Raloxifene suppresses experimental autoimmune encephalomyelitis and NF-κB-dependent CCL20 expression in reactive astrocytes

PLoS One. 2014 Apr 10;9(4):e94320. doi: 10.1371/journal.pone.0094320. eCollection 2014.

Abstract

Recent clinical data have led to the consideration of sexual steroids as new potential therapeutic tools for multiple sclerosis. Selective estrogen receptor modulators can exhibit neuroprotective effects like estrogen, with fewer systemic estrogen side effects than estrogen, offering a more promising therapeutic modality for multiple sclerosis. The important role of astrocytes in a proinflammatory effect mediated by CCL20 signaling on inflammatory cells has been documented. Their potential contribution to selective estrogen receptor modulator-mediated protection is still unknown. Using a mouse model of chronic neuroinflammation, we report that raloxifene, a selective estrogen receptor modulator, alleviated experimental autoimmune encephalomyelitis-an animal model of multiple sclerosis-and decreased astrocytic production of CCL20. Enzyme-linked immunosorbent assay, immunohistochemistry imaging and transwell migration assays revealed that reactive astrocytes express CCL20, which promotes Th17 cell migration. In cultured rodent astrocytes, raloxifene inhibited IL-1β-induced CCL20 expression and chemotaxis ability for Th17 migration, whereas the estrogen receptor antagonist ICI 182,780 blocked this effect. Western blotting further indicated that raloxifene suppresses IL-1β-induced NF-κB activation (phosphorylation of p65) and translocation but does not affect phosphorylation of IκB. In conclusion, these data demonstrate that raloxifene provides robust neuroprotection against experimental autoimmune encephalomyelitis, partially via an inhibitory action on CCL20 expression and NF-κB pathways in reactive astrocytes. Our results contribute to a better understanding of the critical roles of raloxifene in treating experimental autoimmune encephalomyelitis and uncover reactive astrocytes as a new target for the inhibitory action of estrogen receptors on chemokine CCL20 expression.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Astrocytes / drug effects*
  • Astrocytes / pathology
  • Cell Movement / drug effects
  • Cells, Cultured
  • Chemokine CCL20 / antagonists & inhibitors
  • Chemokine CCL20 / genetics*
  • Chemokine CCL20 / immunology
  • Disease Models, Animal
  • Encephalomyelitis, Autoimmune, Experimental / drug therapy*
  • Encephalomyelitis, Autoimmune, Experimental / genetics
  • Encephalomyelitis, Autoimmune, Experimental / pathology
  • Estradiol / analogs & derivatives
  • Estradiol / pharmacology
  • Female
  • Fulvestrant
  • Gene Expression Regulation
  • Mice
  • Mice, Inbred C57BL
  • Multiple Sclerosis / drug therapy
  • Multiple Sclerosis / genetics
  • Multiple Sclerosis / pathology
  • NF-kappa B / genetics
  • NF-kappa B / metabolism
  • Raloxifene Hydrochloride / pharmacology*
  • Receptors, Estrogen / antagonists & inhibitors
  • Receptors, Estrogen / genetics
  • Receptors, Estrogen / immunology
  • Selective Estrogen Receptor Modulators / pharmacology*
  • Signal Transduction
  • Th17 Cells / drug effects
  • Th17 Cells / pathology

Substances

  • CCL20 protein, mouse
  • Chemokine CCL20
  • NF-kappa B
  • Receptors, Estrogen
  • Selective Estrogen Receptor Modulators
  • Fulvestrant
  • Raloxifene Hydrochloride
  • Estradiol

Grants and funding

This study was supported by grants from the National Science Foundation of China (No. 81171126). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.