The goal of this research is to elucidate the molecular basis of S100B protein's involvement in the neuroinflammatory responses of activated glia (astrocytes and microglia). S100B is a glial-derived cytokine that is significantly elevated in the brains of Alzheimer's disease (AD) patients. S100B induces pro- inflammatory cytokines (e.g., IL-1beta and TNFalpha) and oxidative stress related enzymes (e.g. iNOS) in activated glia, and enhances the ability of other stimuli, such as beta-amyloid, to activate glia. These neuroinflammatory responses, in turn, can lead to neuronal dysfunction and further glial activation. Thus, S100B contributes to a vicious cycle of glial activation/ neuroinflammation/neuronal dysfunction that provides the potential for self-propagation of neurodegenerative events and chronic glial activation seen in AD. However, very little is known about the molecular mechanisms by which S100B induces iNOS and pro-inflammatory cytokines, how modulation of these specific molecular pathways in activated glia affects neuronal viability, or how modulation of S100B itself influences glial and neuronal responses. The hypothesis is that responses of glial cells to S100B contribute to and environment that enhances the progression of neuropathology, and that knowledge of the molecular basis of S100B action on glia will provide the potential for blocking specific glial activation pathways that have neurotoxic consequences.
Specific aim 1 will address the mechanisms by which S100B activates glia.
Aim 1 A will examine if S100B stimulates iNOS through a cytokine-dependent, NFkappaB regulated pathway.
Aim 1 B will determine if p38 and ERK MAP kinases are important for S100B-induced IL-1beta production in activated microglia.
Aim 1 C will elucidate the potential role of RAGE, a new receptor that can interact with S100B, in mediating the S100B-induced neuroinflammatory responses.
Aim 2 will determine the contribution of specific signal transduction pathways leading to iNOS induction on neuronal dysfunction, by utilizing a defined co-culture system of primary astrocytes and neurons where NO- dependent neuronal death occurs. Pathways will be modulated by selective pharmacological agents or molecular constructs, and effects on neuronal viability determined. Astrocytes from genetically engineered mice with overexpression or knockout of specific components of glial signaling pathways will also be used to ascertain how an in vivo alteration in a specific pathway affects neuronal function.
Aim 3 will use a discovery approach employing cell-based screens of chemical libraries and recursive synthetic chemistry refinement to discover ligands that modulate S100B production in activated astrocytes. These studies will provide new insight into glial-neuronal interactions and the knowledge base necessary for pursuit of novel strategies to block glial activation and its neurotoxic consequences.

Agency
National Institute of Health (NIH)
Institute
National Institute on Aging (NIA)
Type
Research Project (R01)
Project #
5R01AG020243-03
Application #
6771718
Study Section
Special Emphasis Panel (ZRG1-BDCN-4 (01))
Program Officer
Wise, Bradley C
Project Start
2002-07-01
Project End
2006-06-30
Budget Start
2004-07-15
Budget End
2006-06-30
Support Year
3
Fiscal Year
2004
Total Cost
$253,570
Indirect Cost
Name
Northwestern University at Chicago
Department
Anatomy/Cell Biology
Type
Schools of Medicine
DUNS #
005436803
City
Chicago
State
IL
Country
United States
Zip Code
60611
Barone, Eugenio; Di Domenico, Fabio; Sultana, Rukhsana et al. (2012) Heme oxygenase-1 posttranslational modifications in the brain of subjects with Alzheimer disease and mild cognitive impairment. Free Radic Biol Med 52:2292-301
Sultana, Rukhsana; Robinson, Renã A S; Di Domenico, Fabio et al. (2011) Proteomic identification of specifically carbonylated brain proteins in APP(NLh)/APP(NLh) × PS-1(P264L)/PS-1(P264L) human double mutant knock-in mice model of Alzheimer disease as a function of age. J Proteomics 74:2430-40
Crist, Richard C; Roth, Jacquelyn J; Baran, Amy A et al. (2010) The armadillo repeat domain of Apc suppresses intestinal tumorigenesis. Mamm Genome 21:450-7
Wing, Laura K; Behanna, Heather A; Van Eldik, Linda J et al. (2006) De novo and molecular target-independent discovery of orally bioavailable lead compounds for neurological disorders. Curr Alzheimer Res 3:205-14
Craft, Jeffrey M; Watterson, D Martin; Van Eldik, Linda J (2006) Human amyloid beta-induced neuroinflammation is an early event in neurodegeneration. Glia 53:484-90
Craft, Jeffrey M; Watterson, D Martin; Van Eldik, Linda J (2005) Neuroinflammation: a potential therapeutic target. Expert Opin Ther Targets 9:887-900
Craft, Jeffrey M; Watterson, D Martin; Marks, Alexander et al. (2005) Enhanced susceptibility of S-100B transgenic mice to neuroinflammation and neuronal dysfunction induced by intracerebroventricular infusion of human beta-amyloid. Glia 51:209-16
Liu, Ling; Li, Yuekui; Van Eldik, Linda J et al. (2005) S100B-induced microglial and neuronal IL-1 expression is mediated by cell type-specific transcription factors. J Neurochem 92:546-53
Craft, Jeffrey M; Van Eldik, Linda J; Zasadzki, Magdalena et al. (2004) Aminopyridazines attenuate hippocampus-dependent behavioral deficits induced by human beta-amyloid in a murine model of neuroinflammation. J Mol Neurosci 24:115-22
Craft, Jeffrey M; Watterson, D Martin; Frautschy, Sally A et al. (2004) Aminopyridazines inhibit beta-amyloid-induced glial activation and neuronal damage in vivo. Neurobiol Aging 25:1283-92

Showing the most recent 10 out of 14 publications