Pan-PPAR modulation effectively protects APP/PS1 mice from amyloid deposition and cognitive deficits


BIBLIOGRAPHIC THERAPEUTIC AGENT ANIMAL MODEL EXPERIMENTAL DESIGN OUTCOMES

Bibliographic

Year of Publication:
2015
Contact PI Name:
Michael T. Heneka
Contact PI Affiliation:
Clinical Neuroscience Unit, Department of Neurology, University of Bonn, Bonn, Germany
Co-Authors:
Markus P. Kummer, Rafael Schwarzenberger, Sakina Sayah-Jeanne, Mathieu Dubernet, Robert Walczak, Dean W. Hum, Stephanie Schwartz, Daisy Axt
Primary Reference (PubMED ID):
Funding Source:
Deutsche Forschungsgemeinschaft/German Research Foundation
European Union INMiND Project
Study Goal and Principal Findings:

Alzheimer’s disease (AD) is a neurodegenerative condition that leads to neuronal death and memory dysfunction. In the past, specific peroxisome proliferator-activated receptor (PPAR)γ-agonists, such as pioglitazone, have been tested with limited success to improve AD pathology. Here, we investigated the therapeutic efficacy of GFT1803, a novel potent PPAR agonist that activates all the three PPAR isoforms (α/δ/γ) in the APP/PS1 mouse model in comparison to the selective PPARγ-agonist pioglitazone. Both compounds showed similar brain/plasma partitioning ratios, although whole body and brain exposure to GFT1803 was significantly lower as compared to pioglitazone, at doses used in this study. Oral treatment of APP/PS1 mice with GFT1803 decreased microglial activation, amyloid β (Aβ) plaque area, Aβ levels in sodium dodecyl sulfate- and formic acid-soluble fractions in a concentration-dependent manner. With a single exception of Aβ38 and Aβ40 levels, measured by ELISA, these effects were not observed in mice treated with pioglitazone. Both ligands decreased glial fibrillary acidic protein (GFAP) expression to similar extent and did not affect ApoE expression. Finally, GFT1803 increased insulin-degrading enzyme expression. Analysis of spatial memory formation in the Morris water maze demonstrated that both compounds were able to partially revert the phenotype of APP/PS1 mice in comparison to wild-type mice with GFT1803 being most effective. As compared to pioglitazone, GFT1803 (pan-PPAR agonist) produced both quantitatively superior and qualitatively different therapeutic effects with respect to amyloid plaque burden, insoluble Aβ content, and neuroinflammation at significantly lower whole body and brain exposure rates.

Therapeutic Agent

Therapeutic Information:
Therapy Type:
Small Molecule
Therapeutic Agent:
GFT1803
Therapeutic Target:
Peroxisome Proliferator-Activated Receptor alpha (PPAR alpha)
Therapeutic Target:
Peroxisome Proliferator-Activated Receptor delta (PPAR delta)
Therapeutic Target:
Peroxisome Proliferator-Activated Receptor gamma (PPAR gamma)
Therapy Type:
Small Molecule
Therapeutic Agent:
Pioglitazone
Therapeutic Target:
Peroxisome Proliferator-Activated Receptor gamma (PPAR gamma)
Therapeutic Notes:
GFT1803 is a pan-PPAR agonist that activates all the three PPAR isoforms (α/δ/γ).

Peroxisome Proliferator-Activated Receptor alpha (PPAR alpha) has been nominated as a potential target for AD. Nominated targets are obtained from several sources, including the National Institute on Aging's Accelerating Medicines Partnership in Alzheimer's Disease (AMP-AD) consortium. Targets have been identified using computational analyses of high-dimensional genomic, proteomic and/or metabolomic data derived from human samples. See Agora link for more information.

Animal Model

Model Information:
Species:
Mouse
Model Type:
APPxPS1
Strain/Genetic Background:
C57BL/6
Species:
Mouse
Model Type:
Non-transgenic
Strain/Genetic Background:
Swiss

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
Experiment Notes

Female APPxPS1 mice and male Swiss mice were used in this study. Neither group was balanced for sex.

Outcomes

Outcome Measured
Outcome Parameters
Behavioral
Exploratory Activity
Morris Water Maze
Open Field Test
Motor Function
Locomotor Activity
Path Length
Histopathology
Activated Astrocytes
Activated Microglia
beta Amyloid Deposits
beta Amyloid Load
Colocalization-Astrocytes/Microglia/Amyloid Plaques
Biochemical
Brain-Buffer Soluble beta Amyloid Peptide 38
Brain-Buffer Soluble beta Amyloid Peptide 40
Brain-Buffer Soluble beta Amyloid Peptide 42
Brain-Detergent Soluble beta Amyloid Peptide 38
Brain-Detergent Soluble beta Amyloid Peptide 40
Brain-Detergent Soluble beta Amyloid Peptide 42
Brain-Formic Acid Soluble beta Amyloid Peptide 38
Brain-Formic Acid Soluble beta Amyloid Peptide 40
Brain-Formic Acid Soluble beta Amyloid Peptide 42
Amyloid Precursor Protein (APP)
APP-CTF83 (CTF alpha)
APP-CTF99 (CTF beta)
Soluble Amyloid Precursor Protein alpha (sAPP alpha)
Apolipoprotein E (ApoE)
Glial Fibrillary Acidic Protein (GFAP)
Insulin Degrading Enzyme (IDE)
EC50
Emax
Immunochemistry
beta Amyloid Load
Brain-beta Amyloid Deposits
Glial Fibrillary Acidic Protein (GFAP)
Ionized Calcium Binding Adaptor Molecule 1 (Iba1)
Spectroscopy
Liquid Chromatography with Tandem Mass Spectrometry (LC/MS/MS)
Pharmacokinetics
Area Under the Curve (AUC)
Drug Concentration-Brain
Brain/Plasma Ratio

Source URL: http://alzped.nia.nih.gov/pan-ppar-modulation