protein Transport/drug effects , Animals , Biological Factors/pharmacology , Biological Factors/therapeutic use , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Prealbumin/antagonists & inhibitors , Prealbumin/chemistry , Prealbumin/metabolism , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Protein Folding/drug effects , Structure-Activity Relationship , Treatment Outcome
13.
J Am Chem Soc ; 137(23): 7404-14, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-26051248

ABSTRACT

Fluorogenic probes, due to their often greater spatial and temporal sensitivity in comparison to permanently fluorescent small molecules, represent powerful tools to study protein localization and function in the context of living systems. Herein, we report fluorogenic probe 4, a 1,3,4-oxadiazole designed to bind selectively to transthyretin (TTR). Probe 4 comprises a fluorosulfate group not previously used in an environment-sensitive fluorophore. The fluorosulfate functional group does not react covalently with TTR on the time scale required for cellular imaging, but does red shift the emission maximum of probe 4 in comparison to its nonfluorosulfated analogue. We demonstrate that probe 4 is dark in aqueous buffers, whereas the TTR·4 complex exhibits a fluorescence emission maximum at 481 nm. The addition of probe 4 to living HEK293T cells allows efficient binding to and imaging of exogenous TTR within intracellular organelles, including the mitochondria and the endoplasmic reticulum. Furthermore, live Caenorhabditis elegans expressing human TTR transgenically and treated with probe 4 display TTR·4 fluorescence in macrophage-like coelomocytes. An analogue of fluorosulfate probe 4 does react selectively with TTR without labeling the remainder of the cellular proteome. Studies on this analogue suggest that certain aryl fluorosulfates, due to their cell and organelle permeability and activatable reactivity, could be considered for the development of protein-selective covalent probes.


Subject(s)
Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Fluorescent Dyes/chemistry , Fluorides/chemistry , Organelles/metabolism , Prealbumin/analysis , Sulfuric Acids/chemistry , Animals , Cell Survival , Cells, Cultured , HEK293 Cells , Humans , Models, Molecular , Molecular Structure , Prealbumin/biosynthesis , Prealbumin/chemistry
14.
Neurobiol Aging ; 36(7): 2241-2247, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25911278

ABSTRACT

Endogenous murine amyloid-ß peptide (Aß) is expressed in most Aß precursor protein (APP) transgenic mouse models of Alzheimer's disease but its contribution to ß-amyloidosis remains unclear. We demonstrate ∼ 35% increased cerebral Aß load in APP23 transgenic mice compared with age-matched APP23 mice on an App-null background. No such difference was found for the much faster Aß-depositing APPPS1 transgenic mouse model between animals with or without the murine App gene. Nevertheless, both APP23 and APPPS1 mice codeposited murine Aß, and immunoelectron microscopy revealed a tight association of murine Aß with human Aß fibrils. Deposition of murine Aß was considerably less efficient compared with the deposition of human Aß indicating a lower amyloidogenic potential of murine Aß in vivo. The amyloid dyes Pittsburgh Compound-B and pentamer formyl thiophene acetic acid did not differentiate between amyloid deposits consisting of human Aß and deposits of mixed human-murine Aß. Our data demonstrate a differential effect of murine Aß on human Aß deposition in different APP transgenic mice. The mechanistically complex interaction of human and mouse Aß may affect pathogenesis of the models and should be considered when models are used for translational preclinical studies.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Brain/metabolism , Animals , Disease Models, Animal , Humans , Mice, Transgenic
15.
Brain Pathol ; 25(6): 743-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25677332

ABSTRACT

An important early event in the pathogenesis of Alzheimer's disease (AD) is the aberrant polymerization and extracellular accumulation of amyloid-ß peptide (Aß). In young transgenic mice expressing the human Aß-precursor protein (APP), deposits of Aß can be induced by the inoculation of minute amounts of brain extract containing Aß aggregates ("Aß seeds"), indicative of a prion-like seeding phenomenon. Moreover, focal intracerebral injection of Aß seeds can induce deposits not only in the immediate vicinity of the injection site, but, with time, also in distal regions of the brain. However, it remains uncertain whether the spatial progression of Aß deposits occurs via nonsystematic diffusion from the injection site to proximal regions or via directed transit along neuroanatomical pathways. To address this question, we analyzed the spatiotemporal emergence of Aß deposits in two different APP-transgenic mouse models that had been previously inoculated with Aß seeds into the hippocampal formation. The results revealed a specific, neuroanatomically constrained pattern of induced Aß deposits in structures corresponding to the limbic connectome, supporting the hypothesis that neuronal pathways act as conduits for the movement of proteopathic agents among brain regions, thereby facilitating the progression of disease.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/metabolism , Amyloidosis/pathology , Connectome , Limbic System/metabolism , Limbic System/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Disease Progression , Humans , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Mutation
17.
J Neurosci ; 34(31): 10264-73, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25080588

ABSTRACT

Deposition of aggregated amyloid-ß (Aß) peptide in brain is an early event and hallmark pathology of Alzheimer's disease and cerebral Aß angiopathy. Experimental evidence supports the concept that Aß multimers can act as seeds and structurally corrupt other Aß peptides by a self-propagating mechanism. Here we compare the induction of cerebral ß-amyloidosis by intraperitoneal applications of Aß-containing brain extracts in three Aß-precursor protein (APP) transgenic mouse lines that differ in levels of transgene expression in brain and periphery (APP23 mice, APP23 mice lacking murine APP, and R1.40 mice). Results revealed that beta-amyloidosis induction, which could be blocked with an anti-Aß antibody, was dependent on the amount of inoculated brain extract and on the level of APP/Aß expression in the brain but not in the periphery. The induced Aß deposits in brain occurred in a characteristic pattern consistent with the entry of Aß seeds at multiple brain locations. Intraperitoneally injected Aß could be detected in blood monocytes and some peripheral tissues (liver, spleen) up to 30 d after the injection but escaped histological and biochemical detection thereafter. These results suggest that intraperitoneally inoculated Aß seeds are transported from the periphery to the brain in which corruptive templating of host Aß occurs at multiple sites, most efficiently in regions with high availability of soluble Aß.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Amyloidosis , Cerebral Cortex/pathology , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Amyloidosis/chemically induced , Amyloidosis/genetics , Amyloidosis/pathology , Animals , Antibodies/pharmacology , Blood Cells/metabolism , Blood Cells/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Drug Administration Routes , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Peritoneal Cavity/pathology , Plaque, Amyloid/pathology , Time Factors
18.
PLoS One ; 9(2): e90523, 2014.
Article in English | MEDLINE | ID: mdl-24587388

ABSTRACT

Alzheimer's disease (AD) is characterized by neurotoxic amyloid-ß plaque formation in brain parenchyma and cerebral blood vessels known as cerebral amyloid angiopathy (CAA). Besides CAA, AD is strongly related to vascular diseases such as stroke and atherosclerosis. Cerebrovascular dysfunction occurs in AD patients leading to alterations in blood flow that might play an important role in AD pathology with neuronal loss and memory deficits. Platelets are the major players in hemostasis and thrombosis, but are also involved in neuroinflammatory diseases like AD. For many years, platelets were accepted as peripheral model to study the pathophysiology of AD because platelets display the enzymatic activities to generate amyloid-ß (Aß) peptides. In addition, platelets are considered to be a biomarker for early diagnosis of AD. Effects of Aß peptides on platelets and the impact of platelets in the progression of AD remained, however, ill-defined. The present study explored the cellular mechanisms triggered by Aß in platelets. Treatment of platelets with Aß led to platelet activation and enhanced generation of reactive oxygen species (ROS) and membrane scrambling, suggesting enhanced platelet apoptosis. More important, platelets modulate soluble Aß into fibrillar structures that were absorbed by apoptotic but not vital platelets. This together with enhanced platelet adhesion under flow ex vivo and in vivo and platelet accumulation at amyloid deposits of cerebral vessels of AD transgenic mice suggested that platelets are major contributors of CAA inducing platelet thrombus formation at vascular amyloid plaques leading to vessel occlusion critical for cerebrovascular events like stroke.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Blood Platelets/metabolism , Adult , Alzheimer Disease/blood , Alzheimer Disease/genetics , Amyloid/ultrastructure , Amyloid beta-Peptides/pharmacology , Animals , Apoptosis/drug effects , Blood Platelets/drug effects , Blood Platelets/ultrastructure , Cells, Cultured , Cerebral Amyloid Angiopathy/blood , Cerebral Amyloid Angiopathy/genetics , Cerebral Amyloid Angiopathy/metabolism , Disease Models, Animal , Disease Progression , Humans , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Microscopy, Fluorescence , Middle Aged , Platelet Activation/drug effects , Reactive Oxygen Species/metabolism , Young Adult
19.
EMBO Rep ; 14(11): 1017-22, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23999102

ABSTRACT

The polymorphic ß-amyloid lesions present in individuals with Alzheimer's disease are collectively known as cerebral ß-amyloidosis. Amyloid precursor protein (APP) transgenic mouse models similarly develop ß-amyloid depositions that differ in morphology, binding of amyloid conformation-sensitive dyes, and Aß40/Aß42 peptide ratio. To determine the nature of such ß-amyloid morphotypes, ß-amyloid-containing brain extracts from either aged APP23 brains or aged APPPS1 brains were intracerebrally injected into the hippocampus of young APP23 or APPPS1 transgenic mice. APPPS1 brain extract injected into young APP23 mice induced ß-amyloid deposition with the morphological, conformational, and Aß40/Aß42 ratio characteristics of ß-amyloid deposits in aged APPPS1 mice, whereas APP23 brain extract injected into young APP23 mice induced ß-amyloid deposits with the characteristics of ß-amyloid deposits in aged APP23 mice. Injecting the two extracts into the APPPS1 host revealed a similar difference between the induced ß-amyloid deposits, although less prominent, and the induced deposits were similar to the ß-amyloid deposits found in aged APPPS1 hosts. These results indicate that the molecular composition and conformation of aggregated Aß in APP transgenic mice can be maintained by seeded conversion.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Brain/pathology , Animals , Brain/drug effects , Mice , Mice, Transgenic , Polymers/pharmacology , Presenilin-1/metabolism , Spectrum Analysis , Thiophenes/pharmacology
20.
Brain Pathol ; 23(3): 333-41, 2013 May.
Article in English | MEDLINE | ID: mdl-23587139

ABSTRACT

Accumulation, aggregation and deposition of Aß peptides are pathological hallmarks in the brains of individuals affected by Alzheimer's disease (AD) or by cerebral ß-amyloid angiopathy (Aß-CAA). While Aß is a peptide of yet largely unknown function, it is constantly produced in the human brain where it normally remains in a soluble state. However, Aß peptides are aggregation prone by their intrinsic ability to adopt alternative conformations rich in ß-sheet structure that aggregate into oligomeric as well as fibrillar formations. This transition from soluble to aggregated state has been hypothesized to initiate the pathological cascade and is therefore subject to intensive research. Mounting evidence suggests prion-like templated misfolding as the biochemical phenomenon responsible for promoting progressive Aß aggregation. Here, we review studies in vitro and in vivo that suggest that cerebral Aß aggregation may indeed progress via prion-like templated misfolding. The implications of these findings are discussed with respect to understanding initiation and progression of the disease and to developing therapeutics.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Prion Diseases/metabolism , Proteostasis Deficiencies/metabolism , Alzheimer Disease/pathology , Humans , Prion Diseases/pathology , Protein Structure, Secondary , Proteostasis Deficiencies/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...