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










Database
Language
Publication year range
1.
J Neurosci ; 39(10): 1910-1929, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30626696

ABSTRACT

The intracellular tyrosine kinase Pyk2 (PTK2B) is related to focal adhesion kinase and localizes to postsynaptic sites in brain. Pyk2 genetic variation contributes to late onset Alzheimer's disease (AD) risk. We recently observed that Pyk2 is required for synapse loss and for learning deficits in a transgenic mouse model of AD. Here, we explore the cellular and biochemical basis for the action of Pyk2 tyrosine kinase in amyloid-ß oligomer (Aßo)-induced dendritic spine loss. Overexpression of Pyk2 reduces dendritic spine density of hippocampal neurons by a kinase-dependent mechanism. Biochemical isolation of Pyk2-interacting proteins from brain identifies Graf1c, a RhoA GTPase-activating protein inhibited by Pyk2. Aßo-induced reductions in dendritic spine motility and chronic spine loss require both Pyk2 kinase and RhoA activation. Thus, Pyk2 functions at postsynaptic sites to modulate F-actin control by RhoA and regulate synapse maintenance of relevance to AD risk.SIGNIFICANCE STATEMENT Genetic variation at the Pyk2 locus is a risk for Alzheimer's disease. We have observed that Pyk2 is required for AD transgenic synapse loss and memory dysfunction. However, the cellular and biochemical basis for Pyk2 function related to AD is not defined. Here, we show that brain Pyk2 interacts with the RhoGAP protein Graf1 to alter dendritic spine stability via RhoA GTPase. Amyloid-ß oligomer-induced dendritic spine loss requires the Pyk2/Graf1 pathway.


Subject(s)
Amyloid beta-Peptides/metabolism , Dendritic Spines/metabolism , Focal Adhesion Kinase 2/metabolism , GTPase-Activating Proteins/metabolism , Hippocampus/metabolism , Synapses/metabolism , Synapses/pathology , rhoA GTP-Binding Protein/metabolism , Animals , Female , Focal Adhesion Kinase 2/genetics , HEK293 Cells , Hippocampus/pathology , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
2.
J Neurosci ; 39(4): 758-772, 2019 01 23.
Article in English | MEDLINE | ID: mdl-30518596

ABSTRACT

Dozens of genes have been implicated in late onset Alzheimer's disease (AD) risk, but none has a defined mechanism of action in neurons. Here, we show that the risk factor Pyk2 (PTK2B) localizes specifically to neurons in adult brain. Absence of Pyk2 has no major effect on synapse formation or the basal parameters of synaptic transmission in the hippocampal Schaffer collateral pathway. However, the induction of synaptic LTD is suppressed in Pyk2-null slices. In contrast, deletion of Pyk2 expression does not alter LTP under control conditions. Of relevance for AD pathophysiology, Pyk2-/- slices are protected from amyloid-ß-oligomer (Aßo)-induced suppression of LTP in hippocampal slices. Acutely, a Pyk2 kinase inhibitor also prevents Aßo-induced suppression of LTP in WT slices. Female and male transgenic AD model mice expressing APPswe/PSEN1ΔE9 require Pyk2 for age-dependent loss of synaptic markers and for impairment of learning and memory. However, absence of Pyk2 does not alter Aß accumulation or gliosis. Therefore, the Pyk2 risk gene is directly implicated in a neuronal Aßo signaling pathway impairing synaptic anatomy and function.SIGNIFICANCE STATEMENT Genetic variation at the Pyk2 (PTK2B) locus is a risk for late onset Alzheimer's disease (AD), but the pathophysiological role of Pyk2 is not clear. Here, we studied Pyk2 neuronal function in mice lacking expression with and without transgenes generating amyloid-ß (Aß) plaque pathology. Pyk2 is not required for basal synaptic transmission or LTP, but participates in LTD. Hippocampal slices lacking Pyk2 are protected from AD-related Aß oligomer suppression of synaptic plasticity. In transgenic AD model mice, deletion of Pyk2 rescues synaptic loss and learning/memory deficits. Therefore, Pyk2 plays a central role in AD-related synaptic dysfunction mediating Aß-triggered dysfunction.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Focal Adhesion Kinase 2/genetics , Synapses/pathology , Animals , Behavior, Animal , Female , Gliosis/genetics , Gliosis/pathology , Learning/physiology , Long-Term Potentiation/genetics , Long-Term Synaptic Depression/genetics , Male , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Risk Factors , Signal Transduction/genetics
3.
J Neurosci ; 37(38): 9207-9221, 2017 09 20.
Article in English | MEDLINE | ID: mdl-28842420

ABSTRACT

Biochemical and genetic evidence implicate soluble oligomeric amyloid-ß (Aßo) in triggering Alzheimer's disease (AD) pathophysiology. Moreover, constitutive deletion of the Aßo-binding cellular prion protein (PrPC) prevents development of memory deficits in APPswe/PS1ΔE9 mice, a model of familial AD. Here, we define the role of PrPC to rescue or halt established AD endophenotypes in a therapeutic disease-modifying time window after symptom onset. Deletion of Prnp at either 12 or 16 months of age fully reverses hippocampal synapse loss and completely rescues preexisting behavioral deficits by 17 months. In contrast, but consistent with a neuronal function for Aßo/PrPC signaling, plaque density, microgliosis, and astrocytosis are not altered. Degeneration of catecholaminergic neurons remains unchanged by PrPC reduction after disease onset. These results define the potential of targeting PrPC as a disease-modifying therapy for certain AD-related phenotypes after disease onset.SIGNIFICANCE STATEMENT The study presented here further elucidates our understanding of the soluble oligomeric amyloid-ß-Aßo-binding cellular prion protein (PrPC) signaling pathway in a familial form of Alzheimer's disease (AD) by implicating PrPC as a potential therapeutic target for AD. In particular, genetic deletion of Prnp rescued several familial AD (FAD)-associated phenotypes after disease onset in a mouse model of FAD. This study underscores the therapeutic potential of PrPC deletion given that patients already present symptoms at the time of diagnosis.


Subject(s)
Alzheimer Disease/physiopathology , Brain/physiopathology , Mental Disorders/physiopathology , Prion Proteins/metabolism , Synapses/metabolism , Synaptic Transmission , Alzheimer Disease/complications , Alzheimer Disease/pathology , Animals , Animals, Genetically Modified , Brain/pathology , Disease Progression , Female , Gene Deletion , Male , Mental Disorders/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Synapses/pathology
4.
Cell Rep ; 20(1): 76-88, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28683325

ABSTRACT

Metabotropic glutamate receptor 5 (mGluR5) has been implicated in Alzheimer's disease (AD) pathology. We sought to understand whether mGluR5's role in AD requires glutamate signaling. We used a potent mGluR5 silent allosteric modulator (SAM, BMS-984923) to separate its well-known physiological role in glutamate signaling from a pathological role in mediating amyloid-ß oligomer (Aßo) action. Binding of the SAM to mGluR5 does not change glutamate signaling but strongly reduces mGluR5 interaction with cellular prion protein (PrPC) bound to Aßo. The SAM compound prevents Aßo-induced signal transduction in brain slices and in an AD transgenic mouse model, the APPswe/PS1ΔE9 strain. Critically, 4 weeks of SAM treatment rescues memory deficits and synaptic depletion in the APPswe/PS1ΔE9 transgenic mouse brain. Our data show that mGluR5's role in Aßo-dependent AD phenotypes is separate from its role in glutamate signaling and silent allosteric modulation of mGluR5 has promise as a disease-modifying AD intervention with a broad therapeutic window.


Subject(s)
Alzheimer Disease/drug therapy , Neuroprotective Agents/pharmacology , Receptor, Metabotropic Glutamate 5/metabolism , Synaptic Transmission/drug effects , Allosteric Regulation , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/drug effects , Brain/metabolism , HEK293 Cells , Humans , Memory , Mice , Neuroprotective Agents/therapeutic use , Phenotype , Presenilins/genetics , Prions/metabolism , Protein Binding
5.
Biochem Biophys Res Commun ; 483(4): 1143-1147, 2017 02 19.
Article in English | MEDLINE | ID: mdl-27639648

ABSTRACT

Soluble oligomers of amyloid-beta (Aßo) are implicated by biochemical and genetic evidence as a trigger for Alzheimer's disease (AD) pathophysiology. A key step is Aßo interaction with the neuronal surface to initiate a cascade of altered signal transduction leading to synaptic dysfunction and damage. This review discusses neuronal cell surface molecules with high affinity selectively for oligomeric disease-associated states of Aß, with a particular focus on the role of cellular prion protein (PrPC) in this process. Additional receptors may contribute to mediation of Aßo action, but PrPC appears to play a primary role in a number of systems. The specificity of binding, the genetic necessity in mouse models of disease and downstream signaling pathways are considered. Signal transduction downstream of Aßo complexes with PrPC involves metabotropic glutamate receptor 5 (mGluR5), Fyn kinase and Pyk2 kinase, with deleterious effects on synaptic transmission and maintenance. Current data support the hypothesis that a substantial portion of Aßo toxicity in AD is mediated after initial interaction with PrPC on the neuronal surface. As such, the interaction of Aßo with PrPC is a potential therapeutic intervention site for AD.


Subject(s)
Amyloid beta-Peptides/metabolism , PrPC Proteins/metabolism , Animals , Humans , Mice , Protein Binding , Signal Transduction
6.
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
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
SELECTION OF CITATIONS
SEARCH DETAIL
...