Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) characterized by focal T cell and myeloid cell infiltrates leading to demyelination and loss of neurologic function. Activated microglia and macrophages are the predominant inflammatory cells in active or chronic MS plaques, and they persist in secondary-progressive MS. Despite advances in our understanding of MS pathophysiology, there are minimal disease-modifying treatments or preventions for innate-mediated, secondary-progressive forms of MS. We recently found that mutually antagonistic pathways driven by TGF? and APOE signaling in microglia, dictate a phenotypic switch between homeostatic (M0) and neurodegenerative (MGnD) phenotypes. Using a systems biology approach, we identified major transcriptional and epigenetic regulators of M0- and MGnD- microglia. Finally, we reveal a new role for TGF?-IFN? signaling in the reprogramming of peripheral monocytes into microglia-like cells with the M0-molecular signature. Based on our published and preliminary data, we hypothesize that modulating the APOE-TGF?/IFN? pathway will restore the homeostatic-tolerogenic microglia and ameliorate EAE. We will address our hypothesis in the following aims:
Aim 1 : Replenish M0-homeostatic microglia via APOE-TGF?/IFN? signaling in EAE In this Aim, we will determine 1) the impact of TGF?/IFN? signaling on the replenishment of M0-microglia in EAE; 2) the regulatory networks controlled by APOE-TGF? signaling underlying microglia phenotype regulation in EAE; and 3) fate-map analysis of microglia phenotype switch during peak and recovery stage of EAE.
Aim 2 : Reprogram monocytes into MG-like cells via APOE-TGF?/IFN? pathway in EAE and from MS patients In this Aim, we will determine 1) whether targeting Apoe in Ly6CHi monocytes facilitates the acquisition of the M0-microglial transcriptional program; 2) the molecular mechanisms underlying TGF?/IFN?-mediated reprogramming of Ly6CHi monocytes; and 3) whether modulation of the APOE-TGF?/IFN? pathway in CD14+/CD16? monocytes isolated from MS subjects induces the M0-microglial transcriptional program to serve as a novel therapeutic approach for progressive MS. Successful completion will result in: 1) identification of molecular mechanisms mediated by APOE-TGF?/IFN? signaling in microglia and monocyte phenotype regulation that contribute to the development, progression and resolution of EAE, and 2) understanding the role of APOE-TGF?/IFN? signaling in reprogramming of peripheral monocytes into homeostatic microglia-like cells in EAE and from MS patients.

Public Health Relevance

This application will investigate major regulatory signaling in brain (microglia) and peripheral (monocytes) immune cells and as targets to treat progressive Multiple Sclerosis (MS). We identified a new mechanism which reprograms and restores the protective microglia phenotype in a mouse model of MS. We will implement our novel tools to specifically modulate the APOE-TGF?/IFN? pathway to restore functional microglia and ameliorate disease progression in a mouse model of MS.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
2R01NS088137-06
Application #
9818237
Study Section
Clinical Neuroimmunology and Brain Tumors Study Section (CNBT)
Program Officer
Utz, Ursula
Project Start
2014-09-30
Project End
2024-06-30
Budget Start
2019-09-15
Budget End
2020-06-30
Support Year
6
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Brigham and Women's Hospital
Department
Type
DUNS #
030811269
City
Boston
State
MA
Country
United States
Zip Code
02115
Rubino, Stephen J; Mayo, Lior; Wimmer, Isabella et al. (2018) Acute microglia ablation induces neurodegeneration in the somatosensory system. Nat Commun 9:4578
Drokhlyansky, Eugene; Göz Aytürk, Didem; Soh, Timothy K et al. (2017) The brain parenchyma has a type I interferon response that can limit virus spread. Proc Natl Acad Sci U S A 114:E95-E104
Shi, Yang; Yamada, Kaoru; Liddelow, Shane Antony et al. (2017) ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature 549:523-527
Zrzavy, Tobias; Hametner, Simon; Wimmer, Isabella et al. (2017) Loss of 'homeostatic' microglia and patterns of their activation in active multiple sclerosis. Brain 140:1900-1913
Mazaheri, Fargol; Snaidero, Nicolas; Kleinberger, Gernot et al. (2017) TREM2 deficiency impairs chemotaxis and microglial responses to neuronal injury. EMBO Rep 18:1186-1198
Colonna, Marco; Butovsky, Oleg (2017) Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu Rev Immunol 35:441-468
Krasemann, Susanne; Madore, Charlotte; Cialic, Ron et al. (2017) The TREM2-APOE Pathway Drives the Transcriptional Phenotype of Dysfunctional Microglia in Neurodegenerative Diseases. Immunity 47:566-581.e9
Makker, Preet G S; Duffy, Samuel S; Lees, Justin G et al. (2017) Characterisation of Immune and Neuroinflammatory Changes Associated with Chemotherapy-Induced Peripheral Neuropathy. PLoS One 12:e0170814
Delpech, Jean-Christophe; Wei, Lan; Hao, Jin et al. (2016) Early life stress perturbs the maturation of microglia in the developing hippocampus. Brain Behav Immun 57:79-93
Bisht, Kanchan; Sharma, Kaushik P; Lecours, Cynthia et al. (2016) Dark microglia: A new phenotype predominantly associated with pathological states. Glia 64:826-39

Showing the most recent 10 out of 18 publications