Activation of sphingosine-1-phosphate receptor subtype 1 in the central nervous system contributes to morphine-induced hyperalgesia and antinociceptive tolerance in rodents.

TitleActivation of sphingosine-1-phosphate receptor subtype 1 in the central nervous system contributes to morphine-induced hyperalgesia and antinociceptive tolerance in rodents.
Publication TypeJournal Article
Year of Publication2020
AuthorsDoyle TM, Janes K, Chen Z, Grace PM, Esposito E, Cuzzocrea S, Largent-Milnes TM, Neumann WL, Watkins LR, Spiegel S, Vanderah TW, Salvemini D
JournalPain
Volume161
Issue9
Pagination2107-2118
Date Published2020 09 01
ISSN1872-6623
KeywordsAnalgesics, Animals, Female, Hyperalgesia, Male, Mice, Morphine, Rodentia, Sphingosine-1-Phosphate Receptors
Abstract

ABSTRACT: Morphine-induced alterations in sphingolipid metabolism in the spinal cord and increased formation of the bioactive sphingolipid metabolite sphingosine-1-phosphate (S1P) have been implicated in the development of morphine-induced hyperalgesia (OIH; increased pain sensitivity) and antinociceptive tolerance. These adverse effects hamper opioid use for treating chronic pain and contribute to dependence and abuse. S1P produces distinct effects through 5 G-protein-coupled receptors (S1PR1-5) and several intracellular targets. How S1P exerts its effects in response to morphine remains unknown. Here, we report that S1P contributes to the development of morphine-induced hyperalgesia and tolerance through S1P receptor subtype 1 (S1PR1) signaling in uninjured male and female rodents, which can be blocked by targeting S1PR1 with S1PR1 antagonists or RNA silencing. In mouse neuropathic pain models, S1PR1 antagonists blocked the development of tolerance to the antiallodynic effects of morphine without altering morphine pharmacokinetics and prevented prolonged morphine-induced neuropathic pain. Targeting S1PR1 reduced morphine-induced neuroinflammatory events in the dorsal horn of the spinal cord: increased glial marker expression, mitogen-activated protein kinase p38 and nuclear factor κB activation, and increased inflammatory cytokine expression, such as interleukin-1β, a cytokine central in the modulation of opioid-induced neural plasticity. Our results identify S1PR1 as a critical path for S1P signaling in response to sustained morphine and reveal downstream neuroinflammatory pathways impacted by S1PR1 activation. Our data support investigating S1PR1 antagonists as a clinical approach to mitigate opioid-induced adverse effects and repurposing the functional S1PR1 antagonist FTY720, which is FDA-approved for multiple sclerosis, as an opioid adjunct.

DOI10.1097/j.pain.0000000000001888
Alternate JournalPain
PubMed ID32301840
PubMed Central IDPMC7554181
Grant ListR01 DA043543 / DA / NIDA NIH HHS / United States
R21 DA023056 / DA / NIDA NIH HHS / United States
Faculty Member Reference: 
Tally Largent-Milnes, PhD
Todd Vanderah, PhD