Skip to main content
U.S. flag

An official website of the United States government

Phosphodiesterase III inhibitor promotes drainage of cerebrovascular β-amyloid

Bibliographic

Year of Publication:
2014
Contact PI Name:
Masafumi Ihara
Contact PI Affiliation:
Department of Stroke and Cerebrovascular Diseases, National Cerebral and Cardiovascular Center, Osaka, Japan
Co-Authors:
T. Maki, Y. Okamoto, R.O. Carare, Y. Hase, Y. Hattori, C.A. Hawkes, S. Saito, Y. Yamamoto, Y. Terasaki, H. Ishibashi-Ueda, A. Taguchi, R. Takahashi, T. Miyakawa, R.N. Kalaria, E.H. Lo, K. Arai
Primary Reference (PubMED ID):
Funding Source:
Japanese Ministry of Health, Labour and Welfare
Japan Ministry of Education
Takeda Science Foundation
Alzheimer's Research UK
Japan Society for the Promotion of Sciences (JSPS)
National Institute of Neurological Disorders and Stroke (NINDS)
Study Goal and Principal Findings:

 Brain amyloidosis is a key feature of Alzheimer’s disease (AD). It also incorporates cerebrovascular amyloid b (Aβ) in the form of cerebral amyloid angiopathy (CAA) involving neurovascular dysfunction. Studies recently showed by retrospective analysis that patients with mild cognitive impairment receiving a vasoactive drug cilostazol, a selective inhibitor of phosphodiesterase (PDE) III, exhibit significantly reduced cognitive decline. This study tested whether cilostazol protects against the disruption of the neurovascular unit and facilitates the arterial pulsation-driven perivascular drainage of Aβ in AD/CAA. Results showed that PDE III is abnormally upregulated in cerebral blood vessels of AD and CAA subjects and closely correlates with vascular amyloid burden. Cilostazol treatment maintained cerebral hyperemic and vasodilative responses to hypercapnia and acetylcholine, suppressed degeneration of pericytes and vascular smooth muscle cells, promoted perivascular drainage of soluble fluorescent Aβ1-40, and rescued cognitive deficits in Tg-SwDI mice. Although cilostazol decreased endogenous Aβ production in cultured neurons, C-terminal fragment of amyloid precursor protein expression was not altered in cilostazol-treated Tg-SwDI mice.The predominant action of cilostazol on Aβ metabolism is likely to facilitate Aβ clearance due to the sustained cerebrovascular function in vivo. These findings mechanistically demonstrate that cilostazol is a promising therapeutic approach for AD and CAA.

Therapeutic Agent

Therapeutic Information:
Therapy Type:
Small Molecule
Therapeutic Agent:
Cilostazol
Therapeutic Target:
Phosphodiesterase Type 3 (PDE3)

Animal Model

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

Outcomes

Outcome Measured
Outcome Parameters
Behavioral
Y Maze
Grooming Performance
Histopathology
beta Amyloid Deposits
Cerebral Amyloid Angiopathy (CAA)
Microhemorrhages
beta Amyloid Load
Biochemical
APP-CTFs
Amyloid Precursor Protein (APP)
Brain-beta Amyloid Peptide 40
Brain-beta Amyloid Peptide 42
Immunochemistry
Brain-beta Amyloid Peptides
Phosphodiesterase 3 (PDE3)
Electron Microscopy
Microvessels
Imaging
Cerebral Blood Flow (CBF)
Cell Biology
Cell Viability
Biomarker
Cerebral Blood Flow (CBF)
Toxicology
Physical Appearance
Microhemorrhage