Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Mol Cell ; 72(3): 426-443.e12, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30401430

ABSTRACT

Protein phase separation by low-complexity, intrinsically disordered domains generates membraneless organelles and links to neurodegeneration. Cellular prion protein (PrPC) contains such domains, causes spongiform degeneration, and is a receptor for Alzheimer's amyloid-ß oligomers (Aßo). Here, we show that PrPC separates as a liquid phase, in which α-helical Thr become unfolded. At the cell surface, PrPC Lys residues interact with Aßo to create a hydrogel containing immobile Aßo and relatively mobile PrPC. The Aßo/PrP hydrogel has a well-defined stoichiometry and dissociates with excess Aßo. NMR studies of hydrogel PrPC reveal a distinct α-helical conformation for natively unfolded amino-terminal Gly and Ala residues. Aßo/PrP hydrogel traps signal-transducing mGluR5 on the plasma membrane. Recombinant PrPC extracts endogenous Aßo from human Alzheimer's soluble brain lysates into hydrogel, and a PrPC antagonist releases Aßo from endogenous brain hydrogel. Thus, coupled phase and conformational transitions of PrPC are driven by Aß species from Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/physiology , PrPC Proteins/chemistry , PrPC Proteins/physiology , Alzheimer Disease/metabolism , Animals , Brain , COS Cells , Cell Line , Cell Membrane , Chlorocebus aethiops , HEK293 Cells , Humans , Hydrogels , Magnetic Resonance Imaging/methods , Molecular Conformation , Neurons , Prions/chemistry , Prions/physiology , Protein Binding , Receptor, Metabotropic Glutamate 5 , Signal Transduction
2.
J Biol Chem ; 294(15): 6042-6053, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30787106

ABSTRACT

Oligomeric assemblies of amyloid-ß (Aß) peptide (Aßo) in the brains of individuals with Alzheimer's disease (AD) are toxic to neuronal synapses. More than a dozen Aß receptor candidates have been suggested to be responsible for various aspects of the molecular pathology and memory impairment in mouse models of AD. A lack of consistent experimental design among previous studies of different receptor candidates limits evaluation of the relative roles of these candidates, producing some controversy within the field. Here, using cell-based assays with several Aß species, including Aßo from AD brains obtained by autopsy, we directly compared the Aß-binding capacity of multiple receptor candidates while accounting for variation in expression and confirming cell surface expression. In a survey of 15 reported Aß receptors, only cellular prion protein (PrPC), Nogo receptor 1 (NgR1), and leukocyte immunoglobulin-like receptor subfamily B member 2 (LilrB2) exhibited direct binding to synaptotoxic assemblies of synthetic Aß. Both PrPC and NgR1 preferentially bound synaptotoxic oligomers rather than nontoxic monomers, and the method of oligomer preparation did not significantly alter our binding results. Hippocampal neurons lacking both NgR1 and LilrB2 exhibited a partial reduction of Aßo binding, but this reduction was lower than in neurons lacking PrPC under the same conditions. Finally, binding studies with soluble Aßo from human AD brains revealed a strong affinity for PrPC, weak affinity for NgR1, and no detectable affinity for LilrB2. These findings clarify the relative contributions of previously reported Aß receptors under controlled conditions and highlight the prominence of PrPC as an Aß-binding site.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Membrane Glycoproteins/metabolism , Nogo Receptor 1/metabolism , PrPC Proteins/metabolism , Receptors, Immunologic/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , COS Cells , Chlorocebus aethiops , Disease Models, Animal , Female , HEK293 Cells , Humans , Male , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Nogo Receptor 1/genetics , PrPC Proteins/genetics , Receptors, Immunologic/genetics
3.
Cereb Cortex ; 27(7): 3660-3674, 2017 07 01.
Article in English | MEDLINE | ID: mdl-27365298

ABSTRACT

Synaptic loss is critical in Alzheimer's disease (AD), but the dynamics of synapse turnover are poorly defined. We imaged dendritic spines in transgenic APPswe/PSen1∆E9 (APP/PS1) cerebral cortex. Dendritic spine turnover is increased far from plaque in aged APP/PS1 mice, and in young APP/PS1 mice prior to plaque formation. Dysregulation occurs in the presence of soluble Aß oligomer and requires cellular prion protein (PrPC). APP/PS1 mice lack responsiveness of spine turnover to sensory stimulation. Critically, enhanced spine turnover is coupled with the loss of persistent spines starting early and continuing with age. To evaluate mechanisms of experience-independent supranormal spine turnover, we analyzed the transcriptome of young APP/PS1 mouse brain when turnover is altered but synapse density and memory are normal, and plaque and inflammation are absent. Early PrPC-dependent expression changes occur in synaptic and lipid-metabolizing genes. Thus, pathologic synaptic dysregulation underlying AD begins at a young age prior to Aß plaque.


Subject(s)
Alzheimer Disease/pathology , Cerebral Cortex/pathology , Dendritic Spines/pathology , Hippocampus/pathology , Plaque, Amyloid/pathology , Sensory Deprivation , Age Factors , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , Animals , Dendritic Spines/ultrastructure , Disease Models, Animal , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Imaging, Three-Dimensional , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neuroimaging , Plaque, Amyloid/etiology , Presenilin-1/genetics , Prion Proteins/genetics , Prion Proteins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Time Factors , Vibrissae/innervation
4.
Acta Neuropathol ; 133(5): 785-807, 2017 05.
Article in English | MEDLINE | ID: mdl-28070672

ABSTRACT

Progranulin (PGRN) is implicated in Alzheimer's disease (AD) as well as frontotemporal lobar degeneration. Genetic studies demonstrate an association of the common GRN rs5848 variant that results in reduced PGRN levels with increased risk for AD. However, the mechanisms by which PGRN reduction from the GRN AD risk variant or mutation exacerbates AD pathophysiology remain ill defined. Here, we show that the GRN AD risk variant has no significant effects on florbetapir positron emission tomographic amyloid imaging and cerebrospinal fluid (CSF) Aß levels, whereas it is associated with increased CSF tau levels in human subjects of the Alzheimer's disease neuroimaging initiative studies. Consistent with the human data, subsequent analyses using the APPswe/PS1ΔE9 (APP/PS1) mouse model of cerebral amyloidosis show that PGRN deficiency has no exacerbating effects on Aß pathology. In contrast and unexpectedly, PGRN deficiency significantly reduces diffuse Aß plaque growth in these APP/PS1 mice. This protective effect is due, at least in part, to enhanced microglial Aß phagocytosis caused by PGRN deficiency-induced expression of TYROBP network genes (TNG) including an AD risk factor Trem2. PGRN-deficient APP/PS1 mice also exhibit less severe axonal dystrophy and partially improved behavior phenotypes. While PGRN deficiency reduces these amyloidosis-related phenotypes, other neuronal injury mechanisms are increased by loss of PGRN, revealing a multidimensional interaction of GRN with AD. For example, C1q complement deposition at synapses is enhanced in APP/PS1 mice lacking PGRN. Moreover, PGRN deficiency increases tau AT8 and AT180 pathologies in human P301L tau-expressing mice. These human and rodent data suggest that global PGRN reduction induces microglial TNG expression and increases AD risk by exacerbating neuronal injury and tau pathology, rather than by accelerating Aß pathology.


Subject(s)
Alzheimer Disease/metabolism , Amyloidogenic Proteins/metabolism , Frontotemporal Lobar Degeneration/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Plaque, Amyloid/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Amyloidosis/metabolism , Animals , Disease Models, Animal , Frontotemporal Lobar Degeneration/pathology , Granulins , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Mice , Mice, Transgenic , Microglia/pathology , Plaque, Amyloid/pathology , Progranulins
5.
Brain ; 139(Pt 2): 526-46, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26667279

ABSTRACT

Alzheimer's disease-related phenotypes in mice can be rescued by blockade of either cellular prion protein or metabotropic glutamate receptor 5. We sought genetic and biochemical evidence that these proteins function cooperatively as an obligate complex in the brain. We show that cellular prion protein associates via transmembrane metabotropic glutamate receptor 5 with the intracellular protein mediators Homer1b/c, calcium/calmodulin-dependent protein kinase II, and the Alzheimer's disease risk gene product protein tyrosine kinase 2 beta. Coupling of cellular prion protein to these intracellular proteins is modified by soluble amyloid-ß oligomers, by mouse brain Alzheimer's disease transgenes or by human Alzheimer's disease pathology. Amyloid-ß oligomer-triggered phosphorylation of intracellular protein mediators and impairment of synaptic plasticity in vitro requires Prnp-Grm5 genetic interaction, being absent in transheterozygous loss-of-function, but present in either single heterozygote. Importantly, genetic coupling between Prnp and Grm5 is also responsible for signalling, for survival and for synapse loss in Alzheimer's disease transgenic model mice. Thus, the interaction between metabotropic glutamate receptor 5 and cellular prion protein has a central role in Alzheimer's disease pathogenesis, and the complex is a potential target for disease-modifying intervention.


Subject(s)
Alzheimer Disease/metabolism , Intracellular Fluid/metabolism , Prions/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Signal Transduction/physiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Frontal Lobe/metabolism , Frontal Lobe/pathology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Prion Proteins , Prions/genetics , Protein Binding/physiology , Receptor, Metabotropic Glutamate 5/genetics
6.
J Biol Chem ; 290(28): 17415-38, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26018073

ABSTRACT

Alzheimer disease (AD) is characterized by amyloid-ß accumulation, with soluble oligomers (Aßo) being the most synaptotoxic. However, the multivalent and unstable nature of Aßo limits molecular characterization and hinders research reproducibility. Here, we characterized multiple Aßo forms throughout the life span of various AD mice and in post-mortem human brain. Aßo exists in several populations, where prion protein (PrP(C))-interacting Aßo is a high molecular weight Aß assembly present in multiple mice and humans with AD. Levels of PrP(C)-interacting Aßo match closely with mouse memory and are equal or superior to other Aß measures in predicting behavioral impairment. However, Aßo metrics vary considerably between mouse strains. Deleting PrP(C) expression in mice with relatively low PrP(C)-interacting Aßo (Tg2576) results in partial rescue of cognitive performance as opposed to complete recovery in animals with a high percentage of PrP(C)-interacting Aßo (APP/PSEN1). These findings highlight the relative contributions and interplay of Aßo forms in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Memory Disorders/metabolism , Prions/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/etiology , Alzheimer Disease/psychology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Animals , Behavior, Animal , Disease Models, Animal , Female , Humans , Male , Memory Disorders/etiology , Memory Disorders/psychology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Middle Aged , Molecular Weight , PrPC Proteins/chemistry , PrPC Proteins/genetics , PrPC Proteins/metabolism , Prefrontal Cortex/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Prions/chemistry , Prions/genetics , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
7.
Ann Neurol ; 77(6): 953-71, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25707991

ABSTRACT

OBJECTIVE: Currently no effective disease-modifying agents exist for the treatment of Alzheimer disease (AD). The Fyn tyrosine kinase is implicated in AD pathology triggered by amyloid-ß oligomers (Aßo) and propagated by Tau. Thus, Fyn inhibition may prevent or delay disease progression. Here, we sought to repurpose the Src family kinase inhibitor oncology compound, AZD0530, for AD. METHODS: The pharmacokinetics and distribution of AZD0530 were evaluated in mice. Inhibition of Aßo signaling to Fyn, Pyk2, and Glu receptors by AZD0530 was tested by brain slice assays. After AZD0530 or vehicle treatment of wild-type and AD transgenic mice, memory was assessed by Morris water maze and novel object recognition. For these cohorts, amyloid precursor protein (APP) metabolism, synaptic markers (SV2 and PSD-95), and targets of Fyn (Pyk2 and Tau) were studied by immunohistochemistry and by immunoblotting. RESULTS: AZD0530 potently inhibits Fyn and prevents both Aßo-induced Fyn signaling and downstream phosphorylation of the AD risk gene product Pyk2, and of NR2B Glu receptors in brain slices. After 4 weeks of treatment, AZD0530 dosing of APP/PS1 transgenic mice fully rescues spatial memory deficits and synaptic depletion, without altering APP or Aß metabolism. AZD0530 treatment also reduces microglial activation in APP/PS1 mice, and rescues Tau phosphorylation and deposition abnormalities in APP/PS1/Tau transgenic mice. There is no evidence of AZD0530 chronic toxicity. INTERPRETATION: Targeting Fyn can reverse memory deficits found in AD mouse models, and rescue synapse density loss characteristic of the disease. Thus, AZD0530 is a promising candidate to test as a potential therapy for AD.


Subject(s)
Alzheimer Disease/drug therapy , Behavior, Animal/drug effects , Benzodioxoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Quinazolines/pharmacology , Signal Transduction/drug effects , Amyloid beta-Peptides/drug effects , Animals , Benzodioxoles/pharmacokinetics , Disease Models, Animal , Focal Adhesion Kinase 2/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/pharmacokinetics
8.
J Biol Chem ; 289(41): 28460-77, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25148681

ABSTRACT

Soluble Amyloid-ß oligomers (Aßo) can trigger Alzheimer disease (AD) pathophysiology by binding to cell surface cellular prion protein (PrP(C)). PrP(C) interacts physically with metabotropic glutamate receptor 5 (mGluR5), and this interaction controls the transmission of neurotoxic signals to intracellular substrates. Because the interruption of the signal transduction from PrP(C) to mGluR5 has therapeutic potential for AD, we developed assays to explore the effect of endogenous ligands, agonists/antagonists, and antibodies on the interaction between PrP(C) and mGluR5 in cell lines and mouse brain. We show that the PrP(C) segment of amino acids 91-153 mediates the interaction with mGluR5. Agonists of mGluR5 increase the mGluR5-PrP(C) interaction, whereas mGluR5 antagonists suppress protein association. Synthetic Aßo promotes the protein interaction in mouse brain and transfected HEK-293 cell membrane preparations. The interaction of PrP(C) and mGluR5 is enhanced dramatically in the brains of familial AD transgenic model mice. In brain homogenates with Aßo, the interaction of PrP(C) and mGluR5 is reversed by mGluR5-directed antagonists or antibodies directed against the PrP(C) segment of amino acids 91-153. Silent allosteric modulators of mGluR5 do not alter Glu or basal mGluR5 activity, but they disrupt the Aßo-induced interaction of mGluR5 with PrP(C). The assays described here have the potential to identify and develop new compounds that inhibit the interaction of PrP(C) and mGluR5, which plays a pivotal role in the pathogenesis of Alzheimer disease by transmitting the signal from extracellular Aßo into the cytosol.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Antibodies/pharmacology , PrPC Proteins/antagonists & inhibitors , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Binding Sites , Biological Assay , Brain Chemistry , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Disease Models, Animal , Gene Expression Regulation , HEK293 Cells , Humans , Ligands , Mice , Mice, Transgenic , Peptide Mapping , PrPC Proteins/chemistry , PrPC Proteins/genetics , PrPC Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Receptor, Metabotropic Glutamate 5/chemistry , Receptor, Metabotropic Glutamate 5/genetics , Receptor, Metabotropic Glutamate 5/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
9.
Cell Rep ; 26(1): 145-158.e8, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30605671

ABSTRACT

Cellular prion protein (PrPC) binds the scrapie conformation of PrP (PrPSc) and oligomeric ß-amyloid peptide (Aßo) to mediate transmissible spongiform encephalopathy (TSE) and Alzheimer's disease (AD), respectively. We conducted cellular and biochemical screens for compounds blocking PrPC interaction with Aßo. A polymeric degradant of an antibiotic targets Aßo binding sites on PrPC with low nanomolar affinity and prevents Aßo-induced pathophysiology. We then identified a range of negatively charged polymers with specific PrPC affinity in the low to sub-nanomolar range, from both biological (melanin) and synthetic (poly [4-styrenesulfonic acid-co-maleic acid], PSCMA) origin. Association of PSCMA with PrPC prevents Aßo/PrPC-hydrogel formation, blocks Aßo binding to neurons, and abrogates PrPSc production by ScN2a cells. We show that oral PSCMA yields effective brain concentrations and rescues APPswe/PS1ΔE9 transgenic mice from AD-related synapse loss and memory deficits. Thus, an orally active PrPC-directed polymeric agent provides a potential therapeutic approach to address neurodegeneration in AD and TSE.


Subject(s)
Alzheimer Disease/physiopathology , Prion Proteins/antagonists & inhibitors , Animals , Mice , Mice, Transgenic , Signal Transduction
10.
Alzheimers Res Ther ; 7(1): 25, 2015.
Article in English | MEDLINE | ID: mdl-25945128

ABSTRACT

INTRODUCTION: Recent studies have shown that several strains of transgenic Alzheimer's disease (AD) mice overexpressing the amyloid precursor protein (APP) have cortical hyperexcitability, and their results have suggested that this aberrant network activity may be a mechanism by which amyloid-ß (Aß) causes more widespread neuronal dysfunction. Specific anticonvulsant therapy reverses memory impairments in various transgenic mouse strains, but it is not known whether reduction of epileptiform activity might serve as a surrogate marker of drug efficacy for memory improvement in AD mouse models. METHODS: Transgenic AD mice (APP/PS1 and 3xTg-AD) were chronically implanted with dural electroencephalography electrodes, and epileptiform activity was correlated with spatial memory function and transgene-specific pathology. The antiepileptic drugs ethosuximide and brivaracetam were tested for their ability to suppress epileptiform activity and to reverse memory impairments and synapse loss in APP/PS1 mice. RESULTS: We report that in two transgenic mouse models of AD (APP/PS1 and 3xTg-AD), the presence of spike-wave discharges (SWDs) correlated with impairments in spatial memory. Both ethosuximide and brivaracetam reduce mouse SWDs, but only brivaracetam reverses memory impairments in APP/PS1 mice. CONCLUSIONS: Our data confirm an intriguing therapeutic role of anticonvulsant drugs targeting synaptic vesicle protein 2A across AD mouse models. Chronic ethosuximide dosing did not reverse spatial memory impairments in APP/PS1 mice, despite reduction of SWDs. Our data indicate that SWDs are not a reliable surrogate marker of appropriate target engagement for reversal of memory dysfunction in APP/PS1 mice.

12.
Nat Neurosci ; 15(9): 1227-35, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22820466

ABSTRACT

Amyloid-beta (Aß) oligomers are thought to trigger Alzheimer's disease pathophysiology. Cellular prion protein (PrP(C)) selectively binds oligomeric Aß and can mediate Alzheimer's disease-related phenotypes. We examined the specificity, distribution and signaling of Aß-PrP(C) complexes, seeking to understand how they might alter the function of NMDA receptors (NMDARs) in neurons. PrP(C) is enriched in postsynaptic densities, and Aß-PrP(C) interaction leads to Fyn kinase activation. Soluble Aß assemblies derived from the brains of individuals with Alzheimer's disease interacted with PrP(C) to activate Fyn. Aß engagement of PrP(C)-Fyn signaling yielded phosphorylation of the NR2B subunit of NMDARs, which was coupled to an initial increase and then a loss of surface NMDARs. Aß-induced dendritic spine loss and lactate dehydrogenase release required both PrP(C) and Fyn, and human familial Alzheimer's disease transgene-induced convulsive seizures did not occur in mice lacking PrP(C). These results delineate an Aß oligomer signal transduction pathway that requires PrP(C) and Fyn to alter synaptic function, with deleterious consequences in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Neurons/physiology , PrPC Proteins/metabolism , Proto-Oncogene Proteins c-fyn/physiology , Synapses/physiology , Alzheimer Disease/physiopathology , Animals , Blotting, Western , Calcium Signaling/physiology , Cell Line , Dendritic Spines/metabolism , Electroencephalography , Enzyme Activation , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphorylation , PrPC Proteins/genetics , Protein Binding , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Seizures/genetics , Seizures/prevention & control , Synapses/drug effects , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL