Skip to main content
U.S. flag

An official website of the United States government

Kv1.3 inhibition as a potential microglia-targeted therapy for Alzheimer’s disease: preclinical proof of concept

Bibliographic

Year of Publication:
2018
Contact PI Name:
Izumi Maezawa
Contact PI Affiliation:
Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, California, USA
Co-Authors:
Hai M. Nguyen, Jacopo Di Lucente, David Paul Jenkins, Vikrant Singh, Silvia Hilt, Kyoungmi Kim, Srikant Rangaraju, Allan I. Levey, Heike Wulff, Lee-Way Jin
Primary Reference (PubMED ID):
Funding Source:
National Institute of Neurological Disorders and Stroke (NINDS)
National Institute on Aging (NIA)
Alzheimer's Association
Study Goal and Principal Findings:

Microglia significantly contribute to the pathophysiology of Alzheimer’s disease but an effective microglia-targeted therapeutic approach is not yet available clinically. The potassium channels Kv1.3 and Kir2.1 play important roles in regulating immune cell functions and have been implicated by in vitro studies in the ‘M1-like pro-inflammatory’ or ‘M2-like anti-inflammatory’ state of microglia, respectively. We here found that amyloid-β oligomer-induced expression of Kv1.3 and Kir2.1 in cultured primary microglia. Likewise, ex vivo microglia acutely isolated from the Alzheimer’s model 5xFAD mice co-expressed Kv1.3 and Kir2.1 as well as markers traditionally associated with M1 and M2 activation suggesting that amyloid-β oligomer induces a microglial activation state that is more complex than previously thought. Using the orally available, brain penetrant small molecule Kv1.3 blocker PAP-1 as a tool, we showed that pro-inflammatory and neurotoxic microglial responses induced by amyloid-β oligomer required Kv1.3 activity in vitro and in hippocampal slices. Since we further observed that Kv1.3 was highly expressed in microglia of transgenic Alzheimer’s mouse models and human Alzheimer’s disease brains, we hypothesized that pharmacological Kv1.3 inhibition could mitigate the pathology induced by amyloid-β aggregates. Indeed, treating APP/PS1 transgenic mice with a 5-month oral regimen of PAP-1, starting at 9 months of age, when the animals already manifest cognitive deficits and amyloid pathology, reduced neuroinflammation, decreased cerebral amyloid load, enhanced hippocampal neuronal plasticity, and improved behavioural deficits. The observed decrease in cerebral amyloid deposition was consistent with the in vitro finding that PAP-1 enhanced amyloid-β uptake by microglia. Collectively, these results provide proof-of-concept data to advance Kv1.3 blockers to Alzheimer’s disease clinical trials.

Bibliographic Notes:
Heike Wulff (Department of Pharmacology, University of California Davis, Davis, CA, USA) and Izumi Maezawa and Lee-Way Jin (Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, CA, USA) are corresponding authors on this paper.

Therapeutic Agent

Therapeutic Information:
Therapy Type:
Small Molecule
Therapeutic Agent:
PAP-1
Therapeutic Target:
Voltage-Gated Potassium Kv1.3 Channel

Animal Model

Model Information:
Species:
Mouse
Model Type:
APPxPS1
Strain/Genetic Background:
C57BL/6
Species:
Mouse
Model Type:
APPxPS1
Strain/Genetic Background:
Not Reported

Experimental Design

Is the following information reported in the study?:
Power/Sample Size Calculation
Randomized into Groups
Blinded for Treatment
Blinded for Outcome Measures
Pharmacokinetic Measures
Pharmacodynamic Measures
Toxicology Measures
ADME Measures
Biomarkers
Dose
Formulation
Route of Delivery
Duration of Treatment
Frequency of Administration
Age of Animal at the Beginning of Treatment
Age of Animal at the End of Treatment
Sex as a Biological Variable
Study Balanced for Sex as a Biological Variable
Number of Premature Deaths
Number of Excluded Animals
Statistical Plan
Genetic Background
Inclusion/Exclusion Criteria Included
Conflict of Interest

Outcomes

Outcome Measured
Outcome Parameters
Behavioral
Exploratory Activity
Novel Object Recognition Test (NORT)
Open Field Test
Spontaneous Activity
Step-Through Passive Avoidance Test
Motor Function
Locomotor Activity
Histopathology
Activated Microglia
beta Amyloid Deposits
beta Amyloid Load
Colocalization-Astrocytes/Microglia/Amyloid Plaques
Biochemical
Brain-Buffer Insoluble beta Amyloid Peptide 42
Brain-Buffer Soluble beta Amyloid Peptide 42
CD86 mRNA
CD206 mRNA
Interleukin 1 beta (IL-1 beta) mRNA
Interleukin 6 (IL-6) mRNA
Inducible Nitric Oxide Synthase (iNOS/NOS2) mRNA
Nitric Oxide (NO)
Potassium Channel Kv1.3 mRNA
Tumor Necrosis Factor alpha (TNF alpha) mRNA
p38 Mitogen-Activated Protein Kinase (p38 MAPK)
phospho-p38 Mitogen-Activated Protein Kinase (phospho-p38 MAPK)
Immunochemistry
Brain-beta Amyloid Deposits
beta Amyloid Load
CD11b
CD68
Ionized Calcium Binding Adaptor Molecule 1 (Iba1)
Potassium Channel Kv1.3
Neuronal Marker NeuN
phospho-Nuclear Factor kappa B (phospho-NFkB)
Microscopy
Cell Count
Cell Survival
Spectroscopy
Liquid Chromatography-Mass Spectrometry (LC/MS)
Cell Biology
beta Amyloid Peptide Phagocytosis
Cell Growth
Microglial Proliferation
Electrophysiology
Capacitance
Current Density
Current-Voltage (I-V) Curve
Inward Rectifying Current (Kir)
Outward Rectifying Current
Potassium Channel (Kv) Current
Peak Current
Use-Dependent Inactivation
Voltage Clamp Recording
Voltage Steps
Whole Cell Patch Clamp Recording
field Excitatory Postsynaptic Potential (fEPSP)
Long Term Potentiation (LTP)
Pharmacokinetics
Blood Brain Barrier Penetration
Drug Concentration-Brain
Drug Concentration-Plasma