Skip to main content
U.S. flag

An official website of the United States government

CD40L disruption enhances Aβ vaccine-mediated reduction of cerebral amyloidosis while minimizing cerebral amyloid angiopathy and inflammation

Bibliographic

Year of Publication:
2008
Contact PI Name:
Jun Tan
Contact PI Affiliation:
Neuroimmunology Laboratory, Department of Psychiatry and Behavioral Medicine, University of South Florida, Tampa, Florida, USA
Co-Authors:
D. Obregon, H. Hou, Y. Bai, W.V. Nikolic, T. Mori, Deyan Luo, J. Zeng, J. Ehrhart, F. Fernandez, D. Morgan, B. Giunta, T. Town
Primary Reference (PubMED ID):
Funding Source:
Alzheimer's Association
National Institute on Aging (NIA)
National Institute of Neurological Disorders and Stroke (NINDS)
Study Goal and Principal Findings:

Amyloid-β (Aβ) immunization efficiently reduces amyloid plaque load and memory impairment in transgenic mouse models of Alzheimer’s disease (AD) (Schenk et al., 1999; Morgan et al., 2000). Active Aβ immunization has also yielded favorable results in a subset of AD patients (Hock et al., 2003). However, a small percentage of patients developed severe aseptic meningoencephalitis associated with brain inflammation and infiltration of T-cells (Nicoll et al., 2003; Orgogozo et al., 2003). We and others have shown that blocking the CD40-CD40 ligand (L) interaction mitigates Aβ-induced inflammatory responses and enhances Aβ clearance (Tan et al., 2002b; Townsend et al., 2005). Here, we utilized genetic and pharmacologic approaches to test whether CD40-CD40L blockade could enhance the efficacy of Aβ1–42 immunization, while limiting potentially damaging inflammatory responses. We show that genetic or pharmacologic interruption of CD40-CD40L interaction enhanced Aβ1–42 immunization efficacy to reduce cerebral amyloidosis in the PSAPP and Tg2576 mouse models of AD. Potentially deleterious pro-inflammatory immune responses, cerebral amyloid angiopathy (CAA) and cerebral microhemorrhage were reduced or absent in these combined approaches. Pharmacologic blockade of CD40L decreased T-cell neurotoxicity to Aβ-producing neurons. Further reduction of cerebral amyloidosis in Aβ-immunized PSAPP mice completely deficient for CD40 occurred in the absence of Aβ immunoglobulin G (IgG) antibodies or efflux of Aβ from brain to blood, but was rather correlated with anti-inflammatory cytokine profiles and reduced plasma soluble CD40L. These results suggest CD40-CD40L blockade promotes anti-inflammatory cellular immune responses, likely resulting in promotion of microglial phagocytic activity and Aβ clearance while precluding generation of neurotoxic Aβ-reactive T-cells. Thus, combined approaches of Aβ immunotherapy and CD40-CD40L blockade may provide for safer and more effective Aβ vaccine.

Bibliographic Notes:
Jun Tan (Neuroimmunology Laboratory, Department of Psychiatry and Behavioral Medicine, University of South Florida, Tampa, FL, USA) and Terrence Town (Neurosurgical Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA) are corresponding authors on this paper.

Therapeutic Agent

Therapeutic Information:
Therapy Type:
Biologic - Immunotherapy(active)
Therapeutic Agent:
beta Amyloid Peptide 1-42
Therapeutic Target:
beta Amyloid Peptide
Therapy Type:
Biologic - Immunotherapy(passive)
Therapeutic Agent:
anti-CD40L Antibody
Therapeutic Target:
CD40LG

Animal Model

Model Information:
Species:
Mouse
Model Type:
APPxPS1
Strain/Genetic Background:
C57BL/6
Species:
Mouse
Model Type:
APP
Strain/Genetic Background:
Not Reported
Species:
Mouse
Model Type:
APPxPS1
Strain/Genetic Background:
Not Reported
Species:
Mouse
Model Type:
APPxPS1
Strain/Genetic Background:
Not Reported
Animal Model Notes:
APPswe/PSEN1dE9 mice were crossed with CD40 KO mice (https://www.jax.org/strain/002928) to generate PSAPP/CD40+/- and PSAPP/CD40-/- mice for this study.

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
Histopathology
Activated Microglia
beta Amyloid Deposits
beta Amyloid Load
Congophillic Amyloid Deposits
Cerebral Amyloid Angiopathy (CAA)
Biochemical
Brain-Detergent Soluble beta Amyloid Peptide 40
Brain-Detergent Soluble beta Amyloid Peptide 42
Brain-Guanidine Soluble beta Amyloid Peptide 40
Brain-Guanidine Soluble beta Amyloid Peptide 42
Interleukin 1 beta (IL-1 beta)
Interleukin 2 (IL-2)
Interleukin 4 (IL-4)
Interleukin 10 (IL-10)
Interferon (IFN) gamma
Tumor Growth Factor beta (TGF beta)
Tumor Necrosis Factor alpha (TNF alpha)
Immunochemistry
Brain-beta Amyloid Deposits
CD45
Ionized Calcium Binding Adaptor Molecule 1 (Iba1)
Major Histocompatibility Antigens Class 2 (MHC II)
Microglia Morphology
Cell Biology
Cell Viability
Cytokines
Immunology
Anti-beta Amyloid Antibody Titers
Antibody Isotypes
Biomarker
Plasma-beta Amyloid Peptide 40
Plasma-beta Amyloid Peptide 42
Pharmacodynamics
Target Engagement (Reduction beta Amyloid Deposits)
Toxicology
General Health