Skip to main content
U.S. flag

An official website of the United States government

Glutaminyl cyclase inhibition attenuates pyroglutamate Aβ and Alzheimer’s disease–like pathology

Bibliographic

Year of Publication:
2008
Contact PI Name:
Hans-Ulrich Demuth
Contact PI Affiliation:
Probiodrug AG, Halle, Germany
Co-Authors:
Stephan Schilling, Ulrike Zeitschel, Torsten Hoffmann, Ulrich Heiser, Mike Francke, Astrid Kehlen, Max Holzer, Birgit Hutter-Paier, Manuela Prokesch, Manfred Windisch, Wolfgang Jagla, Dagmar Schlenzig, Christiane Lindner, Thomas Rudolph, Gunter Reuter
Primary Reference (PubMED ID):
Funding Source:
German Federal Ministry of Education and Research (BMBF)
Study Goal and Principal Findings:

Because of their abundance, resistance to proteolysis, rapid aggregation and neurotoxicity, N-terminally truncated and, in particular, pyroglutamate (pE)-modified Aβ peptides have been suggested as being important in the initiation of pathological cascades resulting in the development of Alzheimer’s disease. This study found that the N-terminal pE-formation is catalyzed by glutaminyl cyclase in vivo. Glutaminyl cyclase expression was upregulated in the cortices of individuals with Alzheimer’s disease and correlated with the appearance of pE-modified Aβ. Oral application of a glutaminyl cyclase inhibitor resulted in reduced Aβ3(pE)–42 burden in two different transgenic mouse models of Alzheimer’s disease and in a new Drosophila model. Treatment of mice was accompanied by reductions in Aβx–40/42, diminished plaque formation and gliosis and improved performance in context memory and spatial learning tests. These observations are consistent with the hypothesis that Aβ3(pE)–42 acts as a seed for Aβ aggregation by self-aggregation and co-aggregation with Aβ1–40/42. Therefore, Aβ3(pE)–40/42 peptides seem to represent Aβ forms with exceptional potency for disturbing neuronal function. The reduction of brain pE-Aβ by inhibition of glutaminyl cyclase offers a new therapeutic option for the treatment of Alzheimer’s disease and provides implications for other amyloidoses, such as familial Danish dementia.

Therapeutic Agent

Therapeutic Information:
Therapy Type:
Small Molecule
Therapeutic Agent:
PBD150
Therapeutic Target:
Glutaminyl Cyclase (QC)

Animal Model

Model Information:
Species:
Mouse
Model Type:
APP
Strain/Genetic Background:
B6/SJL
Species:
Mouse
Model Type:
APP
Strain/Genetic Background:
C57/Bl6

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
Fear Conditioning Response
Morris Water Maze
Histopathology
beta Amyloid Deposits
Activated Microglia
Activated Astrocytes
Biochemical
Glutaminyl Cyclase
Pyroglutamate Modified beta Amyloid Peptides
beta-Site Amyloid Precursor Protein Cleaving Enzyme 1 (BACE1) Activity
Insulin Degrading Enzyme (IDE) Activity
Amyloid Precursor Protein (APP)
Neprilysin
Aminopeptidase N
Brain-beta Amyloid Peptide-Total
Immunochemistry
Pyroglutamate Modified beta Amyloid Peptides
Microglia
Glial Fibrillary Acidic Protein (GFAP)
Brain-beta Amyloid Peptides
Electron Microscopy
Aggregated beta Amyloid Peptide
Pharmacokinetics
Drug Concentration-Brain
Drug Concentration-Organs
Pharmacodynamics
Target Engagement (Inhibition Glutaminyl Cyclase)
Toxicology
Body Weight
Mortality