(S)-lacosamide inhibition of CRMP2 phosphorylation reduces postoperative and neuropathic pain behaviors through distinct classes of sensory neurons identified by constellation pharmacology

Authors
Moutal, AubinChew, Lindsey A.Yang, XiaofangWang, YueYeon, Seul KiTelemi, EdwinMeroueh, SeeneenPark, Ki DukShrinivasan, RaghuramanGilbraith, Kerry B.Qu, ChaolingXie, Jennifer Y.Patwardhan, AmolVanderah, Todd W.Khanna, MayPorreca, FrankKhanna, Rajesh
Issue Date
2016-07
Publisher
LIPPINCOTT WILLIAMS & WILKINS
Citation
PAIN, v.157, no.7, pp.1448 - 1463
Abstract
Chronic pain affects the life of millions of people. Current treatments have deleterious side effects. We have advanced a strategy for targeting protein interactions which regulate the N-type voltage-gated calcium (CaV2.2) channel as an alternative to direct channel block. Peptides uncoupling CaV2.2 interactions with the axonal collapsin response mediator protein 2 (CRMP2) were antinociceptive without effects on memory, depression, and reward/addiction. A search for small molecules that could recapitulate uncoupling of the CaV2.2-CRMP2 interaction identified (S)-lacosamide [(S)-LCM], the inactive enantiomer of the Food and Drug Administration-approved antiepileptic drug (R)-lacosamide [(R)-LCM, Vimpat]. We show that (S)-LCM, but not (R)-LCM, inhibits CRMP2 phosphorylation by cyclin dependent kinase 5, a step necessary for driving CaV2.2 activity, in sensory neurons. (S)-lacosamide inhibited depolarization-induced Ca2+ influx with a low micromolar IC50. Voltage-clamp electrophysiology experiments demonstrated a commensurate reduction in Ca2+ currents in sensory neurons after an acute application of (S)-LCM. Using constellation pharmacology, a recently described high content phenotypic screening platform for functional fingerprinting of neurons that uses subtype-selective pharmacological agents to elucidate cell-specific combinations (constellations) of key signaling proteins that define specific cell types, we investigated if (S)-LCM preferentially acts on certain types of neurons. (S)-lacosamide decreased the dorsal root ganglion neurons responding to mustard oil, and increased the number of cells responding to menthol. Finally, (S)-LCM reversed thermal hypersensitivity and mechanical allodynia in a model of postoperative pain, and 2 models of neuropathic pain. Thus, using (S)-LCM to inhibit CRMP2 phosphorylation is a novel and efficient strategy to treat pain, which works by targeting specific sensory neuron populations.
Keywords
RESPONSE MEDIATOR PROTEIN-2; GATED CALCIUM-CHANNELS; ANTIEPILEPTIC DRUG LACOSAMIDE; SPARED NERVE INJURY; SPINAL-CORD-INJURY; INTRATHECAL ZICONOTIDE; RAT MODEL; SODIUM-CHANNELS; MECHANICAL HYPERSENSITIVITY; PERIPHERAL NEUROPATHY; RESPONSE MEDIATOR PROTEIN-2; GATED CALCIUM-CHANNELS; ANTIEPILEPTIC DRUG LACOSAMIDE; SPARED NERVE INJURY; SPINAL-CORD-INJURY; INTRATHECAL ZICONOTIDE; RAT MODEL; SODIUM-CHANNELS; MECHANICAL HYPERSENSITIVITY; PERIPHERAL NEUROPATHY; CaV2.2; CRMP2; (S)-lacosamide; Constellation pharmacology; Calcium imaging; Postoperative pain; Neuropathic pain
ISSN
0304-3959
URI
https://pubs.kist.re.kr/handle/201004/123896
DOI
10.1097/j.pain.0000000000000555
Appears in Collections:
KIST Article > 2016
Files in This Item:
There are no files associated with this item.
Export
RIS (EndNote)
XLS (Excel)
XML

qrcode

Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.

BROWSE