Abstract

BackgroundFTY720 (fingolimod, Gilenya™) is a daily oral therapy for multiple sclerosis that readily accesses the central nervous system (CNS). FTY720 is a structural analog to the sphingolipid sphingosine-1-phosphate (S1P) and is a cognate ligand for the S1P G-protein coupled receptors (S1PR). Studies in experimental autoimmune encephalomyelitis using mice with conditionally deleted S1P1R from astrocytes indicate that one beneficial effect of FTY720 in this model is via downregulating external receptors, which inhibits responses induced by the natural ligand. Another proposed effect of FTY720 on neuroinflammation is its ability to maintain persistent signaling in cells via internalized S1P1R resulting in functional responses that include suppressing intracellular calcium release. We used human fetal astrocytes to investigate potential dual inhibitory- and function-inducing effects of daily FTY720 on responses relevant to neuroinflammation. For the inhibitory effects, we used signaling and proliferation induced by the natural ligand S1P. For the function-inducing responses, we measured inhibition of intracellular calcium release stimulated by the proinflammatory cytokine, interleukin (IL)-1β.MethodsAstrocytes derived from human fetal CNS specimens and maintained in dissociated cultures were exposed to 100 nM of the biologically active form of FTY720 over a dosing regimen that ranged from a single exposure (with or without washout after 1 h) to daily exposures up to 5 days. Responses measured include: phosphorylation of extracellular-signal-regulated kinases (pERK1/2) by Western blotting, Ki-67 immunolabeling for cell proliferation, IL-1β-induced calcium release by ratiometric fluorescence, and cytokine/chemokine (IL-6, CXCL10) secretions by ELISA.ResultsWe observed that a single addition of FTY720 inhibited subsequent S1PR ligand-induced pERK1/2 signaling for >24 h. Daily FTY720 treatments (3-5 days) maintained this effect together with a loss of proliferative responses to the natural ligand S1P. Repeated FTY720 dosing concurrently maintained a functional cell response as measured by the inhibition of intracellular calcium release when stimulated by the cytokine IL-1β. Recurrent FTY720 treatments did not inhibit serum- or IL-1β-induced pERK1/2. The secretions of IL-6 and CXCL10 in response to IL-1β were unaffected by FTY720 treatment(s).ConclusionOur results indicate that daily FTY720 exposures may regulate specific neuroinflammatory responses by desensitizing astrocytes to external S1PR stimuli while sustaining cellular influences that are independent of new surface S1PR activation.

Highlights

  • FTY720 is a daily oral therapy for multiple sclerosis that readily accesses the central nervous system (CNS)

  • We studied how FTY720 could influence neuroinflammationrelevant responses via its dual role in inhibiting surface S1P G-protein coupled receptors (S1PR) signaling and proliferation while sustaining active responses in the cells as measured by the inhibition of intracellular calcium release when stimulated by the cytokine interleukin (IL)-1β

  • In this study, we investigated the functional effects of repeated daily doses of FTY720 on human fetal astrocytes in vitro

Read more

Summary

Introduction

FTY720 (fingolimod, GilenyaTM) is a daily oral therapy for multiple sclerosis that readily accesses the central nervous system (CNS). Studies in experimental autoimmune encephalomyelitis using mice with conditionally deleted S1P1R from astrocytes indicate that one beneficial effect of FTY720 in this model is via downregulating external receptors, which inhibits responses induced by the natural ligand Another proposed effect of FTY720 on neuroinflammation is its ability to maintain persistent signaling in cells via internalized S1P1R resulting in functional responses that include suppressing intracellular calcium release. Systemic administrations of FTY720 to immunodeficient animals enhanced functional recovery following traumatic spinal cord injury [5]; of note is that the effects in both animal models implicate drug interactions with S1PR expressed by astrocytes It remains unclear, if the observed tissue protection/repair processes are the results of inhibiting astrocyte responses to the natural ligand and/or inducing cellular signaling. Zu Heringdorf et al demonstrated that activating S1P1R in stably transfected (non-neural) cell lines negatively regulates intracellular calcium (Ca2+) release and such a release could have a number of neuroinflammationrelevant consequences including mitochondrial stress, production of free radicals, and proteases/phospholipases activation [9,10]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call