Combined effects of hematopoietic progenitor cell mobilization from bone marrow by granulocyte colony stimulating factor and AMD3100 and chemotaxis into the brain using stromal cell-derived factor-1α in an Alzheimer's disease mouse model


BIBLIOGRAPHIC THERAPEUTIC AGENT ANIMAL MODEL EXPERIMENTAL DESIGN OUTCOMES

Bibliographic

Year of Publication:
2011
Contact PI Name:
Jae-Sung Bae
Contact PI Affiliation:
School of Medicine, Kyungpook National University, Jung‐Gu, Daegu, South Korea
Co-Authors:
Ji-Woong Shin, Jong Kil Lee, Jeong Eun Lee, Woo-Kie Min, Edward H. Schuchman, Hee Kyung Jin
Primary Reference (PubMED ID):
Funding Source:
National Research Foundation of Korea (NRF)
Korean Ministry of Science and Technology (MoST)
Study Goal and Principal Findings:

Transplantation of bone marrow‐derived stem cells (BMSCs) has been suggested as a potential therapeutic approach to prevent neurodegenerative diseases, but it remains problematic due to issues of engraftment, potential toxicities, and other factors. An alternative strategy is pharmacological‐induced recruitment of endogenous BMSCs into an injured site by systemic administration of growth factors or chemokines. Therefore, the aim of this study was to examine the effects of therapy involving granulocyte colony stimulating factor (G‐CSF)/AMD3100 (CXCR4 antagonist) and stromal cell‐derived factor‐1α (SDF‐1α) on endogenous BM‐derived hematopoietic progenitor cell (BM‐HPC) recruitment into the brain of an Alzheimer's disease (AD) mouse model. To mobilize BM‐HPCs, G‐CSF was injected intraperitoneally and boosted by AMD3100. Simultaneously, these mice received an intracerebral injection with SDF‐1α to induce migration of mobilized BM‐HPCs into brain. We found that the memory deficit in the AD mice was significantly improved by these treatments, but amyloid β deposition was unchanged. Interestingly, microglial activation was increased with alternative activation of microglia to a neuroprotective phenotype. Furthermore, by generating an amyloid precursor protein/presenilin 1‐green fluorescent protein (GFP) chimeric mouse, we ascertained that the GFP positive microglia identified in the brain were BM‐derived. Additionally, increased hippocampal neurogenesis and improved memory was observed in mice receiving combined G‐CSF/AMD3100 and SDF‐1α, but not in controls or animals receiving each treatment alone. These results suggest that SDF‐1α is an effective adjuvant in inducing migration into brain of the endogenous BM‐HPCs, mobilized by G‐CSF/AMD3100, and that the two can act synergistically to produce a therapeutic effect. This approach warrants further investigation as a potential therapeutic option for the treatment of AD patients in the future.

Therapeutic Agent

Therapeutic Information:
Therapy Type:
Biologic - Protein
Therapeutic Agent:
Human Recombinant G‐CSF
Therapeutic Target:
Granulocyte Colony-Stimulating Factor Receptor (G-CSFR)
Therapy Type:
Small Molecule
Therapeutic Agent:
AMD3100
Therapeutic Target:
Chemokine C-X-C Motif Receptor 4 (CXCR4)
Therapy Type:
Biologic - Peptide
Therapeutic Agent:
Stromal Cell‐Derived Factor‐1α (SDF‐1α)
Therapeutic Target:
Chemokine C-X-C Motif Receptor 4 (CXCR4)
Therapeutic Notes:
Granulocyte Colony-Stimulating Factor Receptor (G-CSFR) has been nominated as a potential target for AD. Nominated targets are obtained from several sources, including the National Institute on Aging's Accelerating Medicines Partnership in Alzheimer's Disease (AMP-AD) consortium. Targets have been identified using computational analyses of high-dimensional genomic, proteomic and/or metabolomic data derived from human samples. See Agora link for more information.

Animal Model

Model Information:
Species:
Mouse
Model Type:
APPxPS1
Strain/Genetic Background:
Not Reported
Animal Model Notes:
APP/PS1 double transgenic mice and nontransgenic (NT) control littermates were generated by mating single transgenic mice expressing human mutant APP and mutant PS1. Six‐month‐old APP/PS1 mice were used as recipients to create APP/PS1‐green fluorescent protein (GFP) chimeric mice. The BM donor animals were 6‐week‐old homozygous C57BL/6‐Tg (CAG‐EGFP)1osb/J mice on the C57BL/6 background (The Jackson Laboratory, Bar Harbor, Maine).

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
beta Amyloid Load
beta Amyloid Deposits
Activated Microglia
Biochemical
Tumor Necrosis Factor alpha (TNF alpha)
Tumor Necrosis Factor Receptor 1 (TNFR1) mRNA
Interleukin 4 (IL-4)
Immunochemistry
Ionized Calcium Binding Adaptor Molecule 1 (Iba1)
Interleukin 1 beta (IL-1 beta)
Tumor Necrosis Factor alpha (TNF alpha)
Interleukin 4 (IL-4)
Chitinase 3-Like Protein 3 (YM1)
CD11b
Neuronal Marker NeuN
5-bromo-2’-deoxyuridine (BrdU)

Source URL: http://alzped.nia.nih.gov/combined-effects-hematopoietic