Females are more to susceptible many autoimmune diseases. In multiple sclerosis (MS), not only is there a 3-4 fold increase in disease incidence, but there are sex-determined differences in the average age of onset and clinical course. Yet the cellular and molecular underpinnings of this sex-dimorphism have remained undefined. The SJL mouse model of MS recapitulates these differences in that female mice exhibit higher incidence, more severe disease, and a more consistent relapsing-remitting pattern than their male counterparts. This difference in disease susceptibility corresponds to qualitatively distinct anti-myelin Th cell cytokine responses. Whereas females generate pro-inflammatory Th1/Th17-dominant responses, the response in males is Th2-skewed and non-pathogenic. In this application we provide evidence that type 2 innate lymphoid cells (ILC2s) exert a male-specific protective influence. Best studied in allergic airway models, ILC2s are c-kit+ and are essential for inducing Th2 immunity through production of IL-13. We propose that mast cell activation in immunized in male mice elicits production of ILC2 activating factors such as IL-33 that promote ILC2 functionality. The inability to generate a robust IL-33 response in females leads to a functional deficit in ILC2 activity. Mast cells (c-kit+ Fc?R1+) are one important source of IL-33 in vivo and testosterone directly induces Il33 exclusively in male-derived cells, despite equivalent androgen receptor expression by female-derived mast cells. These data suggest a cellular and molecular target of testosterone and identify a potential mechanism of action for testosterone-mediated protection in CNS autoimmune disease.
Specific Aim 1 : Determine the IL-33 expression kinetics, cellular source (s) and its requirement for protection in immunized male mice. Using IL-33- reporter mice (Il33Cit/+) or IL-33-deficient mice (Il33Cit/Cit (Il33-deficient) these experiments will define the IL-33-expressing cells, when and where it is produced.
Specific Aim 2 : Determine how testosterone influences the expression of IL-33 and other factors that regulate sex-dimorphic EAE protection. IL-33 gene expression will be evaluated in mice treated with testosterone or androgen receptor (AR) antagonists. We will ask if testosterone induces epigenetic changes at the Il33 locus conferring changes in chromatin accessibility. Other AR target genes will also be examined.
Specific Aim 3 : Explore the contributions of sex determining genes on ILC2 and mast cell function in EAE. The potential effect of sex chromosomes independent of hormonal influences on ILC2 and mast cell gene expression and function will be examined using four core genotype mice.

Public Health Relevance

Women are 3-4 times more likely to develop autoimmune disease but the basis of this sex-bias is not known. Our studies using a mouse model of multiple sclerosis, an autoimmune disease that results from immune destruction of nerve insulating structures, show differences in the activity of Group 2 innate lymphoid cells (ILC2s), cells that suppress immune destruction in males but not females. This application will determine the basis of this sex-determined action of ILC2s.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI128292-02
Application #
9522165
Study Section
Hypersensitivity, Autoimmune, and Immune-mediated Diseases Study Section (HAI)
Program Officer
Esch, Thomas R
Project Start
2017-07-14
Project End
2022-06-30
Budget Start
2018-07-01
Budget End
2019-06-30
Support Year
2
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Northwestern University at Chicago
Department
Microbiology/Immun/Virology
Type
Schools of Medicine
DUNS #
005436803
City
Chicago
State
IL
Country
United States
Zip Code
60611