In patients with multiple sclerosis (MS), there is a distinct gender bias, with approximately twice as many affected females as males. Sex hormones may contribute to susceptibility or resistance to MS by influencing development and function of potentially pathogenic T cells specific for central nervous system (CNS) antigens, as well as regulatory T cells that might modify the course of disease. Previously, we reported that low doses of 17beta-estradiol (E2) can reduce severity of EAE by inhibiting activation, cytokine and chemokine production, and encephalitogenicity of murine T cells specific for myelin oligodendrocyte glycoprotein (MOG), proteolipid protein (PLP), or myelin basic protein (MBP), and by inhibiting I recruitment of inflammatory cells into the CNS. Of particular importance, estrogen treatment profoundly reduced intracellular production of TNF-alpha, known as a critical inflammatory cytokine in EAE and MS. Recent evaluation of E2 effects on EAE using microarray analysis of splenocyte gene expression confirmed potent inhibition of TNF-alpha, and further identified several previously unsuspected immune-associated candidate genes appearing to be strongly affected by E2 treatment in vivo. These modulatory effects of estrogen on pathogenic, recruited, and regulatory cells in EAE are likely mediated y direct interaction with estrogen receptors (ER), which include the classical ER-alpha and ER-beta that are internal f receptors (iER), and possibly membrane ERs (mER) that may be distinct from iERs. This proposal will test the hypothesis that modulation of EAE by estrogen involves receptor-mediated regulation of TNF-alpha and several other novel immunerelated genes, thereby inhibiting inflammatory effects of macrophages, dendritic cells and T cells and enhancing regulatory NKT cell activity. Our primary goals are to determine if the inhibitory effects of E2 on EAE can be observed in spontaneous EAE in the absence of regulatory T and NKT cells, and if inhibitory effects of E2 on EAE are mediated through iER-alpha and /or iERbeta, or in contrast, by neither of the classical iERs. Using newly developed ER-alpha and ER-beta knockout mice we will for the first time associate E2- dependent regulation with either or both iERs, or alternatively with mER if E2 effects persist in double KO mice. Moreover, for each novel E2-affected gene implicated from the microarray analysis, we will investigate the cellular source, effects of E2 on transcription and protein production, and contribution to EAE induction in vivo. Genes found to be important in EAE will be further evaluated in blood cells from women with different levels of E2. The work proposed will identify key estrogen-sensitive genes, including TNFalpha, contributing to EAE induction and resistance. Changes in these genes can be followed during estrogen treatment as surrogate markers to verify effective doses of estrogen. From this research, we will develop a solid basis for using estrogen replacement therapy for MS.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
1R01NS045445-01
Application #
6487971
Study Section
Special Emphasis Panel (ZAI1-NN-I (J2))
Program Officer
Utz, Ursula
Project Start
2002-08-15
Project End
2006-07-31
Budget Start
2002-08-15
Budget End
2003-07-31
Support Year
1
Fiscal Year
2002
Total Cost
$247,479
Indirect Cost
Name
Oregon Health and Science University
Department
Neurology
Type
Schools of Medicine
DUNS #
009584210
City
Portland
State
OR
Country
United States
Zip Code
97239
Bodhankar, Sheetal; Vandenbark, Arthur A; Offner, Halina (2012) Oestrogen treatment of experimental autoimmune encephalomyelitis requires 17ýý-oestradiol-receptor-positive B cells that up-regulate PD-1 on CD4+ Foxp3+ regulatory T cells. Immunology 137:282-93
Bodhankar, Sheetal; Offner, Halina (2011) GPR30 FORMS AN INTEGRAL PART OF E2-PROTECTIVE PATHWAY IN EXPERIMENTAL AUTOIMMUNE ENCEPHALOMYELITIS. Immunol Endocr Metab Agents Med Chem 11:262-274
Bodhankar, Sheetal; Wang, Chunhe; Vandenbark, Arthur A et al. (2011) Estrogen-induced protection against experimental autoimmune encephalomyelitis is abrogated in the absence of B cells. Eur J Immunol 41:1165-75
Jones, Richard E; Kaler, Laurie; Murphy, Stephanie et al. (2010) Tissue-Dependent Expression of Estrogen Receptor ? in 17?-Estradiol-Mediated Attenuation of Autoimmune CNS Inflammation. Open Autoimmun J 2:197-204
Yates, M A; Li, Y; Chlebeck, P et al. (2010) Progesterone treatment reduces disease severity and increases IL-10 in experimental autoimmune encephalomyelitis. J Neuroimmunol 220:136-9
Subramanian, Sandhya; Ayala, Patricia; Wadsworth, Teri L et al. (2010) CCR6: a biomarker for Alzheimer's-like disease in a triple transgenic mouse model. J Alzheimers Dis 22:619-29
Wang, Chunhe; Li, Yuexin; Proctor, Thomas M et al. (2010) Down-modulation of programmed death 1 alters regulatory T cells and promotes experimental autoimmune encephalomyelitis. J Neurosci Res 88:7-15
Yates, Melissa A; Li, Yuexin; Chlebeck, Peter J et al. (2010) GPR30, but not estrogen receptor-alpha, is crucial in the treatment of experimental autoimmune encephalomyelitis by oral ethinyl estradiol. BMC Immunol 11:20
Wang, Chunhe; Dehghani, Babak; Li, Yuexin et al. (2009) Membrane estrogen receptor regulates experimental autoimmune encephalomyelitis through up-regulation of programmed death 1. J Immunol 182:3294-303
Wang, Chunhe; Dehghani, Babak; Li, Yuexin et al. (2009) Oestrogen modulates experimental autoimmune encephalomyelitis and interleukin-17 production via programmed death 1. Immunology 126:329-35

Showing the most recent 10 out of 19 publications