Skip to main content
U.S. flag

An official website of the United States government

Chronic rapamycin restores brain vascular integrity and function through NO synthase activation and improves memory in symptomatic mice modeling Alzheimer’s disease

Bibliographic

Year of Publication:
2013
Contact PI Name:
Veronica Galvan
Contact PI Affiliation:
Department of Physiology and the Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
Co-Authors:
Ai-Ling Lin, Wei Zheng, Jonathan J. Halloran, Raquel R. Burbank, Stacy A. Hussong, Matthew J. Hart, Martin Javors, Yen-Yu Ian Shih, Eric Muir, Rene Solano Fonseca, Randy Strong, Arlan G. Richardson, James D. Lechleiter, Peter T. Fox
Primary Reference (PubMED ID):
Funding Source:
Ellison Medical Foundation
William and Ella Owens Medical Research Foundation
National Institute on Aging (NIA)
United States Department of Veterans Affairs (VA)
Study Goal and Principal Findings:

Vascular pathology is a major feature of Alzheimer's disease (AD) and other dementias. We recently showed that chronic administration of the target-of-rapamycin (TOR) inhibitor rapamycin, which extends lifespan and delays aging, halts the progression of AD-like disease in transgenic human (h)APP mice modeling AD when administered before disease onset. Here we demonstrate that chronic reduction of TOR activity by rapamycin treatment started after disease onset restored cerebral blood flow (CBF) and brain vascular density, reduced cerebral amyloid angiopathy and microhemorrhages, decreased amyloid burden, and improved cognitive function in symptomatic hAPP (AD) mice. Like acetylcholine (ACh), a potent vasodilator, acute rapamycin treatment induced the phosphorylation of endothelial nitric oxide (NO) synthase (eNOS) and NO release in brain endothelium. Administration of the NOS inhibitor L-NG-Nitroarginine methyl ester reversed vasodilation as well as the protective effects of rapamycin on CBF and vasculature integrity, indicating that rapamycin preserves vascular density and CBF in AD mouse brains through NOS activation. Taken together, our data suggest that chronic reduction of TOR activity by rapamycin blocked the progression of AD-like cognitive and histopathological deficits by preserving brain vascular integrity and function. Drugs that inhibit the TOR pathway may have promise as a therapy for AD and possibly for vascular dementias.

Therapeutic Agent

Therapeutic Information:
Therapy Type:
Small Molecule
Therapeutic Agent:
Rapamycin (Rap)
Therapeutic Target:
Mechanistic Target of Rapamycin (mTOR)
Therapy Type:
Small Molecule
Therapeutic Agent:
N(G)-Nitro-L-arginine Methyl Ester (LNAME)
Therapeutic Target:
Nitric Oxide Synthase (NOS)
Therapeutic Notes:
Acetylcholine (Ach) (300 μL, 7.5 μg/mL solution in phosphate-buffered saline), as a positive control for vasodilation, together with Rhodamine-dextran and DAF-FM was injected intravenously via tail vein. In addition, Monocrystalline iron oxide nanoparticle was injected through the tail vein at 30 mg/kg for vascular density measurement.

Animal Model

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

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
Morris Water Maze
Histopathology
Microhemorrhages
Cerebral Amyloid Angiopathy (CAA)
Dense-core/Compact Plaques
Biochemical
Endothelial Nitric Oxide Synthase (eNOS/NOS3)
phospho-Endothelial Nitric Oxide Synthase (phospho-eNOS/NOS3)
Plasma-beta Amyloid Peptide 40
Plasma-beta Amyloid Peptide 42
Immunochemistry
Vascular beta Amyloid
beta Amyloid Load
Lectin
Microscopy
Microhemorrhages
Vascular Structure
Spectroscopy
Liquid Chromatography with Tandem Mass Spectrometry (LC/MS/MS)
Imaging
Cerebral Blood Flow (CBF)
Cerebral Metabolic Rates of Glucose Uptake (CMRglc)
MRI-Vessel Function Map
Magnetic Resonance Imaging (MRI)
In Vivo Fluorescence Imaging
In Vivo Positron Emission Tomographic (PET) Imaging
Biomarker
Cerebral Blood Flow (CBF)
Cerebral Metabolic Rates of Glucose Uptake (CMRglc)
Plasma-beta Amyloid Peptide 40
Plasma-beta Amyloid Peptide 42
Toxicology
Body Weight
Anxiety
Physiology
Heart Rate
Respiration Rate
Cerebral Blood Flow (CBF)