ArticlePDF Available

Neuroexcitatory effects of morphine-3-glucuronide are dependent on Toll-like receptor 4 signaling

Authors:

Abstract and Figures

Background: Multiple adverse events are associated with the use of morphine for the treatment of chronic non-cancer pain, including opioid-induced hyperalgesia (OIH). Mechanisms of OIH are independent of opioid tolerance and may involve the morphine metabolite morphine-3-glucuronide (M3G). M3G exhibits limited affinity for opioid receptors and no analgesic effect. Previous reports suggest that M3G can act via the Toll-like receptor 4 (TLR4)/myeloid differentiation protein-2 (MD-2) heterodimer in the central nervous system to elicit pain. Methods: Immunoblot and immunocytochemistry methods were used to characterize the protein expression of TLR4 present in lumbar dorsal root ganglion (DRG). Using in vitro intracellular calcium and current clamp techniques, we determined whether TLR4 activation as elicited by the prototypical agonists of TLR4, lipopolysaccharide (LPS) and M3G, contributed to changes in intracellular calcium and increased excitation. Rodents were also injected with M3G to determine the degree to which M3G-induced tactile hyperalgesia could be diminished using either a small molecule inhibitor of the MD-2/TLR4 complex in rats or TLR4 knockout mice. Whole cell voltage-clamp recordings were made from small- and medium-diameter DRG neurons (25 μm < DRG diameter <45 μm) for both control and M3G-treated neurons to determine the potential influence on voltage-gated sodium channels (NaVs). Results: We observed that TLR4 immunoreactivity was present in peptidergic and non-peptidergic sensory neurons in the DRG. Non-neuronal cells in the DRG lacked evidence of TLR4 expression. Approximately 15% of assayed small- and medium-diameter sensory neurons exhibited a change in intracellular calcium following LPS administration. Both nociceptive and non-nociceptive neurons were observed to respond, and approximately 40% of these cells were capsaicin-insensitive. Increased excitability observed in sensory neurons following LPS or M3G could be eliminated using Compound 15, a small molecule inhibitor of the TLR4/MD-2 complex. Likewise, systemic injection of M3G induced rapid tactile, but not thermal, nociceptive behavioral changes in the rat, which were prevented by pre-treating animals with Compound 15. Unlike TLR4 wild-type mice, TLR4 knockout mice did not exhibit M3G-induced hyperalgesia. As abnormal pain sensitivity is often associated with NaVs, we predicted that M3G acting via the MD-2/TLR4 complex may affect the density and gating of NaVs in sensory neurons. We show that M3G increases tetrodotoxin-sensitive and tetrodotoxin-resistant (NaV1.9) current densities. Conclusions: These outcomes provide evidence that M3G may play a role in OIH via the TLR4/MD-2 heterodimer complex and biophysical properties of tetrodotoxin-sensitive and tetrodotoxin-resistant NaV currents.
Content may be subject to copyright.
This Provisional PDF corresponds to the article as it appeared upon acceptance. Fully formatted
PDF and full text (HTML) versions will be made available soon.
Neuroexcitatory effects of morphine-3-glucuronide are dependent on Toll-like
receptor 4 signaling
Journal of Neuroinflammation 2012, 9:200 doi:10.1186/1742-2094-9-200
Michael R Due (mrdue@iupui.edu})
Andrew D Piekarz (piekarza@gmail.com})
Natalie Wilson (wilsnat@gmail.com})
Polina Feldman (pfeldman@iupui.edu})
Matthew S Ripsch (mripsch@iupui.edu})
Sherry Chavez (chavez.sherry@gmail.com})
Hang Yin (Hubert.Yin@colorado.edu})
Rajesh Khanna (khanna5@iupui.edu})
Fletcher A White (fawhite@iupui.edu})
ISSN 1742-2094
Article type Research
Submission date 6 June 2012
Acceptance date 31 July 2012
Publication date 16 August 2012
Article URL http://www.jneuroinflammation.com/content/9/1/200
This peer-reviewed article can be downloaded, printed and distributed freely for any purposes (see
copyright notice below).
Articles in JNI are listed in PubMed and archived at PubMed Central.
For information about publishing your research in JNI or any BioMed Central journal, go to
http://www.jneuroinflammation.com/authors/instructions/
For information about other BioMed Central publications go to
http://www.biomedcentral.com/
Journal of Neuroinflammation
© 2012 Due et al. ; licensee BioMed Central Ltd.
This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0),
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Neuroexcitatory effects of morphine-3-glucuronide
are dependent on Toll-like receptor 4 signaling
Michael R Due1,†
Email: mrdue@iupui.edu
Andrew D Piekarz2,†
Email: piekarza@gmail.com
Natalie Wilson1
Email: wilsnat@gmail.com
Polina Feldman3
Email: pfeldman@iupui.edu
Matthew S Ripsch1
Email: mripsch@iupui.edu
Sherry Chavez4
Email: chavez.sherry@gmail.com
Hang Yin4
Email: Hubert.Yin@colorado.edu
Rajesh Khanna2
Email: khanna5@iupui.edu
Fletcher A White1,5,*
Email: fawhite@iupui.edu
1 Department of Anesthesia, Indiana University School of Medicine, 950 Walnut
St, R2, Indianapolis, IN 46202, USA
2 Department of Pharmacology, Indiana University School of Medicine, 950
Walnut St, R2, Indianapolis, IN 46202, USA
3 Medical Neuroscience Program, Indiana University School of Medicine, 950
Walnut St, R2, Indianapolis, IN 46202, USA
4 Department of Chemistry and Biochemistry, University of Colorado at Boulder,
JSCBB A224, Boulder, CO 80302, USA
5 Department of Anesthesia, Stark Neurosciences Research Institute, Indiana
University School of Medicine, 950 Walnut St, R2, Rm 427, Indianapolis, IN
46202, USA
* Corresponding author. Department of Anesthesia, Stark Neurosciences Research
Institute, Indiana University School of Medicine, 950 Walnut St, R2, Rm 427,
Indianapolis, IN 46202, USA
Equal contributors.
Abstract
Background
Multiple adverse events are associated with the use of morphine for the treatment of chronic
non-cancer pain, including opioid-induced hyperalgesia (OIH). Mechanisms of OIH are
independent of opioid tolerance and may involve the morphine metabolite morphine-3-
glucuronide (M3G). M3G exhibits limited affinity for opioid receptors and no analgesic
effect. Previous reports suggest that M3G can act via the Toll-like receptor 4 (TLR4)/myeloid
differentiation protein-2 (MD-2) heterodimer in the central nervous system to elicit pain.
Methods
Immunoblot and immunocytochemistry methods were used to characterize the protein
expression of TLR4 present in lumbar dorsal root ganglion (DRG). Using in vitro
intracellular calcium and current clamp techniques, we determined whether TLR4 activation
as elicited by the prototypical agonists of TLR4, lipopolysaccharide (LPS) and M3G,
contributed to changes in intracellular calcium and increased excitation. Rodents were also
injected with M3G to determine the degree to which M3G-induced tactile hyperalgesia could
be diminished using either a small molecule inhibitor of the MD-2/TLR4 complex in rats or
TLR4 knockout mice. Whole cell voltage-clamp recordings were made from small- and
medium-diameter DRG neurons (25 μm<DRG diameter <45 μm) for both control and M3G-
treated neurons to determine the potential influence on voltage-gated sodium channels
(NaVs).
Results
We observed that TLR4 immunoreactivity was present in peptidergic and non-peptidergic
sensory neurons in the DRG. Non-neuronal cells in the DRG lacked evidence of TLR4
expression. Approximately 15% of assayed small- and medium-diameter sensory neurons
exhibited a change in intracellular calcium following LPS administration. Both nociceptive
and non-nociceptive neurons were observed to respond, and approximately 40% of these cells
were capsaicin-insensitive. Increased excitability observed in sensory neurons following LPS
or M3G could be eliminated using Compound 15, a small molecule inhibitor of the
TLR4/MD-2 complex. Likewise, systemic injection of M3G induced rapid tactile, but not
thermal, nociceptive behavioral changes in the rat, which were prevented by pre-treating
animals with Compound 15. Unlike TLR4 wild-type mice, TLR4 knockout mice did not
exhibit M3G-induced hyperalgesia. As abnormal pain sensitivity is often associated with
NaVs, we predicted that M3G acting via the MD-2/TLR4 complex may affect the density and
gating of NaVs in sensory neurons. We show that M3G increases tetrodotoxin-sensitive and
tetrodotoxin-resistant (NaV1.9) current densities.
Conclusions
These outcomes provide evidence that M3G may play a role in OIH via the TLR4/MD-2
heterodimer complex and biophysical properties of tetrodotoxin-sensitive and tetrodotoxin-
resistant NaV currents.
Introduction
Opioids such as morphine have been and continue to be used for the treatment of chronic
pain. Adverse events such as opioid-induced hyperalgesia (OIH) often complicate the clinical
course of pain treatment and may be due to the morphine metabolite morphine-3-glucuronide
(M3G). Morphine is metabolized primarily in the mammalian liver into two metabolites,
M3G and morphine-6-glucouronide (M6G) via glucuronidation [1,2]. Approximately 44 to
55% of morphine is converted to M3G and 9 to 15% to M6G [3,4]. M6G is thought to be
responsible for much of the pain-relieving effects of morphine by acting on opioid receptors
[2,5]. Due to low affinity for opioid receptors, M3G has effectively no analgesic effect [6,7].
However, administration of M3G in rodents elicits myoclonus and stimulus-dependent
hyperalgesia [8,9].
The mechanism or receptor by which M3G induces these excitatory effects is unknown.
However, actions of M3G in the nervous system are thought to occur through the Toll-like
receptor 4 (TLR4) and the accessory secreted glycoprotein myeloid differentiation protein-2
(MD-2) complex to elicit proinflammatory glial activation [9,10]. More importantly, TLR4
knockout mice exhibit a three-fold leftward shift in the systemic dose of morphine necessary
for analgesia when compared with wild-type mice [10]. Further evidence to support M3G as
a novel TLR4 agonist is needed - particularly evidence of a direct effect on states of neuronal
excitation and/or nociceptive behavior.
Agonist-induced activation of the TLR4/MD-2 complex is not limited to cells of the immune
system or glial cells since the prototypical ligand of TLR4/MD-2 complex,
lipopolysaccharide (LPS), can elicit the release of the neuropeptide calcitonin gene-related
peptide (CGRP) from cultured dorsal root ganglion (DRG) and trigeminal root ganglion
(TRG) sensory neurons and sensitize the transient receptor potential cation channel subfamily
V member 1 (TRPV1) [11,12]. Neuronal release of CGRP is likely due to a combination of
LPS-evoked TRPV1 sensitization and inward ion current changes; however, it is unknown
whether the novel TLR4 agonist M3G has direct effects on states of neuronal excitation
and/or nociceptive behavior.
The aim of our investigation was to test whether M3G administration increases sensory
neuron excitation and alters nociceptive behavior in rodents. The M3G-dependent
depolarization of sensory neurons would imply that there may be a number of ion channels
downstream of TLR4 activation which may contribute to changes in nociceptive behavior.
Using current clamp and whole-cell voltage-clamp recordings, we found that M3G increased
excitability in small- and medium-sized acutely dissociated sensory neurons and enhanced
tetrodotoxin-sensitive (TTX-S) and tetrodotoxin-resistant (TTX-R) voltage-gated sodium
channel (NaV) densities in sensory neurons. We also demonstrated that M3G acting through
the neuronal TLR4/MD-2 complex is likely responsible for rapid induction of tactile, but not
thermal, nociceptive behavior following systemic M3G administration.
Materials and methods
Animals
Pathogen-free, adult female SpragueDawley rats (150 to 200 g; Harlan Laboratories,
Madison, WI, USA) and adult male C57BL/10ScNJ and C57BL/6 mice (20 to 25 g; Jackson
Laboratory, Bar Harbor, ME, USA) were used for all experiments. C57BL/10ScNJ mice
exhibit a homozygous deletion of 74 kb at the Tlr4 locus. Mice and rats were housed in
temperature (23±3 °C) and light (12 hour:12 hour light:dark cycle; lights on at 07.00)
controlled rooms with standard rodent chow and water available ad libitum. These
experiments were approved by the IACUC of Indiana University/Purdue University in
Indianapolis. All procedures were conducted in accordance with the Guide for Care and Use
of Laboratory Animals published by the National Institutes of Health and the ethical
guidelines of the International Association for the Study of Pain. Animals were randomly
assigned to treatment or control groups.
Drugs
All drugs were freshly prepared in saline on the day of the experiment and administered by
intraperitoneal (i.p.) injections. A TLR4 small molecule inhibitor (Compound 15) was
synthesized as described in detail in [13]. A stock solution of lipopolysaccharide (LPS) was
reconstituted in sterile 0.1% BSA/PBS to 5 mg/ml, and aliquots were stored at −20 °C
(Sigma-Aldrich, St Louis, MO, USA). The concentration used was 1 μg/mL. Morphine-3-β-
D-glucuronide (M3G) was supplied by NIH/NIDA Drug Supply Program and utilized at a
concentration (3 μM) that is significantly less than the dose necessary to elicit responses in
rodent central nervous system neurons [14,15].
Tissue processing and immunocytochemistry for neural tissue
Naïve rat lumbar DRG tissue was collected after animals were sacrificed and transcardially
perfused with Zamboni fixative. Primary antiserum used for immunocytochemical procedures
[16] was anti-TLR4 goat L14 extracellular monoclonal antibody (1:200 dilution; Santa Cruz
Biotechnology Inc., Santa Cruz, CA, USA), monoclonal anti-NeuN, polyclonal anti-CGRP
and IB4-FITC (Sigma-Aldrich). After incubation with primary antibodies, 14 μm thick tissue
sections were incubated in secondary antibodies (anti-goat made in horse conjugated to CY3,
Jackson ImmunoResearch, West Grove, PA, USA). Positive control immunocytochemistry
staining for TLR4 was conducted in rat spleen sections. Specific labeling of white pulp was
observed (data not shown).
Cell counts
Images were taken with an intensified charged coupled device camera (CoolSnap HQ2,
Photometrics, Tucson, AZ, USA coupled to a Nikon microscope (Nikon Eclipse Ti; Nikon
Instruments Inc., Melville, NY, USA) using Nikon Elements Software (Nikon Instruments
Inc.). TLR4 immunopositive cell counts were conducted using Image Pro Software (Media
Cybernetics, Bethesda, MD, USA). TLR4 cell counts were taken from at least eight serial
tissue sections per L5 ganglia (70 μm between sections) and combined to reach the total
percentage of neurons.
Tactile behavioral assessment
Von Frey filaments were used to test mechanical sensitivity before and after M3G and/or
Compound 15 administration. Prior to initial von Frey tactile testing, all rodents were
habituated to testing chambers for at least 2 days. Animals were tested for baseline responses
at least twice before initiation of the injection paradigm using previously published methods
[16]. Briefly, the rat was placed on a metal mesh floor and covered with a transparent plastic
dome where the animal rested quietly after an initial few minutes of exploration.
Animals were habituated to this testing apparatus for 15 minutes a day, 2 days prior to pre-
injection behavioral testing. Following acclimatization, each filament was applied to six spots
spaced across the glabrous side of the hind paw; two distinct spots for the distribution of each
nerve branch (saphenous, tibial and sural). Mechanical stimuli were applied with seven
filaments, each differing in the bending force delivered (10, 20, 40, 60, 80, 100, and 120
mN), but each fitted with a flat tip and a fixed diameter of 0.2 mm. The force equivalence of
mN to grams is 100 mN=10.197 g. The filaments were tested in order of ascending force,
with each filament delivered for 1 second in sequence from the 1st to the 6th spot alternately
from one paw to the other. The interstimulus interval was 10 to 15 seconds. A cutoff value of
120 mN was used; animals that did not respond at 120 mN were assigned that value. Stimuli
were applied randomly to left and right hind paws to determine the stimulus intensity
threshold stiffness required to elicit a paw withdrawal response.
The incidence of foot withdrawal was expressed as a percentage of six applications of each
filament as a function of force. A Hill equation was fitted to the function (Origin version 6.0,
Microcal Software Northampton, MA USA) relating the percentage of indentations eliciting a
withdrawal to the force of indentation. From this equation, the threshold force was obtained
and defined as the force corresponding to a 50% withdrawal rate. Mouse behavior was
conducted in a similar fashion using a probe fitted with a flat tip and a fixed diameter of 0.1
mm. However, mechanical stimuli were applied to only one location on the glabrous side of
the hind paw. All behavioral testing was performed by laboratory assistants who were blinded
to the experimental conditions and unfamiliar with the experimental aims.
Thermal behavioral assessment
Thermal hyperalgesia was determined by measuring foot withdrawal latency and duration of
the response to heat stimulation [16]. Each rat was placed in a box (22 x 12 x 12 cm)
containing a smooth glass floor. A heat source (UgoBasile Plantar™ Analgesia Instrument,
Trappe PA, USA) was focused on a portion of the hind paw, which is flush against the glass,
and a radiant thermal stimulus was delivered to that site. The stimulus shuts off automatically
when the hind paw moves (or after 20 seconds to prevent tissue damage). The intensity of the
heat stimulus was constant throughout all experiments. A thermal stimulus was delivered six
times to each hind paw at 5-minute intervals. The value for the response based on thermal
latency and duration of paw withdrawal was obtained by averaging five of six measurements
per animal. The baseline response for right and left hind paws were tested for 2 days prior to
initiation of the injection paradigm.
Immunoblot methodology
Fresh frozen L3-L6 DRGs and TRGs were homogenized in modified RIPA buffer with
protease/phosphatase inhibitors (USBio, Swampscott, MA, USA). Samples (40 μg/lane) were
resolved by 10% SDS-PAGE and transferred to a nitrocellulose membrane. After incubation
in 10% non-fat milk blocking solution overnight at 4 °C, the membrane was incubated with
primary antisera for 1 hour (anti-TLR4 goat M16; 1:1,000; Santa Cruz Biotechnology Inc.,
Santa Cruz, CA, USA). The membrane was reprobed with a monoclonal anti-β actin
(1:10,000; Sigma-Aldrich). Immunopositive bands were detected by enhanced
chemiluminescence (ThermoScientific, Rockford Ill, USA) using donkey anti-goat (Santa
Cruz Biotechnology Inc.) or rabbit anti-mouse (Jackson ImmunoResearch) horseradish
peroxidase-conjugated secondary antibodies.
Preparation of acutely dissociated dorsal root ganglion neurons
Lumbar DRGs were acutely dissociated using methods previously described [16]. Briefly,
L1-L6 DRGs were removed from naïve animals. The DRGs were treated with collagenase A
and collagenase D in HBSS for 20 minutes (1 mg/ml; Roche Applied Science, Indianapolis,
IN, USA), followed by treatment with papain (30 units/ml, Worthington Biochemical,
Lakewood, NJ, USA) in HBSS containing 0.5 mM EDTA and cysteine at 35 °C. The cells
were then dissociated via mechanical trituration in culture media containing 1 mg/ml BSA
and trypsin inhibitor (1 mg/ml, Sigma-Aldrich). The culture media was DMEM, Ham’s F12
mixture, supplemented with 10% fetal bovine serum, penicillin and streptomycin (100 μg/ml
and 100 U/ml) and N2 (Life Technologies, Carlsbad CA, USA). The cells were plated on
coverslips coated with poly-L lysine and laminin (1 mg/ml) and incubated for 2 to 3 hours
before additional culture media was added to the wells. The cells were then allowed to sit
undisturbed for 12 to 15 hours to adhere at 37 °C (with 5% CO2).
Intracellular calcium imaging
Acute dissociation of lumbar DRG and intracellular calcium imaging was performed using
methods previously described [16]. The dissociated DRG cells were loaded with fura-2 AM
(3 μM, Molecular Probes/Invitrogen Corporation, Carlsbad, CA, USA) for 25 minutes at
room temperature in a balanced sterile salt solution (BSS; NaCl (140 mM), Hepes (10 mM),
CaCl2 (2 mM), MgCl2 (1 mM), glucose (10 mM), KCl (5 mM)). The cells were rinsed with
the BSS and mounted onto a chamber that was placed onto the inverted microscope.
Intracellular calcium was measured by digital video microfluorometry with an intensified
CCD camera (CoolSnap HQ2, Photometrics) coupled to a Nikon microscope (Nikon Eclipse
Ti) and Nikon Elements Software. Cells were illuminated with a Lamda DG-4 175 W xenon
lamp (Sutter Instruments, Novato, CA, USA), and the excitation wavelengths of the fura-2
(340/380 nm) were selected by a filter changer (Sutter Instruments). Sterile solution was
applied to cells prior to LPS application, and any cells that responded to buffer alone were
not used in LPS responsive counts. Compounds were applied directly into the coverslip
bathing solution. LPS (1 μg/mL) was applied first, after which capsaicin (3 nM; Sigma-
Aldrich), high K+ (50 μM) and ATP (3 nM) were added. Only calcium imaging traces that
reflected at least a 50% increase over baseline were included in the analysis. All data were
analyzed by two independent analyzers and only responses that were in agreement between
the two individuals were used in the responsive cell counts.
Electrophysiology
Sharp-electrode intracellular recordings were obtained from 4 to 18 hours after acute
dissociation of lumbar DRG as previously described [17]. Coverslips were transferred to a
recording chamber that was mounted on the stage of an inverted microscope. The chamber
was perfused with 2 mL bath solution containing: NaCl, 120 mM; KCl, 3 mM; CaCl2, 1 mM;
MgCl2, 1 mM; Hepes, 10 mM; glucose, 10 mM; adjusted to pH 7.4 and osmolarity 300
mosM. The recordings were obtained at room temperature. Electrodes were filled with 1.0 M
KCl (impedance: 40 to 80 MΩ) and positioned by a micromanipulator (Newport Corporation,
Irvine, CA, USA). A −0.1 nA current injection was used to bridge-balance the electrode
resistance. Prior to electrode impalement, the size of the soma to be recorded was classified
by eye according to its diameter as small (≤30 μm), medium (31–45 μm) and large (≥45 μm).
Electrophysiological recordings were performed with continuous current-clamp in bridge
mode using an AxoClamp-2B amplifier, stored digitally via a Digidata 1322A interface, and
analyzed offline with pClamp 9 software (Axon Instruments, Union City, CA, USA). Only
neurons with resting membrane potential more negative than −45 mV were analyzed.
Neuronal excitability of small and medium, dissociated DRG sensory neurons was measured
by injecting 1-second current pulses into the soma every 30 seconds. Current was adjusted in
order to elicit two to four APs per current injection under baseline conditions. Following
three control current injections, LPS (1 μg/mL recording solution) or M3G (3 μM) was
applied to the coverslip and current injections continued every 30 seconds. Neuronal
excitability was measured as the number of APs elicited per current pulse before and after
addition of LPS or M3G. An attempt to reverse changes in excitability generated by LPS and
M3G was performed using the TLR4 inhibitor, Compound 15 [13]. Compound 15 (50 μM)
was added following two experimental current pulses in the presence of LPS or M3G in
experiments where these ligands increased neuronal excitability.
Whole cell voltage-clamp recordings were made from small- and medium-diameter DRG
neurons (25 μm<DRG diameter <45 μm) for both control and M3G treated neurons using a
HEKA EPC10 amplifier as previously described [18]. To ensure the fidelity of the voltage-
clamp during data acquisition all recordings were made using an extracellular bath solution
containing reduced sodium (70 mM). TTX-S current densities were estimated following post
hoc subtraction of the slow-inactivating TTX-R current [18]. TTX-R current densities were
measured from recordings obtained in the presence of 500 nM TTX. NaV1.8 currents were
estimated from the current elicited from a 150 ms pulse to 0 mV from a holding potential of
−100 mV and NaV1.9 currents were estimated as the current remaining during the last 10%
of a 150 ms test pulse to −60 mV from a holding potential of −100 mV.
Statistics
GraphPad Software (LaJolla, CA, USA) was used to determine the statistical significance.
The statistical significance of differences between means was determined by Student’s t-test
or a one-way analysis of variance (ANOVA) followed by post hoc, pair-wise comparisons
(Bonferroni’s method). Logistic regression was used to determine differences in the
percentages of different groups of cells.
Results
Toll-like receptor 4 present on sensory neurons
We first investigated the extent to which TLR4 protein expression is present in lumbar and
trigeminal sensory ganglia (Figure 1A). TLR4 expression present within the DRG is localized
exclusively in sensory neurons and averages 28±5.6% of total sensory neurons per DRG
assayed (1 L5 DRG per animal; n=6 rodents). TLR4-immunopositive sensory neurons
exhibit peptidergic and non-peptidergic neuronal phenotypes in the rat lumbar DRG (Figure
1B-M).
Figure 1 Toll-like receptor 4 expression in sensory neurons. (A) Western blot analysis of
Toll-like receptor 4 (TLR4) isolated from trigeminal root ganglion (TRG) and lumbar L3-L6
dorsal root ganglion (DRG) lysates. (B-M) Immunofluorescent images of TLR4 in
peptidergic and non-peptidergic DRG populations from sectioned naïve L4 or L5 DRGs.
NeuN is a neuronal marker (B, green arrowheads), which co-labeled with TLR4 (C, red
arrowheads). Nuclei are stained with DAPI (D, H, L, blue). Merged images demonstrate co-
labeling of NeuN containing neurons with TLR4 (E, yellow arrowheads). Calcitonin gene-
related peptide (CGRP) is a marker for peptidergic-containing sensory neurons (F, green
arrowheads), which co-labeled with TLR4 (G, red arrowheads). Merged images demonstrate
co-labeling of CGRP-containing neurons with TLR4 (I, yellow arrowheads). Isolectin B4
(IB4) is a marker for non-peptidergic sensory neurons (J, green arrowheads), which co-
labeled with TLR4 (K, red arrowheads). Merged images show co-labeling of IB4 non-
peptidergic sensory neurons with TLR4 (M, yellow arrowheads). Scale bar is 60 μm (B-M)
Lipopolysaccharide elicits intracellular calcium mobilization in dissociated
sensory neurons
Acute dissociation of DRG cells does not appear to alter the neuronal expression of TLR4
(Figure 2A). To functionally characterize neuronal TLR4 signaling we monitored
intracellular calcium mobilization [Ca2+]i following acute LPS administration (Figure 2B).
The resultant cellular responses were categorized into three neuronal and non-neuronal cell
types: non-capsaicin-sensitive neurons (high K and ATP responsive), capsaicin-sensitive
neurons (capsaicin, high K, and ATP responsive), and glia (ATP responsive only). A
moderate number of sensory neurons (approximately 14%) exhibited an LPS-induced [Ca2+]i
flux, of which 8.3% were capsaicin-sensitive (n=85)and 5.9% were non-capsaicin-sensitive
(n=72). Likewise, LPS-responsive non-neuronal cells were limited in number (6.8%, n=44)
(Table 1).
Figure 2 Calcium imaging of functional Toll-like receptor 4 signaling in cultured dorsal
root ganglion neurons. (A) Toll-like receptor 4 (TLR4)-IR (green) is largely restricted to
neurons and not non-neuronal cell types (blue, DAPI) in dissociated dorsal root ganglion
(DRG) cultures. (B) Representative recording of a transient intracellular calcium increase in a
dissociated DRG sensory neuron via lipopolysaccharide (LPS) administration (1 μg/mL)
Table 1 LPS response profile of acutely dissociated DRG cells from rats
Non-capsaicin-sensitive neuron
6% (5/85)
Capsaicin-sensitive neuron
8% (6/72)
Glia
7% (3/44)
Lipopolysaccharide and morphine-3-glucuronide-induced sensory neuron
excitation can be blocked by Toll-like receptor 4/myeloid differentiation
protein-2 small molecule inhibitor
LPS has been shown to elicit an inward current in trigeminal sensory neurons [12] and
enhance sensory neuron excitability in colonic nociceptive neurons [19]. Based on these
observations, agonists of TLR4 should conceivably increase the excitability of sensory
neurons in response to repeated current injection of the same amplitude. To determine the
degree to which LPS-induced intracellular calcium mobilization reflects changes in sensory
neuron excitation, we examined neuronal response using sharp electrodes in current clamp
mode. Following repeated current pulse combined with LPS administration, we observed a
significant increase in the excitability of small- to medium-diameter sensory neurons when
compared to baseline levels (15.8% neurons responded, 3.2±0.3 action potentials (APs) for
control vs. 11.7±1.7 APs for LPS, n=38) (Figure 3). Representative recordings (Figure 3A)
and grouped data (Figure 3B) demonstrate that the excitability of these neurons was
significantly increased by LPS when compared with control levels. We utilized a previously
reported small molecule inhibitor, Compound 15 [13,20], as a chemical probe to further
investigate the molecular mechanism of M3G-induced neuron response. The ability of
Compound 15 to disrupt the TLR4/MD-2 complex formation and selectively block TLR4
signal transduction without affecting other homologous TLR family proteins has been
previously demonstrated [13]. Further, Compound 15 was screened against a panel of 12
representative kinases, showing negligible non-specific inhibitory effects [20]. Importantly,
Compound 15 demonstrated high specificity and low toxicity both in vitro and in vivo [21],
providing an excellent probing tool to study the TLR4-specific signal transduction. The
specificity of this compound was utilized in our studies. Additionally, Compound 15
completely blocked the increased excitability of LPS suggesting that the observed increase in
excitation is mediated through TLR4/MD-2 (100%, 4.5±0.5 APs for LPS+Compound 15,
n=4; ANOVA, interaction F(2,18)=21.29, P<0.0001; Bonferroni multiple comparison test,
*P<0.05) (Figure 3A,B).
Figure 3 Lipopolysaccharide and morphine-3-glucuronide increase the excitability of
nociceptive dorsal root ganglion neurons. Current clamp recordings were performed on
small (≥30 μm) to medium (≥40 μm) dorsal root ganglion (DRG) neurons (L1-6) from naïve
rats. Firing of two to four action potentials (APs) was elicited by a 1 second depolarizing
current injection (ranging from 0.1 to 0.6 nA depending on the cell) every 30 seconds. (A)
Representative recordings demonstrating that application of 2 μg/mL lipopolysaccharide
(LPS) increases the number of elicited APs and Compound 15 can reverse this effect. (B)
Group data demonstrating that LPS caused a significant increase in DRG AP firing that is
reversed with Compound 15. (C) Representative recordings demonstrating that application of
3 μM morphine-3-glucuronide (M3G) increases the number of elicited APs and Compound
15 can reverse this effect. (D) Group data showing that M3G caused a significant increase in
DRG AP firing that is reversed by Compound 15
Given the ability of LPS to elicit increased excitability when combined with a depolarizing
current injection in small- and medium-diameter sensory neurons, we tested whether M3G
could produce similar effects in sensory neurons. The combination of repeated current pulse
combined with M3G administration significantly increased the excitability of small- to
medium-diameter sensory neurons when compared to baseline levels (19.6% neurons
responded, 2.8±0.7 APs for control vs. 14.2±2.4 APs for M3G, n=46) (Figure 3C).
Subsequent treatment with the TLR4/MD-2 small molecule inhibitor Compound 15
completely blocked M3G-dependent excitability in sensory neurons (100%, 2.0±0.6 APs for
M3G+Compound 15, n=4; ANOVA, interaction F(2,22)=24.16, P<0.0001; Bonferroni
multiple comparison test, P<0.05) (Figure 3C,D).
Pretreatment of rodents with Toll-like receptor 4/myeloid differentiation
protein-2 small molecule inhibitor prevents the rapid induction of tactile
hyperalgesia due to systemic morphine-3-glucuronide administration
Direct administration of M3G into the central nervous system by intracerebroventricular or
intrathecal routes produces a range of behaviors, including reductions in tail flick latencies,
touch-evoked agitation and thermal hyperalgesia [8-10,22]. However, there is limited
information regarding the effects of systemic M3G administration on noxious thermal or
tactile stimulus-dependent behavioral outcomes. Though M3G (10 mg/kg, i.p.) failed to elicit
changes in thermal latencies or duration (data not shown, n=4; Student’s t-test, P>0.05 for
both), M3G produced significant reductions in the paw withdrawal threshold to tactile
stimulus (Figure 4A; n=6, P<0.05). In contrast, paw withdrawal threshold to tactile stimulus
in rodents pretreated with Compound 15 (10 mg/kg, i.p., 1 hour), a small molecule inhibitor
of TLR4/MD-2 complex, prior to M3G administration (10 mg/kg, i.p.) did not differ from
baseline thresholds or the combination of Compound 15 and vehicle (Figure 4A; n=6,
ANOVA, interaction F(3,20)=69.33, P<0.05; Bonferroni multiple comparison test,
P<0.05).
Figure 4 Systemic morphine-3-glucuronide administration fails to elicit tactile
hyperalgesia in rats pretreated with the small molecule inhibitor of Toll-like receptor
4/myeloid differentiation protein-2 complex, Compound 15, or in Toll-like receptor 4
knockout mice. (A) Systemic morphine-3-glucuronide (M3G) (10 mg/kg, intraperitoneally)
produced robust tactile hyperalgesia (n=6, ANOVA, Bonferroni multiple comparison test,
*P<0.05), which could be prevented by pretreatment with Compound 15 (10 mg/kg,
intraperitoneally, n=6). (B) Shown is the force necessary to elicit paw withdrawal in Toll-
like receptor 4 (Tlr4) wild-type and Tlr4 knockout mice. Systemic M3G (25 mg/kg,
intraperitoneally) produced robust tactile hyperalgesia in Tlr4 wild-type mice (n=6,
*P<0.05). Tlr4 knockout mice failed to display a decrease in paw withdrawal force
following systemic M3G (n=6)
Toll-like receptor 4 knockout fails to exhibit morphine-3-glucuronide-induced
tactile hyperalgesia
To define whether M3G is dependent on the presence of a functional TLR4/MD-2 complex,
we tested the degree to which M3G induces tactile hyperalgesia in a mouse line that exhibits
a spontaneous mutation of the Tlr4 gene [23]. Like rats treated with M3G, the Tlr4 wild-type
mice displayed a significant increase in tactile hyperalgesia (Figure 4B; n=6, t-test, t=71.55,
df=10, P<0.05). However, Tlr4 knockout mice failed to display tactile hyperalgesia
following M3G administration (Figure 4B; n=6, t-test, t=0.278, df=10, P>0.05).
Morphine-3-glucuronide increases sodium current density in both
tetrodotoxin-sensitive and tetrodotoxin-resistant sodium currents
TTX-S and TTX-R sodium currents have been shown to contribute to neuronal excitability in
peripheral sensory neurons [24,25]. Here, we tested the possibility that TTX-S currents
(Figure 5A) and TTX-R currents (NaV1.8 and NaV1.9; Figure 5B) could potentially be
modulated by M3G and thus contribute to the increased hyperexcitability of sensory neurons
in the presence of M3G. We compared untreated DRG neurons to neurons exposed acutely
(approximately 5 minutes) to 3 μM M3G. We determined that TTX-S current density is
increased approximately 2.5 times (Figure 5C, n=12, P<0.05) in the presence of M3G, and
persistent TTX-R (NaV1.9) current density is increased approximately five times (Figure 5E,
n=12, P<0.05) in the presence of M3G (P<0.05). In contrast, NaV1.8 TTX-R current
density is not affected by M3G (Figure 5D, n=12, P>0.05).
Figure 5 Morphine-3-glucuronide-induced sensory neuron excitation is likely due to
effects on voltage-gated sodium channels. (A-B) Representative current traces from acutely
dissociated control dorsal root ganglion (DRG) neurons evoked by 200-ms steps in 5-mV
increments applied from a holding potential of −100 mV. (C-E) Peak current densities
(pA/pF) of DRGs exposed to extracellular recording solution (control) or 3 μM morphine-3-
glucuronide (M3G) for 5 minutes. Tetrodotoxin-sensitive (TTX-S) current densities were
estimated using a pre-pulse inactivation protocol (500 ms pre-pulses) with a 0 mV test pulse
as well as using post hoc kinetic subtraction [18]. Tetrodotoxin-resistant (TTX-R) current
densities were made in the presence of 500 nM TTX to pharmacologically isolate the
properties of voltage-gated sodium channel (NaV)1.8 and NaV1.9 currents. TTX-R NaV1.8
currents were estimated from the current elicited from a 150 ms pulse to 0 mV from a holding
potential of −100 mV whereas TTX-R NaV1.9 currents were estimated as the current
remaining during the last 15 ms of a 150 msec test pulse to −60 mV from a holding potential
of −100 mV(*P<0.05, versus control). Error bars indicate mean±SE from at least 10 cells
per condition. Small- and medium-diameter DRG neurons were used for these experiments
Discussion
The production of the M3G metabolite following glucuronidation of morphine contributes to
many off-target side effects of the opioid, including pain. Our results provide the first
demonstration that M3G directly evokes sensory neuron excitability. The receptor
responsible for this neuroexcitatory event appears to be the pattern recognition receptor,
TLR4, as we have shown that treating sensory neurons with an inhibitor of the TLR4/MD-2
interaction, Compound 15, completely blocks the increased neuronal excitability evoked by
M3G and effectively prevents systemic M3G-induced changes in tactile hyperalgesia. More
importantly, whole-cell voltage-clamp recordings demonstrated that M3G increased TTX-S
and TTX-R NaV densities in sensory neurons.
Little is known about the manner in which TLR4 contributes to pain. Broadly speaking,
increased spinal microglial TLR4 activation correlates with both onset of behavioral
hypersensitivity in rodent models of neuropathy and following administration of opioids
[21,26]. However, the degree to which activation of TLR4 present on sensory neurons
contributes to pain behavior is unknown. Clearly the ability of LPS administration to elicit
intracellular calcium mobilization in both capsaicin- and non-capsaicin-sensitive neurons
suggests that local administration of LPS to the plantar hind paw might produce thermal and
mechanical sensitivities [27,28]. Likewise, the fact that M3G elicits neuronal excitation in
both small- and medium-diameter sensory neurons might predict similar thermal- and tactile-
dependent behavioral outcomes. Despite these expectations, systemic M3G administration
evoked behavioral changes that were limited only to TLR4/MD-2-dependent mechanical
sensitivities. This outcome suggests that short-term M3G exposure-mediated changes in
tactile-mediated nociceptive behavior through TLR4 are independent of inflammation.
The apparent effects of M3G via TLR4/MD-2-mediated changes in tactile-dependent
nociceptive behavior and non-capsaicin responsive cells may require unique ionic
mechanisms such as modulation of voltage-gated sodium and/or potassium currents in
addition to voltage-dependent calcium channels [29]. Here, we demonstrate that M3G
increased TTX-S currents approximately 2.5 times. The TTX-S current in small-diameter
neurons is most likely a combination of NaV1.6 and Nav1.7 [25]. These TTX-S channels can
directly contribute to neuronal excitability [25], with NaV1.6 attributing to the resurgent
sodium current and NaV1.7 attributing to amplifying subthreshold generator potentials by
producing a prominent ramp current [30-32]. Interestingly, the LPS-induced activation of
microglia appears to be dependent on TTX-S NaV1.6 channels [33].
The TTX-R sodium channel NaV1.9 has been shown to contribute to the genesis of heat and
mechanical pain hypersensitivity [34]. In addition, NaV1.9 knockout mice have been shown
to lack acute visceral hypersensitivity [35]. Given the five-fold increase in NaV1.9 current
density, the action of M3G on this sodium channel isoform may be critical in the ability of
M3G to generate hyperexcitability in sensory neurons. Supporting evidence includes
observations that NaV1.9 enhances and prolongs the response to depolarizations that are
subthreshold for AP electrogenesis [36]. NaV1.9 also functions to lower threshold for single
AP and repetitive firing [37]. Thus, NaV1.9 may serve as an anion channel target for
reducing M3G-induced pain sensitivity associated with morphine administration. Moreover,
the ability of TLR4 signaling to modulate sodium channels elucidates a novel mechanism by
which TLR4 agonists can influence neuronal states of excitation.
Mechanisms of excitation in primary sensory neurons via the TLR4/MD-2 complex remain
elusive. However, due to the fact that IL- is known to increase the excitability of
nociceptors via enhancing persistent TTX-R current [38,39], and IL- receptors signal
through a common Toll/IL-1 receptor domain [40], it may come as no surprise that
TLR4/MD-2 complex activation is capable of exciting neurons. Many other possible
mechanisms exist which may also contribute to M3G-elicited neuronal excitation. It is
possible that, in capsaicin-sensitive sensory neurons, LPS as a surrogate for M3G induces
transactivation of TRPV1 via phospholipase C and results in the production of inositol 1,4,5-
triphosphage and subsequent [Ca2+]i release cascade [41,42]. Subsequently, the increase in
LPS-induced [Ca2+]i may depend on calcium influx via PKA (N-type calcium channels) or
PKC pathways (non-N-type calcium channels) [12,19,43]. However, non-capsaicin-
responsive cells would require other ionic mechanisms, such as modulation of voltage-gated
sodium and/or potassium currents in addition to voltage-dependent calcium channels [29].
That the systemic M3G-induced behavioral outcome observed by our studies was limited to
mechanical pain sensitivity may reflect unique capabilities of a TLR4/MD-2 dependent,
capsaicin-insensitive sensory neuron population. Few experimental studies exist that
delineate such a population with the exception of a subpopulation of unmyelinated polymodal
nociceptive sensory neurons that express Mas-related G protein-coupled receptor member D
(Mrgprd). Mrgprd appears to be selectively expressed in non-peptidergic nociceptors which
when genetically ablated in the adult animal fail to respond to noxious mechanosensation
whereas thermosensation is unaffected [44,45]. Whether the TLR4 agonist M3G is dependent
on Mrgprd-positive sensory neurons for the changes in mechanical nociceptive behavior is
unknown. Another subpopulation of sensory neurons that may be responsive to the
administration of M3G is the non-peptidergic primary afferents that exhibit the delta opioid
receptor and functionally contribute to mechanical pain [46].
In conclusion, TLR4 signaling via M3G within sensory neurons may provide a critical
element for understanding the development of tolerance and paradoxical hyperalgesia that
can occur following morphine-based therapies. The relationship between TLR4 expression in
DRGs, tactile hyperalgesia and neuronal hyperexcitability via the morphine metabolite M3G
may imply that TLR4-sensitized neurons may serve a vital signal to dampen the analgesic
effects of morphine. Better understanding of these TLR4-mediated events in sensory neurons
may provide the necessary framework for the design of agents that not only counteract
deleterious opioid-induced cellular adaptations but also effectively reduce analgesic
tolerance.
Abbreviations
ANOVA, analysis of variance; AP, action potential; CGRP, calcitonin gene-related peptide;
DRG, dorsal root ganglion; IL, interleukin; i.p., intraperitoneal; LPS, lipopolysaccharide;
M3G, morphine-3-glucuronide; M6G, morphine-6-glucouronide; MD, myeloid
differentiation protein; Mrgprd, Mas-related G protein-coupled receptor member D; NaV,
voltage-gated sodium channel; OIH, opioid-induced hyperalgesia; TLR, Toll-like receptor;
TRG, trigeminal root ganglion; TRPV1, transient receptor potential cation channel subfamily
V member 1; TTX-R, tetrodotoxin-resistant; TTX-S, tetrodotoxin-sensitive
Competing interests
The authors declare that they have no competing interests.
Authors’ contributions
MRD performed the experiments, analyzed the data, and wrote the manuscript; AP performed
the experiments, analyzed the data; NMW performed the experiments, analyzed the data; PF
performed the experiments, analyzed the data; SC synthesized Compound 15; HY
synthesized Compound 15; RK participated in the study design and data interpretation; FAW
was the main investigator of this work, and was in charge of the study design, analysis and
interpretation of results, and wrote the manuscript. All authors read and approved the final
manuscript.
Acknowledgements
This work was supported by a Indiana Spinal Cord & Brain Injury Research Grant (FAW)
and the NIH (NS049136 and DA026040 to FAW; NS067425 and DA026950 to HY),the
National Scientist Development from the American Heart Association (SDG5280023 to RK),
and the Neurofibromatosis New Investigator Award from the DOD/CDMRP (NF1000099 to
RK).
References
1. Coughtrie MW, Ask B, Rane A, Burchell B, Hume R: The enantioselective
glucuronidation of morphine in rats and humans. Evidence for the involvement of more
than one UDP-glucuronosyltransferase isoenzyme. Biochem Pharmacol 1989, 38:3273
3280.
2. Zelcer N, van de Wetering K, Hillebrand M, Sarton E, Kuil A, Wielinga PR, Tephly T,
Dahan A, Beijnen JH, Borst P: Mice lacking multidrug resistance protein 3 show altered
morphine pharmacokinetics and morphine-6-glucuronide antinociception. Proc Natl
Acad Sci U S A 2005, 102:72747279.
3. Osborne R, Joel S, Trew D, Slevin M: Morphine and metabolite behavior after
different routes of morphine administration: demonstration of the importance of the
active metabolite morphine-6-glucuronide. Clin Pharmacol Ther 1990, 47:1219.
4. Hasselstrom J, Sawe J: Morphine pharmacokinetics and metabolism in humans.
Enterohepatic cycling and relative contribution of metabolites to active opioid
concentrations. Clin Pharmacokinet 1993, 24:344354.
5. van Dorp EL, Romberg R, Sarton E, Bovill JG, Dahan A: Morphine-6-glucuronide:
morphine’s successor for postoperative pain relief? Anesth Analg 2006, 102:17891797.
6. Labella FS, Pinsky C, Havlicek V: Morphine derivatives with diminished opiate
receptor potency show enhanced central excitatory activity. Brain Res 1979, 174:263
271.
7. Hewett K, Dickenson AH, McQuay HJ: Lack of effect of morphine-3-glucuronide on
the spinal antinociceptive actions of morphine in the rat: an electrophysiological study.
Pain 1993, 53:5963.
8. Bartlett SE, Cramond T, Smith MT: The excitatory effects of morphine-3-glucuronide
are attenuated by LY274614, a competitive NMDA receptor antagonist, and by
midazolam, an agonist at the benzodiazepine site on the GABAA receptor complex. Life
Sci 1994, 54:687694.
9. Lewis SS, Hutchinson MR, Rezvani N, Loram LC, Zhang Y, Maier SF, Rice KC, Watkins
LR: Evidence that intrathecal morphine-3-glucuronide may cause pain enhancement via
toll-like receptor 4/MD-2 and interleukin-1beta. Neuroscience 2010, 165:569583.
10. Hutchinson MR, Zhang Y, Shridhar M, Evans JH, Buchanan MM, Zhao TX, Slivka PF,
Coats BD, Rezvani N, Wieseler J, et al: Evidence that opioids may have toll-like receptor
4 and MD-2 effects. Brain Behav Immun 2010, 24:8395.
11. Hua XY, Chen P, Fox A, Myers RR: Involvement of cytokines in lipopolysaccharide-
induced facilitation of CGRP release from capsaicin-sensitive nerves in the trachea:
studies with interleukin-1beta and tumor necrosis factor-alpha. J Neurosci 1996,
16:47424748.
12. Diogenes A, Ferraz CC, Akopian AN, Henry MA, Hargreaves KM: LPS sensitizes
TRPV1 via activation of TLR4 in trigeminal sensory neurons. J Dent Res 2011, 90:759
764.
13. Bevan DE, Martinko AJ, Loram LC, Stahl JA, Taylor FR, Joshee S, Watkins LR, Yin H:
Selection, Preparation, and Evaluation of Small- Molecule Inhibitors of Toll-Like
Receptor 4. ACS Med Chem Lett 2010, 1:194198.
14. Moran TD, Smith PA: Morphine-3beta-D-glucuronide suppresses inhibitory synaptic
transmission in rat substantia gelatinosa. J Pharmacol Exp Ther 2002, 302:568576.
15. Hemstapat K, Monteith GR, Smith D, Smith MT: Morphine-3-glucuronide’s neuro-
excitatory effects are mediated via indirect activation of N-methyl-D-aspartic acid
receptors: mechanistic studies in embryonic cultured hippocampal neurones. Anesth
Analg 2003, 97:494505. table of contents.
16. Bhangoo S, Ren D, Miller RJ, Henry KJ, Lineswala J, Hamdouchi C, Li B, Monahan PE,
Chan DM, Ripsch MS, White FA: Delayed functional expression of neuronal chemokine
receptors following focal nerve demyelination in the rat: a mechanism for the
development of chronic sensitization of peripheral nociceptors. Molecular Pain 2007,
3:38.
17. Ma C, LaMotte RH: Enhanced excitability of dissociated primary sensory neurons
after chronic compression of the dorsal root ganglion in the rat. Pain 2005, 113:106112.
18. Cummins TR, Waxman SG: Downregulation of tetrodotoxin-resistant sodium
currents and upregulation of a rapidly repriming tetrodotoxin-sensitive sodium current
in small spinal sensory neurons after nerve injury. J Neurosci 1997, 17:35033514.
19. Ochoa-Cortes F, Ramos-Lomas T, Miranda-Morales M, Spreadbury I, Ibeakanma C,
Barajas-Lopez C, Vanner S: Bacterial cell products signal to mouse colonic nociceptive
dorsal root ganglia neurons. Am J Physiol Gastrointest Liver Physiol 2010, 299:G723
G732.
20. Chavez SA, Martinko AJ, Lau C, Pham MN, Cheng K, Bevan DE, Mollnes TE, Yin H:
Development of beta-amino alcohol derivatives that inhibit Toll-like receptor 4
mediated inflammatory response as potential antiseptics. J Med Chem 2011, 54:4659
4669.
21. Wang X, Loram LC, Ramos K, de Jesus AJ, Thomas J, Cheng K, Reddy A, Somogyi AA,
Hutchinson MR, Watkins LR, Yin H: Morphine activates neuroinflammation in a manner
parallel to endotoxin. Proc Natl Acad Sci U S A 2012, Apr 17, 109(16):63256330.
22. Smith MT, Watt JA, Cramond T: Morphine-3-glucuronidea potent antagonist of
morphine analgesia. Life Sci 1990, 47:579585.
23. Poltorak A, Merlin T, Nielsen PJ, Sandra O, Smirnova I, Schupp I, Boehm T, Galanos C,
Freudenberg MA: A point mutation in the IL-12R beta 2 gene underlies the IL-12
unresponsiveness of Lps-defective C57BL/10ScCr mice. J Immunol 2001, 167:21062111.
24. Roberson DP, Binshtok AM, Blasl F, Bean BP, Woolf CJ: Targeting of sodium channel
blockers into nociceptors to produce long-duration analgesia: a systematic study and
review. Br J Pharmacol 2011, 164:4858.
25. Rush AM, Cummins TR, Waxman SG: Multiple sodium channels and their roles in
electrogenesis within dorsal root ganglion neurons. J Physiol 2007, 579:114.
26. Tanga FY, Nutile-McMenemy N, DeLeo JA: The CNS role of Toll-like receptor 4 in
innate neuroimmunity and painful neuropathy. Proc Natl Acad Sci U S A 2005,
102:58565861.
27. Kanaan SA, Saade NE, Haddad JJ, Abdelnoor AM, Atweh SF, Jabbur SJ, Safieh-
Garabedian B: Endotoxin-induced local inflammation and hyperalgesia in rats and mice:
a new model for inflammatory pain. Pain 1996, 66:373379.
28. Maier SF, Wiertelak EP, Martin D, Watkins LR: Interleukin-1 mediates the behavioral
hyperalgesia produced by lithium chloride and endotoxin. Brain Res 1993, 623:321324.
29. Qin X, Hou L, Wang X: Lipopolysaccharide evoked peptide release by calcium-
induced calcium release. Neuroreport 2004, 15:10031006.
30. Cummins TR, Dib-Hajj SD, Herzog RI, Waxman SG: Nav1.6 channels generate
resurgent sodium currents in spinal sensory neurons. FEBS Lett 2005, 579:21662170.
31. Cummins TR, Howe JR, Waxman SG: Slow closed-state inactivation: a novel
mechanism underlying ramp currents in cells expressing the hNE/PN1 sodium channel.
J Neurosci 1998, 18:96079619.
32. Herzog RI, Cummins TR, Ghassemi F, Dib-Hajj SD, Waxman SG: Distinct repriming
and closed-state inactivation kinetics of Nav1.6 and Nav1.7 sodium channels in mouse
spinal sensory neurons. J Physiol 2003, 551:741750.
33. Black JA, Liu S, Waxman SG: Sodium channel activity modulates multiple functions
in microglia. Glia 2009, 57:10721081.
34. Lolignier S, Amsalem M, Maingret F, Padilla F, Gabriac M, Chapuy E, Eschalier A,
Delmas P, Busserolles J: Nav1.9 channel contributes to mechanical and heat pain
hypersensitivity induced by subacute and chronic inflammation. PLoS One 2011,
6:e23083.
35. Martinez V, Melgar S: Lack of colonic-inflammation-induced acute visceral
hypersensitivity to colorectal distension in Na(v)1.9 knockout mice. Eur J Pain 2008,
12:934944.
36. Herzog RI, Cummins TR, Waxman SG: Persistent TTX-resistant Na+current affects
resting potential and response to depolarization in simulated spinal sensory neurons. J
Neurophysiol 2001, 86:13511364.
37. Baker MD, Chandra SY, Ding Y, Waxman SG, Wood JN: GTP-induced tetrodotoxin-
resistant Na+current regulates excitability in mouse and rat small diameter sensory
neurones. J Physiol 2003, 548:373382.
38. Binshtok AM, Wang H, Zimmermann K, Amaya F, Vardeh D, Shi L, Brenner GJ, Ji RR,
Bean BP, Woolf CJ, Samad TA: Nociceptors are interleukin-1beta sensors. J Neurosci
2008, 28:1406214073.
39. Amaya F, Wang H, Costigan M, Allchorne AJ, Hatcher JP, Egerton J, Stean T, Morisset
V, Grose D, Gunthorpe MJ, et al: The voltage-gated sodium channel Na(v)1.9 is an
effector of peripheral inflammatory pain hypersensitivity. J Neurosci 2006, 26:12852
12860.
40. Vezzani A, Maroso M, Balosso S, Sanchez MA, Bartfai T: IL-1 receptor/Toll-like
receptor signaling in infection, inflammation, stress and neurodegeneration couples
hyperexcitability and seizures. Brain Behavior and Immunity 2011, 25:12811289.
41. Chiang C, Veckman V, Limmer K, David M: Phospholipase C gamma-2 and
intracellular Calcium are required for LPS-induced Toll-Receptor 4 (TLR4)
endocytosis and Interferon Regulatory Factor 3 (IRF3) activation. J Biol Chem 2012,
287(6):37043709.
42. Rohacs T, Thyagarajan B, Lukacs V: Phospholipase C mediated modulation of TRPV1
channels. Mol Neurobiol 2008, 37:153163.
43. Hou L, Wang X: PKC and PKA, but not PKG mediate LPS-induced CGRP release
and [Ca(2+)](i) elevation in DRG neurons of neonatal rats. J Neurosci Res 2001, 66:592
600.
44. Cavanaugh DJ, Lee H, Lo L, Shields SD, Zylka MJ, Basbaum AI, Anderson DJ: Distinct
subsets of unmyelinated primary sensory fibers mediate behavioral responses to noxious
thermal and mechanical stimuli. Proc Natl Acad Sci U S A 2009, 106:90759080.
45. Shields SD, Cavanaugh DJ, Lee H, Anderson DJ, Basbaum AI: Pain behavior in the
formalin test persists after ablation of the great majority of C-fiber nociceptors. Pain
2010, 151:422429.
46. Scherrer G, Imamachi N, Cao YQ, Contet C, Mennicken F, O’Donnell D, Kieffer BL,
Basbaum AI: Dissociation of the opioid receptor mechanisms that control mechanical
and heat pain. Cell 2009, 137:11481159.
Figure 1
Figure 2
Figure 3
Figure 5
... Voltage-gated sodium channels (NaVs) play a key role in the initiation and propagation of action potentials in electrically excitable nociceptive neurons. Specific NaVs in these neurons can be activated by nonclassical opioid receptors [11,13]. This activation and augmentation of NaV-dependent pain sensing appears to counter a few classical opioid effects [14][15][16]. ...
... This activation and augmentation of NaV-dependent pain sensing appears to counter a few classical opioid effects [14][15][16]. Therefore, neuronal activation of NaV1.7 could be one of the mechanisms that limit opioid effectiveness [11,13,15,16]. The activation of these channels can be blocked with an existing class of Food and Drug Administration (FDA)-approved antiepileptic seizure drugs that modulate NaVs [11,[17][18][19][20]. ...
... The basic drug mechanism is that nonclassical opioid receptors function to increase the NaV1.7 current in nociceptive sensory neurons, leading to drug-induced pain, or OIH. The combination of an opioid and specific antiseizure drugs such as carbamazepine, oxcarbazepine, or lacosamide effectively diminishes the opioid-induced NaV1.7 current [11,13,15]. These preclinical observations in several neuropathic pain-induced injury models lay the foundation for changing the management of chronic pain from empirical symptom control to personalized targeting of specific mechanisms responsible for pain. ...
Article
Full-text available
Background Chronic abdominal pain is the hallmark symptom of chronic pancreatitis (CP), with 50% to 80% of patients seeking medical attention for pain control. Although several management options are available, outcomes are often disappointing, and opioids remain a mainstay of therapy. Opioid-induced hyperalgesia is a phenomenon resulting in dose escalation, which may occur partly because of the effects of opioids on voltage-gated sodium channels associated with pain. Preclinical observations demonstrate that the combination of an opioid and the antiseizure drug lacosamide diminishes opioid-induced hyperalgesia and improves pain control. Objective In this phase 1 trial, we aim to determine the safety, tolerability, and dose-limiting toxicity of adding lacosamide to opioids for the treatment of painful CP and assess the feasibility of performance of a pilot study of adding lacosamide to opioid therapy in patients with CP. As an exploratory aim, we will assess the efficacy of adding lacosamide to opioid therapy in patients with painful CP. Methods Using the Bayesian optimal interval design, we will conduct a dose-escalation trial of adding lacosamide to opioid therapy in patients with painful CP enrolled in cohorts of size 3. The initial dose will be 50 mg taken orally twice a day, followed by incremental increases to a maximum dose of 400 mg/day, with lacosamide administered for 7 days at each dose level. Adverse events will be documented according to Common Terminology Criteria for Adverse Events (version 5.0). Results As of December 2023, we have currently enrolled 6 participants. The minimum number of participants to be enrolled is 12 with a maximum of 24. We expect to publish the results by March 2025. Conclusions This trial will test the feasibility of the study design and provide reassurance regarding the tolerability and safety of opioids in treating painful CP. It is anticipated that lacosamide will prove to be safe and well tolerated, supporting a subsequent phase 2 trial assessing the efficacy of lacosamide+opioid therapy in patients with painful CP, and that lacosamide combined with opiates will lower the opioid dose necessary for pain relief and improve the safety profile of opioid use in treating painful CP. Trial Registration Clinicaltrials.gov NCT05603702; https://clinicaltrials.gov/study/NCT05603702 International Registered Report Identifier (IRRID) PRR1-10.2196/50513
... Experiments in animal pain models have demonstrated that Nav blockers such as carbamazepine potentiate the morphine analgesia of postoperative pain in morphine-dependent rats [93] and in model of neuropathic pain [94]; that intrathecally co-administered lamotrigine attenuates antinociceptive tolerance to morphine in rats [95]; and that topiramate and lamotrigine synergistically interact with tramadol in a nociceptive pain model [96]. The potential molecular mechanisms by which anticonvulsants enhance opioid-induced analgesia may involve the blockade of overexpressed and overactive Nav1.7 in injured neurons [97] and Nav 1.6 in microglia [24]. Moreover, the activation of neuronal and glial toll-like receptor 4 (TLR4) by morphine and its metabolite morphine-3-glucuronide may additionally contribute to the pathological function of several subtypes of Nav [97] and contribute to the decreased effectiveness of morphine in neuropathic pain models. ...
... The potential molecular mechanisms by which anticonvulsants enhance opioid-induced analgesia may involve the blockade of overexpressed and overactive Nav1.7 in injured neurons [97] and Nav 1.6 in microglia [24]. Moreover, the activation of neuronal and glial toll-like receptor 4 (TLR4) by morphine and its metabolite morphine-3-glucuronide may additionally contribute to the pathological function of several subtypes of Nav [97] and contribute to the decreased effectiveness of morphine in neuropathic pain models. Thus, PHT, by silencing the activated microglia and reducing abnormal activity of Nav in injured neurons, may enhance the antinociceptive effect of morphine observed in our study. ...
Article
Full-text available
Neuropathic pain remains a clinical challenge due to its complex and not yet fully understood pathomechanism, which result in limited analgesic effectiveness of the management offered, particularly for patients with acute, refractory neuropathic pain states. In addition to the introduction of several modern therapeutic approaches, such as neuromodulation or novel anti-neuropathic drugs, significant efforts have been made in the repurposing of well-known substances such as phenytoin. Although its main mechanism of action occurs at sodium channels in excitable and non-excitable cells and is well documented, how the drug affects the disturbed neuropathic interactions at the spinal cord level and how it influences morphine-induced analgesia have not been clarified, both being crucial from a clinical perspective. We demonstrated that single and repeated systemic administrations of phenytoin decreased tactile and thermal hypersensitivity in an animal model of neuropathic pain. Importantly, we observed an increase in the antinociceptive effect on thermal stimuli with repeated administrations of phenytoin. This is the first study to report that phenytoin improves morphine-induced antinociceptive effects and influences microglia/macrophage activity at the spinal cord and dorsal root ganglion levels in a neuropathic pain model. Our findings support the hypothesis that phenytoin may represent an effective strategy for neuropathic pain management in clinical practice, particularly when combination with opioids is needed.
... As a pain score of greater than 3 is considered as "unacceptable pain", its association with higher serum M3G concentrations is clinically significant. This finding has mechanistic plausibility as M3G binds to MD2 the accessory TLR4 protein causing a proinflammatory response [2] and M3G induces hyperalgesia through the MD2/TLR4 complex in rats through changes in the functioning of voltage-gated sodium channels [26]. That M6G was also associated with "unacceptable pain", although statistically weaker (P = 0.02) may simply be due to its hepatic formation from morphine and its renal elimination being Table 2. Co-incidence of adverse events reported by cancer pain patients receiving slow-release oral morphine. ...
Article
Full-text available
The aim was to determine if opioid neuroimmunopharmacology pathway gene polymorphisms alter serum morphine, morphine-3-glucuronide and morphine-6-glucuronide concentration-response relationships in 506 cancer patients receiving controlled-release oral morphine. Morphine-3-glucuronide concentrations (standardised to 11 h post-dose) were higher in patients without pain control (median (interquartile range) 1.2 (0.7–2.3) versus 1.0 (0.5–1.9) μM, P = 0.006), whereas morphine concentrations were higher in patients with cognitive dysfunction (40 (20–81) versus 29 (14–60) nM, P = 0.02). TLR2 rs3804100 variant carriers had reduced odds (adjusted odds ratio (95% confidence interval) 0.42 (0.22–0.82), P = 0.01) of opioid adverse events. IL2 rs2069762 G/G (0.20 (0.06-0.52)), BDNF rs6265 A/A (0.15 (0.02–0.63)) and IL6R rs8192284 carrier (0.55 (0.34–0.90)) genotypes had decreased, and IL6 rs10499563 C/C increased (3.3 (1.2–9.3)), odds of sickness response (P ≤ 0.02). The study has limitations in heterogeneity in doses, sampling times and diagnoses but still suggests that pharmacokinetics and immune genetics co-contribute to morphine pain control and adverse effects in cancer patients.
... morphine, fentanyl, remifentanil, and oxycodone) bind to and activate TLR4 even at low, subanalgesic concentrations. [3][4][5] And, the systemic administration of sub-analgesic doses of opioid analgesics can, paradoxically, produce hyperalgesia (OIH), 6 an effect that is mediated either directly, by activation of TLR4, which is present on nociceptors, 7,8 or indirectly by the action of opioids at TLR4 on non-neuronal cells, including the immune system that, in turn, release pronociceptive mediators. 9,10 The primary afferent nociceptor has been suggested to play a key role in OIH, 11 which is attenuated by TLR4 antagonists, 12 as well as by intrathecal administration of an oligodeoxynucleotide (ODN) antisense for TLR4 mRNA. ...
Article
Full-text available
While opioids remain amongst the most effective treatments for moderate-to-severe pain, their substantial side effect profile remains a major limitation to broader clinical use. One such side effect is opioid-induced hyperalgesia (OIH), which includes a transition from opioid-induced analgesia to pain enhancement. Evidence in rodents supports the suggestion that OIH may be produced by the action of opioids at Toll-like Receptor 4 (TLR4) either on immune cells that, in turn, produce pronociceptive mediators to act on nociceptors, or by a direct action at nociceptor TLR4. And, sub-analgesic doses of several opioids have been shown to induce hyperalgesia in rodents by their action as TLR4 agonists. In the present in vitro patch-clamp electrophysiology experiments, we demonstrate that low dose morphine directly sensitizes human as well as rodent dorsal root ganglion (DRG) neurons, an effect of this opioid analgesic that is antagonized by LPS-RS Ultrapure, a selective TLR4 antagonist. We found that low concentration (100 nM) of morphine reduced rheobase in human (by 36%) and rat (by 26%) putative C-type nociceptors, an effect of morphine that was markedly attenuated by preincubation with LPS-RS Ultrapure. Our findings support the suggestion that in humans, as in rodents, OIH is mediated by the direct action of opioids at TLR4 on nociceptors.
... Thus, several opioid analgesics (e.g. morphine, fentanyl, remifentanil, and oxycodone) bind to and activate TLR4 even at low, sub-analgesic concentrations 13,24,25 . ...
Preprint
While opioids remain amongst the most effective treatments for moderate-to-severe pain, their substantial side effect profile remains a major limitation to broader clinical use. One such side effect is opioid-induced hyperalgesia (OIH), which includes a transition from opioid-induced analgesia to pain enhancement. Evidence in rodents supports the suggestion that OIH may be produced by the action of opioids at Toll-like Receptor 4 (TLR4) either on immune cells that, in turn, produce pronociceptive mediators to act on nociceptors, or by a direct action at nociceptor TLR4. And, sub-analgesic doses of several opioids have been shown to induce hyperalgesia in rodents by their action as TLR4 agonists. In the present in vitro patch-clamp electrophysiology experiments, we demonstrate that low dose morphine directly sensitizes human as well as rodent dorsal root ganglion (DRG) neurons, an effect of this opioid analgesic that is antagonized by LPS-RS Ultrapure, a selective TLR4 antagonist. We found that morphine (100 nM) reduced rheobase in human (by 36%) and rat (by 26%) putative C-type nociceptors, an effect of morphine that was markedly attenuated by preincubation with LPS-RS Ultrapure. Our findings support the suggestion that in humans, as well as in rodents, OIH is mediated by the direct action of opioids at TLR4 on nociceptors.
Article
Drug addiction is a chronic and debilitating disease that is considered a global health problem. Various cell types in the brain are involved in the progression of drug addiction. Recently, the xenobiotic hypothesis has been proposed, which frames substances of abuse as exogenous molecules that are responded to by the immune system as foreign "invaders", thus triggering protective inflammatory responses. An emerging body of literature reveals that microglia, the primary resident immune cells in the brain, play an important role in the progression of addiction. Repeated cycles of drug administration cause a progressive, persistent induction of neuroinflammation by releasing microglial proinflammatory cytokines and their metabolic products. This contributes to drug addiction via modulation of neuronal function. In this review, we focus on the role of microglia in the etiology of drug addiction. Then, we discuss the dynamic states of microglia and the correlative and causal evidence linking microglia to drug addiction. Finally, possible mechanisms of how microglia sense drug-related stimuli and modulate the addiction state and how microglia-targeted anti-inflammation therapies affect addiction are reviewed. Understanding the role of microglia in drug addiction may help develop new treatment strategies to fight this devastating societal challenge.
Article
Full-text available
About 25 million American adults experience pain daily and one of the most commonly prescribed drugs to treat pain are opioids. Prolonged opioid usage and dose escalations can cause a paradoxical response where patients experience enhanced pain sensitivity. This opioid induced hyperalgesia (OIH) is a major hurdle when treating pain in the clinic because its underlying mechanisms are still not fully understood. OIH is also commonly overlooked and lacks guidelines to prevent its onset. Research on pain disorders and opioid usage have recognized potential epigenetic drivers of disease including DNA methylation, histone modifications, miRNA regulation, but their involvement in OIH has not been well studied. This article discusses epigenetic changes that may contribute to pathogenesis, with an emphasis on miRNA alterations in OIH. There is a crucial gap in knowledge including how multiple epigenetic modulators contribute to OIH. Elucidating the epigenetic changes underlying OIH and the crosstalk among these mechanisms could lead to the development of novel targets for the prevention and treatment of this painful phenomena.
Article
The immune response plays a pivotal role in the pathogenesis of diseases. Toll-like receptor 4 (TLR4), as an intrinsic immune receptor, exhibits widespread in vivo expression and its dysregulation significantly contributes to the onset of various diseases, encompassing cardiovascular disorders, neoplastic conditions, and inflammatory ailments. This comprehensive review centers on elucidating the architectural and distributive characteristics of TLR4, its conventional signaling pathways, and its mode of action in diverse disease contexts. Ultimately, this review aims to propose novel avenues and therapeutic targets for clinical intervention.
Article
Full-text available
Opioids are one of the most potent drugs for treating moderate to severe pain. Despite irrefutable clinical application in chronic pain management, the long‐term use of opioids has been increasingly questioned due to undesired side effects that demand attention. Opioids such as morphine mediate clinically relevant effects primarily through the µ‐opioid receptor that go beyond their classical role as analgesics, causing potentially fatal side effects such as tolerance, dependence, and addiction. Furthermore, there is growing evidence that opioids affect immune system function, cancer progression, metastasis, and recurrence. Though a biological plausibility, the clinical evidence for the action of opioids on cancer is mixed, revealing a more complex picture as researchers struggle to establish a vital link between opioid receptor agonists, cancer progression, and suppression, or both. Thus, in light of the uncertainty of opioid effects on cancer, in this review, a focused overview of the role of opioid receptors in modulating cancer progression, their underlying signaling mechanisms, and the biological activity of opioid receptor agonists and antagonists is provided.
Article
Full-text available
Opioids create a neuroinflammatory response within the CNS, compromising opioid-induced analgesia and contributing to various unwanted actions. How this occurs is unknown but has been assumed to be via classic opioid receptors. Herein, we provide direct evidence that morphine creates neuroinflammation via the activation of an innate immune receptor and not via classic opioid receptors. We demonstrate that morphine binds to an accessory protein of Toll-like receptor 4 (TLR4), myeloid differentiation protein 2 (MD-2), thereby inducing TLR4 oligomerization and triggering proinflammation. Small-molecule inhibitors, RNA interference, and genetic knockout validate the TLR4/MD-2 complex as a feasible target for beneficially modifying morphine actions. Disrupting TLR4/MD-2 protein-protein association potentiated morphine analgesia in vivo and abolished morphine-induced proinflammation in vitro, the latter demonstrating that morphine-induced proinflammation only depends on TLR4, despite the presence of opioid receptors. These results provide an exciting, nonconventional avenue to improving the clinical efficacy of opioids.
Article
Full-text available
Toll-like receptor 4 (TLR4) is unique among the TLRs in its use of multiple adaptor proteins leading to activation of both the interferon regulatory factor 3 (IRF3) and nuclear factor κB (NF-κB) pathways. Previous work has demonstrated that TLR4 initiates NF-κB activation from the plasma membrane, but that subsequent TLR4 translocation to the endosomes is required for IRF3 activation. Here we have characterized several components of the signaling pathway that governs TLR4 translocation and subsequent IRF3 activation. We find that phospholipase C γ2 (PLCγ2) accounts for LPS-induced inositol 1,4,5-trisphosphate (IP3) production and subsequent calcium (Ca2+) release. Blockage of PLCγ2 function by inhibitors or knockdown of PLCγ2 expression by siRNAs in RAW 264.7 macrophages lead to reduced IRF3, but enhanced NF-κB activation. In addition, bone marrow-derived macrophages from PLCγ2-deficient mice showed impaired IRF3 phosphorylation and expression of IRF3-regulated genes after LPS stimulation. Using cell fractionation, we show that PLCγ2-IP3-Ca2+ signaling cascade is required for TLR4 endocytosis following LPS stimulation. In conclusion, our results describe a novel role of the PLCγ2-IP3-Ca2+ cascade in the LPS-induced innate immune response pathway where release of intracellular Ca2+ mediates TLR4 trafficking and subsequent activation of IRF3.
Article
Full-text available
Inflammation is known to be responsible for the sensitization of peripheral sensory neurons, leading to spontaneous pain and invalidating pain hypersensitivity. Given its role in regulating neuronal excitability, the voltage-gated Nav1.9 channel is a potential target for the treatment of pathological pain, but its implication in inflammatory pain is yet not fully described. In the present study, we examined the role of the Nav1.9 channel in acute, subacute and chronic inflammatory pain using Nav1.9-null mice and Nav1.9 knock-down rats. In mice we found that, although the Nav1.9 channel does not contribute to basal pain thresholds, it plays an important role in heat pain hypersensitivity induced by subacute paw inflammation (intraplantar carrageenan) and chronic ankle inflammation (complete Freund's adjuvant-induced monoarthritis). We showed for the first time that Nav1.9 also contributes to mechanical hypersensitivity in both models, as assessed using von Frey and dynamic weight bearing tests. Consistently, antisense-based Nav1.9 gene silencing in rats reduced carrageenan-induced heat and mechanical pain hypersensitivity. While no changes in Nav1.9 mRNA levels were detected in dorsal root ganglia (DRGs) during subacute and chronic inflammation, a significant increase in Nav1.9 immunoreactivity was observed in ipsilateral DRGs 24 hours following carrageenan injection. This was correlated with an increase in Nav1.9 immunolabeling in nerve fibers surrounding the inflamed area. No change in Nav1.9 current density could be detected in the soma of retrolabeled DRG neurons innervating inflamed tissues, suggesting that newly produced channels may be non-functional at this level and rather contribute to the observed increase in axonal transport. Our results provide evidence that Nav1.9 plays a crucial role in the generation of heat and mechanical pain hypersensitivity, both in subacute and chronic inflammatory pain models, and bring new elements for the understanding of its regulation in those models.
Article
Full-text available
We have developed a strategy to target the permanently charged lidocaine derivative lidocaine N-ethyl bromide (QX-314) selectively into nociceptive sensory neurons through the large-pore transient receptor potential cation channel subfamily V (TRPV1) noxious heat detector channel. This involves co-administration of QX-314 and a TRPV1 agonist to produce a long-lasting local analgesia. For potential clinical use we propose using lidocaine as the TRPV1 agonist, because it activates TRPV1 at clinical doses. We conducted experiments in rats to determine optimal concentrations and ratios of lidocaine and QX-314 that produce the greatest degree and duration of pain-selective block when administered nearby the sciatic nerve: reduction in the response to noxious mechanical (pinch) and to radiant heat stimuli, with minimal disruption in motor function (grip strength). A combination of 0.5% QX-314 and 2% lidocaine produced 1 h of non-selective sensory and motor block followed by >9 h of pain-selective block, where grip strength was unimpaired. QX-314 at this concentration had no effect by itself, while 2% lidocaine by itself produced 1 h of non-selective block. The combination of 0.5% QX-314 and 2% lidocaine was the best of the many tested, in terms of the duration and selectivity of local analgesia. Targeting charged sodium channel blockers into specific sets of axons via activation of differentially expressed large-pore channels provides an opportunity to produce prolonged local analgesia, and represents an example of how exploiting ion channels as a drug delivery port can be used to increase the specificity and efficacy of therapeutics.
Article
Morphine, morphine-6-glucuronide (M6G), morphine-3-glucuronide (M3G) and normorphine were analysed with high performance liquid chromatography in plasma and urine, collected over 72h after administration of single intravenous 5mg and oral 20mg doses of morphine to 7 healthy volunteers. Systemic plasma clearance of morphine was on average 21.1 ± 3.4 ml/min/kg (1.27 ± 0.20 L/h/kg), volume of distribution was 2.9 ± 0.8 L/kg and oral bioavailability was 29.2 ± 7.2%. Clearance of morphine to form M3G and M6G comprised 57.3% and 10.4%, respectively, and renal clearance comprised 10.9% of total systemic plasma clearance; hence, more than one-fifth of a dose (20.8%) remained as unidentified residual clearance. On the basis of the area under the plasma concentration-time curves determined after oral and intravenous administration, the ratios of M6G: morphine were 3.6 ± 1.2 and 0.7 ± 0.3, respectively. The corresponding figures for M3G: morphine were 29.9 ± 6.8 and 7.7 ± 1.4. Differences in metabolic ratios between the parenteral and oral routes could be attributed solely to differences in morphine concentrations as evidenced both by plasma concentrations and amounts excreted in urine. An oral: parenteral potency ratio of 1: 3 may, thus, be due to differences in circulating amounts of morphine since the proportions of an administered dose found as M6G and M3G after administration by both routes were equal. A major finding was a slowly declining terminal phase of morphine and metabolites that was evident both in plasma and in urinary excretion versus time curves, where the half-lives of morphine, M3G and M6G were 15.1 ± 6.5h, 11.2 ± 2.7h and 12.9 ± 4.5h, respectively. The terminal half-life of normorphine was 23.9 ± 10.1 h after oral administration. Comparison of oral with intravenous excretion curves showed that a greater part of morphine and metabolites were excreted during the slowly declining phase after the oral dose than the intravenous dose, which is highly suggestive of enterohepatic cycling. The renal clearance of M6G and morphine was seen to exceed creatinine clearance, possibly due to an active secretion process.
Article
Toll-like receptor 4 (TLR4), a membrane-spanning receptor protein that functions in complex with its accessory protein MD-2, is an intriguing target for therapeutic development. Herein, we report the identification of a series of novel TLR4 inhibitors and the development of a robust, enantioselective synthesis using an unprecedented Mannich type reaction to functionalize a pyrazole ring. In silico and cellular assay results demonstrated that compound 1 and its analogues selectively block TLR4 activation in live cells. Animal model tests showed that 1 and its derivatives could potentiate morphine-induced analgesia in vivo, presumably by attenuating the opioid-induced TLR4 activation.Keywords (keywords): Toll-like receptor; glial cells; signal transduction; protein−protein interactions; synthesis
Article
Calcitonin gene-related peptide (CGRP), is produced in dorsal root ganglia (DRG) neurons and released from primary afferent neurons to mediate hemodynamic effects and neurogenic inflammation. In this work, we determined whether lipopolysaccharide (LPS), an inflammatory stimulator, could trigger CGRP release from cultured DRG neurons and if so, which cellular signaling pathway was involved in this response. Cytoplasmic concentration of calcium ([Ca2+]i) plays a key role in neurotransmitter release, therefore [Ca2+]i was also determined in cultured DRG cells using fluo-3/AM. The results showed that LPS (0.1–10 μg/ml) evoked CGRP release in a time- and concentration-dependent manner from DRG neurons. LPS also increased [Ca2+]i in a concentration-dependent manner. The protein kinase C (PKC) inhibitors, calphostin C 0.5 μM or RO-31-8220 0.1 μM, and the cAMP-dependent protein kinase (PKA) specific inhibitor RP-CAMPS 30 μM or nonspecific inhibitor H8 1 μM inhibited 1 μg/ml LPS-evoked CGRP release and [Ca2+]i increase from DRG neurons. The cGMP-dependent protein kinase (PKG) inhibitor Rp-8-pCPT-cGMPS 30 μM did not block the LPS response. These data suggest that LPS may stimulate CGRP release and [Ca2+]i elevation through PKC and PKA, but not PKG signaling pathway in DRG neurons of neonatal rats. J. Neurosci. Res. 66:592–600, 2001. © 2001 Wiley-Liss, Inc.
Article
The sickness-inducing agents lithium chloride (LiCl) and lipopolysacharide (LPS) produce a long-lasting facilitation of the nociceptive tailflick reflex. Many of the behavioral and physiological changes produced by illness are mediated by interleukin-1 (IL-1) released from monocytes stimulated by the pathogenic substance. Monocytes also produce an IL-1 receptor antagonist (IL-1 ra) which has been sequenced and cloned. The present experiments report that IL-1 can itself produce hyperalgesia as assessed by tailflick to radiant heat, and that recombinant IL-1 ra blocks the hyperalgesia produced by LiCl and LPS.
Article
Toll-like receptor 4 (TLR4) induced proinflammatory signaling has been directly implicated in severe sepsis and represents an attractive therapeutic target. Herein, we report our investigations into the structure-activity relationship and preliminary drug metabolism/pharmacokinetics study of β-amino alcohol derivatives that inhibit the TLR4 signaling pathway. Lead compounds were identified from in vitro cellular examination with micromolar potency for their inhibitory effects on TLR4 signaling and subsequently assessed for their ability to suppress the TLR4-induced inflammatory response in an ex vivo whole blood model. In addition, the toxicology, specificity, solubility, brain-blood barrier permeability, and drug metabolism of several compounds were evaluated. Although further optimizations are needed, our findings lay the groundwork for the future drug development of this class of small molecule agents for the treatment of severe sepsis.
Article
Increasing evidence supports the involvement of immune and inflammatory processes in the etiopathogenesis of seizures. In particular, activation of innate immune mechanisms and the subsequent inflammatory responses, that are induced in the brain by infection, febrile seizures, neurotrauma, stroke are well documented conditions associated with acute symptomatic seizures and with a high risk of developing epilepsy. A decade ago, pharmacological experiments showed that elevated brain levels of the anti-inflammatory molecule IL-1 receptor antagonist reduced seizures in epilepsy models. This observation, together with the evidence of in situ induction of inflammatory mediators and their receptors in experimental and human epileptogenic brain tissue, established the proof-of-concept evidence that the activation of innate immunity and inflammation in the brain are intrinsic features of the pathologic hyperexcitable tissue.