Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Ann Clin Transl Neurol ; 6(3): 554-574, 2019 03.
Article in English | MEDLINE | ID: mdl-30911579

ABSTRACT

Objective: Amyloid-beta oligomers (Aßo) trigger the development of Alzheimer's disease (AD) pathophysiology. Cellular prion protein (PrPC) initiates synaptic damage as a high affinity receptor for Aßo. Here, we evaluated the preclinical therapeutic efficacy of a fully human monoclonal antibody against PrPC. This AZ59 antibody selectively targets the Aßo binding site in the amino-terminal unstructured domain of PrPC to avoid any potential risk of direct toxicity. Methods: Potency of AZ59 was evaluated by binding to PrPC, blockade of Aßo interaction and interruption of Aßo signaling. AZ59 was administered to mice by weekly intraperitoneal dosing and brain antibody measured. APP/PS1 transgenic mice were treated with AZ59 and assessed by memory tests, by brain biochemistry and by histochemistry for Aß, gliosis and synaptic density. Results: AZ59 binds PrPC with 100 pmol/L affinity and blocks human brain Aßo binding to PrPC, as well as prevents synaptotoxic signaling. Weekly i.p. dosing of 20 mg/kg AZ59 in a murine form achieves trough brain antibody levels greater than 10 nmol/L. Aged symptomatic APP/PS1 transgenic mice treated with AZ59 for 5-7 weeks show a full rescue of behavioral and synaptic loss phenotypes. This recovery occurs without clearance of plaque pathology or elimination of gliosis. AZ59 treatment also normalizes synaptic signaling abnormalities in transgenic brain. These benefits are dose-dependent and persist for at least 1 month after the last dose. Interpretation: Preclinical data demonstrate that systemic AZ59 therapy rescues central synapses and memory function from transgenic Alzheimer's disease pathology, supporting a disease-modifying therapeutic potential.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Antibodies, Monoclonal/therapeutic use , PrPC Proteins/antagonists & inhibitors , PrPC Proteins/immunology , Amyloid beta-Peptides/metabolism , Animals , Binding Sites , Brain/pathology , COS Cells , Chlorocebus aethiops , Cognition , Disease Models, Animal , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Synapses/pathology
3.
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
4.
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
5.
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
6.
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
7.
Neuron ; 79(5): 887-902, 2013 Sep 04.
Article in English | MEDLINE | ID: mdl-24012003

ABSTRACT

Soluble amyloid-ß oligomers (Aßo) trigger Alzheimer's disease (AD) pathophysiology and bind with high affinity to cellular prion protein (PrP(C)). At the postsynaptic density (PSD), extracellular Aßo bound to lipid-anchored PrP(C) activates intracellular Fyn kinase to disrupt synapses. Here, we screened transmembrane PSD proteins heterologously for the ability to couple Aßo-PrP(C) with Fyn. Only coexpression of the metabotropic glutamate receptor, mGluR5, allowed PrP(C)-bound Aßo to activate Fyn. PrP(C) and mGluR5 interact physically, and cytoplasmic Fyn forms a complex with mGluR5. Aßo-PrP(C) generates mGluR5-mediated increases of intracellular calcium in Xenopus oocytes and in neurons, and the latter is also driven by human AD brain extracts. In addition, signaling by Aßo-PrP(C)-mGluR5 complexes mediates eEF2 phosphorylation and dendritic spine loss. For mice expressing familial AD transgenes, mGluR5 antagonism reverses deficits in learning, memory, and synapse density. Thus, Aßo-PrP(C) complexes at the neuronal surface activate mGluR5 to disrupt neuronal function.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Neurons/metabolism , PrPC Proteins/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/physiology , Alzheimer Disease/physiopathology , Animals , Calcium/metabolism , Cells, Cultured , Elongation Factor 2 Kinase/metabolism , HEK293 Cells , Humans , Mice , Oocytes , Phosphorylation , Post-Synaptic Density/metabolism , Receptor, Metabotropic Glutamate 5 , Xenopus
8.
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
9.
J Neurosci ; 30(18): 6367-74, 2010 May 05.
Article in English | MEDLINE | ID: mdl-20445063

ABSTRACT

Soluble oligomers of the amyloid-beta (Abeta) peptide are thought to play a key role in the pathophysiology of Alzheimer's disease (AD). Recently, we reported that synthetic Abeta oligomers bind to cellular prion protein (PrP(C)) and that this interaction is required for suppression of synaptic plasticity in hippocampal slices by oligomeric Abeta peptide. We hypothesized that PrP(C) is essential for the ability of brain-derived Abeta to suppress cognitive function. Here, we crossed familial AD transgenes encoding APPswe and PSen1DeltaE9 into Prnp-/- mice to examine the necessity of PrP(C) for AD-related phenotypes. Neither APP expression nor Abeta level is altered by PrP(C) absence in this transgenic AD model, and astrogliosis is unchanged. However, deletion of PrP(C) expression rescues 5-HT axonal degeneration, loss of synaptic markers, and early death in APPswe/PSen1DeltaE9 transgenic mice. The AD transgenic mice with intact PrP(C) expression exhibit deficits in spatial learning and memory. Mice lacking PrP(C), but containing Abeta plaque derived from APPswe/PSen1DeltaE9 transgenes, show no detectable impairment of spatial learning and memory. Thus, deletion of PrP(C) expression dissociates Abeta accumulation from behavioral impairment in these AD mice, with the cognitive deficits selectively requiring PrP(C).


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor/metabolism , Memory/physiology , Nerve Degeneration/genetics , PrPC Proteins/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/mortality , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Avoidance Learning , Brain/metabolism , Brain/pathology , Disease Models, Animal , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , PrPC Proteins/genetics , Presenilin-1/genetics , Random Allocation , Serotonin/metabolism , Survival Analysis
10.
J Mol Med (Berl) ; 88(4): 331-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19960174

ABSTRACT

Prefibrillar oligomers of the beta-amyloid peptide (A beta) are recognized as potential mediators of Alzheimer's disease (AD) pathophysiology. Deficits in synaptic function, neurotoxicity, and the progression of AD have all been linked to the oligomeric A beta assemblies rather than to A beta monomers or to amyloid plaques. However, the molecular sites of A beta oligomer action have remained largely unknown. Recently, the cellular prion protein (PrP(C)) has been shown to act as a functional receptor for A beta oligomers in brain slices. Because PrP(C) serves as the substrate for Creutzfeldt-Jakob Disease (CJD), these data suggest mechanistic similarities between the two neurodegenerative diseases. Here, we review the importance of A beta oligomers in AD, commonalities between AD and CJD, and the newly emergent role of PrP(C) as a receptor for A beta oligomers.


Subject(s)
Alzheimer Disease/blood , Amyloid beta-Peptides/genetics , Prions/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Animals , Brain/pathology , COS Cells , Chlorocebus aethiops , Creutzfeldt-Jakob Syndrome/metabolism , Humans , Mice , Mice, Transgenic , Neurodegenerative Diseases/pathology , Peptide Fragments/chemistry , PrPC Proteins/chemistry , Prions/chemistry
11.
Proc Natl Acad Sci U S A ; 100(16): 9608-13, 2003 Aug 05.
Article in English | MEDLINE | ID: mdl-12869696

ABSTRACT

Assays of drug action typically evaluate biochemical activity. However, accurately matching therapeutic efficacy with biochemical activity is a challenge. High-content cellular assays seek to bridge this gap by capturing broad information about the cellular physiology of drug action. Here, we present a method of predicting the general therapeutic classes into which various psychoactive drugs fall, based on high-content statistical categorization of gene expression profiles induced by these drugs. When we used the classification tree and random forest supervised classification algorithms to analyze microarray data, we derived general "efficacy profiles" of biomarker gene expression that correlate with anti-depressant, antipsychotic and opioid drug action on primary human neurons in vitro. These profiles were used as predictive models to classify naïve in vitro drug treatments with 83.3% (random forest) and 88.9% (classification tree) accuracy. Thus, the detailed information contained in genomic expression data is sufficient to match the physiological effect of a novel drug at the cellular level with its clinical relevance. This capacity to identify therapeutic efficacy on the basis of gene expression signatures in vitro has potential utility in drug discovery and drug target validation.


Subject(s)
Oligonucleotide Array Sequence Analysis , Pharmacogenetics/methods , Algorithms , Cluster Analysis , DNA, Complementary/metabolism , Databases, Genetic , Drug Design , Drug Evaluation, Preclinical/methods , Gene Expression , Humans , Models, Statistical , Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...