Multiple Sclerosis (MS) is characterized by focal inflammatory infiltrates of the CNS, demyelinating lesions and axonal damage, leading to neurologic dysfunction. Cells of the innate and adaptive immune system are involved in MS pathogenesis. MS is also characterized by production of cytokines that activate immune cells, including IFN-?, IL-6, GM-CSF, IL-12 and IL-23. The JAK/STAT pathway is used by cytokines for signaling, and is critical for the development, regulation and termination of immune responses. In the previous funding period, we developed a model of Experimental Autoimmune Encephalomyelitis (EAE) in which the JAK/STAT pathway is aberrantly activated in myeloid cells, with functional consequences for Th1 and Th17 cell differentiation. This model presents as a non-resolving form of atypical EAE. In this atypical model with heightened activation of the JAK/STAT pathway, we demonstrate that AZD1480, an inhibitor of the JAK/STAT pathway, suppresses disease severity, prevents entry of immune cells into the CNS, suppresses differentiation of Th1 and Th17 cells, decreases STAT activation, and reduces expression of cytokines/chemokines. Furthermore, we have tested AZD1480 in four other EAE models (Classical, Relapsing-Remitting (RR), Adoptive Transfer of Th1 or Th17 cells) and observed striking clinical efficacy. Another JAK/STAT inhibitor, AZD5776, also inhibits EAE clinical symptoms. We hypothesize that inappropriate activation of the JAK/STAT pathway promotes aberrant responses of cells of the innate and adaptive immune systems, leading to the neuroinflammation seen in MS, and that therapeutic intervention of this pathway will alter the progression of MS. In this revised, competing renewal, we will evaluate two distinct JAK inhibitors, AZD1480 and AZD5776, in five EAE models. AZD1480 is a JAK1 and JAK2 inhibitor, and AZD5776 inhibits JAK1.
In Aim 1, the therapeutic efficacy of targeting the JAK/STAT pathway in active EAE (classical, atypical, RR) will be examined. We will evaluate 1) which JAKs/STATs are involved in disease pathogenesis; 2) in which cell type(s) (CD4+ T-cells, macrophages, microglia, DCs) activation of the JAK/STAT pathway is detrimental; and 3) the molecular basis for the therapeutic benefit of the JAK inhibitors.
In Aim 2, the role of the JAK/STAT pathway in regulating the pathogenic potential of Th1 and Th17 cells will be tested. Both Th1 and Th17 cells have the capacity to induce CNS autoimmunity, but have differential pathogenic mechanisms. Our preliminary results indicate that AZD1480 is effective in inhibiting both Th1-induced and Th17-induced EAE. We will assess how in vivo treatment with AZD1480/AZD5776 in the context of MOG-immunization regulates the developmental program and/or pathogenicity of Th1 and Th17 cells.
In Aim 3, the mechanism by which the JAK inhibitors AZD1480 and AZD5776 regulate immune cell function will be examined. Macrophages and DCs obtained from naive mice treated with JAK inhibitors will be evaluated for their antigen presentation capability, polarization into proinflammatory or anti-inflammatory phenotypes, and potential neuroprotective properties. CD4+ T-cells derived from JAK inhibitor treated mice will be examined for their subsequent ability to differentiate into pathogenic and/or protective T-cell subsets. These studies will establish the pre-clinical foundation necessary to ultimately test inhibitors of the JAK/STAT pathway in human MS.

Public Health Relevance

Our preliminary results indicate that intervention of the JAK/STAT pathway with JAK inhibitors has therapeutic benefit in five different EAE models; this is the first documentation that suppression of this pathway attenuates EAE pathogenesis. The studies proposed in this revised, competitive renewal will provide the first mechanistic data on how JAK inhibitors regulate cells of the innate and adaptive immune systems, how this impacts subsequent neuroinflammatory responses, and how this ultimately ameliorates EAE disease. Collectively, these pre-clinical translational studies in models of MS will inform us of the validiy of targeting the JAK/STAT pathway for abrogation of neuroinflammatory responses, demyelination and axonal damage, which will lead to the development and utilization of rational, new therapeutics for MS patients.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
5R01NS057563-07
Application #
8918028
Study Section
Clinical Neuroimmunology and Brain Tumors Study Section (CNBT)
Program Officer
Utz, Ursula
Project Start
2006-12-01
Project End
2016-08-31
Budget Start
2015-09-01
Budget End
2016-08-31
Support Year
7
Fiscal Year
2015
Total Cost
Indirect Cost
Name
University of Alabama Birmingham
Department
Anatomy/Cell Biology
Type
Schools of Medicine
DUNS #
063690705
City
Birmingham
State
AL
Country
United States
Zip Code
35294
Gibson, Sara A; Yang, Wei; Yan, Zhaoqi et al. (2018) CK2 Controls Th17 and Regulatory T Cell Differentiation Through Inhibition of FoxO1. J Immunol 201:383-392
Yan, Zhaoqi; Gibson, Sara A; Buckley, Jessica A et al. (2018) Role of the JAK/STAT signaling pathway in regulation of innate immunity in neuroinflammatory diseases. Clin Immunol 189:4-13
Gibson, Sara A; Benveniste, Etty N (2018) Protein Kinase CK2: An Emerging Regulator of Immunity. Trends Immunol 39:82-85
Gibson, Sara A; Yang, Wei; Yan, Zhaoqi et al. (2017) Protein Kinase CK2 Controls the Fate between Th17 Cell and Regulatory T Cell Differentiation. J Immunol 198:4244-4254
Qin, Hongwei; Buckley, Jessica A; Li, Xinru et al. (2016) Inhibition of the JAK/STAT Pathway Protects Against ?-Synuclein-Induced Neuroinflammation and Dopaminergic Neurodegeneration. J Neurosci 36:5144-59
Guthrie, Lauren N; Abiraman, Kavitha; Plyler, Emily S et al. (2016) Attenuation of PKR-like ER Kinase (PERK) Signaling Selectively Controls Endoplasmic Reticulum Stress-induced Inflammation Without Compromising Immunological Responses. J Biol Chem 291:15830-40
Park, Keun Woo; Lin, Ching-Yi; Benveniste, Etty N et al. (2016) Mitochondrial STAT3 is negatively regulated by SOCS3 and upregulated after spinal cord injury. Exp Neurol 284:98-105
Zhao, Jiping; Yu, Hao; Liu, Yudong et al. (2016) Protective effect of suppressing STAT3 activity in LPS-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 311:L868-L880
McFarland, Braden C; Marks, Margaret P; Rowse, Amber L et al. (2016) Loss of SOCS3 in myeloid cells prolongs survival in a syngeneic model of glioma. Oncotarget 7:20621-35
Rajbhandari, Rajani; McFarland, Braden C; Patel, Ashish et al. (2015) Loss of tumor suppressive microRNA-31 enhances TRADD/NF-?B signaling in glioblastoma. Oncotarget 6:17805-16

Showing the most recent 10 out of 38 publications