Please use this identifier to cite or link to this item: https://hdl.handle.net/2440/64861
Citations
Scopus Web of Science® Altmetric
?
?
Type: Journal article
Title: Evidence that intrathecal morphine-3-glucuronide may cause pain enhancement via toll-like receptor 4/MD-2 and interleukin-1beta
Author: Lewis, S.
Hutchinson, M.
Rezvani, N.
Loram, L.
Zhang, Y.
Maier, S.
Rice, K.
Watkins, L.
Citation: Neuroscience, 2010; 165(2):569-583
Publisher: Pergamon-Elsevier Science Ltd
Issue Date: 2010
ISSN: 0306-4522
1873-7544
Statement of
Responsibility: 
S. S. Lewis, M. R. Hutchinson, N. Rezvani, L. C. Loram, Y. Zhang, S. F. Maier, K. C. Rice and L. R. Watkins
Abstract: Morphine-3-glucoronide (M3G) is a major morphine metabolite detected in cerebrospinal fluid of humans receiving systemic morphine. M3G has little-to-no affinity for opioid receptors and induces pain by unknown mechanisms. The pain-enhancing effects of M3G have been proposed to significantly and progressively oppose morphine analgesia as metabolism ensues. We have recently documented that morphine activates toll-like receptor 4 (TLR4), beyond its classical actions on μ-opioid receptors. This suggests that M3G may similarly activate TLR4. This activation could provide a novel mechanism for M3G-mediated pain enhancement, as (a) TLR4 is predominantly expressed by microglia in spinal cord and (b) TLR4 activation releases pain-enhancing substances, including interleukin-1 (IL-1). We present in vitro evidence that M3G activates TLR4, an effect blocked by TLR4 inhibitors, and that M3G activates microglia to produce IL-1. In vivo, intrathecal M3G (0.75 μg) induced potent allodynia and hyperalgesia, blocked or reversed by interleukin-1 receptor antagonist, minocycline (microglial inhibitor), and (+)-and (−)-naloxone. This latter study extends our prior demonstrations that TLR4 signaling is inhibited by naloxone nonstereoselectively. These results with (+)-and (−)-naloxone also demonstrate that the effects cannot be accounted for by actions at classical, stereoselective opioid receptors. Hyperalgesia (allodynia was not tested) and in vitro M3G-induced TLR4 signaling were both blocked by 17-DMAG, an inhibitor of heat shock protein 90 (HSP90) that can contribute to TLR4 signaling. Providing further evidence of proinflammatory activation, M3G upregulated TLR4 and CD11b (microglial/macrophage activation marker) mRNAs in dorsal spinal cord as well as IL-1 protein in the lumbosacral cerebrospinal fluid. Finally, in silico and in vivo data support that the glucuronic acid moiety is capable of inducing TLR4/MD-2 activation and enhanced pain. These data provide the first evidence for a TLR4 and IL-1 mediated component to M3G-induced effects, likely of at least microglial origin.
Keywords: Minocycline
17-DMAG
(+)-naloxone
nonstereoselective
allodynia
hyperalgesia
Rights: © 2010 IBRO.
DOI: 10.1016/j.neuroscience.2009.10.011
Grant ID: http://purl.org/au-research/grants/nhmrc/465423
Published version: http://dx.doi.org/10.1016/j.neuroscience.2009.10.011
Appears in Collections:Aurora harvest
Pharmacology publications

Files in This Item:
There are no files associated with this item.


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