Skip to main content
U.S. flag

An official website of the United States government

A novel retro-inverso peptide inhibitor reduces amyloid deposition, oxidation and inflammation and stimulates neurogenesis in the APPswe/PS1DE9 mouse model of Alzheimer’s disease

Bibliographic

Year of Publication:
2013
Contact PI Name:
David Allsop
Contact PI Affiliation:
Division of Biomedical and Life Sciences, University of Lancaster, Lancaster, Lancashire, UK
Co-Authors:
Vadivel Parthsarathy, Paula L. McClean, Christian Hӧlscher, Mark Taylor, Claire Tinker, Glynn Jones, Oleg Kolosov, Elisa Salvati, Maria Gregori, Massimo Masserini
Primary Reference (PubMED ID):
Funding Source:
Alzheimer's Research UK
Study Goal and Principal Findings:

Previously, we have developed a retro-inverso peptide inhibitor (RI-OR2, rGffvlkGr) that blocks the in vitro formation and toxicity of the Aβ oligomers which are thought to be a cause of neurodegeneration and memory loss in Alzheimer’s disease. We have now attached a retro-inverted version of the HIV protein transduction domain ‘TAT’ to RI-OR2 to target this new inhibitor (RI-OR2-TAT, Ac-rGffvlkGrrrrqrrkkrGy-NH2) into the brain. Following its peripheral injection, a fluorescein-labelled version of RI-OR2-TAT was found to cross the blood brain barrier and bind to the amyloid plaques and activated microglial cells present in the cerebral cortex of 17-months-old APPswe/PS1DE9 transgenic mice. Daily intraperitoneal injection of RIOR2-TAT (at 100 nmol/kg) for 21 days into 10-months-old APPswe/PS1DE9 mice resulted in a 25% reduction (p,0.01) in the cerebral cortex of Aβ oligomer levels, a 32% reduction (p,0.0001) of b-amyloid plaque count, a 44% reduction (p,0.0001) in the numbers of activated microglial cells, and a 25% reduction (p,0.0001) in oxidative damage, while the number of young neurons in the dentate gyrus was increased by 210% (p,0.0001), all compared to control APPswe/PS1DE9 mice injected with vehicle (saline) alone. Our data suggest that oxidative damage, inflammation, and inhibition of neurogenesis are all a downstream consequence of Aβ aggregation, and identify a novel brain-penetrant retro-inverso peptide inhibitor of Aβ oligomer formation for further testing in humans as a potential disease-modifying treatment for Alzheimer’s disease.

Therapeutic Agent

Therapeutic Information:
Therapy Type:
Biologic - Peptide
Therapeutic Agent:
RI-OR2-TAT
Therapeutic Target:
beta Amyloid Peptide

Animal Model

Model Information:
Species:
Mouse
Model Type:
APPxPS1
Strain/Genetic Background:
C57BL/6

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

Male mice were used for experiments with Flu-RI-OR2-TAT. Female mice were used for experiments with RI-OR2-TAT.

Outcomes

Outcome Measured
Outcome Parameters
Histopathology
Activated Microglia
beta Amyloid Deposits
beta Amyloid Load
Colocalization-Astrocytes/Microglia/Amyloid Plaques
Biochemical
Brain-beta Amyloid Oligomers
Immunochemistry
8-oxoguanine
beta Amyloid Load
Brain-beta Amyloid Deposits
Doublecortin (DCX)
Ionized Calcium Binding Adaptor Molecule 1 (Iba1)
Microscopy
Atomic Force Microscopy (AFM)
Spectroscopy
Surface Plasmon Resonance (SPR) Spectroscopy
Cell Biology
Cell Viability
Pharmacokinetics
Blood Brain Barrier Penetration
Pharmacodynamics
Target Engagement (Reduction beta Amyloid Deposits)
Target Engagement (Reduction Amyloid Load)
Pharmacology
Binding Affinity
Target Selectivity